1
|
Karapareddy S, Anche VC, Tamatamu SR, Janga MR, Lawrence K, Nyochembeng LM, Todd A, Walker LT, Sripathi VR. Profiling of rhizosphere-associated microbial communities in North Alabama soils infested with varied levels of reniform nematodes. FRONTIERS IN PLANT SCIENCE 2025; 16:1521579. [PMID: 40123958 PMCID: PMC11925883 DOI: 10.3389/fpls.2025.1521579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025]
Abstract
Introduction Plant roots, nematodes, and soil microorganisms have a complex interaction in the rhizosphere by exchanging or communicating through biomolecules or chemicals or signals. Some rhizospheric (including endophytic) microbes process such compounds via biogeochemical cycles to improve soil fertility, promote plant growth and development, and impart stress tolerance in plants. Some rhizospheric microbes can affect negatively on plant parasitic nematodes (PPNs) thus hindering the ability of nematodes in parasitizing the plant roots. Next-generation sequencing is one of the most widely used and cost-effective ways of determining the composition and diversity of microbiomes in such complex environmental samples. Methods This study employed amplicon sequencing (Illumina/NextSeq) of 16S ribosomal RNA (16S rRNA) for bacteria and Internal Transcribed Spacer (ITS2) region for fungi to profile the soil microbiome in the rhizosphere of cotton grown in North Alabama. We isolated DNA (ZymoBIOMICS) from soil samples in triplicates from four representative locations of North Alabama. Based on the level of Reniform Nematode (RN) Infestation, these locations were classified as Group A-RN Not-Detected (ND), Group B-RN Low Infestation (LI), Group C-RN Medium Infestation (MI), and Group D-RN High Infestation (HI) and determined using sieving method and microscopic examination. Results and discussion Our analyses identified 47,893 bacterial and 3,409 fungal Amplicon Sequence Variants (ASVs) across all groups. Among the bacterial ASVs, 12,758, 10,709, 12,153, and 11,360 unique ASVs were determined in Groups A, B, C, and D, respectively. While 663, 887, 480, and 326 unique fungal ASVs were identified in Groups A, B, C, and D, respectively. Also, the five most abundant rhizospheric bacterial genera identified were Gaiella, Conexibacter, Bacillus, Blastococcus, Streptomyces. Moreover, five abundant fungal genera belonging to Fusarium, Aspergillus, Gibberella, Cladosporium, Lactera were identified. The tight clustering of bacterial nodes in Actinobacteria, Acidobacteria, and Proteobacteria shows they are highly similar and often found together. On the other hand, the close association of Ascomycota and Basidiomycota suggesting that they have different ecological roles but occupy similar niches and contribute similar functions within the microbial community. The abundant microbial communities identified in this study had a role in nutrient recycling, soil health, plant resistance to some environmental stress and pests including nematodes, and biogeochemical cycles. Our findings will aid in broadening our understanding of how microbial communities interact with crops and nematodes in the rhizosphere, influencing plant growth and pest management.
Collapse
Affiliation(s)
- Sowndarya Karapareddy
- College of Agricultural, Life & Natural Sciences, Alabama A&M University, Normal, AL, United States
| | - Varsha C. Anche
- College of Agricultural, Life & Natural Sciences, Alabama A&M University, Normal, AL, United States
| | - Sowjanya R. Tamatamu
- College of Agricultural, Life & Natural Sciences, Alabama A&M University, Normal, AL, United States
| | - Madhusudhana R. Janga
- Institute of Genomics for Crop Abiotic Stress Tolerance, Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, United States
| | - Kathy Lawrence
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL, United States
| | - Leopold M. Nyochembeng
- College of Agricultural, Life & Natural Sciences, Alabama A&M University, Normal, AL, United States
| | - Antonette Todd
- Department of Agriculture & Natural Resources, Delaware State University, Dover, DE, United States
| | - Lloyd T. Walker
- College of Agricultural, Life & Natural Sciences, Alabama A&M University, Normal, AL, United States
| | - Venkateswara R. Sripathi
- College of Agricultural, Life & Natural Sciences, Alabama A&M University, Normal, AL, United States
| |
Collapse
|
2
|
Han J, Wang M, Zhou S, Wang Z, Duan D, Li M, Li X, Xin W, Li X. The Joint Contribution of Host Genetics and Probiotics to Pig Growth Performance. Microorganisms 2025; 13:358. [PMID: 40005725 PMCID: PMC11857988 DOI: 10.3390/microorganisms13020358] [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: 12/18/2024] [Revised: 01/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Intestinal probiotics significantly regulate the growth performance of their host, with their composition being influenced by various factors. While many studies have explored how gut microbiota composition affects growth traits such as body weight and BMI, the research on probiotics influenced by host genetic factors, and their subsequent impact on host growth performance, remains limited. To address this research gap, we collected fecal and tissue samples, as well as phenotypic data, from 193 Yunong black pigs at 280 days of age. We then sequenced and genotyped all 193 subjects using the 50K SNP BeadChip, yielding a comprehensive dataset for genetic and microbiome analyses. We then employed microbiome-wide association studies (MWAS), a meta-analysis, and microbiome-wide genetic association studies (MGWASs) to examine the relationship between host genetics, gut microbiota, and growth performance. Four key microbial taxa, namely Coprococcus, Blautia, Ruminococcaceae, and RF16, were identified as being significantly associated with body weight and BMI. The MGWAS analysis revealed that both Coprococcus and Ruminococcaceae were significantly associated with host genomic variations. A total of four important single nucleotide polymorphisms (SNPs) were mapped to two chromosomal regions, corresponding to three candidate genes. Among them, the candidate genes INPP4B, SCOC, and PABPC4L were identified as being related to the abundance of key microbes. This study provides new insights into the joint contributions of host genetics and probiotics to host growth traits, offering theoretical guidance and data support for the development of efficient and targeted breeding strategies.
Collapse
Affiliation(s)
- Jinyi Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Mingyu Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Shenping Zhou
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Zhenyu Wang
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Dongdong Duan
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Mengyu Li
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Xiuling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenshui Xin
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Xinjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| |
Collapse
|
3
|
Xiao Y, Zhao Q, Ni D, Zhang X, Hao W, Yuan Q, Xu W, Mu W, Wu D, Wu X, Wang S. Polymerization of dietary fructans differentially affects interactions among intestinal microbiota of colitis mice. THE ISME JOURNAL 2025; 19:wrae262. [PMID: 39745882 PMCID: PMC11742283 DOI: 10.1093/ismejo/wrae262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/27/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
The intestinal microbiota plays a critical role in maintaining human health and can be modulated by dietary interventions and lifestyle choices. Fructans, a dietary carbohydrate, are selectively utilized by the intestinal microbiota to confer health benefits. However, the specific effects of different fructan types on microbial changes and functions remain incompletely understood. Here, we investigated how the intestinal microbiota responds to fructans with varying degrees of polymerization in the context of gut dysbiosis. Both low molecular weight fructo-oligosaccharides and high molecular weight levan suppressed intestinal inflammation in a colitis mouse model, mitigating intestinal fibrosis and dysbiosis. Although both the effects of fructo-oligosaccharides and levan are microbiota-dependent, distinct modulation patterns of the intestinal microbiota were observed based on the molecular weight of the fructans. Levan had a more pronounced and persistent impact on gut microbiota compared to fructo-oligosaccharides. Levan particularly promoted the abundance of Dubosiella newyorkensis, which exhibited preventive effects against colitis. Our findings highlight the importance of polymerization levels of dietary fructans in microbiota alterations and identify Dubosiella newyorkensis as a potential probiotic for treating inflammatory diseases.
Collapse
Affiliation(s)
- Yaqin Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Qianyun Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, Southwest Medical University, Xianglin Road, Longmatan District, Luzhou, Sichuan 646000, China
| | - Dawei Ni
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Xiaoqi Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Wei Hao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Wei Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Dingtao Wu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, School of Food and Biological Engineering, Chengdu University, Chengluo Avenue, Chengdu, Sichuan 616106, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, Southwest Medical University, Xianglin Road, Longmatan District, Luzhou, Sichuan 646000, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| |
Collapse
|
4
|
Isenberg RY, Holschbach CS, Gao J, Mandel MJ. Functional analysis of cyclic diguanylate-modulating proteins in Vibrio fischeri. mSystems 2024; 9:e0095624. [PMID: 39436151 PMCID: PMC11575326 DOI: 10.1128/msystems.00956-24] [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: 07/16/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
As bacterial symbionts transition from a motile free-living state to a sessile biofilm state, they must coordinate behavior changes suitable to each lifestyle. Cyclic diguanylate (c-di-GMP) is an intracellular signaling molecule that can regulate this transition, and it is synthesized by diguanylate cyclase (DGC) enzymes and degraded by phosphodiesterase (PDE) enzymes. Generally, c-di-GMP inhibits motility and promotes biofilm formation. While c-di-GMP and the enzymes that contribute to its metabolism have been well studied in pathogens, considerably less focus has been placed on c-di-GMP regulation in beneficial symbionts. Vibrio fischeri is the sole beneficial symbiont of the Hawaiian bobtail squid (Euprymna scolopes) light organ, and the bacterium requires both motility and biofilm formation to efficiently colonize. c-di-GMP regulates swimming motility and cellulose exopolysaccharide production in V. fischeri. The genome encodes 50 DGCs and PDEs, and while a few of these proteins have been characterized, the majority have not undergone comprehensive characterization. In this study, we use protein overexpression to systematically characterize the functional potential of all 50 V. fischeri proteins. All 28 predicted DGCs and 10 of the 14 predicted PDEs displayed at least one phenotype consistent with their predicted function, and a majority of each displayed multiple phenotypes. Finally, active site mutant analysis of proteins with the potential for both DGC and PDE activities revealed potential activities for these proteins. This work presents a systems-level functional analysis of a family of signaling proteins in a tractable animal symbiont and will inform future efforts to characterize the roles of individual proteins during lifestyle transitions.IMPORTANCECyclic diguanylate (c-di-GMP) is a critical second messenger that mediates bacterial behaviors, and Vibrio fischeri colonization of its Hawaiian bobtail squid host presents a tractable model in which to interrogate the role of c-di-GMP during animal colonization. This work provides systems-level characterization of the 50 proteins predicted to modulate c-di-GMP levels. By combining multiple assays, we generated a rich understanding of which proteins have the capacity to influence c-di-GMP levels and behaviors. Our functional approach yielded insights into how proteins with domains to both synthesize and degrade c-di-GMP may impact bacterial behaviors. Finally, we integrated published data to provide a broader picture of each of the 50 proteins analyzed. This study will inform future work to define specific pathways by which c-di-GMP regulates symbiotic behaviors and transitions.
Collapse
Affiliation(s)
- Ruth Y Isenberg
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chandler S Holschbach
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jing Gao
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark J Mandel
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Isenberg RY, Holschbach CS, Gao J, Mandel MJ. Functional analysis of cyclic diguanylate-modulating proteins in Vibrio fischeri. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550417. [PMID: 37546929 PMCID: PMC10402110 DOI: 10.1101/2023.07.24.550417] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
As bacterial symbionts transition from a motile free-living state to a sessile biofilm state, they must coordinate behavior changes suitable to each lifestyle. Cyclic diguanylate (c-di-GMP) is an intracellular signaling molecule that can regulate this transition, and it is synthesized by diguanylate cyclase (DGC) enzymes and degraded by phosphodiesterase (PDE) enzymes. Generally, c-di-GMP inhibits motility and promotes biofilm formation. While c-di-GMP and the enzymes that contribute to its metabolism have been well-studied in pathogens, considerably less focus has been placed on c-di-GMP regulation in beneficial symbionts. Vibrio fischeri is the sole beneficial symbiont of the Hawaiian bobtail squid (Euprymna scolopes) light organ, and the bacterium requires both motility and biofilm formation to efficiently colonize. C-di-GMP regulates swimming motility and cellulose exopolysaccharide production in V. fischeri. The genome encodes 50 DGCs and PDEs, and while a few of these proteins have been characterized, the majority have not undergone comprehensive characterization. In this study, we use protein overexpression to systematically characterize the functional potential of all 50 V. fischeri proteins. All 28 predicted DGCs and 14 predicted PDEs displayed at least one phenotype consistent with their predicted function, and a majority of each displayed multiple phenotypes. Finally, active site mutant analysis of proteins with the potential for both DGC and PDE activities revealed potential activities for these proteins. This work presents a systems-level functional analysis of a family of signaling proteins in a tractable animal symbiont and will inform future efforts to characterize the roles of individual proteins during lifestyle transitions.
Collapse
Affiliation(s)
- Ruth Y. Isenberg
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI USA
- Current address: Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN USA
| | - Chandler S. Holschbach
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI USA
| | - Jing Gao
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Mark J. Mandel
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
6
|
Zhang W, Lan F, Zhou Q, Gu S, Li X, Wen C, Yang N, Sun C. Host genetics and gut microbiota synergistically regulate feed utilization in egg-type chickens. J Anim Sci Biotechnol 2024; 15:123. [PMID: 39245742 PMCID: PMC11382517 DOI: 10.1186/s40104-024-01076-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/14/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Feed efficiency is a crucial economic trait in poultry industry. Both host genetics and gut microbiota influence feed efficiency. However, the associations between gut microbiota and host genetics, as well as their combined contributions to feed efficiency in laying hens during the late laying period, remain largely unclear. METHODS In total, 686 laying hens were used for whole-genome resequencing and liver transcriptome sequencing. 16S rRNA gene sequencing was conducted on gut chyme (duodenum, jejunum, ileum, and cecum) and fecal samples from 705 individuals. Bioinformatic analysis was performed by integrating the genome, transcriptome, and microbiome to screen for key genetic variations, genes, and gut microbiota associated with feed efficiency. RESULTS The heritability of feed conversion ratio (FCR) and residual feed intake (RFI) was determined to be 0.28 and 0.48, respectively. The ileal and fecal microbiota accounted for 15% and 10% of the FCR variance, while the jejunal, cecal, and fecal microbiota accounted for 20%, 11%, and 10% of the RFI variance. Through SMR analysis based on summary data from liver eQTL mapping and GWAS, we further identified four protein-coding genes, SUCLA2, TNFSF13B, SERTM1, and MARVELD3, that influence feed efficiency in laying hens. The SUCLA2 and TNFSF13B genes were significantly associated with SNP 1:25664581 and SNP rs312433097, respectively. SERTM1 showed significant associations with rs730958360 and 1:33542680 and is a potential causal gene associated with the abundance of Corynebacteriaceae in feces. MARVELD3 was significantly associated with the 1:135348198 and was significantly correlated with the abundance of Enterococcus in ileum. Specifically, a lower abundance of Enterococcus in ileum and a higher abundance of Corynebacteriaceae in feces were associated with better feed efficiency. CONCLUSIONS This study confirms that both host genetics and gut microbiota can drive variations in feed efficiency. A small portion of the gut microbiota often interacts with host genes, collectively enhancing feed efficiency. Therefore, targeting both the gut microbiota and host genetic variation by supporting more efficient taxa and selective breeding could improve feed efficiency in laying hens during the late laying period.
Collapse
Affiliation(s)
- Wenxin Zhang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Qianqian Zhou
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, 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
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
7
|
Ermencheva P, Kotov G, Shumnalieva R, Velikova T, Monov S. Exploring the Role of the Microbiome in Rheumatoid Arthritis-A Critical Review. Microorganisms 2024; 12:1387. [PMID: 39065155 PMCID: PMC11278530 DOI: 10.3390/microorganisms12071387] [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: 06/03/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, autoimmune rheumatic disease characterized by synovial joint inflammation with subsequent destruction as well as systemic manifestation, leading to impaired mobility and impaired quality of life. The etiopathogenesis of RA is still unknown, with genetic, epigenetic and environmental factors (incl. tobacco smoking) contributing to disease susceptibility. The link between genetic factors like "shared epitope alleles" and the development of RA is well known. However, why only some carriers have a break in self-tolerance and develop autoimmunity still needs to be clarified. The presence of autoantibodies in patients' serum months to years prior to the onset of clinical manifestations of RA has moved the focus to possible epigenetic factors, including environmental triggers that could contribute to the initiation and perpetuation of the inflammatory reaction in RA. Over the past several years, the role of microorganisms at mucosal sites (i.e., microbiome) has emerged as an essential mediator of inflammation in RA. An increasing number of studies have revealed the microbial role in the immunopathogenesis of autoimmune rheumatic diseases. Interaction between the host immune system and microbiota initiates loss of immunological tolerance and autoimmunity. The alteration in microbiome composition, the so-called dysbiosis, is associated with an increasing number of diseases. Immune dysfunction caused by dysbiosis triggers and sustains chronic inflammation. This review aims to provide a critical summary of the literature findings related to the hypothesis of a reciprocal relation between the microbiome and the immune system. Available data from studies reveal the pivotal role of the microbiome in RA pathogenesis.
Collapse
Affiliation(s)
- Plamena Ermencheva
- Clinic of Rheumatology, University Hospital ‘St. Ivan Rilski’, 13 Urvich Str., 1612 Sofia, Bulgaria; (P.E.); (G.K.); (R.S.); (S.M.)
| | - Georgi Kotov
- Clinic of Rheumatology, University Hospital ‘St. Ivan Rilski’, 13 Urvich Str., 1612 Sofia, Bulgaria; (P.E.); (G.K.); (R.S.); (S.M.)
| | - Russka Shumnalieva
- Clinic of Rheumatology, University Hospital ‘St. Ivan Rilski’, 13 Urvich Str., 1612 Sofia, Bulgaria; (P.E.); (G.K.); (R.S.); (S.M.)
- Department of Rheumatology, Medical University of Sofia, 13 Urvich Str., 1612 Sofia, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Kozyak 1, 1407 Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Kozyak 1, 1407 Sofia, Bulgaria
| | - Simeon Monov
- Clinic of Rheumatology, University Hospital ‘St. Ivan Rilski’, 13 Urvich Str., 1612 Sofia, Bulgaria; (P.E.); (G.K.); (R.S.); (S.M.)
- Department of Rheumatology, Medical University of Sofia, 13 Urvich Str., 1612 Sofia, Bulgaria
| |
Collapse
|
8
|
Kotlyarov S. Importance of the gut microbiota in the gut-liver axis in normal and liver disease. World J Hepatol 2024; 16:878-882. [PMID: 38948437 PMCID: PMC11212653 DOI: 10.4254/wjh.v16.i6.878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
The gut microbiota is of growing interest to clinicians and researchers. This is because there is a growing understanding that the gut microbiota performs many different functions, including involvement in metabolic and immune processes that are systemic in nature. The liver, with its important role in detoxifying and metabolizing products from the gut, is at the forefront of interactions with the gut microbiota. Many details of these interactions are not yet known to clinicians and researchers, but there is growing evidence that normal gut microbiota function is important for liver health. At the same time, factors affecting the gut microbiota, including nutrition or medications, may also have an effect through the gut-liver axis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, Ryazan 390026, Russia.
| |
Collapse
|
9
|
HAYASHI K, UCHIDA R, HORIBA T, KAWAGUCHI T, GOMI K, GOTO Y. Soy sauce-like seasoning enhances the growth of Agathobacter rectalis and the production of butyrate, propionate, and lactate. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:275-281. [PMID: 38966053 PMCID: PMC11220332 DOI: 10.12938/bmfh.2023-103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/08/2024] [Indexed: 07/06/2024]
Abstract
The short-chain fatty acids responsible for gut homeostasis are volatile fatty acids produced by commensal bacteria in the gut as fermentation products from undigested food components. Among the short-chain fatty acids, butyrate is important for maintaining intestinal tract anaerobic conditions, promoting epithelial barrier functions, and inducing regulatory T cells that suppress inflammatory bowel disease and allergic diarrhea. However, the type of food-derived molecular components and mechanisms by which they regulate the growth and butyrate production of butyrate-producing bacteria are not clearly understood. Agathobacter rectalis is a butyrate-producing bacterium highly colonized in the gut of the Japanese population. In this study, we investigated the effects on A. rectalis of a soy sauce-like seasoning made by brewing with a low salt concentration. The soy sauce-like seasoning promoted the growth of A. rectalis 2.6-fold. An ethanol precipitate prepared from the soy sauce-like seasoning was critical for promoting the growth of A. rectalis and the production of butyrate, propionate, and lactate. Fourier transform infrared spectroscopy (FT-IR) analysis suggested that polysaccharides were active ingredients in the ethanol precipitate of the soy sauce-like seasoning. Inulin, a representative prebiotic with effects against butyrate-producing bacteria, had a limited effect on the growth of A. rectalis compared with the soy sauce-like seasoning. Our results indicate that polysaccharides in a soy sauce-like seasoning contributed to the growth of A. rectalis and enhanced production of butyrate, propionate, and lactate.
Collapse
Affiliation(s)
- Kanako HAYASHI
- Project for Host-Microbial Interactions in Symbiosis and
Pathogenesis, Division of Molecular Immunology, Medical Mycology Research Center, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8673, Japan
- Reserch and Development Division, Kikkoman Corporation, 338
Noda, Noda-shi, Chiba 278-0037, Japan
| | - Riichiro UCHIDA
- Reserch and Development Division, Kikkoman Corporation, 338
Noda, Noda-shi, Chiba 278-0037, Japan
| | - Taro HORIBA
- Reserch and Development Division, Kikkoman Corporation, 338
Noda, Noda-shi, Chiba 278-0037, Japan
| | - Tomohiro KAWAGUCHI
- Reserch and Development Division, Kikkoman Corporation, 338
Noda, Noda-shi, Chiba 278-0037, Japan
| | - Keiko GOMI
- Reserch and Development Division, Kikkoman Corporation, 338
Noda, Noda-shi, Chiba 278-0037, Japan
| | - Yoshiyuki GOTO
- Project for Host-Microbial Interactions in Symbiosis and
Pathogenesis, Division of Molecular Immunology, Medical Mycology Research Center, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8673, Japan
- Division of Pandemic and Post-disaster Infectious Diseases,
Research Institute of Disaster Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku,
Chiba-shi, Chiba 260-8673, Japan
- Division of Infectious Disease Vaccine R&D, Research
Institute of Disaster Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba
260-8673, Japan
- Chiba University Synergy Institute for Futuristic Mucosal
Vaccine Research and Development (cSIMVa), Chiba University, 1-8-1 Inohana, Chuo-ku,
Chiba-shi, Chiba 260-8673, Japan
| |
Collapse
|
10
|
Li Z, Chen F, Wei M, Zhi L, Su Z, Chong Y, Xiao Z, Wang J. Concurrent impacts of polystyrene nanoplastic exposure and Aeromonas hydrophila infection on oxidative stress, immune response and intestinal microbiota of grass carp (Ctenopharyngodon idella). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169225. [PMID: 38101646 DOI: 10.1016/j.scitotenv.2023.169225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
Research has demonstrated that polystyrene nanoplastics (PS-NPs) can have adverse effects on the immune responses of fish. NPs have the potential to increase the likelihood of infections in fish by pathogenic bacteria, such as the opportunistic pathogen Aeromonas hydrophila, potentially increasing the virulence of pathogenic bacteria infections in fish. The concurrent effects of PS-NPs and A. hydrophila on grass carp intestinal tissues were assessed by exposing grass carp to different concentrations of PS-NPs (10 μg/L, 100 μg/L, 1000 μg/L) after infection with A. hydrophila. As the concentration of PS-NPs in the exposure and the duration of A. hydrophila infection both escalated, intestinal tissues showed damage in the form of disordered breakage of intestinal villi, thinning of the intestinal wall, and reduced necrosis of the cells in the annulus muscle layer. The AHS-PS100 group and AHS-PS1000 group exhibited a substantial rise in the function of CAT, SOD, GST, and MPO, as well as increased MDA content and elevated ROS levels (p < 0.05). In the AHS-PS1000 group, the expression levels of IL-6, IL-8, IL-10, IL-1β, TNF-α, and IFN-γ2 experienced a significant upsurge (p < 0.05). In addition, exposure to PS-NPs and A. hydrophila infection induced modifications in the microbial composition of the grass carp gut, affecting both phylum and genus taxonomic categories. Moreover, an increase in the abundance of Spirochaetota and Bacteroidota was observed not only in the positive control group but also in the AHS-PS100 and AHS-PS1000 groups following A. hydrophila infection. These experimental results indicate that PS-NPs exposure will aggravate the oxidative stress and inflammatory response of grass carp intestinal tissue in response to A. hydrophila infection, and lead to changes in intestinal microbial diversity and abundance. Overall, this study provides valuable hints on the potential concurrent effects of PS-NPs exposure on grass carp's response to A. hydrophila infection.
Collapse
Affiliation(s)
- Zhen Li
- College of Marine Sciences, College of Natural Resources and Environment, Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, South China Agricultural University, Guangzhou 510642, China; College of Biology and Agricultural, Shaoguan University, Shaoguan 512005, China
| | - Fang Chen
- College of Marine Sciences, College of Natural Resources and Environment, Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, South China Agricultural University, Guangzhou 510642, China
| | - Maochun Wei
- Xiamen Key Laboratory of Intelligent Fishery, Xiamen Ocean Vocational College, Xiamen 361100, China
| | - Linyong Zhi
- College of Marine Sciences, College of Natural Resources and Environment, Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, South China Agricultural University, Guangzhou 510642, China
| | - Zeliang Su
- College of Marine Sciences, College of Natural Resources and Environment, Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, South China Agricultural University, Guangzhou 510642, China
| | - Yunxiao Chong
- College of Marine Sciences, College of Natural Resources and Environment, Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, South China Agricultural University, Guangzhou 510642, China.
| | - Zhengzhong Xiao
- College of Biology and Agricultural, Shaoguan University, Shaoguan 512005, China.
| | - Jun Wang
- College of Marine Sciences, College of Natural Resources and Environment, Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, South China Agricultural University, Guangzhou 510642, China; Institute of Eco-Environmental Research, Guangxi Academy of Sciences, Nanning 530007, China; Xiamen Key Laboratory of Intelligent Fishery, Xiamen Ocean Vocational College, Xiamen 361100, China.
| |
Collapse
|
11
|
Chen X, de Vos P. Structure-function relationship and impact on the gut-immune barrier function of non-digestible carbohydrates and human milk oligosaccharides applicable for infant formula. Crit Rev Food Sci Nutr 2023; 64:8325-8345. [PMID: 37035930 DOI: 10.1080/10408398.2023.2199072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Human milk oligosaccharides (hMOs) in mothers' milk play a crucial role in guiding the colonization of microbiota and gut-immune barrier development in infants. Non-digestible carbohydrates (NDCs) such as synthetic single hMOs, galacto-oligosaccharides (GOS), inulin-type fructans and pectin oligomers have been added to infant formula to substitute some hMOs' functions. HMOs and NDCs can modulate the gut-immune barrier, which is a multiple-layered functional unit consisting of microbiota, a mucus layer, gut epithelium, and the immune system. There is increasing evidence that the structures of the complex polysaccharides may influence their efficacy in modulating the gut-immune barrier. This review focuses on the role of different structures of individual hMOs and commonly applied NDCs in infant formulas in (i) direct regulation of the gut-immune barrier in a microbiota-independent manner and in (ii) modulation of microbiota composition and microbial metabolites of these polysaccharides in a microbiota-dependent manner. Both have been shown to be essential for guiding the development of an adequate immune barrier, but the effects are very dependent on the structural features of hMO or NDC. This knowledge might lead to tailored infant formulas for specific target groups.
Collapse
Affiliation(s)
- Xiaochen Chen
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
12
|
Vassey M, Firdaus R, Aslam A, Wheldon LM, Oldfield NJ, Ala’Aldeen DAA, Wooldridge KG. G1 Cell Cycle Arrest Is Induced by the Fourth Extracellular Loop of Meningococcal PorA in Epithelial and Endothelial Cells. Cell Microbiol 2023. [DOI: 10.1155/2023/7480033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Neisseria meningitidis is the most frequent cause of bacterial meningitis and is one of the few bacterial pathogens that can breach the blood-brain barrier (BBB). The 37/67 kDa laminin receptor (LamR) was previously identified as a receptor mediating meningococcal binding to rodent and human brain microvascular endothelial cells, which form part of the BBB. The meningococcal surface proteins PorA and PilQ were identified as ligands for this receptor. Subsequently, the fourth extracellular loop of PorA (PorA-Loop4) was identified as the LamR-binding moiety. Here, we show that PorA-Loop4 targets the 37 kDa laminin receptor precursor (37LRP) on the cell surface by demonstrating that deletion of this loop abrogates the recruitment of 37LRP under meningococcal colonies. Using a circularized peptide corresponding to PorA-Loop4, as well as defined meningococcal mutants, we demonstrate that host cell interaction with PorA-Loop4 results in perturbation of p-CDK4 and Cyclin D1. These changes in cell cycle control proteins are coincident with cellular responses including inhibition of cell migration and a G1 cell cycle arrest. Modulation of the cell cycle of host cells is likely to contribute to the pathogenesis of meningococcal disease.
Collapse
|
13
|
Khelaifia S, Virginie P, Belkacemi S, Tassery H, Terrer E, Aboudharam G. Culturing the Human Oral Microbiota, Updating Methodologies and Cultivation Techniques. Microorganisms 2023; 11:microorganisms11040836. [PMID: 37110259 PMCID: PMC10143722 DOI: 10.3390/microorganisms11040836] [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: 01/24/2023] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Recent years have been marked by a paradigm shift in the study of the human microbiota, with a re-emergence of culture-dependent approaches. Numerous studies have been devoted to the human microbiota, while studies on the oral microbiota still remain limited. Indeed, various techniques described in the literature may enable an exhaustive study of the microbial composition of a complex ecosystem. In this article, we report different methodologies and culture media described in the literature that can be applied to study the oral microbiota by culture. We report on specific methodologies for targeted culture and specific culture techniques and selection methodologies for cultivating members of the three kingdoms of life commonly found in the human oral cavity, namely, eukaryota, bacteria and archaea. This bibliographic review aims to bring together the various techniques described in the literature, enabling a comprehensive study of the oral microbiota in order to demonstrate its involvement in oral health and diseases.
Collapse
Affiliation(s)
- Saber Khelaifia
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Pilliol Virginie
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Souad Belkacemi
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Herve Tassery
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Elodie Terrer
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Gérard Aboudharam
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| |
Collapse
|
14
|
The Phytopathogen Fusarium verticillioides Modifies the Intestinal Morphology of the Sugarcane Borer. Pathogens 2023; 12:pathogens12030443. [PMID: 36986365 PMCID: PMC10056812 DOI: 10.3390/pathogens12030443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Background: In tropical sugarcane crops, the fungus Fusarium verticillioides, the agent responsible for the occurrence of the red rot complex, occurs in association with the sugarcane borer Diatraea saccharalis. This fungus, in addition to being transmitted vertically, can manipulate both the insect and the plant for its own dissemination in the field. Due to the complex interaction between F. verticillioides and D. saccharalis, and the high incidence of the fungus in the intestinal region, our objective was to investigate whether F. verticillioides could alter the intestinal structure of the insect. Methods: We combined analysis of scanning electron microscopy and light microscopy to identify whether the presence of the fungus F. verticillioides, in artificial diets or in sugarcane, could lead to any alteration or regional preference in the insect’s intestinal ultrastructure over the course of its development, or its offspring development, analyzing the wall and microvillous structures of the mid-digestive system. Results: Here, we show that the fungus F. verticillioides alters the intestinal morphology of D. saccharalis, promoting an increase of up to 3.3 times in the thickness of the midgut compared to the control. We also observed that the phytopathogen colonizes the intestinal microvilli for reproduction, suggesting that this region can be considered the gateway of the fungus to the insect’s reproductive organs. In addition, the colonization of this region promoted the elongation of microvillous structures by up to 180% compared to the control, leading to an increase in the area used for colonization. We also used the fungus Colletotrichum falcatum in the tests, and it did not differ from the control in any test, showing that this interaction is specific between D. saccharalis and F. verticillioides. Conclusions: The phytopathogenic host F. verticillioides alters the intestinal morphology of the vector insect in favor of its colonization.
Collapse
|
15
|
Ji J, Wu L, Wei J, Wu J, Guo C. The Gut Microbiome and Ferroptosis in MAFLD. J Clin Transl Hepatol 2023; 11:174-187. [PMID: 36406312 PMCID: PMC9647110 DOI: 10.14218/jcth.2022.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/22/2022] [Accepted: 06/12/2022] [Indexed: 12/04/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a new disease definition, and is proposed to replace the previous name, nonalcoholic fatty liver disease (NAFLD). Globally, MAFLD/NAFLD is the most common liver disease, with an incidence rate ranging from 6% to 35% in adult populations. The pathogenesis of MAFLD/NAFLD is closely related to insulin resistance (IR), and the genetic susceptibility to acquired metabolic stress-associated liver injury. Similarly, the gut microbiota in MAFLD/NAFLD is being revaluated by scientists, as the gut and liver influence each other via the gut-liver axis. Ferroptosis is a novel form of programmed cell death caused by iron-dependent lipid peroxidation. Emerging evidence suggests that ferroptosis has a key role in the pathological progression of MAFLD/NAFLD, and inhibition of ferroptosis may become a novel therapeutic strategy for the treatment of NAFLD. This review focuses on the main mechanisms behind the promotion of MAFLD/NAFLD occurrence and development by the intestinal microbiota and ferroptosis. It outlines new strategies to target the intestinal microbiota and ferroptosis to facilitate future MAFLD/NAFLD therapies.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liwei Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jue Wei
- Department of Gastroenterology Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| |
Collapse
|
16
|
Talyuli OAC, Oliveira JHM, Bottino-Rojas V, Silveira GO, Alvarenga PH, Barletta ABF, Kantor AM, Paiva-Silva GO, Barillas-Mury C, Oliveira PL. The Aedes aegypti peritrophic matrix controls arbovirus vector competence through HPx1, a heme-induced peroxidase. PLoS Pathog 2023; 19:e1011149. [PMID: 36780872 PMCID: PMC9956595 DOI: 10.1371/journal.ppat.1011149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 02/24/2023] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Aedes aegypti mosquitoes are the main vectors of arboviruses. The peritrophic matrix (PM) is an extracellular layer that surrounds the blood bolus. It acts as an immune barrier that prevents direct contact of bacteria with midgut epithelial cells during blood digestion. Here, we describe a heme-dependent peroxidase, hereafter referred to as heme peroxidase 1 (HPx1). HPx1 promotes PM assembly and antioxidant ability, modulating vector competence. Mechanistically, the heme presence in a blood meal induces HPx1 transcriptional activation mediated by the E75 transcription factor. HPx1 knockdown increases midgut reactive oxygen species (ROS) production by the DUOX NADPH oxidase. Elevated ROS levels reduce microbiota growth while enhancing epithelial mitosis, a response to tissue damage. However, simultaneous HPx1 and DUOX silencing was not able to rescue bacterial population growth, as explained by increased expression of antimicrobial peptides (AMPs), which occurred only after double knockdown. This result revealed hierarchical activation of ROS and AMPs to control microbiota. HPx1 knockdown produced a 100-fold decrease in Zika and dengue 2 midgut infection, demonstrating the essential role of the mosquito PM in the modulation of arbovirus vector competence. Our data show that the PM connects blood digestion to midgut immunological sensing of the microbiota and viral infections.
Collapse
Affiliation(s)
- Octavio A. C. Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Henrique M. Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departments of Microbiology and Molecular Genetics and of Molecular Biology and Biochemistry, University of California, Irvine, California, United States of America
| | - Gilbert O. Silveira
- Laboratório de Expressão Genica em Eucariotos, Instituto Butantan and Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Patricia H. Alvarenga
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Ana Beatriz F. Barletta
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Asher M. Kantor
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Gabriela O. Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Pedro L. Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Khan A, Moni SS, Ali M, Mohan S, Jan H, Rasool S, Kamal MA, Alshahrani S, Halawi M, Alhazmi HA. Antifungal Activity of Plant Secondary Metabolites on Candida albicans: An Updated Review. Curr Mol Pharmacol 2023; 16:15-42. [PMID: 35249516 DOI: 10.2174/1874467215666220304143332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/24/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Fungal infections have been increasing continuously worldwide, especially in immunocompromised individuals. Fungi, regarded as eukaryotic pathogens, have many similarities to the host cells, which inhibit anti-fungal drug development progress. Various fungal model systems have been studied, and it was concluded that Candida spp. is the most common disease-causing fungus. Candida species are well known to cause infections not only in our mouth, skin, and vagina, but they are also a frequent cause of life-threatening hospital bloodstream infections. The morphological and developmental pathways of Candida have been studied extensively, providing insight into the fungus development. Candida albicans is known to be the most pathogenic species responsible for a variety of infections in humans. Conventional anti-fungal drugs, mainly azoles drugs available in the market, have been used for years developing resistance in C. albicans. Hence, the production of new anti-fungal drugs, which require detailed molecular knowledge of fungal pathogenesis, needs to be encouraged. Therefore, this review targets the new approach of "Green Medicines" or the phytochemicals and their secondary metabolites as a source of novel anti-fungal agents to overcome the drug resistance of C. albicans, their mechanism of action, and their combined effects with the available anti-fungal drugs.
Collapse
Affiliation(s)
- Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | | | - M Ali
- Department of Pharmacognosy, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, 45142, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Huma Jan
- Department of Clinical Biochemistry, University of Kashmir, Hazratbal, Srinagar -190006, J&K, India
| | - Saiema Rasool
- Department of School Education, Govt. of Jammu & Kashmir, Srinagar, 190001 J&K, India
| | - Mohammad A Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589. Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
- Enzymoics, 7 Peterlee place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Maryam Halawi
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, 45142, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| |
Collapse
|
18
|
Tseng YC, Xue C, Ng IS. Symbiosis culture of probiotic Escherichia coli Nissle 1917 and Lactobacillus rhamnosus GG using lactate utilization protein YkgG. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
19
|
Ganesan R, Wierz JC, Kaltenpoth M, Flórez LV. How It All Begins: Bacterial Factors Mediating the Colonization of Invertebrate Hosts by Beneficial Symbionts. Microbiol Mol Biol Rev 2022; 86:e0012621. [PMID: 36301103 PMCID: PMC9769632 DOI: 10.1128/mmbr.00126-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Beneficial associations with bacteria are widespread across animals, spanning a range of symbiont localizations, transmission routes, and functions. While some of these associations have evolved into obligate relationships with permanent symbiont localization within the host, the majority require colonization of every host generation from the environment or via maternal provisions. Across the broad diversity of host species and tissue types that beneficial bacteria can colonize, there are some highly specialized strategies for establishment yet also some common patterns in the molecular basis of colonization. This review focuses on the mechanisms underlying the early stage of beneficial bacterium-invertebrate associations, from initial contact to the establishment of the symbionts in a specific location of the host's body. We first reflect on general selective pressures that can drive the transition from a free-living to a host-associated lifestyle in bacteria. We then cover bacterial molecular factors for colonization in symbioses from both model and nonmodel invertebrate systems where these have been studied, including terrestrial and aquatic host taxa. Finally, we discuss how interactions between multiple colonizing bacteria and priority effects can influence colonization. Taking the bacterial perspective, we emphasize the importance of developing new experimentally tractable systems to derive general insights into the ecological factors and molecular adaptations underlying the origin and establishment of beneficial symbioses in animals.
Collapse
Affiliation(s)
- Ramya Ganesan
- Department of Evolutionary Ecology, Institute of Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Jürgen C. Wierz
- Department of Evolutionary Ecology, Institute of Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Martin Kaltenpoth
- Department of Evolutionary Ecology, Institute of Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Laura V. Flórez
- Department of Evolutionary Ecology, Institute of Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
- Department of Plant and Environmental Sciences, Section for Organismal Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Cardoso MH, Meneguetti BT, Oliveira-Júnior NG, Macedo MLR, Franco OL. Antimicrobial peptide production in response to gut microbiota imbalance. Peptides 2022; 157:170865. [PMID: 36038014 DOI: 10.1016/j.peptides.2022.170865] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022]
Abstract
The gut microbiota presents essential functions in the immune response. The gut epithelium acts as a protective barrier and, therefore, can produce several antimicrobial peptides (AMPs) that can act against pathogenic microorganisms, including bacteria. Several factors cause a disturbance in gut microbiota, including the exacerbated and erroneous use of antibiotics. Antibiotic therapy has been closely related to bacterial resistance and is also correlated with undesired side-effects to the host, including the eradication of commensal bacteria. Consequently, this results in gut microbiota imbalance and inflammatory bowel diseases (IBD) development. In this context, AMPs in the gut epithelium play a restructuring role for gut microbiota. Some naturally occurring AMPs are selective for pathogenic bacteria, thus preserving the health microbiota. Therefore, AMPs produced by the host's epithelial cells represent effective molecules in treating gut bacterial infections. Bearing this in mind, this review focused on describing the importance of the host's AMPs in gut microbiota modulation and their role as anti-infective agents against pathogenic bacteria.
Collapse
Affiliation(s)
- Marlon H Cardoso
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil; Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900 Campo Grande, Mato Grosso do Sul, Brazil.
| | - Beatriz T Meneguetti
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil
| | - Nelson G Oliveira-Júnior
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil
| | - Maria L R Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900 Campo Grande, Mato Grosso do Sul, Brazil
| | - Octávio L Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil.
| |
Collapse
|
21
|
Yadav U, Bano N, Bag S, Srivastava S, Singh PC. An Insight into the Endophytic Bacterial Community of Tomato after Spray Application of Propiconazole and Bacillus subtilis Strain NBRI-W9. Microbiol Spectr 2022; 10:e0118622. [PMID: 36066253 PMCID: PMC9602357 DOI: 10.1128/spectrum.01186-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/04/2022] [Indexed: 12/30/2022] Open
Abstract
Propiconazole (PCZ) is a commonly sprayed fungicide against fungal pathogens. Being systemic in action, it reaches subcellular layers and impacts the endophytes. Although PCZ is a fungicide, it is hypothesized to exert an inhibitory effect on the bacterial endophytes. Therefore, this study aims to get an insight into the perturbations caused by the systemically acting antifungal agents PCZ and Bacillus subtilis (W9) and the consequences thereof. The current study compared the 16S rRNA microbial diversity, abundance, and functions of the endophytic bacterial community of tomato in response to PCZ, W9, and PCZ+W9 application. The implications of these treatments on the development of bacterial speck disease by Pseudomonas syringae were also studied. The culturable endophyte population fluctuated after (bio)fungicide application and stabilized by 72 h. At 72 h, the endophyte population was ~3.6 × 103 CFUg-1 in control and ~3.6 × 104 in W9, ~3.0 × 102 in PCZ, and ~5.3 × 103 in PCZ+W9 treatment. A bacterial community analysis showed a higher relative abundance of Bacillales, Burkholderiales, Rhizobiales, Pseudomonadales, and Actinomycetales in the W9 treatment compared with that in the PCZ treatment and control. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) analysis showed enhanced metabolic pathways related to secretion, stress, chemotaxis, and mineral nutrition in the W9 treatment. Disease severity was greater in PCZ than that in the W9 treatment. Disease severity on tomato plants showed strong negative correlations with Sphingomonas (r = -0.860) and Janthinobacterium (r = -0.810), indicating that the natural biocontrol communities are agents of plant resistance to diseases. Outcomes show that systemic chemicals are a potential threat to the nontarget endophytes and that plants became susceptible to disease on endophyte decline; this issue could be overcome by the application of microbial inoculums. IMPORTANCE Endophytes are plant inhabitants acting as its extended genome. The present study highlights the importance of maintaining plant endophytes for sustainable disease resistance in plants. The impact of chemical fungicides and biofungicides was shown on tomato endophytes, in addition to their implications on plant susceptibility to bacterial speck disease. The observations point toward the deleterious effects of systemic pesticide application on endophyte niches that disrupt their diversity and functions compromising plant immunity.
Collapse
Affiliation(s)
- Udit Yadav
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Nasreen Bano
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Sumit Bag
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Suchi Srivastava
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Poonam C. Singh
- CSIR-National Botanical Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
22
|
Sivaraman K, Shanthi C. Purified fish skin collagen hydrolysate attenuates TNF-α induced barrier dysfunction in-vitro and DSS induced colitis in-vivo model. Int J Biol Macromol 2022; 222:448-461. [PMID: 36116587 DOI: 10.1016/j.ijbiomac.2022.09.122] [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: 05/04/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/05/2022]
Abstract
Inflammatory mediators are key components in establishing pathogenesis in inflammatory bowel disease. Balanced expression of anti-inflammatory and pro-inflammatory cytokines is an important cue in maintaining gut native and adaptive immunity. In the present study, purified hydrolysate fraction of fish skin collagen from Clarias batrachus and Pangasius pangasius was evaluated as a treatment agent against TNF-α induced barrier dysfunction in Caco-2 cell line model and DSS induced colitis in mice model. Cell adhesion on purified hydrolysate fraction coated surfaces was found to be enhanced with increasing concentration in both Clarias batrachus and Pangasius pangasius. Alkaline phosphatase activity was enhanced in a concentration-dependent manner. The paracellular permeability assay demonstrated that Pangasius pangasius purified hydrolysate fraction had countered TNF-α induced barrier dysfunction. Analysis of the tight junction proteins (occludin, zonulae occluden, and claudin) by RT PCR, immunofluorescence, and western blot, further confirmed the effectiveness of Pangasius pangasius purified hydrolysate fraction against TNF-α. The Pangasius pangasius purified hydrolysate fraction was further evaluated for efficacy in DSS-induced colitis mice model. Two concentration of Pangasius pangasius purified hydrolysate was chosen based on in-vitro experiments, 80 μg/kg and 200 μg/kg BW of Balb/C male mice administered through intra-rectal route along with fish skin collagen 80 μg/kg BW. Pangasius pangasius purified hydrolysate fraction treatment improved the clinical signs of colitis such as body weight, rectal bleeding, colon length, and stool consistency caused by DSS administration. Immunofluorescence of colon tissue section showed that Pangasius pangasius purified hydrolysate fraction enhanced the expression of occludin protein. This study hints at the use of Pangasius pangasius purified hydrolysate fraction as a potential nutraceutical or treatment agent in healing ulcers of the mucosa.
Collapse
Affiliation(s)
- K Sivaraman
- Department of Biotechnology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - C Shanthi
- Department of Biotechnology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India.
| |
Collapse
|
23
|
Yu H, Mi C, Wang Q, Dai G, Zhang T, Zhang G, Xie K, Zhao Z. Long noncoding RNA profiling reveals that LncRNA BTN3A2 inhibits the host inflammatory response to Eimeria tenella infection in chickens. Front Immunol 2022; 13:891001. [PMID: 36091044 PMCID: PMC9452752 DOI: 10.3389/fimmu.2022.891001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/05/2022] [Indexed: 02/02/2023] Open
Abstract
Coccidiosis is a widespread parasitic disease that causes serious economic losses to the poultry industry every year. Long noncoding RNAs (lncRNAs) play important roles in transcriptional regulation and are involved in a variety of diseases and immune responses. However, the lncRNAs associated with Eimeria tenella (E. tenella) resistance have not been identified in chickens. In addition, the expression profiles and functions of lncRNAs during E. tenella infection remain unclear. In the present study, high-throughput sequencing was applied to identify lncRNAs in chicken cecal tissues from control (JC), resistant (JR), and susceptible (JS) groups on day 4.5 post-infection (pi), and functional tests were performed. A total of 564 lncRNAs were differentially expressed, including 263 lncRNAs between the JS and JC groups, 192 between the JR and JS groups, and 109 between the JR and JC groups. Functional analyses indicated that these differentially expressed lncRNAs were involved in pathways related to E. tenella infection, including the NF-kappa B signaling, B cell receptor signaling and natural killer cell-mediated cytotoxicity pathways. Moreover, through cis regulation network analysis of the differentially expressed lncRNAs, we found that a novel lncRNA termed lncRNA BTN3A2 was significantly increased in both cecum tissue and DF-1 cells after coccidia infection or sporozoite stimulation. Functional test data showed that the overexpression of lncRNA BTN3A2 reduced the production of inflammatory cytokines, including IL-6, IL-1β, TNF-α and IL-8, while lncRNA BTN3A2 knockdown promoted the production of these inflammatory cytokines. Taken together, this study identify the differentially expressed lncRNAs during E. tenella infection in chickens for the first time and provide the direct evidence that lncRNA BTN3A2 regulates the host immune response to coccidia infection.
Collapse
Affiliation(s)
- Hailiang Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Changhao Mi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Qi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Guojun Dai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- *Correspondence: Guojun Dai,
| | - Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Genxi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Kaizhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhenhua Zhao
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| |
Collapse
|
24
|
Gough EK. The impact of mass drug administration of antibiotics on the gut microbiota of target populations. Infect Dis Poverty 2022; 11:76. [PMID: 35773678 PMCID: PMC9245274 DOI: 10.1186/s40249-022-00999-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
Antibiotics have become a mainstay of healthcare in the past century due to their activity against pathogens. This manuscript reviews the impact of antibiotic use on the intestinal microbiota in the context of mass drug administration (MDA). The importance of the gut microbiota to human metabolism and physiology is now well established, and antibiotic exposure may impact host health via collateral effects on the microbiota and its functions. To gain further insight into how gut microbiota respond to antibiotic perturbation and the implications for public health, factors that influence the impact of antibiotic exposure on the microbiota, potential health outcomes of antibiotic-induced microbiota alterations, and strategies that have the potential to ameliorate these wider antibiotic-associated microbiota perturbations are also reviewed.
Collapse
Affiliation(s)
- Ethan K Gough
- Department of International Health, Human Nutrition Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
25
|
Rahman MM, Islam F, -Or-Rashid MH, Mamun AA, Rahaman MS, Islam MM, Meem AFK, Sutradhar PR, Mitra S, Mimi AA, Emran TB, Fatimawali, Idroes R, Tallei TE, Ahmed M, Cavalu S. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front Cell Infect Microbiol 2022; 12:903570. [PMID: 35795187 PMCID: PMC9251340 DOI: 10.3389/fcimb.2022.903570] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
In the last two decades, considerable interest has been shown in understanding the development of the gut microbiota and its internal and external effects on the intestine, as well as the risk factors for cardiovascular diseases (CVDs) such as metabolic syndrome. The intestinal microbiota plays a pivotal role in human health and disease. Recent studies revealed that the gut microbiota can affect the host body. CVDs are a leading cause of morbidity and mortality, and patients favor death over chronic kidney disease. For the function of gut microbiota in the host, molecules have to penetrate the intestinal epithelium or the surface cells of the host. Gut microbiota can utilize trimethylamine, N-oxide, short-chain fatty acids, and primary and secondary bile acid pathways. By affecting these living cells, the gut microbiota can cause heart failure, atherosclerosis, hypertension, myocardial fibrosis, myocardial infarction, and coronary artery disease. Previous studies of the gut microbiota and its relation to stroke pathogenesis and its consequences can provide new therapeutic prospects. This review highlights the interplay between the microbiota and its metabolites and addresses related interventions for the treatment of CVDs.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Harun -Or-Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Md. Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Mohaimenul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Atkia Farzana Khan Meem
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Popy Rani Sutradhar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Anjuman Ara Mimi
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Fatimawali
- Pharmacy Study Program, Faculty of Mathematics and Natural Sciences, University of Sam Ratulangi, Manado, Indonesia
| | - Rinaldi Idroes
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Syiah Kuala, Banda Aceh, Indonesia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, University of Sam Ratulangi, Manado, Indonesia
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
26
|
Wegierska AE, Charitos IA, Topi S, Potenza MA, Montagnani M, Santacroce L. The Connection Between Physical Exercise and Gut Microbiota: Implications for Competitive Sports Athletes. Sports Med 2022; 52:2355-2369. [PMID: 35596883 PMCID: PMC9474385 DOI: 10.1007/s40279-022-01696-x] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2022] [Indexed: 12/16/2022]
Abstract
Gut microbiota refers to those microorganisms in the human digestive tract that display activities fundamental in human life. With at least 4 million different bacterial types, the gut microbiota is composed of bacteria that are present at levels sixfold greater than the total number of cells in the entire human body. Among its multiple functions, the microbiota helps promote the bioavailability of some nutrients and the metabolization of food, and protects the intestinal mucosa from the aggression of pathogenic microorganisms. Moreover, by stimulating the production of intestinal mediators able to reach the central nervous system (gut/brain axis), the gut microbiota participates in the modulation of human moods and behaviors. Several endogenous and exogenous factors can cause dysbiosis with important consequences on the composition and functions of the microbiota. Recent research underlines the importance of appropriate physical activity (such as sports), nutrition, and a healthy lifestyle to ensure the presence of a functional physiological microbiota working to maintain the health of the whole human organism. Indeed, in addition to bowel disturbances, variations in the qualitative and quantitative microbial composition of the gastrointestinal tract might have systemic negative effects. Here, we review recent studies on the effects of physical activity on gut microbiota with the aim of identifying potential mechanisms by which exercise could affect gut microbiota composition and function. Whether physical exercise of variable work intensity might reflect changes in intestinal health is analyzed.
Collapse
Affiliation(s)
- Angelika Elzbieta Wegierska
- Interdisciplinary Department of Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy.,Italian Athletics Federation (FIDAL), Rome, Italy
| | - Ioannis Alexandros Charitos
- Emergency/Urgent Department, National Poisoning Center, Riuniti University Hospital of Foggia, Foggia, Italy
| | - Skender Topi
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan "A. Xhuvani", Elbasan, Albania
| | - Maria Assunta Potenza
- Department of Biomedical Sciences and Human Oncology-Section of Pharmacology, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology-Section of Pharmacology, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
27
|
Ramires LC, Santos GS, Ramires RP, da Fonseca LF, Jeyaraman M, Muthu S, Lana AV, Azzini G, Smith CS, Lana JF. The Association between Gut Microbiota and Osteoarthritis: Does the Disease Begin in the Gut? Int J Mol Sci 2022; 23:1494. [PMID: 35163417 PMCID: PMC8835947 DOI: 10.3390/ijms23031494] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/11/2022] [Accepted: 01/25/2022] [Indexed: 02/05/2023] Open
Abstract
Some say that all diseases begin in the gut. Interestingly, this concept is actually quite old, since it is attributed to the Ancient Greek physician Hippocrates, who proposed the hypothesis nearly 2500 years ago. The continuous breakthroughs in modern medicine have transformed our classic understanding of the gastrointestinal tract (GIT) and human health. Although the gut microbiota (GMB) has proven to be a core component of human health under standard metabolic conditions, there is now also a strong link connecting the composition and function of the GMB to the development of numerous diseases, especially the ones of musculoskeletal nature. The symbiotic microbes that reside in the gastrointestinal tract are very sensitive to biochemical stimuli and may respond in many different ways depending on the nature of these biological signals. Certain variables such as nutrition and physical modulation can either enhance or disrupt the equilibrium between the various species of gut microbes. In fact, fat-rich diets can cause dysbiosis, which decreases the number of protective bacteria and compromises the integrity of the epithelial barrier in the GIT. Overgrowth of pathogenic microbes then release higher quantities of toxic metabolites into the circulatory system, especially the pro-inflammatory cytokines detected in osteoarthritis (OA), thereby promoting inflammation and the initiation of many disease processes throughout the body. Although many studies link OA with GMB perturbations, further research is still needed.
Collapse
Affiliation(s)
- Luciano C. Ramires
- Department of Orthopaedics and Sports Medicine, Mãe de Deus Hospital, Porto Alegre 90110-270, RS, Brazil;
| | - Gabriel Silva Santos
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, SP, Brazil; (G.A.); (J.F.L.)
| | - Rafaela Pereira Ramires
- Department of Biology, Cellular, Molecular and Biomedical Science, Boise State University, 1910 W University Drive, Boise, ID 83725, USA;
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, The Federal University of São Paulo, São Paulo 04024-002, SP, Brazil
| | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine, Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India;
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, Tamil Nadu, India;
| | - Anna Vitória Lana
- Department of Medicine, Max Planck University Center, Indaiatuba 13343-060, SP, Brazil;
| | - Gabriel Azzini
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, SP, Brazil; (G.A.); (J.F.L.)
| | - Curtis Scott Smith
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 83703, USA;
| | - José Fábio Lana
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, SP, Brazil; (G.A.); (J.F.L.)
| |
Collapse
|
28
|
Wickramasuriya SS, Park I, Lee K, Lee Y, Kim WH, Nam H, Lillehoj HS. Role of Physiology, Immunity, Microbiota, and Infectious Diseases in the Gut Health of Poultry. Vaccines (Basel) 2022; 10:vaccines10020172. [PMID: 35214631 PMCID: PMC8875638 DOI: 10.3390/vaccines10020172] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/10/2023] Open
Abstract
“Gut health” refers to the physical state and physiological function of the gastrointestinal tract and in the livestock system; this topic is often focused on the complex interacting components of the intestinal system that influence animal growth performance and host-microbial homeostasis. Regardless, there is an increasing need to better understand the complexity of the intestinal system and the various factors that influence gut health, since the intestine is the largest immune and neuroendocrine organ that interacts with the most complex microbiome population. As we face the post-antibiotic growth promoters (AGP) era in many countries of the world, livestock need more options to deal with food security, food safety, and antibiotic resilience to maintain agricultural sustainability to feed the increasing human population. Furthermore, developing novel antibiotic alternative strategies needs a comprehensive understanding of how this complex system maintains homeostasis as we face unpredictable changes in external factors like antibiotic-resistant microbes, farming practices, climate changes, and consumers’ preferences for food. In this review, we attempt to assemble and summarize all the relevant information on chicken gut health to provide deeper insights into various aspects of gut health. Due to the broad and complex nature of the concept of “gut health”, we have highlighted the most pertinent factors related to the field performance of broiler chickens.
Collapse
Affiliation(s)
- Samiru S. Wickramasuriya
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
| | - Inkyung Park
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
| | - Kyungwoo Lee
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
- Department of Animal Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Youngsub Lee
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
| | - Woo H. Kim
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
- College of Veterinary Medicine and Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Hyoyoun Nam
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
| | - Hyun S. Lillehoj
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA; (S.S.W.); (I.P.); (K.L.); (Y.L.); (W.H.K.); (H.N.)
- Correspondence: ; Tel.: +1-301-504-8771
| |
Collapse
|
29
|
Murphy EA, Velázquez KT. The role of diet and physical activity in influencing the microbiota/microbiome. DIET, INFLAMMATION, AND HEALTH 2022:693-745. [DOI: 10.1016/b978-0-12-822130-3.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
30
|
Yang X, Shi A, Song Y, Niu C, Yu X, Shi X, Pang Y, Ma X, Cheng Y. The effects of ammonia-N stress on immune parameters, antioxidant capacity, digestive function, and intestinal microflora of Chinese mitten crab, Eriocheir sinensis, and the protective effect of dietary supplement of melatonin. Comp Biochem Physiol C Toxicol Pharmacol 2021; 250:109127. [PMID: 34252579 DOI: 10.1016/j.cbpc.2021.109127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/18/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022]
Abstract
Ammonia nitrogen pollution seriously affects the economic benefits of Chinese mitten crab (Eriocheir sinensis) farming. In this study, we first evaluated the protective effects of melatonin (MT) on immune parameters, antioxidant capacity, and digestive enzymes of E. sinensis under acute ammonia nitrogen stress. The results showed that ammonia-N stress significantly decreased the antibacterial ability of crabs, nevertheless MT could significantly improve it under ammonia-N stress (P < 0.05). Ammonia-N group hemolymph antioxidant capacity indicators (T-AOC, T-SOD, GSH-Px) were significantly decreased than control (p < 0.05), while the MT ammonia-N group hemolymph T-SOD activity significantly increased than ammonia-N group (p < 0.05). For hepatopancreas, ammonia-N group GSH-PX activity significantly decreased than control group, but MT ammonia-N group was significant increased than ammonia-N (p < 0.05). Ammonia-N stress has significantly increased the content of MDA in hemolymph and hepatopancreas (p < 0.05), but MT ammonia-N treatment significantly decreased than ammonia-N group (p < 0.05). Compared with the control group, ammonia-N significantly reduced the activities of Trypsin in the intestine and hepatopancreas (p < 0.05), while MT ammonia-N group can significantly improve the intestinal trypsin activity than ammonia-N (p < 0.05). The intestinal microbiota of E. sinensis results showed that ammonia-N stress significantly decreased the relative abundance of Bacteroidetes (p < 0.05). Ammonia-N stress significantly decreased the Dysgonomonas and Rubellimicrobium, and the Citrobacter significantly increased. In summary, melatonin has a protective effect on E. sinensis under ammonia-N stress. Acute ammonia-N stress may lead to the decrease of probiotics and the increase of pathogenic bacteria, which may be closely related to the impairment of digestive function and immune function.
Collapse
Affiliation(s)
- Xiaozhen Yang
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| | - Aoya Shi
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yameng Song
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Chao Niu
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Xiaowen Yu
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Xingliang Shi
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yangyang Pang
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Xueli Ma
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yongxu Cheng
- National Demonstration Center for Experimental Fisheries Science Education, Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
31
|
Li J, Gálvez EJC, Amend L, Almási É, Iljazovic A, Lesker TR, Bielecka AA, Schorr EM, Strowig T. A versatile genetic toolbox for Prevotella copri enables studying polysaccharide utilization systems. EMBO J 2021; 40:e108287. [PMID: 34676563 PMCID: PMC8634118 DOI: 10.15252/embj.2021108287] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/08/2021] [Accepted: 09/24/2021] [Indexed: 12/30/2022] Open
Abstract
Prevotella copri is a prevalent inhabitant of the human gut and has been associated with plant‐rich diet consumption and diverse health states. The underlying genetic basis of these associations remains enigmatic due to the lack of genetic tools. Here, we developed a novel versatile genetic toolbox for rapid and efficient genetic insertion and allelic exchange applicable to P. copri strains from multiple clades. Enabled by the genetic platform, we systematically investigated the specificity of polysaccharide utilization loci (PULs) and identified four highly conserved PULs for utilizing arabinan, pectic galactan, arabinoxylan, and inulin, respectively. Further genetic and functional analysis of arabinan utilization systems illustrate that P. copri has evolved two distinct types of arabinan‐processing PULs (PULAra) and that the type‐II PULAra is significantly enriched in individuals consuming a vegan diet compared to other diets. In summary, this genetic toolbox will enable functional genetic studies for P. copri in future.
Collapse
Affiliation(s)
- Jing Li
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eric J C Gálvez
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Hannover Medical School, Hannover, Germany
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Éva Almási
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Aida Iljazovic
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till R Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Agata A Bielecka
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eva-Magdalena Schorr
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Hannover Medical School, Hannover, Germany.,Centre for Individualized Infection Medicine, Hannover, Germany
| |
Collapse
|
32
|
Wen C, Yan W, Mai C, Duan Z, Zheng J, Sun C, Yang N. Joint contributions of the gut microbiota and host genetics to feed efficiency in chickens. MICROBIOME 2021; 9:126. [PMID: 34074340 PMCID: PMC8171024 DOI: 10.1186/s40168-021-01040-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/22/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Feed contributes most to livestock production costs. Improving feed efficiency is crucial to increase profitability and sustainability for animal production. Host genetics and the gut microbiota can both influence the host phenotype. However, the association between the gut microbiota and host genetics and their joint contribution to feed efficiency in chickens is largely unclear. RESULTS Here, we examined microbial data from the duodenum, jejunum, ileum, cecum, and feces in 206 chickens and their host genotypes and confirmed that the microbial phenotypes and co-occurrence networks exhibited dramatic spatial heterogeneity along the digestive tract. The correlations between host genetic kinship and gut microbial similarities within different sampling sites were weak, with coefficients ranging from - 0.07 to 0.08. However, microbial genome-wide analysis revealed that genetic markers near or inside the genes MTHFD1L and LARGE1 were associated with the abundances of cecal Megasphaera and Parabacteroides, respectively. The effect of host genetics on residual feed intake (RFI) was 39%. We further identified three independent genetic variations that were related to feed efficiency and had a modest effect on the gut microbiota. The contributions of the gut microbiota from the different parts of the intestinal tract on RFI were distinct. The cecal microbiota accounted for 28% of the RFI variance, a value higher than that explained by the duodenal, jejunal, ileal, and fecal microbiota. Additionally, six bacteria exhibited significant associations with RFI. Specifically, lower abundances of duodenal Akkermansia muciniphila and cecal Parabacteroides and higher abundances of cecal Lactobacillus, Corynebacterium, Coprobacillus, and Slackia were related to better feed efficiency. CONCLUSIONS Our findings solidified the notion that both host genetics and the gut microbiota, especially the cecal microbiota, can drive the variation in feed efficiency. Although host genetics has a limited effect on the entire microbial community, a small fraction of gut microorganisms tends to interact with host genes, jointly contributing to feed efficiency. Therefore, the gut microbiota and host genetic variations can be simultaneously targeted by favoring more-efficient taxa and selective breeding to improve feed efficiency in chickens. Video abstract.
Collapse
Affiliation(s)
- Chaoliang Wen
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
| | - Wei Yan
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
| | - Chunning Mai
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
| | - Zhongyi Duan
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- National Animal Husbandry Service, Beijing, 100125, China
| | - Jiangxia Zheng
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China.
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
33
|
Geng R, Liu H, Tan K, Wang Z, Wang W. RNase1 can modulate gut microbiota and metabolome after Aeromonas hydrophila infection in blunt snout bream. Environ Microbiol 2021; 23:5258-5272. [PMID: 33973327 DOI: 10.1111/1462-2920.15564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/18/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022]
Abstract
Pancreatic ribonuclease (RNase1) of Megalobrama amblycephala exhibits both antimicrobial and digestive activity. The gut microbiome improve the digestion and metabolic capacity and enhance the functioning of the immune system of the host against pathogenic bacteria. In this study, we aimed to assess the protective effect of RNase1 on Aeromonas hydrophila-induced inflammation and intestinal microbial metabolism. Megalobrama amblycephala were randomly divided into three groups: control (injected PBS), infection (A. hydrophila-injected), and treatment group (RNase1 pretreatment 24 h before the A. hydrophila injection). The morphological symptoms were significantly alleviated by RNase1. RNase1 reshaped the perturbed gut microbiota by upregulating Proteobacteria and Vibrio richness and downregulating Firmicutes, Chlamydiae, Bacillus, and Gemmobacter richness. The lysophosphatidylcholine, (±) 17 HETE, D- (+) -cellobiose, and PC (20:5) in the treatment group were restored by RNase 1 protein treatment to the level of the control group. In the treatment group, phospholipid metabolism, fatty acid metabolism, glucose metabolism and lipid metabolism were different from the control and infection groups. The proinflammatory factors concentration in intestinal samples significantly increased after A. hydrophila infection. Our results revealed that RNase1 plays an important role in resistance to pathogen invasion, reducing inflammation, and improving intestinal function, thus inhibiting the occurrence of disease.
Collapse
Affiliation(s)
- Ruijing Geng
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
| | - Han Liu
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China.,Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Engineering Technology Research Center for Fish Breeding and Culture in Hubei Province, Wuhan, 430070, China
| | - Kianann Tan
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhiqiang Wang
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
| | - Weimin Wang
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
34
|
Myers S, Do T, Meade JL, Tugnait A, Vernon JJ, Pistolic J, Hancock REW, Marsh PD, Trivedi HM, Chen D, Devine DA. Immunomodulatory streptococci that inhibit CXCL8 secretion and NFκB activation are common members of the oral microbiota. J Med Microbiol 2021; 70. [PMID: 33734952 PMCID: PMC8346732 DOI: 10.1099/jmm.0.001329] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Introduction Oral tissues are generally homeostatic despite exposure to many potential inflammatory agents including the resident microbiota. This requires the balancing of inflammation by regulatory mechanisms and/or anti-inflammatory commensal bacteria. Thus, the levels of anti-inflammatory commensal bacteria in resident populations may be critical in maintaining this homeostatic balance. Hypothesis/Gap Statement The incidence of immunosuppressive streptococci in the oral cavity is not well established. Determining the proportion of these organisms and the mechanisms involved may help to understand host-microbe homeostasis and inform development of probiotics or prebiotics in the maintenance of oral health. Aim To determine the incidence and potential modes of action of immunosuppressive capacity in resident oral streptococci. Methodology Supragingival plaque was collected from five healthy participants and supragingival and subgingival plaque from five with gingivitis. Twenty streptococci from each sample were co-cultured with epithelial cells±flagellin or LL-37. CXCL8 secretion was detected by ELISA, induction of cytotoxicity in human epithelial cells by lactate dehydrogenase release and NFκB-activation using a reporter cell line. Bacterial identification was achieved through partial 16S rRNA gene sequencing and next-generation sequencing. Results CXCL8 secretion was inhibited by 94/300 isolates. Immunosuppressive isolates were detected in supragingival plaque from healthy (4/5) and gingivitis (4/5) samples, and in 2/5 subgingival (gingivitis) plaque samples. Most were Streptococcus mitis/oralis. Seventeen representative immunosuppressive isolates all inhibited NFκB activation. The immunosuppressive mechanism was strain specific, often mediated by ultra-violet light-labile factors, whilst bacterial viability was essential in certain species. Conclusion Many streptococci isolated from plaque suppressed epithelial cell CXCL8 secretion, via inhibition of NFκB. This phenomenon may play an important role in oral host-microbe homeostasis.
Collapse
Affiliation(s)
- Sarah Myers
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - Thuy Do
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - Josephine L Meade
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - Aradhna Tugnait
- Division of Restorative Dentistry, University of Leeds, Leeds, UK
| | - Jon J Vernon
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - Jelena Pistolic
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Columbia, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Columbia, Canada
| | - Philip D Marsh
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | | | | | - Deirdre A Devine
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| |
Collapse
|
35
|
Talyuli OAC, Bottino-Rojas V, Polycarpo CR, Oliveira PL, Paiva-Silva GO. Non-immune Traits Triggered by Blood Intake Impact Vectorial Competence. Front Physiol 2021; 12:638033. [PMID: 33737885 PMCID: PMC7960658 DOI: 10.3389/fphys.2021.638033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
Blood-feeding arthropods are considered an enormous public health threat. They are vectors of a plethora of infectious agents that cause potentially fatal diseases like Malaria, Dengue fever, Leishmaniasis, and Lyme disease. These vectors shine due to their own physiological idiosyncrasies, but one biological aspect brings them all together: the requirement of blood intake for development and reproduction. It is through blood-feeding that they acquire pathogens and during blood digestion that they summon a collection of multisystemic events critical for vector competence. The literature is focused on how classical immune pathways (Toll, IMD, and JAK/Stat) are elicited throughout the course of vector infection. Still, they are not the sole determinants of host permissiveness. The dramatic changes that are the hallmark of the insect physiology after a blood meal intake are the landscape where a successful infection takes place. Dominant processes that occur in response to a blood meal are not canonical immunological traits yet are critical in establishing vector competence. These include hormonal circuitries and reproductive physiology, midgut permeability barriers, midgut homeostasis, energy metabolism, and proteolytic activity. On the other hand, the parasites themselves have a role in the outcome of these blood triggered physiological events, consistently using them in their favor. Here, to enlighten the knowledge on vector-pathogen interaction beyond the immune pathways, we will explore different aspects of the vector physiology, discussing how they give support to these long-dated host-parasite relationships.
Collapse
Affiliation(s)
- Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla R Polycarpo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Soopramanien M, Khan NA, Siddiqui R. Gut microbiota of animals living in polluted environments are a potential resource of anticancer molecules. J Appl Microbiol 2021; 131:1039-1055. [PMID: 33368930 DOI: 10.1111/jam.14981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/09/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022]
Abstract
Cancer is a prominent cause of morbidity and mortality worldwide, in spite of advances in therapeutic interventions and supportive care. In 2018 alone, there were 18·1 million new cancer cases and 9·6 million deaths indicating the need for novel anticancer agents. Plant-based products have often been linked with protective effects against communicable and non-communicable diseases. Recently, we have shown that animals such as crocodiles thrive in polluted environments and are often exposed to carcinogenic agents, but still benefit from prolonged lifespan. The protective mechanisms shielding them from cancer could be attributed to the immune system, and/or it is possible that their gut microbiota produce anticancer molecules. In support, several lines of evidence suggest that gut microbiota plays a critical role in the physiology of its host. Here, we reviewed the available literature to assess whether the gut microbiota of animals thriving in polluted environment possess anticancer molecules.
Collapse
Affiliation(s)
- M Soopramanien
- Department of Biological Sciences, Sunway University, Bandar Sunway, Malaysia
| | - N A Khan
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - R Siddiqui
- College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| |
Collapse
|
37
|
Sun X, Liu Y, Jiang P, Song S, Ai C. Interaction of sulfated polysaccharides with intestinal Bacteroidales plays an important role in its biological activities. Int J Biol Macromol 2020; 168:496-506. [PMID: 33321137 DOI: 10.1016/j.ijbiomac.2020.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/19/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022]
Abstract
The bioactivities of sulfated polysaccharides have shown to be associated with the gut microbiota, but the underlying mechanisms remain unclear. In this study, the effect of sulfated polysaccharides from pacific abalone (AGSP) on the human gut microbiota was analyzed via an in vitro fermentation model. The results revealed that AGSP altered the overall structure of the gut microbiota and increased relative abundances of some Bacteroidales members, implying that intestinal Bacteroidales can play important roles in the bioactivities of AGSP. To elucidate the underlying mechanisms, some species from the Bacteroides and Parabacteroides within Bacteroidales were isolated, and their characteristics on AGSP utilization were analyzed. It showed that AGSP utilization by intestinal Bacteroidales was species-dependent, and some species that liberated AGSP breakdown products promoted the growth of others unable to live in AGSP, forming an AGSP utilization network. The in vitro cell model showed that AGSP oligosaccharides had better anti-inflammatory activity and weaker cytotoxicity, implying that microbial degradation of AGSP can influence its reaction with host cells. These results indicated that the interaction between polysaccharides and gut microbes can together determine the beneficial effects of polysaccharides on the host health.
Collapse
Affiliation(s)
- Xiaona Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Yili Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Pingrui Jiang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
38
|
Bajinka O, Darboe A, Tan Y, Abdelhalim KA, Cham LB. Gut microbiota and the human gut physiological changes. ANN MICROBIOL 2020. [DOI: 10.1186/s13213-020-01608-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
The human gut can be colonized by number of microorganisms. The most studied are bacteria, which changes from birth to newborn born into adult-like gut microbiota. Much is known about the effects of dietary, medications, and lifestyles on the bacterial composition. However, the host physiological changes influencing the gut microbiota, the immediate consequences, and the possible gut microbiota therapy are not studied at length. This review is based profoundly on animal model studies through experimentation and some human clinical trials for the past 20 years.
Forward
The physiological factors studied to influences gut microbiota are bacterial mucosal receptors, mucin glycosylation, mucus, epithelial microvilli, and tight junction. Host secretions and immune response such as immunity, secretory A (sIgA), inflammasome, innate immunity, immune response, glycans, bile acids, peristalsis, microRNA, and adhesion to intestinal glycans are as well found to confer variety of alterations on gut microbial flora.
Conclusion
Despite the resilience of the gut microbiota in response to changes, chain of events causes the imbalance microbiota. Increased pro-inflammatory potential with the help of cell barriers, host secretions, and immune response mediate gut recovery.
Collapse
|
39
|
黄 嘉, 王 利, 吴 小, 陈 焕, 付 秀, 陈 少, 刘 涛. [Analysis of intestinal flora in patients with chronic rhinosinusitis based on highthroughput sequencing]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1319-1324. [PMID: 32990228 PMCID: PMC7544583 DOI: 10.12122/j.issn.1673-4254.2020.09.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the changes in diversity, relative abundance and distribution of intestinal flora in patients with chronic rhinosinusitis and nasal polyps (CRSwNP) using high-throughput sequencing technology identify the intestinal flora significantly related to pathogenesis and progression of CRSwNP. METHODS Ten patients with CRSwNP hospitalized in the Department of Otolaryngology-Head and Neck Surgery of Guangdong Provincial People's Hospital were selected as the case group with 10 healthy volunteers recruited in the same period as the control group. Fecal genomic DNA extraction kit was used to extract the DNA in the fecal samples, and the DNA fragment length was measured and quantified. The V3 and V4 highly variable regions of the 16S rDNA gene of prokaryotes were amplified followed by library construction, Illumina MiSeq sequencing, sequence alignment and species identification analysis. The relative abundance, diversity and distribution characteristics of the intestinal flora were analyzed, and the relevant metabolic pathways were predicted. RESULTS Compared with the control group, the patients with CRSwNP had significant changes in the overall structure of the intestinal flora, highlighted by increased abundance of Saccharopolyspora and decreased contents of Ruminococcae, Coprococcus, Collinsella and Dialister. Among the metabolic pathways predicted to be associated with CRSwNP, 9 showed significant changes in patients with CRSwNP as compared with the control group (P < 0.05). CONCLUSIONS Patients with CRSwNP have significant changes in the structural characteristics of intestinal flora related with multiple metabolic pathways, and these changes may play an important role in the development of chronic rhinosinusitis.
Collapse
Affiliation(s)
- 嘉裕 黄
- 广东省人民医院//广东省医学科学院耳鼻咽喉头颈外科,广东 广州 510080Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- 汕头大学医学院,广东 汕头 515063Shantou University Medical College, Shantou 515063, China
| | - 利平 王
- 广东省人民医院//广东省医学科学院耳鼻咽喉头颈外科,广东 广州 510080Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - 小琴 吴
- 广东省人民医院//广东省医学科学院耳鼻咽喉头颈外科,广东 广州 510080Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - 焕钧 陈
- 广东省英德市人民医院耳鼻咽喉科,广东 英德 513000Department of Otolaryngology, People's Hospital of Yingde City, Yingde 513000, China
| | - 秀丽 付
- 广东省英德市人民医院耳鼻咽喉科,广东 英德 513000Department of Otolaryngology, People's Hospital of Yingde City, Yingde 513000, China
| | - 少华 陈
- 广东省人民医院//广东省医学科学院耳鼻咽喉头颈外科,广东 广州 510080Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - 涛 刘
- 广东省人民医院//广东省医学科学院耳鼻咽喉头颈外科,广东 广州 510080Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
40
|
The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins (Basel) 2020; 12:toxins12100619. [PMID: 32998222 PMCID: PMC7600953 DOI: 10.3390/toxins12100619] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Mycotoxins are fungal metabolites that occur in human foods and animal feeds, potentially threatening human and animal health. The intestine is considered as the first barrier against these external contaminants, and it consists of interconnected physical, chemical, immunological, and microbial barriers. In this context, based on in vitro, ex vivo, and in vivo models, we summarize the literature for compromised intestinal barrier issues caused by various mycotoxins, and we reviewed events related to disrupted intestinal integrity (physical barrier), thinned mucus layer (chemical barrier), imbalanced inflammatory factors (immunological barrier), and dysfunctional bacterial homeostasis (microbial barrier). We also provide important information on deoxynivalenol, a leading mycotoxin implicated in intestinal dysfunction, and other adverse intestinal effects induced by other mycotoxins, including aflatoxins and ochratoxin A. In addition, intestinal perturbations caused by mycotoxins may also contribute to the development of mycotoxicosis, including human chronic intestinal inflammatory diseases. Therefore, we provide a clear understanding of compromised intestinal barrier induced by mycotoxins, with a view to potentially develop innovative strategies to prevent and treat mycotoxicosis. In addition, because of increased combinatorial interactions between mycotoxins, we explore the interactive effects of multiple mycotoxins in this review.
Collapse
|
41
|
Lema I, Araújo JR, Rolhion N, Demignot S. Jejunum: The understudied meeting place of dietary lipids and the microbiota. Biochimie 2020; 178:124-136. [PMID: 32949677 DOI: 10.1016/j.biochi.2020.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Although the jejunum is the main intestinal compartment responsible for lipid digestion and absorption, most of the studies assessing the impact of dietary lipids on the intestinal microbiota have been performed in the ileum, colon and faeces. This lack of interest in the jejunum is due to the much lower number of microbes present in this intestinal region and to the difficulty in accessing its lumen, which requires invasive methods. Recently, several recent publications highlighted that the whole jejunal microbiota or specific bacterial members are able to modulate lipid absorption and metabolism in enterocytes. This information reveals new strategies in the development of bacterial- and metabolite-based therapeutic interventions or nutraceutical recommendations to treat or prevent metabolic-related disorders, including obesity, cardiovascular diseases and malnutrition. This review is strictly focused on the following triad: dietary lipids, the jejunal epithelium and the jejunal microbiota. First, we will describe each member of the triad: the structure and functions of the jejunum, the composition of the jejunal microbiota, and dietary lipid handling by enterocytes and by microorganisms. Then, we will present the mechanisms leading to lipid malabsorption in small intestinal bacterial overgrowth (SIBO), a disease in which the jejunal microbiota is altered and which highlights the strong interactions among this triad. We will finally review the recent literature about the interactions among members of the triad, which should encourage research teams to further explore the mechanisms by which specific microbial strains or metabolites, alone or in concert, can mediate, control or modulate lipid absorption in the jejunum.
Collapse
Affiliation(s)
- Ingrid Lema
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, UMR_S 938, F-75012, Paris, France; EPHE, PSL University, F-75014, Paris, France
| | - João Ricardo Araújo
- Nutrition and Metabolism, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal; Center for Health Technology Services Research (CINTESIS), Oporto, Portugal
| | - Nathalie Rolhion
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, UMR_S 938, F-75012, Paris, France
| | - Sylvie Demignot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, UMR_S 938, F-75012, Paris, France; EPHE, PSL University, F-75014, Paris, France.
| |
Collapse
|
42
|
Gerardo NM, Hoang KL, Stoy KS. Evolution of animal immunity in the light of beneficial symbioses. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190601. [PMID: 32772666 DOI: 10.1098/rstb.2019.0601] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immune system processes serve as the backbone of animal defences against pathogens and thus have evolved under strong selection and coevolutionary dynamics. Most microorganisms that animals encounter, however, are not harmful, and many are actually beneficial. Selection should act on hosts to maintain these associations while preventing exploitation of within-host resources. Here, we consider how several key aspects of beneficial symbiotic associations may shape host immune system evolution. When host immunity is used to regulate symbiont populations, there should be selection to evolve and maintain targeted immune responses that recognize symbionts and suppress but not eliminate symbiont populations. Associating with protective symbionts could relax selection on the maintenance of redundant host-derived immune responses. Alternatively, symbionts could facilitate the evolution of host immune responses if symbiont-conferred protection allows for persistence of host populations that can then adapt. The trajectory of immune system evolution will likely differ based on the type of immunity involved, the symbiont transmission mode and the costs and benefits of immune system function. Overall, the expected influence of beneficial symbiosis on immunity evolution depends on how the host immune system interacts with symbionts, with some interactions leading to constraints while others possibly relax selection on immune system maintenance. This article is part of the theme issue 'The role of the microbiome in host evolution'.
Collapse
Affiliation(s)
- Nicole M Gerardo
- Department of Biology, Emory University, O. Wayne Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322, USA
| | - Kim L Hoang
- Department of Biology, Emory University, O. Wayne Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322, USA
| | - Kayla S Stoy
- Department of Biology, Emory University, O. Wayne Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322, USA
| |
Collapse
|
43
|
Lyu Y, Su C, Verbrugghe A, Van de Wiele T, Martos Martinez-Caja A, Hesta M. Past, Present, and Future of Gastrointestinal Microbiota Research in Cats. Front Microbiol 2020; 11:1661. [PMID: 32793152 PMCID: PMC7393142 DOI: 10.3389/fmicb.2020.01661] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
The relationship between microbial community and host has profound effects on the health of animals. A balanced gastrointestinal (GI) microbial population provides nutritional and metabolic benefits to its host, regulates the immune system and various signaling molecules, protects the intestine from pathogen invasion, and promotes a healthy intestinal structure and an optimal intestinal function. With the fast development of next-generation sequencing, molecular techniques have become standard tools for microbiota research, having been used to demonstrate the complex intestinal ecosystem. Similarly to other mammals, the vast majority of GI microbiota in cats (over 99%) is composed of the predominant bacterial phyla Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria. Many nutritional and clinical studies have shown that cats' microbiota can be affected by several different factors including body condition, age, diet, and inflammatory diseases. All these factors have different size effects, and some of these may be very minor, and it is currently unknown how important these are. Further research is needed to determine the functional variations in the microbiome in disease states and in response to environmental and/or dietary modulations. Additionally, further studies are also needed to explain the intricate relationship between GI microbiota and the genetics and immunity of its host. This review summarizes past and present knowledge of the feline GI microbiota and looks into the future possibilities and challenges of the field.
Collapse
Affiliation(s)
- Yang Lyu
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chunxia Su
- Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Adronie Verbrugghe
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Tom Van de Wiele
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Ana Martos Martinez-Caja
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Myriam Hesta
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
44
|
Slavin M, Li HA, Frankenfeld C, Cheskin LJ. What is Needed for Evidence-Based Dietary Recommendations for Migraine: A Call to Action for Nutrition and Microbiome Research. Headache 2020; 59:1566-1581. [PMID: 31603554 DOI: 10.1111/head.13658] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The gastrointestinal symptoms of migraine attacks have invited numerous dietary hypotheses for migraine etiology through the centuries. Substantial efforts have been dedicated to identifying dietary interventions for migraine attack prevention, with limited success. Meanwhile, mounting evidence suggests that the reverse relationship may also exist - that the biological mechanisms of migraine may influence dietary intake. More likely, the truth involves some combination of both, where the disease influences food intake, and the foods eaten impact the manifestations of the disease. In addition, the gut's microbiota is increasingly suspected to influence the migraine brain via the gut-brain axis, though these hypotheses remain largely unsubstantiated. OBJECTIVE This paper presents an overview of the strength of existing evidence for food-based dietary interventions for migraine, noting that there is frequently evidence to suggest that a dietary risk factor for migraine exists but no evidence for how to best intervene; in fact, our intuitive assumptions on interventions are being challenged with new evidence. We then look to the future for promising avenues of research, notably the gut microbiome. CONCLUSION The evidence supports a call to action for high-quality dietary and microbiome research in migraine, both to substantiate hypothesized relationships and build the evidence base regarding nutrition's potential impact on migraine attack prevention and treatment.
Collapse
Affiliation(s)
- Margaret Slavin
- Department of Nutrition and Food Studies, George Mason University, Fairfax, VA, USA
| | - Huilun Amber Li
- Department of Nutrition and Food Studies, George Mason University, Fairfax, VA, USA
| | - Cara Frankenfeld
- Department of Global and Community Health, George Mason University, Fairfax, VA, USA
| | - Lawrence J Cheskin
- Department of Nutrition and Food Studies, George Mason University, Fairfax, VA, USA
| |
Collapse
|
45
|
Bergman P, Raqib R, Rekha RS, Agerberth B, Gudmundsson GH. Host Directed Therapy Against Infection by Boosting Innate Immunity. Front Immunol 2020; 11:1209. [PMID: 32595649 PMCID: PMC7304486 DOI: 10.3389/fimmu.2020.01209] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022] Open
Abstract
The innate immune system constitutes the first line of defense against invading pathogens, regulating the normal microbiota and contributes to homeostasis. Today we have obtained detailed knowledge on receptors, signaling pathways, and effector molecules of innate immunity. Our research constellation has focused on ways to induce the expression of antimicrobial peptides (AMPs), the production of oxygen species (ROS and NO), and to activate autophagy, during the last two decades. These innate effectors, with different mechanisms of action, constitute a powerful defense armament in phagocytes and in epithelial cells. Innate immunity does not only protect the host from invading pathogens, but also regulates the composition of the microbiota, which is an area of intense research. Notably, some virulent bacteria have the capacity to downregulate innate defenses and can thereby cause invasive disease. Understanding the detailed mechanisms behind pathogen-mediated suppression of innate effectors are currently in progress. This information can be of importance for the development of novel treatments based on counteraction of the downregulation; we have designated this type of treatment as host directed therapy (HDT). The concept to boost innate immunity may be particularly relevant as many pathogens are developing resistance against classical antibiotics. Many pathogens that are resistant to antibiotics are sensitive to the endogenous effectors included in early host defenses, which contain multiple effectors working in cooperation to control infections. Here, we review recent data related to downregulation of AMPs by pathogenic bacteria, induction of innate effector mechanisms, including cytokine-mediated effects, repurposed drugs and the role of antibiotics as direct modulators of host responses. These findings can form a platform for the development of novel treatment strategies against infection and/or inflammation.
Collapse
Affiliation(s)
- Peter Bergman
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,The Immunodeficiency Unit, Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Rubhana Raqib
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rokeya Sultana Rekha
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Agerberth
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gudmundur H Gudmundsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Biomedical Center, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
46
|
Ma N, Zhang J, Reiter RJ, Ma X. Melatonin mediates mucosal immune cells, microbial metabolism, and rhythm crosstalk: A therapeutic target to reduce intestinal inflammation. Med Res Rev 2020; 40:606-632. [PMID: 31420885 DOI: 10.1002/med.21628] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
Nowadays, melatonin, previously considered only as a pharmaceutical product for rhythm regulation and sleep aiding, has shown its potential as a co-adjuvant treatment in intestinal diseases, however, its mechanism is still not very clear. A firm connection between melatonin at a physiologically relevant concentration and the gut microbiota and inflammation has recently established. Herein, we summarize their crosstalk and focus on four novelties. First, how melatonin is synthesized and degraded in the gut and exerts potentially diverse phenotypic effects through its diverse metabolites. Second, how melatonin mediates the activation and proliferation of intestinal mucosal immune cells with paracrine and autocrine properties. By modulating T/B cells, mast cells, macrophages and dendritic cells, melatonin immunomodulatory involved in regulating T-cell differentiation, intervening T/B cell interaction and attenuating the production of pro-inflammatory factors, achieving its antioxidant action via specific receptors. Third, how melatonin exerts antimicrobial action and modulates microbial components, such as lipopolysaccharide, amyloid-β peptides via nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) or signal transducers and activators of transcription (STAT1) pathway to modulate intestinal immune function in immune-pineal axis. The last, how melatonin mediates the effect of intestinal bacterial activity signals on the body rhythm system through the NF-κB pathway and influences the mucosal epithelium oscillation via clock gene expression. These processes are achieved at mitochondrial and nuclear levels to control the host immune cell development. Considering unclear mechanisms and undiscovered actions of melatonin in gut-microbiome-immune axis, it's time to reveal them and provide new insight for the outlook of melatonin as a potential therapeutic target in the treatment and management of intestinal diseases.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jie Zhang
- Animal Husbandry and Veterinary Department, Beijing Vocational College of Agriculture, Beijing, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Department of Internal Medicine and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
47
|
Malaguarnera L. Vitamin D and microbiota: Two sides of the same coin in the immunomodulatory aspects. Int Immunopharmacol 2019; 79:106112. [PMID: 31877495 DOI: 10.1016/j.intimp.2019.106112] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
Abstract
The gut microbiota is crucial for host immune response, vitamin synthesis, short chain fatty acids (SCFAs) production, intestinal permeability, nutrient digestion energy metabolism and protection from pathogens. Therefore, gut microbiota guarantees the host's predisposition to gastrointestinal diseases. Intestinal microbiota may be damaged by environmental components with negative health conditions. Dysbiosis consisting in alteration in the gut microbiota has been involved in several disorders including inflammation, allergic reactions, autoimmune diseases, heart diseases, obesity, and metabolic syndrome and even in the state of malignant carcinogenesis existing in humans. Several epidemiological studies have shown that inadequate solar exposure results in vitamin D insufficiency/deficiency which has a strong impact on different immune responses and the occurrence of a wide range of pathological conditions. Additionally, new evidence indicates that the vitamin D pathway plays a key role in gut homeostasis. Due to the strong connection between vitamin D and microbiota, herein we focus on the new findings about intestinal bacteria-immune crosstalk and the impact of vitamin D in gut microbiota regulation, in order to offer new clarifications on their interaction. Understanding the mechanism by which vitamin D can affect the gut microbiota composition and its dynamic activities, as well as the innate and adaptive state of the immune system, is not only a fundamental research but also an opportunity to improve health status.
Collapse
Affiliation(s)
- Lucia Malaguarnera
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia, 97, Catania, Italy.
| |
Collapse
|
48
|
Marcolla CS, Alvarado CS, Willing BP. Early life microbial exposure shapes subsequent animal health. CANADIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1139/cjas-2019-0029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biosecurity standards and farming practices have profoundly changed the way domestic animals interact with the environment and themselves. Farm intensification processes resemble the lifestyle changes that humans underwent post industrialization, which have been linked to the occurrence of immune-mediated and metabolic disorders. Modern rearing practices reduce maternal and offspring interactions, promote changes in diet, restrict animals indoors, and rely on the use of antibiotics and vaccines to maintain animal health. These practices may hinder the proper colonization of the gastrointestinal tract with commensal organisms that co-evolved with livestock species. The gut microbiota aids nutrient digestion, stimulates immune and intestinal development and maturation, and promotes the competitive exclusion of pathogens. Microbial colonization in early life is critical for host metabolic and immune programming, and disruptions of gut microbial community stability can lead to development of metabolic and immune disorders seen at later stages of life. Identifying how farming practices influence microbial composition and the potential effects on host physiology, metabolism, and disease resistance is necessary to guide intervention strategies to promote beneficial microbial–host interactions, and improve animal health and performance.
Collapse
Affiliation(s)
- Camila Schultz Marcolla
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Carla Sosa Alvarado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Benjamin Peter Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
49
|
Ecological impact of a rare sugar on grapevine phyllosphere microbial communities. Microbiol Res 2019; 232:126387. [PMID: 31790975 DOI: 10.1016/j.micres.2019.126387] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 01/14/2023]
Abstract
Plants host a complex microbiota inside or outside their tissues, and phyllosphere microorganisms can be influenced by environmental, nutritional and agronomic factors. Rare sugars are defined as monosaccharides with limited availability in nature and they are metabolised by only few certain microbial taxa. Among rare sugars, tagatose (TAG) is a low-calories sweetener that stimulates and inhibits beneficial and pathogenic bacteria in the human gut microbiota, respectively. Based on this differential effect on human-associated microorganisms, we investigated the effect of TAG treatments on the grapevine phyllosphere microorganisms to evaluate whether it can engineer the microbiota and modify the ratio between beneficial and pathogenic plant-associated microorganisms. TAG treatments changed the structure of the leaf microbiota and they successfully reduced leaf infections of downy mildew (caused by Plasmopara viticola) and powdery mildew (caused by Erysiphe necator) under field conditions. TAG increased the relative abundance of indigenous beneficial microorganisms, such as some potential biocontrol agents, which could partially contribute to disease control. The taxonomic composition of fungal and bacterial leaf populations differed according to grapevine locations, therefore TAG effects on the microbial structure were influenced by the composition of the originally residing microbiota. TAG is a promising biopesticide that could shift the balance of pathogenic and beneficial plant-associated microorganisms, suggesting selective nutritional/anti-nutritional properties for some specific taxa. More specifically, TAG displayed possible plant prebiotic effects on the phyllosphere microbiota and this mechanism of action could represent a novel strategy that can be further developed for sustainable plant protection.
Collapse
|
50
|
Zhang YG, Zhu X, Lu R, Messer JS, Xia Y, Chang EB, Sun J. Intestinal epithelial HMGB1 inhibits bacterial infection via STAT3 regulation of autophagy. Autophagy 2019; 15:1935-1953. [PMID: 30894054 PMCID: PMC6844505 DOI: 10.1080/15548627.2019.1596485] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 12/30/2022] Open
Abstract
Extracellular HMGB1 (high mobility group box 1) is considered as a damage-associated molecular pattern protein. However, little is known about its intracellular role. We studied the mechanism whereby intestinal epithelial HMGB1 contributes to host defense, using cell culture, colonoids, conditional intestinal epithelial HMGB1-knockout mice with Salmonella-colitis, il10-/- mice, and human samples. We report that intestinal HMGB1 is an important contributor to host protection from inflammation and infection. We identified a physical interaction between HMGB1 and STAT3. Lacking intestinal epithelial HMGB1 led to redistribution of STAT3 and activation of STAT3 post bacterial infection. Indeed, Salmonella-infected HMGB1-deficient cells exhibited less macroautophagy/autophagy due to decreased expression of autophagy proteins and transcriptional repression by activated STAT3. Then, increased p-STAT3 and extranuclear STAT3 reduced autophagic responses and increased inflammation. STAT3 inhibition restored autophagic responses and reduced bacterial invasion in vitro and in vivo. Moreover, low level of HMGB1 was correlated with reduced nuclear STAT3 and enhanced p-STAT3 in inflamed intestine of il10-/- mice and inflammatory bowel disease (IBD) patients. We revealed that colonic epithelial HMGB1 was directly involved in the suppression of STAT3 activation and the protection of intestine from bacterial infection and injury. Abbreviations: ATG16L1: autophagy-related 16-like 1 (S. cerevisiae); DAMP: damage-associated molecular pattern; HBSS: Hanks balanced salt solution; HMGB1: high mobility group box 1; IBD: inflammatory bowel disease; IL1B/Il-1β: interleukin 1 beta; IL10: interleukin 10; IL17/IL-17: interleukin 17; MEFs: mouse embryonic fibroblasts; STAT3: signal transducer and activator of transcription 3; TLR: toll-like receptor; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Yong-Guo Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Xiaorong Zhu
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Rong Lu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jeannette S. Messer
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Yinglin Xia
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Eugene B. Chang
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|