1
|
Saranya S, Prathiviraj R, Chellapandi P. Evolutionary Transitions of DNA Replication Origins Between Archaea and Bacteria. J Basic Microbiol 2024:e2400527. [PMID: 39663550 DOI: 10.1002/jobm.202400527] [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: 08/17/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
DNA replication origins play a crucial role in cellular division and are evolutionarily conserved across domains. This study investigated the evolutionary transitions of replication origins between archaea and bacteria by analyzing 2733 bacterial and 257 archaeal genomes. Our findings revealed that certain methanogens and bacteria share phylogenetic proximity, suggesting evolutionary interactions across diverse ecological systems. Evolutionary transitions in replication origins may have occurred between gut methanogens and bacteria, haloarchaea (Halogeometricum borinquense DSM 11551 and Halovivax ruber XH-70), halobacteria, and sulfur-reducing archaea. Methanosarcina barkeri (M. barkeri), Methanosaeta thermophila, and Methanococcoides burtonii (M. burtonii) were closely related to respiratory tract bacteria in humans. Methanohalobium evestigatum (M. evestigatum) is strongly linked to the animal gut pathogen Mycoplasma putrefaciens (M. putrefaciens). Several thermophilic hydrogenotrophic methanogens clustered with oral and fish pathogens. Pyrococcus furiosus (P. furiosus) was evolutionarily related to the replication origin of plant pathogens. This study sheds light on the ecological drivers of DNA replication origin evolution and their role in microbial speciation and adaptation. Our findings highlight the influence of mutualistic and parasitic relationships on these evolutionary transitions. It could have significant implications in biotechnology and medicine, such as developing novel antimicrobial strategies and understanding host-pathogen dynamics.
Collapse
Affiliation(s)
- S Saranya
- Department of Bioinformatics, Industrial Systems Biology Lab, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - R Prathiviraj
- Department of Bioinformatics, Industrial Systems Biology Lab, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - P Chellapandi
- Department of Bioinformatics, Industrial Systems Biology Lab, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| |
Collapse
|
2
|
Bouaka Tsakeng CU, Melachio Tanekou TT, Ngambia Freitas FS, Tirados I, Tsagmo Ngoune JM, Bigoga JD, Njiokou F, Wondji CS. Patterns of microbiome composition in tsetse fly Glossina palpalis palpalis during vector control using Tiny Targets in Campo, South Cameroon. Microbiol Spectr 2024; 12:e0093524. [PMID: 39297636 PMCID: PMC11540164 DOI: 10.1128/spectrum.00935-24] [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: 04/24/2024] [Accepted: 08/22/2024] [Indexed: 11/08/2024] Open
Abstract
Novel vector control tools against African trypanosomiases require a deep understanding of the factors driving tsetse vector fitness or population resilience in their ecosystems. Following evidence of microbiota-mediated host fitness or traits shaping, including insecticide resistance in arthropod populations, we undertook a comparative study of the microbiota in wild-caught tsetse flies during vector control with deltamethrin-impregnated traps called Tiny Targets. The bacterial microbiome composition of tsetse flies collected before and after 6, 12, and 18 months of vector control were characterized using high-throughput sequencing of the V3-V4 hypervariable region of the bacterial 16S rRNA gene and compared. Overall, 48 bacterial genera and five phyla were identified. The primary symbiont Wigglesworthia dominated almost all the samples with an overall relative abundance of 71.76%. A significant increase was observed in microbiome diversities over the vector control with new taxa identified. Interestingly, few genera, like Curvibacter for instance, displayed a regularly increasing abundance, from 0.57% to 0.65%, 4.73%, and 8.57% after 6, 12, and 18 months of tsetse control, respectively. This study provided preliminary for further investigation into the role and mechanism of action of microbiota in tsetse fly fitness under selective pressure like insecticides.IMPORTANCEThe interest in vector control in the fight against African trypanosomiases has been reinforced in recent years, with the development of small insecticide-impregnated screens, known as "Tiny Targets". As some tsetse biotopes are difficult to access for their installation, other tools are under consideration that involve using bacteria harbored by the tsetse vector to block the development of trypanosomes or impair the tsetse's fitness in its natural environment. Several bacterial symbionts were previously described as important for tsetse fly development, and some like Burkholderia and Citrobacter also found in tsetse flies were found associated with insecticide tolerance in other arthropods. In this research, we found the bacterial genera, Curvibacter and Acinetobacter, increased in abundance in tsetse flies during vector control. These bacteria deserve further attention to determine if they can interfere with insecticides used to control tsetse fly populations.
Collapse
Affiliation(s)
- Calmes Ursain Bouaka Tsakeng
- Centre for Research in
Infectious Diseases (CRID),
Yaoundé, Cameroon
- Department of
Biochemistry, Faculty of Science, University of Yaoundé
I, Yaoundé,
Cameroon
| | - Tito Tresor Melachio Tanekou
- Centre for Research in
Infectious Diseases (CRID),
Yaoundé, Cameroon
- Department of
Microbiology and Parasitology, Faculty of Science, University of
Bamenda, Bamenda,
Cameroon
| | | | - Inaki Tirados
- Department of Vector
Biology, Liverpool School of Tropical Medicine (LSTM), Pembroke
Place, Liverpool,
United Kingdom
| | - Jean Marc Tsagmo Ngoune
- Department of
Parasites and Insect Vectors, Trypanosome Transmission Group,
Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur,
Université Paris Cité,
Paris, France
| | - Jude Daiga Bigoga
- Department of
Biochemistry, Faculty of Science, University of Yaoundé
I, Yaoundé,
Cameroon
| | - Flobert Njiokou
- Department of Animal
Biology and Physiology, Faculty of Science, University of Yaoundé
I, Yaoundé,
Cameroon
| | - Charles Sinclair Wondji
- Centre for Research in
Infectious Diseases (CRID),
Yaoundé, Cameroon
- Department of Vector
Biology, Liverpool School of Tropical Medicine (LSTM), Pembroke
Place, Liverpool,
United Kingdom
| |
Collapse
|
3
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
4
|
Li C, Zhu C, Tu G, Chen Z, Mo Z, Luo C. Impact of Altered Gut Microbiota on Ketamine-Induced Conditioned Place Preference in Mice. Neuropsychiatr Dis Treat 2024; 20:1725-1740. [PMID: 39318552 PMCID: PMC11421448 DOI: 10.2147/ndt.s476420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Objects Ketamine is a drug of abuse worldwide and current treatments for ketamine abuse are inadequate. It is an urgent need to develop novel anti-addictive strategy. Since gut microbiota plays a crucial role in drug abuse, the present study investigates the impact and mechanisms of the gut microbiota in addictive behaviors induced by ketamine addiction. Methods Conditioned place preference (CPP) was employed to assess addiction, followed by 16S rRNA gene sequencing to elucidate alterations in the gut microbiota. Furthermore, qRT-PCR, ELISA, and immunohistochemistry were conducted to evaluate the expression levels of crucial genes and proteins associated with the gut-brain axis. Additionally, we investigated whether ketamine addiction is regulated through the gut microbiota by orally administering antibiotics to establish pseudo-germ-free mice. Results We found that repeated ketamine administration (20 mg/kg) induced CPP and significantly altered gut microbiota diversity and composition, as revealed by 16S rRNA gene sequencing. Compared to the control group, ketamine exposure exhibited differences in the relative abundance of 5 microbial families, with 4 (Lachnospiraceae, Ruminococcaceae, Desulfovibrionaceae and Family-XIII) showing increases, while one (Prevotellaceae) displayed a decrease. At the genus level, five genera were upregulated, while one was downregulated. Furthermore, COG analysis revealed significant differences in protein functionality between the two groups. Additionally, axis series studies showed that ketamine dependence reduced levels of tight junction proteins, GABA and GABRA1, while increasing BDNF and 5-HT. Moreover, an oral antibiotic cocktail simulating pseudo germ-free conditions in mice did not enhance the addictive behavior induced by ketamine. Conclusion Our study supports the hypothesis that ketamine-induced CPP is mediated through the gut microbiota. The present study provides new insights into improvement of efficient strategy for addiction treatment.
Collapse
Affiliation(s)
- Chan Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
- School of Life Sciences, Guangzhou University, Guangzhou, People's Republic of China
| | - Chen Zhu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Genghong Tu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, People's Republic of China
| | - Zhijie Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhixian Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, People's Republic of China
| | - Chaohua Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
5
|
Babajanyan SG, Garushyants SK, Wolf YI, Koonin EV. Microbial diversity and ecological complexity emerging from environmental variation and horizontal gene transfer in a simple mathematical model. BMC Biol 2024; 22:148. [PMID: 38965531 PMCID: PMC11225191 DOI: 10.1186/s12915-024-01937-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/13/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Microbiomes are generally characterized by high diversity of coexisting microbial species and strains, and microbiome composition typically remains stable across a broad range of conditions. However, under fixed conditions, microbial ecology conforms with the exclusion principle under which two populations competing for the same resource within the same niche cannot coexist because the less fit population inevitably goes extinct. Therefore, the long-term persistence of microbiome diversity calls for an explanation. RESULTS To explore the conditions for stabilization of microbial diversity, we developed a simple mathematical model consisting of two competing populations that could exchange a single gene allele via horizontal gene transfer (HGT). We found that, although in a fixed environment, with unbiased HGT, the system obeyed the exclusion principle, in an oscillating environment, within large regions of the phase space bounded by the rates of reproduction and HGT, the two populations coexist. Moreover, depending on the parameter combination, all three major types of symbiosis were obtained, namely, pure competition, host-parasite relationship, and mutualism. In each of these regimes, certain parameter combinations provided for synergy, that is, a greater total abundance of both populations compared to the abundance of the winning population in the fixed environment. CONCLUSIONS The results of this modeling study show that basic phenomena that are universal in microbial communities, namely, environmental variation and HGT, provide for stabilization and persistence of microbial diversity, and emergence of ecological complexity.
Collapse
Affiliation(s)
- Sanasar G Babajanyan
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, 20894, MD, USA.
| | - Sofya K Garushyants
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, 20894, MD, USA
| | - Yuri I Wolf
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, 20894, MD, USA
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, 20894, MD, USA.
| |
Collapse
|
6
|
Jiang Y, Jiang Y, Li L, Liu X, Hou X, Wang W. High-Molecular-Weight Hyaluronic Acid Can Be Used as a Food Additive to Improve the Symptoms of Persistent Inflammation, Immunosuppression and Catabolism Syndrome (PICS). BIOLOGY 2024; 13:319. [PMID: 38785801 PMCID: PMC11118101 DOI: 10.3390/biology13050319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Hyaluronic acid (HA) is a new functional food additive which has the potential to ameliorate persistent inflammation, immunosuppression and catabolism syndrome (PICS), but the biological effects of HA with various molecular weights differ dramatically. To systematically investigate the efficacy of HA in altering PICS symptoms, medium-molecular-weight (MMW) HA was specifically selected to test its intervention effect on a PICS mouse model induced by CLP through oral administration, with high-molecular-weight (HMW) and low-molecular-weight (LMW) HA also participating in the experimental validation process. The results of pathological observations and gut flora showed that MMW HA rapidly alleviated lung lesions and intestinal structural changes in PICS mice in the short term. However, although long-term MMW HA administration significantly reduced the proportions of harmful bacteria in gut flora, inflammatory responses in the intestines and lungs of PICS mice were significantly higher in the MMW HA group than in the HMW HA and LMW HA groups. The use of HMW HA not only rapidly reduced the mortality rate of PICS mice but also improved their grip strength and the recovery of spleen and thymus indices. Furthermore, it consistently promoted the recovery of lung and intestinal tissues in PICS mice, and it also assisted in the sustained restoration of their gut microbiota. These effects were superior to those of LMW HA and MMW HA. The experimental results indicate that HMW weight HA has the greatest potential to be an adjunct in alleviating PICS as a food additive, while the safety of other HAs requires further attention.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoming Hou
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (Y.J.); (L.L.); (X.L.)
| | - Wenfei Wang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.J.); (Y.J.); (L.L.); (X.L.)
| |
Collapse
|
7
|
Zhang Y, Gacesa R, Fu J. Implications of blood type in personalised microbiome therapy. Clin Transl Med 2024; 14:e1618. [PMID: 38468485 PMCID: PMC10928326 DOI: 10.1002/ctm2.1618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 03/13/2024] Open
Affiliation(s)
- Yue Zhang
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Ranko Gacesa
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
- Department of Gastroenterology and HepatologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Jingyuan Fu
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
- Department of PediatricsUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
8
|
Babajanyan SG, Garushyants SK, Wolf YI, Koonin EV. Microbial diversity and ecological complexity emerging from environmental variation and horizontal gene transfer in a simple mathematical model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576128. [PMID: 38313259 PMCID: PMC10836074 DOI: 10.1101/2024.01.17.576128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Microbiomes are generally characterized by high diversity of coexisting microbial species and strains that remains stable within a broad range of conditions. However, under fixed conditions, microbial ecology conforms with the exclusion principle under which two populations competing for the same resource within the same niche cannot coexist because the less fit population inevitably goes extinct. To explore the conditions for stabilization of microbial diversity, we developed a simple mathematical model consisting of two competing populations that could exchange a single gene allele via horizontal gene transfer (HGT). We found that, although in a fixed environment, with unbiased HGT, the system obeyed the exclusion principle, in an oscillating environment, within large regions of the phase space bounded by the rates of reproduction and HGT, the two populations coexist. Moreover, depending on the parameter combination, all three major types of symbiosis obtained, namely, pure competition, host-parasite relationship and mutualism. In each of these regimes, certain parameter combinations provided for synergy, that is, a greater total abundance of both populations compared to the abundance of the winning population in the fixed environments. These findings show that basic phenomena that are universal in microbial communities, environmental variation and HGT, provide for stabilization of microbial diversity and ecological complexity.
Collapse
Affiliation(s)
- Sanasar G. Babajanyan
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Sofya K. Garushyants
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Yuri I. Wolf
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| |
Collapse
|
9
|
Wang S, Yin F, Guo Z, Li R, Sun W, Wang Y, Geng Y, Sun C, Sun D. Association between gut microbiota and glioblastoma: a Mendelian randomization study. Front Genet 2024; 14:1308263. [PMID: 38239850 PMCID: PMC10794655 DOI: 10.3389/fgene.2023.1308263] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/01/2023] [Indexed: 01/22/2024] Open
Abstract
Background: Glioblastoma (GBM) is the most prevalent malignant brain tumor, significantly impacting the physical and mental wellbeing of patients. Several studies have demonstrated a close association between gut microbiota and the development of GBM. In this investigation, Mendelian randomization (MR) was employed to rigorously evaluate the potential causal relationship between gut microbiota and GBM. Methods: We utilized summary statistics derived from genome-wide association studies (GWAS) encompassing 211 gut microbiota and GBM. The causal association between gut microbiota and GBM was scrutinized using Inverse Variance Weighted (IVW), MR-Egger, and Weighted Median (WM) methods. Cochrane's Q statistic was employed to conduct a heterogeneity test. MR-Pleiotropic Residuals and Outliers (MR-PRESSO) were applied to identify and eliminate SNPs with horizontal pleiotropic outliers. Additionally, Reverse MR was employed to assess the causal relationship between GBM and pertinent gut microbiota. Results: The MR study estimates suggest that the nine gut microbiota remain stable, considering heterogeneity and sensitivity methods. Among these, the family.Peptostreptococcaceae and genus.Eubacterium brachy group were associated with an increased risk of GBM, whereas family.Ruminococcaceae, genus.Anaerostipes, genus.Faecalibacterium, genus.LachnospiraceaeUCG004, genus.Phascolarctobacterium, genus.Prevotella7, and genus.Streptococcus were associated with a reduced risk of GBM. Following Benjamini and Hochberg (BH) correction, family.Ruminococcaceae (OR = 0.04, 95% CI: 0.01-0.19, FDR = 0.003) was identified as playing a protective role against GBM. Conclusion: This groundbreaking study is the first to demonstrate that family.Ruminococcaceae is significantly associated with a reduced risk of GBM. The modulation of family_Ruminococcaceae for the treatment of GBM holds considerable potential clinical significance.
Collapse
Affiliation(s)
- Song Wang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Fangxu Yin
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zheng Guo
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Rui Li
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuchao Wang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yichen Geng
- Nursing College of Binzhou Medical University, Yantai, Shandong, China
| | - Chao Sun
- Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Zhernakova DV, Wang D, Liu L, Andreu-Sánchez S, Zhang Y, Ruiz-Moreno AJ, Peng H, Plomp N, Del Castillo-Izquierdo Á, Gacesa R, Lopera-Maya EA, Temba GS, Kullaya VI, van Leeuwen SS, Xavier RJ, de Mast Q, Joosten LAB, Riksen NP, Rutten JHW, Netea MG, Sanna S, Wijmenga C, Weersma RK, Zhernakova A, Harmsen HJM, Fu J. Host genetic regulation of human gut microbial structural variation. Nature 2024; 625:813-821. [PMID: 38172637 PMCID: PMC10808065 DOI: 10.1038/s41586-023-06893-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 11/23/2023] [Indexed: 01/05/2024]
Abstract
Although the impact of host genetics on gut microbial diversity and the abundance of specific taxa is well established1-6, little is known about how host genetics regulates the genetic diversity of gut microorganisms. Here we conducted a meta-analysis of associations between human genetic variation and gut microbial structural variation in 9,015 individuals from four Dutch cohorts. Strikingly, the presence rate of a structural variation segment in Faecalibacterium prausnitzii that harbours an N-acetylgalactosamine (GalNAc) utilization gene cluster is higher in individuals who secrete the type A oligosaccharide antigen terminating in GalNAc, a feature that is jointly determined by human ABO and FUT2 genotypes, and we could replicate this association in a Tanzanian cohort. In vitro experiments demonstrated that GalNAc can be used as the sole carbohydrate source for F. prausnitzii strains that carry the GalNAc-metabolizing pathway. Further in silico and in vitro studies demonstrated that other ABO-associated species can also utilize GalNAc, particularly Collinsella aerofaciens. The GalNAc utilization genes are also associated with the host's cardiometabolic health, particularly in individuals with mucosal A-antigen. Together, the findings of our study demonstrate that genetic associations across the human genome and bacterial metagenome can provide functional insights into the reciprocal host-microbiome relationship.
Collapse
Affiliation(s)
- Daria V Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Daoming Wang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Lei Liu
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Yue Zhang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Angel J Ruiz-Moreno
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Haoran Peng
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Niels Plomp
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Ángela Del Castillo-Izquierdo
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Ranko Gacesa
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Esteban A Lopera-Maya
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Godfrey S Temba
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vesla I Kullaya
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Sander S van Leeuwen
- University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen, The Netherlands
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Serena Sanna
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- Institute for Genetic and Biomedical Research, National Research Council, Cagliari, Italy
| | - Cisca Wijmenga
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Rinse K Weersma
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Hermie J M Harmsen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands.
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands.
| |
Collapse
|
11
|
Ma C, Zhang Y, Jiang S, Teng F, Huang S, Zhang J. Cross-cohort single-nucleotide-variant profiling of gut microbiota suggests a novel gut-health assessment approach. mSystems 2023; 8:e0082823. [PMID: 37905808 PMCID: PMC10734426 DOI: 10.1128/msystems.00828-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/21/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE Most studies focused much on the change in abundance and often failed to explain the microbiome variation related to disease conditions, Herein, we argue that microbial genetic changes can precede the ecological changes associated with the host physiological changes and, thus, would offer a new information layer from metagenomic data for predictive modeling of diseases. Interestingly, we preliminarily found a few genetic biomarkers on SCFA production can cover most chronic diseases involved in the meta-analysis. In the future, it is of both scientific and clinical significance to further explore the dynamic interactions between adaptive evolution and ecology of gut microbiota associated with host health status.
Collapse
Affiliation(s)
- Chenchen Ma
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yufeng Zhang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Shuaiming Jiang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Fei Teng
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Jiachao Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, Hainan, China
| |
Collapse
|
12
|
Zhang Y, Wang X, Li W, Yang Y, Wu Z, Lyu Y, Yue C. Intestinal microbiota: a new perspective on delaying aging? Front Microbiol 2023; 14:1268142. [PMID: 38098677 PMCID: PMC10720643 DOI: 10.3389/fmicb.2023.1268142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
The global aging situation is severe, and the medical pressures associated with aging issues should not be underestimated. The need and feasibility of studying aging and intervening in aging have been confirmed. Aging is a complex natural physiological progression, which involves the irreversible deterioration of body cells, tissues, and organs with age, leading to enhanced risk of disease and ultimately death. The intestinal microbiota has a significant role in sustaining host dynamic balance, and the study of bidirectional communication networks such as the brain-gut axis provides important directions for human disease research. Moreover, the intestinal microbiota is intimately linked to aging. This review describes the intestinal microbiota changes in human aging and analyzes the causal controversy between gut microbiota changes and aging, which are believed to be mutually causal, mutually reinforcing, and inextricably linked. Finally, from an anti-aging perspective, this study summarizes how to achieve delayed aging by targeting the intestinal microbiota. Accordingly, the study aims to provide guidance for further research on the intestinal microbiota and aging.
Collapse
Affiliation(s)
- Yuemeng Zhang
- Yan’an Key Laboratory of Microbial Drug Innovation and Transformation, School of Basic Medicine, Yan’an University, Yan’an, Shaanxi, China
| | - Xiaomei Wang
- Yan’an University of Physical Education, Yan’an University, Yan’an, Shaanxi, China
| | - Wujuan Li
- Yan’an Key Laboratory of Microbial Drug Innovation and Transformation, School of Basic Medicine, Yan’an University, Yan’an, Shaanxi, China
| | - Yi Yang
- Yan’an Key Laboratory of Microbial Drug Innovation and Transformation, School of Basic Medicine, Yan’an University, Yan’an, Shaanxi, China
| | - Zhuoxuan Wu
- Yan’an Key Laboratory of Microbial Drug Innovation and Transformation, School of Basic Medicine, Yan’an University, Yan’an, Shaanxi, China
| | - Yuhong Lyu
- Yan’an Key Laboratory of Microbial Drug Innovation and Transformation, School of Basic Medicine, Yan’an University, Yan’an, Shaanxi, China
| | - Changwu Yue
- Yan’an Key Laboratory of Microbial Drug Innovation and Transformation, School of Basic Medicine, Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
13
|
Good BH, Rosenfeld LB. Eco-evolutionary feedbacks in the human gut microbiome. Nat Commun 2023; 14:7146. [PMID: 37932275 PMCID: PMC10628149 DOI: 10.1038/s41467-023-42769-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
Gut microbiota can evolve within their hosts on human-relevant timescales, but little is known about how these changes influence (or are influenced by) the composition of their local community. Here, by combining ecological and evolutionary analyses of a large cohort of human gut metagenomes, we show that the short-term evolution of the microbiota is linked with shifts in its ecological structure. These correlations are not simply explained by expansions of the evolving species, and often involve additional fluctuations in distantly related taxa. We show that similar feedbacks naturally emerge in simple resource competition models, even in the absence of cross-feeding or predation. These results suggest that the structure and function of host microbiota may be shaped by their local evolutionary history, which could have important implications for personalized medicine and microbiome engineering.
Collapse
Affiliation(s)
- Benjamin H Good
- Department of Applied Physics, Stanford University, Stanford, CA, 94305, USA.
- Department of Biology, Stanford University, Stanford, CA, 94305, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, 94158, USA.
| | - Layton B Rosenfeld
- Department of Computer Science, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
14
|
Xu Z, Xu H, Chen X, Huang X, Tian J, Zhao J, Liu B, Shi F, Wu J, Pu J. CCDC103 as a Prognostic Biomarker Correlated with Tumor Progression and Immune Infiltration in Glioma. Onco Targets Ther 2023; 16:819-837. [PMID: 37873495 PMCID: PMC10590567 DOI: 10.2147/ott.s429958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/03/2023] [Indexed: 10/25/2023] Open
Abstract
Background The Coiled-coil domain-containing proteins (CCDCs) are expressed in many cancers, but the role of Coiled-coil domain-containing protein 103 (CCDC103) in cancers remains unclear. Further investigations are necessary to ascertain its diagnostic significance and understand its biological function in cancers. This study aims to elucidate the biological functionalities of CCDC103 in glioma and evaluate the correlation between CCDC103 expression with glioma progression. Methods Clinical data on glioma patients were acquired from The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), and the Gene Expression Omnibus (GEO). The evaluation encompassed the examination of correlations between CCDC103 expression, pathological characteristics, and clinical outcomes. Furthermore, the analysis included the assessment of the correlations between CCDC103 expression and immune cell infiltration as well as glioma progression. Results Gliomas have higher levels of CCDC103 expression than the para-carcinoma tissues. Poorer prognosis, unfavorable histological characteristics, the absence of IDH gene mutations, and the absence of chromosome 1p and 19q deletions were all associated with higher expression of CCDC103 in gliomas. In addition to patient age, tumor grade, the absence of IDH mutations, and the absence of chromosome 1p and 19q deletions, univariate and multivariate Cox analyses showed that CCDC103 expression was independently prognostic of overall survival, disease-free survival, and progression-free survival in patients with glioma. Furthermore, tumor infiltration of B cells, neutrophils, macrophages, and dendritic cells were all linked with elevated expression of CCDC103. High CCDC103 expression was linked to immune response-related signaling pathways and cell proliferation, according to gene set enrichment analysis (GSEA). Notably, the knockdown of CCDC103 in glioma cell lines resulted in a significant reduction in cell proliferation and migration. Conclusion The correlation between CCDC103 expression and both glioma progression and immune cell infiltration implies that CCDC103 expression holds promise as a valuable prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Zhixing Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
- Department of Neurosurgery, The Pu’er People’s Hospital, Pu’er, 665000, People’s Republic of China
| | - Haitao Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| | - Xi Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| | - Xiaobing Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| | - Jintao Tian
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| | - Jinxi Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| | - Bohu Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| | - Fengcai Shi
- Department of Neurosurgery, The Pu’er People’s Hospital, Pu’er, 665000, People’s Republic of China
| | - Jin Wu
- Department of Neurosurgery, The Pu’er People’s Hospital, Pu’er, 665000, People’s Republic of China
| | - Jun Pu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, People’s Republic of China
| |
Collapse
|
15
|
Majumdar A, Siva Venkatesh IP, Basu A. Short-Chain Fatty Acids in the Microbiota-Gut-Brain Axis: Role in Neurodegenerative Disorders and Viral Infections. ACS Chem Neurosci 2023; 14:1045-1062. [PMID: 36868874 DOI: 10.1021/acschemneuro.2c00803] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
The gut-brain axis (GBA) is the umbrella term to include all bidirectional communication between the brain and gastrointestinal (GI) tract in the mammalian body. Evidence from over two centuries describes a significant role of GI microbiome in health and disease states of the host organism. Short-chain fatty acids (SCFAs), mainly acetate, butyrate, and propionate that are the physiological forms of acetic acid, butyric acid, and propionic acid respectively, are GI bacteria derived metabolites. SCFAs have been reported to influence cellular function in multiple neurodegenerative diseases (NDDs). In addition, the inflammation modulating properties of SCFAs make them suitable therapeutic candidates in neuroinflammatory conditions. This review provides a historical background of the GBA and current knowledge of the GI microbiome and role of individual SCFAs in central nervous system (CNS) disorders. Recently, a few reports have also identified the effects of GI metabolites in the case of viral infections. Among these viruses, the flaviviridae family is associated with neuroinflammation and deterioration of CNS functions. In this context, we additionally introduce SCFA based mechanisms in different viral pathogenesis to understand the former's potential as agents against flaviviral disease.
Collapse
Affiliation(s)
- Atreye Majumdar
- National Brain Research Centre, Manesar, Haryana 122052, India
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana 122052, India
| |
Collapse
|
16
|
Eroglu A, Al'Abri IS, Kopec RE, Crook N, Bohn T. Carotenoids and Their Health Benefits as Derived via Their Interactions with Gut Microbiota. Adv Nutr 2023; 14:238-255. [PMID: 36775788 DOI: 10.1016/j.advnut.2022.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/21/2022] [Accepted: 10/28/2022] [Indexed: 12/23/2022] Open
Abstract
Carotenoids have been related to a number of health benefits. Their dietary intake and circulating levels have been associated with a reduced incidence of obesity, diabetes, certain types of cancer, and even lower total mortality. Their potential interaction with the gut microbiota (GM) has been generally overlooked but may be of relevance, as carotenoids largely bypass absorption in the small intestine and are passed on to the colon, where they appear to be in part degraded into unknown metabolites. These may include apo-carotenoids that may have biological effects because of higher aqueous solubility and higher electrophilicity that could better target transcription factors, i.e., NF-κB, PPARγ, and RAR/RXRs. If absorbed in the colon, they could have both local and systemic effects. Certain microbes that may be supplemented were also reported to produce carotenoids in the colon. Although some bactericidal aspects of carotenoids have been shown in vitro, a few studies have also demonstrated a prebiotic-like effect, resulting in bacterial shifts with health-associated properties. Also, stimulation of IgA could play a role in this respect. Carotenoids may further contribute to mucosal and gut barrier health, such as stabilizing tight junctions. This review highlights potential gut-related health-beneficial effects of carotenoids and emphasizes the current research gaps regarding carotenoid-GM interactions.
Collapse
Affiliation(s)
- Abdulkerim Eroglu
- Department of Molecular and Structural Biochemistry, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA; Plants for Human Health Institute, North Carolina Research Campus, North Carolina State University, Kannapolis, NC, USA.
| | - Ibrahim S Al'Abri
- Department of Chemical and Biomolecular Engineering, College of Engineering, North Carolina State University, Raleigh, NC, USA
| | - Rachel E Kopec
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, USA; Foods for Health Discovery Theme, The Ohio State University, Columbus, OH, USA
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, College of Engineering, North Carolina State University, Raleigh, NC, USA
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, rue 1 A-B, Thomas Edison, L-1445 Strassen, Luxembourg.
| |
Collapse
|
17
|
Zhang XX, Lv QB, Yan QL, Zhang Y, Guo RC, Meng JX, Ma H, Qin SY, Zhu QH, Li CQ, Liu R, Liu G, Li SH, Sun DB, Ni HB. A Catalog of over 5,000 Metagenome-Assembled Microbial Genomes from the Caprinae Gut Microbiota. Microbiol Spectr 2022; 10:e0221122. [PMID: 36321901 PMCID: PMC9769736 DOI: 10.1128/spectrum.02211-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Most microbiome studies regarding the ruminant digestive tract have focused on the rumen microbiota, whereas only a few studies were performed on investigating the gut microbiota of ruminants, which limits our understanding of this important component. Herein, the gut microbiota of 30 Caprinae animals (sheep and goats) from six provinces in China was characterized using ultradeep (>100 Gbp per sample) metagenome shotgun sequencing. An inventory of Caprinae gut microbial species containing 5,046 metagenomic assembly genomes (MAGs) was constructed. Particularly, 2,530 of the genomes belonged to uncultured candidate species. These genomes largely expanded the genomic repository of the current microbes in the Caprinae gut. Several enzymes and biosynthetic gene clusters encoded by these Caprinae gut species were identified. In summary, our study extends the gut microbiota characteristics of Caprinae and provides a basis for future studies on animal production and animal health. IMPORTANCE We constructed a microbiota catalog containing 5,046 MAGs from Caprinae gut from six regions of China. Most of the MAGs do not overlap known databases and appear to be potentially new species. We also characterized the functional spectrum of these MAGs and analyzed the differences between different regions. Our study enriches the understanding of taxonomic, functional, and metabolic diversity of Caprinae gut microbiota. We are confident that the manuscript will be of utmost interest to a wide range of readers and be widely applied in future research.
Collapse
Affiliation(s)
- Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
- Heilongjiang Provincial Key Laboratory of the Prevention and Control of Bovine Diseases, College of Animal Science, Heilongjiang Bayi Agriculture University, Daqing, Heilongjiang Province, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People's Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Qing-Bo Lv
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
- Heilongjiang Provincial Key Laboratory of the Prevention and Control of Bovine Diseases, College of Animal Science, Heilongjiang Bayi Agriculture University, Daqing, Heilongjiang Province, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People's Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Qiu-Long Yan
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning Province, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, Hubei Province, China
| | - Ruo-Chun Guo
- Puensum Genetech Institute, Wuhan, Hubei Province, China
| | - Jin-Xin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
| | - Si-Yuan Qin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
- Center for Biological Disaster Prevention and Control, National Forestry and Grassland Administration, Shenyang, Liaoning Province, China
| | - Qing-He Zhu
- Heilongjiang Provincial Key Laboratory of the Prevention and Control of Bovine Diseases, College of Animal Science, Heilongjiang Bayi Agriculture University, Daqing, Heilongjiang Province, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People's Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Chun-Qiu Li
- Heilongjiang Provincial Key Laboratory of the Prevention and Control of Bovine Diseases, College of Animal Science, Heilongjiang Bayi Agriculture University, Daqing, Heilongjiang Province, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People's Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
| | - Gang Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
| | - Sheng-Hui Li
- Puensum Genetech Institute, Wuhan, Hubei Province, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Dong-Bo Sun
- Heilongjiang Provincial Key Laboratory of the Prevention and Control of Bovine Diseases, College of Animal Science, Heilongjiang Bayi Agriculture University, Daqing, Heilongjiang Province, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People's Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hong-Bo Ni
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People's Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
18
|
Li P, Ma X, Liu D, Wei Y, Li P, Hou H, Yao J, Chen A, Liang Y, Zhou Z, Wang P. A microbiome abundant environment remodels the intestinal microbiota and improves resistance to obesity induced by chlorpyrifos in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120415. [PMID: 36257564 DOI: 10.1016/j.envpol.2022.120415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/23/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
There is a growing consensus that the appropriate microbiome abundant environment actuates microbiota changes to influence human health. Whether living environment reacts on the threat of contaminants and the underlying mechanism remain largely unknown. Therefore, we constructed microbiome abundant environment models, focusing on their regulatory effects on the obesity induced by the exogenous chemical chlorpyrifos (CPF) and the related mechanisms. The results uncovered that the constructed farm and woodland microbiome abundant environment could protect mice against CPF-induced obesity effectively. The microbiome abundant environment regulated CPF-induced microbiota imbalance, characterized by an increase in Lactobacillus abundance. These altered microbiotas modified the intestinal immune system by increasing the expression of Foxp3 and IL-10, and mitigated intestinal barrier injury by upregulating the expression of IL-22 and intestinal tight junction proteins. Fecal microbiota transplantation could receive similar phenotypes on alleviating CPF-induced obesity development. Our results demonstrate that the microbiome abundant environment attenuates exogenous chemical-induced health risks by remodeling the intestinal microbiota, improving the intestinal ecosystem, and preventing intestinal epithelial leakage.
Collapse
Affiliation(s)
- Peize Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Xiaoran Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Yimu Wei
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Pengxi Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Haonan Hou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Jianing Yao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Aisong Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Yiran Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, China.
| |
Collapse
|
19
|
Wang Y, Zhang Z, Li B, He B, Li L, Nice EC, Zhang W, Xu J. New Insights into the Gut Microbiota in Neurodegenerative Diseases from the Perspective of Redox Homeostasis. Antioxidants (Basel) 2022; 11:2287. [PMID: 36421473 PMCID: PMC9687622 DOI: 10.3390/antiox11112287] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 08/27/2023] Open
Abstract
An imbalance between oxidants and antioxidants in the body can lead to oxidative stress, which is one of the major causes of neurodegenerative diseases. The gut microbiota contains trillions of beneficial bacteria that play an important role in maintaining redox homeostasis. In the last decade, the microbiota-gut-brain axis has emerged as a new field that has revolutionized the study of the pathology, diagnosis, and treatment of neurodegenerative diseases. Indeed, a growing number of studies have found that communication between the brain and the gut microbiota can be accomplished through the endocrine, immune, and nervous systems. Importantly, dysregulation of the gut microbiota has been strongly associated with the development of oxidative stress-mediated neurodegenerative diseases. Therefore, a deeper understanding of the relationship between the gut microbiota and redox homeostasis will help explain the pathogenesis of neurodegenerative diseases from a new perspective and provide a theoretical basis for proposing new therapeutic strategies for neurodegenerative diseases. In this review, we will describe the role of oxidative stress and the gut microbiota in neurodegenerative diseases and the underlying mechanisms by which the gut microbiota affects redox homeostasis in the brain, leading to neurodegenerative diseases. In addition, we will discuss the potential applications of maintaining redox homeostasis by modulating the gut microbiota to treat neurodegenerative diseases, which could open the door for new therapeutic approaches to combat neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Wang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhe Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Li
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Jia Xu
- School of Medicine, Ningbo University, Ningbo 315211, China
| |
Collapse
|
20
|
Zhang X, Zhao L, Zhang H, Zhang Y, Ju H, Wang X, Ren H, Zhu X, Dong Y. The immunosuppressive microenvironment and immunotherapy in human glioblastoma. Front Immunol 2022; 13:1003651. [PMID: 36466873 PMCID: PMC9712217 DOI: 10.3389/fimmu.2022.1003651] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 08/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant intracranial tumor in adults, characterized by extensive infiltrative growth, high vascularization, and resistance to multiple therapeutic approaches. Among the many factors affecting the therapeutic effect, the immunosuppressive GBM microenvironment that is created by cells and associated molecules via complex mechanisms plays a particularly important role in facilitating evasion of the tumor from the immune response. Accumulating evidence is also revealing a close association of the gut microbiota with the challenges in the treatment of GBM. The gut microbiota establishes a connection with the central nervous system through bidirectional signals of the gut-brain axis, thus affecting the occurrence and development of GBM. In this review, we discuss the key immunosuppressive components in the tumor microenvironment, along with the regulatory mechanism of the gut microbiota involved in immunity and metabolism in the GBM microenvironment. Lastly, we concentrate on the immunotherapeutic strategies currently under investigation, which hold promise to overcome the hurdles of the immunosuppressive tumor microenvironment and improve the therapeutic outcome for patients with GBM.
Collapse
Affiliation(s)
- Xuehua Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Leilei Zhao
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - He Zhang
- Department of Immunology, Qiqihar Medical University, Qiqihar, China
| | - Yurui Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Huanyu Ju
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Xiaoyu Wang
- Department of Neurology, Hongda Hospital, Jinxiang, China
| | - Huan Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, China
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, China
| |
Collapse
|
21
|
Lieberman TD. Detecting bacterial adaptation within individual microbiomes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210243. [PMID: 35989602 PMCID: PMC9393564 DOI: 10.1098/rstb.2021.0243] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/17/2022] [Indexed: 12/11/2022] Open
Abstract
The human microbiome harbours a large capacity for within-person adaptive mutations. Commensal bacterial strains can stably colonize a person for decades, and billions of mutations are generated daily within each person's microbiome. Adaptive mutations emerging during health might be driven by selective forces that vary across individuals, vary within an individual, or are completely novel to the human population. Mutations emerging within individual microbiomes might impact the immune system, the metabolism of nutrients or drugs, and the stability of the community to perturbations. Despite this potential, relatively little attention has been paid to the possibility of adaptive evolution within complex human-associated microbiomes. This review discusses the promise of studying within-microbiome adaptation, the conceptual and technical limitations that may have contributed to an underappreciation of adaptive de novo mutations occurring within microbiomes to date, and methods for detecting recent adaptive evolution. This article is part of a discussion meeting issue 'Genomic population structures of microbial pathogens'.
Collapse
Affiliation(s)
- Tami D. Lieberman
- Department of Civil and Environmental Engineering, Institute for Medical Engineering and Science,Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- Ragon Institute, Cambridge, MA, USA
| |
Collapse
|
22
|
Chai S, Zhou L, Chi Y, Chen L, Pei S, Chen B. Enhanced antibacterial activity with increasing P doping ratio in CQDs. RSC Adv 2022; 12:27709-27715. [PMID: 36320288 PMCID: PMC9516558 DOI: 10.1039/d2ra04809d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022] Open
Abstract
It is an urgent challenge to develop efficient antibacterial agents against resistant bacteria in the treatment of infectious diseases. Carbon quantum dots (CQDs) have attracted much attention owing to their good stability, low toxicity and excellent biocompatibility. In this work, CQDs doped with different contents of the element phosphorus (P) were prepared by a simple hydrothermal method using valine as a carbon source, triethylamine as a nitrogen source and different volumes of phosphoric acid as a phosphorus source. The average diameter and the surface charge could be regulated from 2.89 nm to 1.56 nm and +2.58 mV to +5.47 mV by increasing the content of the element P in these CQDs. Importantly, these CQDs showed effective bacterial inhibition against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). The minimal inhibitory concentration (MIC) decreased from 0.71, to 0.51 to 0.18 mg mL-1 on E. coli and S. aureus with the increase of P element content. Furthermore, the morphologies of E. coli cells and S. aureus were damaged and became irregular upon treatment with these CQDs. The results of singlet oxygen (1O2) detection demonstrated that intracellular 1O2 was generated during the antibacterial process. We speculated that bacterial inhibition induced by these CQDs was accompanied by disruption of permeability and structural integrity, owing to strong electrostatic interactions between negatively charged bacteria and positively charged CQDs and production of singlet oxygen of CQDs. Together, this study indicates that the CQDs can be a candidate to treat resistant bacterial infections and may improve the understanding of killing pathogens by antibacterial CQD drugs.
Collapse
Affiliation(s)
- Shuiqin Chai
- Chongqing Key Laboratory of Industrial Fermentation Microorganism, Chongqing University of Science and Technology Chongqing 401331 P. R. China
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology Chongqing 401331 P. R. China
| | - Lijia Zhou
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology Chongqing 401331 P. R. China
| | - Yuting Chi
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology Chongqing 401331 P. R. China
| | - Linshuo Chen
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology Chongqing 401331 P. R. China
| | - Shuchen Pei
- Chongqing Key Laboratory of Industrial Fermentation Microorganism, Chongqing University of Science and Technology Chongqing 401331 P. R. China
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology Chongqing 401331 P. R. China
| | - Bin Chen
- Chongqing Key Laboratory of Non-linear Circuit and Intelligent Information Processing, College of Electronic and Information Engineering, Southwest University Chongqing 400715 P. R. China
| |
Collapse
|
23
|
Kang JTL, Teo JJY, Bertrand D, Ng A, Ravikrishnan A, Yong M, Ng OT, Marimuthu K, Chen SL, Chng KR, Gan YH, Nagarajan N. Long-term ecological and evolutionary dynamics in the gut microbiomes of carbapenemase-producing Enterobacteriaceae colonized subjects. Nat Microbiol 2022; 7:1516-1524. [PMID: 36109646 PMCID: PMC9519440 DOI: 10.1038/s41564-022-01221-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 07/29/2022] [Indexed: 11/09/2022]
Abstract
AbstractLong-term colonization of the gut microbiome by carbapenemase-producing Enterobacteriaceae (CPE) is a growing area of public health concern as it can lead to community transmission and rapid increase in cases of life-threatening CPE infections. Here, leveraging the observation that many subjects are decolonized without interventions within a year, we used longitudinal shotgun metagenomics (up to 12 timepoints) for detailed characterization of ecological and evolutionary dynamics in the gut microbiome of a cohort of CPE-colonized subjects and family members (n = 46; 361 samples). Subjects who underwent decolonization exhibited a distinct ecological shift marked by recovery of microbial diversity, key commensals and anti-inflammatory pathways. In addition, colonization was marked by elevated but unstable Enterobacteriaceae abundances, which exhibited distinct strain-level dynamics for different species (Escherichia coli and Klebsiella pneumoniae). Finally, comparative analysis with whole-genome sequencing data from CPE isolates (n = 159) helped identify substrain variation in key functional genes and the presence of highly similar E. coli and K. pneumoniae strains with variable resistance profiles and plasmid sharing. These results provide an enhanced view into how colonization by multi-drug-resistant bacteria associates with altered gut ecology and can enable transfer of resistance genes, even in the absence of overt infection and antibiotic usage.
Collapse
|
24
|
Emergent evolutionary forces in spatial models of luminal growth and their application to the human gut microbiota. Proc Natl Acad Sci U S A 2022; 119:e2114931119. [PMID: 35787046 PMCID: PMC9282425 DOI: 10.1073/pnas.2114931119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The genetic composition of the gut microbiota is constantly reshaped by ecological and evolutionary forces. These strain-level dynamics are challenging to understand because they depend on complex spatial growth processes that take place within a host. Here we introduce a population genetic framework to predict how stochastic evolutionary forces emerge from simple models of microbial growth in spatially extended environments like the intestinal lumen. Our framework shows how fluid flow and longitudinal variation in growth rate combine to shape the frequencies of genetic variants in simulated fecal samples, yielding analytical expressions for the effective generation times, selection coefficients, and rates of genetic drift. We find that over longer timescales, the emergent evolutionary dynamics can often be captured by well-mixed models that lack explicit spatial structure, even when there is substantial spatial variation in species-level composition. By applying these results to the human colon, we find that continuous fluid flow and simple forms of wall growth alone are unlikely to create sufficient bottlenecks to allow large fluctuations in mutant frequencies within a host. We also find that the effective generation times may be significantly shorter than expected from traditional average growth rate estimates. Our results provide a starting point for quantifying genetic turnover in spatially extended settings like the gut microbiota and may be relevant for other microbial ecosystems where unidirectional fluid flow plays an important role.
Collapse
|
25
|
Zhen Y, Ge L, Chen Q, Xu J, Duan Z, Loor JJ, Wang M. Latent Benefits and Toxicity Risks Transmission Chain of High Dietary Copper along the Livestock-Environment-Plant-Human Health Axis and Microbial Homeostasis: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6943-6962. [PMID: 35666880 DOI: 10.1021/acs.jafc.2c01367] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The extensive use of high-concentration copper (Cu) in feed additives, fertilizers, pesticides, and nanoparticles (NPs) inevitably causes significant pollution in the ecological environment. This type of chain pollution begins with animal husbandry: first, Cu accumulation in animals poisons them; second, high Cu enters the soil and water sources with the feces and urine to cause toxicity, which may further lead to crop and plant pollution; third, this process ultimately endangers human health through consumption of livestock products, aquatic foods, plants, and even drinking water. High Cu potentially alters the antibiotic resistance of soil and water sources and further aggravates human disease risks. Thus, it is necessary to formulate reasonable Cu emission regulations because the benefits of Cu for livestock and plants cannot be ignored. The present review evaluates the potential hazards and benefits of high Cu in livestock, the environment, the plant industry, and human health. We also discuss aspects related to bacterial and fungal resistance and homeostasis and perspectives on the application of Cu-NPs and microbial high-Cu removal technology to reduce the spread of toxicity risks to humans.
Collapse
Affiliation(s)
- Yongkang Zhen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, Xinjiang 832000, China
| | - Ling Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Qiaoqing Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jun Xu
- Institute for Quality and Safety and Standards of Agricultural Products Research, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330000, China
| | - Zhenyu Duan
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, Xinjiang 832000, China
| | - Juan J Loor
- Mammalian Nutrition Physiology Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Mengzhi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, Xinjiang 832000, China
| |
Collapse
|
26
|
Soheili M, Alinaghipour A, Salami M. Good bacteria, oxidative stress and neurological disorders: Possible therapeutical considerations. Life Sci 2022; 301:120605. [DOI: 10.1016/j.lfs.2022.120605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022]
|
27
|
The Relationship between Physical Activity, Physical Exercise, and Human Gut Microbiota in Healthy and Unhealthy Subjects: A Systematic Review. BIOLOGY 2022; 11:biology11030479. [PMID: 35336852 PMCID: PMC8945171 DOI: 10.3390/biology11030479] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/06/2023]
Abstract
Simple Summary To date, the influence that physical activity (PA)/physical exercise (PE) can exert on the human gut microbiota (GM) is still poorly understood. Several issues arise in structuring research in this area, starting from the association between PA/PE and diet. Indeed, the diet of an individual is a key factor for the composition of the GM and those who regularly practice PA/PE, generally, have dietary patterns favorable to the creation of an ideal environment for the proliferation of a GM capable of contributing to the host’s health. It is therefore difficult to establish with certainty whether the effects generated on the GM are due to a PA protocol, the type of diet followed, or to both. In addition, most of the available studies use animal models to investigate a possible correlation between PA/PE and changes in the GM, which may be not necessarily applied to humans. Evidence suggests that aerobic PA/PE seems capable of producing significant changes in GM; training parameters, likewise, can differentially influence the GM in young or elderly people and these changes appear to be transient and reversible. Abstract Several studies have been conducted to find at least an association between physical activity (PA)/ physical exercise (PE) and the possibility to modulate the gut microbiome (GM). However, the specific effects produced on the human GM by different types of PA/PE, different training modalities, and their age-related effects are not yet fully understood. Therefore, this systematic review aims to evaluate and summarize the current scientific evidence investigating the bi-directional relationship between PA/PE and the human GM, with a specific focus on the different types/variables of PA/PE and age-related effects, in healthy and unhealthy people. A systematic search was conducted across four databases (Web of Science, Medline (PubMed), Google Scholar, and Cochrane Library). Information was extracted using the populations, exposure, intervention, comparison, outcomes (PICOS) format. The Oxford Quality Scoring System Scale, the Risk of Bias in Non-Randomized Studies of Interventions (ROBINS-I) tool, and the JBI Critical Appraisal Checklist for Analytical Cross-Sectional Studies were used as a qualitative measure of the review. The protocol was registered in PROSPERO (code: CRD42022302725). The following data items were extracted: author, year of publication, study design, number and age of participants, type of PA/PE carried out, protocol/workload and diet assessment, duration of intervention, measurement tools used, and main outcomes. Two team authors reviewed 694 abstracts for inclusion and at the end of the screening process, only 76 full texts were analyzed. Lastly, only 25 research articles met the eligibility criteria. The synthesis of these findings suggests that GM diversity is associated with aerobic exercise contrary to resistance training; abundance of Prevotella genus seems to be correlated with training duration; no significant change in GM richness and diversity are detected when exercising according to the minimum dose recommended by the World Health Organizations; intense and prolonged PE can induce a higher abundance of pro-inflammatory bacteria; PA does not lead to significant GM α/β-diversity in elderly people (60+ years). The heterogeneity of the training parameters used in the studies, diet control, and different sequencing methods are the main confounders. Thus, this systematic review can provide an in-depth overview of the relationship between PA/PE and the human intestinal microbiota and, at the same time, provide indications from the athletic and health perspective.
Collapse
|
28
|
Conwill A, Kuan AC, Damerla R, Poret AJ, Baker JS, Tripp AD, Alm EJ, Lieberman TD. Anatomy promotes neutral coexistence of strains in the human skin microbiome. Cell Host Microbe 2022; 30:171-182.e7. [PMID: 34995483 PMCID: PMC8831475 DOI: 10.1016/j.chom.2021.12.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/18/2021] [Accepted: 12/09/2021] [Indexed: 01/04/2023]
Abstract
What enables strains of the same species to coexist in a microbiome? Here, we investigate whether host anatomy can explain strain co-residence of Cutibacterium acnes, the most abundant species on human skin. We reconstruct on-person evolution and migration using whole-genome sequencing of C. acnes colonies acquired from healthy subjects, including from individual skin pores, and find considerable spatial structure at the level of pores. Although lineages (sets of colonies separated by <100 mutations) with in vitro fitness differences coexist within centimeter-scale regions, each pore is dominated by a single lineage. Moreover, colonies from a pore typically have identical genomes. An absence of adaptive signatures suggests a genotype-independent source of low within-pore diversity. We therefore propose that pore anatomy imposes random single-cell bottlenecks; the resulting population fragmentation reduces competition and promotes coexistence. Our findings suggest that therapeutic interventions involving pore-dwelling species might focus on removing resident populations over optimizing probiotic fitness.
Collapse
Affiliation(s)
- Arolyn Conwill
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anne C Kuan
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ravalika Damerla
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexandra J Poret
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jacob S Baker
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - A Delphine Tripp
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Eric J Alm
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Tami D Lieberman
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
29
|
Park SY, Rao C, Coyte KZ, Kuziel GA, Zhang Y, Huang W, Franzosa EA, Weng JK, Huttenhower C, Rakoff-Nahoum S. Strain-level fitness in the gut microbiome is an emergent property of glycans and a single metabolite. Cell 2022; 185:513-529.e21. [PMID: 35120663 PMCID: PMC8896310 DOI: 10.1016/j.cell.2022.01.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/07/2021] [Accepted: 01/05/2022] [Indexed: 02/05/2023]
Abstract
The human gut microbiota resides within a diverse chemical environment challenging our ability to understand the forces shaping this ecosystem. Here, we reveal that fitness of the Bacteroidales, the dominant order of bacteria in the human gut, is an emergent property of glycans and one specific metabolite, butyrate. Distinct sugars serve as strain-variable fitness switches activating context-dependent inhibitory functions of butyrate. Differential fitness effects of butyrate within the Bacteroides are mediated by species-level variation in Acyl-CoA thioesterase activity and nucleotide polymorphisms regulating an Acyl-CoA transferase. Using in vivo multi-omic profiles, we demonstrate Bacteroides fitness in the human gut is associated together, but not independently, with Acyl-CoA transferase expression and butyrate. Our data reveal that each strain of the Bacteroides exists within a unique fitness landscape based on the interaction of chemical components unpredictable by the effect of each part alone mediated by flexibility in the core genome.
Collapse
Affiliation(s)
- Sun-Yang Park
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Chitong Rao
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Katharine Z Coyte
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Gavin A Kuziel
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Yancong Zhang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wentao Huang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric A Franzosa
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jing-Ke Weng
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seth Rakoff-Nahoum
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Microbiology, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Wu X, Wei Q, Wang X, Shang Y, Zhang H. Evolutionary and dietary relationships of wild mammals based on the gut microbiome. Gene 2022; 808:145999. [PMID: 34627942 DOI: 10.1016/j.gene.2021.145999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/10/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023]
Abstract
Gut microbiome influence the health and evolution of mammals and multiple factors modulate the structure and function of gut microbiome. However, the specific changes of the diets and phylogeny on the gut microbiome were unclear. Here, we compared the gut microbiome of 16 rare wild mammals. All data (>200G 16S rRNA gene sequences) were generated using a high-throughput sequencing platform. Firmicutes and Bacteroidetes were the most predominant phyla in all mammals. However, Proteobacteria was an additionally dominant phylum specifically detected in the microbiome of carnivores and omnivores. Moreover, the dominant phyla in canids were Firmicutes, Bacteroidetes, Proteobacteria, and Fusobacteria. Phylogenetic reconstructions based on the gut microbiome and mitochondrial genome of these mammals were similar. The impact of the host on the microbiome community composition was most evident when considering conspecific and congeneric relationships. Similarity clustering showed that the gut microbiome of herbivores was clustered together, and the other clade comprised both omnivores and carnivores. Collectively, these results revealed that phylogenetic relationships and diet have an important impact on the gut microbiome, and thus the gut microbiome community composition may reflect both the phylogenetic relationships and diets. This study provides valuable basic data to facilitate future efforts related to animal conservation and health.
Collapse
Affiliation(s)
- Xiaoyang Wu
- School of Life Science, Qufu Normal University, Qufu, Shandong, PR China
| | - Qinguo Wei
- School of Life Science, Qufu Normal University, Qufu, Shandong, PR China
| | - Xibao Wang
- School of Life Science, Qufu Normal University, Qufu, Shandong, PR China
| | - Yongquan Shang
- School of Life Science, Qufu Normal University, Qufu, Shandong, PR China
| | - Honghai Zhang
- School of Life Science, Qufu Normal University, Qufu, Shandong, PR China.
| |
Collapse
|
31
|
Sharma R. Emerging Interrelationship Between the Gut Microbiome and Cellular Senescence in the Context of Aging and Disease: Perspectives and Therapeutic Opportunities. Probiotics Antimicrob Proteins 2022; 14:648-663. [PMID: 34985682 PMCID: PMC8728710 DOI: 10.1007/s12602-021-09903-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 12/12/2022]
Abstract
The significance of diversity, composition, and functional attributes of the gut microbiota in shaping human health is well recognized. Studies have shown that gut microbiota is closely linked to human aging, and changes in the gut microbiome can predict human survival and longevity. In addition, a causal relationship between gut microbiota dysbiosis and chronic age-related disorders is also becoming apparent. Recent advances in our understanding of the cellular and molecular aspects of biological aging have revealed a cellular senescence-centric view of the aging process. However, the association between the gut microbiome and cellular senescence is only beginning to be understood. The present review provides an integrative view of the evolving relationship between the gut microbiome and cellular senescence in aging and disease. Evidence relating to microbiome-mediated modulation of senescent cells, as well as senescent cells-mediated changes in intestinal homeostasis and diseases, have been discussed. Unanswered questions and future research directions have also been deliberated to truly ascertain the relationship between the gut microbiome and cellular senescence for developing microbiome-based age-delaying and longevity-promoting therapies.
Collapse
Affiliation(s)
- Rohit Sharma
- Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan, 173229, India.
| |
Collapse
|
32
|
Mazzucco R, Schlötterer C. Long-term gut microbiome dynamics in Drosophila melanogaster reveal environment-specific associations between bacterial taxa at the family level. Proc Biol Sci 2021; 288:20212193. [PMID: 34905708 PMCID: PMC8670958 DOI: 10.1098/rspb.2021.2193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The influence of the microbiome on its host is well-documented, but the interplay of its members is not yet well-understood. Even for simple microbiomes, the interaction among members of the microbiome is difficult to study. Longitudinal studies provide a promising approach to studying such interactions through the temporal covariation of different taxonomic units. By contrast to most longitudinal studies, which span only a single host generation, we here present a post hoc analysis of a whole-genome dataset of 81 samples that follows microbiome composition for up to 180 host generations, which cover nearly 10 years. The microbiome diversity remained rather stable in replicated Drosophila melanogaster populations exposed to two different temperature regimes. The composition changed, however, systematically across replicates of the two temperature regimes. Significant associations between families, mostly specific to one temperature regime, indicate functional interdependence of different microbiome components. These associations also involve moderately abundant families, which emphasizes their functional importance, and highlights the importance of looking beyond the common constituents of the Drosophila microbiome.
Collapse
Affiliation(s)
- Rupert Mazzucco
- Institut für Populationsgenetik, Veterinärmedizinische Universität Wien, Veterinärplatz 1, Wien 1210, Austria
| | - Christian Schlötterer
- Institut für Populationsgenetik, Veterinärmedizinische Universität Wien, Veterinärplatz 1, Wien 1210, Austria
| |
Collapse
|
33
|
Haraoui LP. Networked collective microbiomes and the rise of subcellular 'units of life'. Trends Microbiol 2021; 30:112-119. [PMID: 34696928 DOI: 10.1016/j.tim.2021.09.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
Microbiomes are generally conceived of as one element of a pair - their partner being the habitat they occupy. I call this common scientific practice 'pair-thinking'. Research into antimicrobial resistance and its underlying anthropogenic drivers highlights the growing footprint occupied by mobile genetic elements (MGEs). Furthermore, these MGEs are known to circulate widely between microbiomes. Using a pluralistic framework anchored within a processual microbial ontology, these observations point to a reframing of microbiomes as networked and collective, thus challenging pair-thinking. Such a shift has implications for the future of microbiome research, from conceptual and methodological perspectives, and exposes the impacts of anthropogenic forces on the evolution of microbiomes and the functions they carry out.
Collapse
Affiliation(s)
- Louis-Patrick Haraoui
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
34
|
Ma C, Zhang C, Chen D, Jiang S, Shen S, Huo D, Huang S, Zhai Q, Zhang J. Probiotic consumption influences universal adaptive mutations in indigenous human and mouse gut microbiota. Commun Biol 2021; 4:1198. [PMID: 34663913 PMCID: PMC8523657 DOI: 10.1038/s42003-021-02724-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 09/21/2021] [Indexed: 11/30/2022] Open
Abstract
The adaptive evolution in indigenous intestinal microbes derived from probiotics is critical to safety and efficacy evaluation of probiotics, yet it is still largely underexplored. Here, through 11 publicly accessible datasets, we demonstrated that probiotic consumption can lead to widespread single-nucleotide variants (SNVs) in the native microbiota. Interestingly, the same probiotic strains introduced far more SNVs in mouse gut than humans. Furthermore, the pattern of probiotics-induced SNVs was highly probiotic-strain specific, and 17 common SNVs in Faecalibacterium prausnitzii genome were identified cross studies, which might lead to changes in bacterial protein structure. Further, nearly 50% of F. prausnitzii SNVs can be inherited for six months in an independent human cohort, whereas the other half only transiently occurred. Collectively, our study substantially extended our understanding of co-evolution of the probiotics and the indigenous gut microbiota, highlighting the importance of assessment of probiotics efficacy and safety in an integrated manner. Chenchen Ma, Chengcheng Zhang, and Denghui Chen et al. examine how probiotic consumption impacts gut microbiota composition in human and mice through a global, cross-cohort metagenomic analysis. Their results suggest that probiotic consumption may result in widespread variation among the native microbiota in both the human and mouse gut.
Collapse
Affiliation(s)
- Chenchen Ma
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, 570228, Haikou, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 214122, Wuxi, China
| | - Denghui Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shuaiming Jiang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, 570228, Haikou, China
| | - Siyuan Shen
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, 570228, Haikou, China
| | - Dongxue Huo
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, 570228, Haikou, China
| | - Shi Huang
- Department of Pediatrics and Center for Microbiome Innovation at Jacobs School of Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 214122, Wuxi, China.
| | - Jiachao Zhang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, 570228, Haikou, China.
| |
Collapse
|
35
|
Peralta-Marzal LN, Prince N, Bajic D, Roussin L, Naudon L, Rabot S, Garssen J, Kraneveld AD, Perez-Pardo P. The Impact of Gut Microbiota-Derived Metabolites in Autism Spectrum Disorders. Int J Mol Sci 2021; 22:10052. [PMID: 34576216 PMCID: PMC8470471 DOI: 10.3390/ijms221810052] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a set of neurodevelopmental disorders characterised by behavioural impairment and deficiencies in social interaction and communication. A recent study estimated that 1 in 89 children have developed some form of ASD in European countries. Moreover, there is no specific treatment and since ASD is not a single clinical entity, the identification of molecular biomarkers for diagnosis remains challenging. Besides behavioural deficiencies, individuals with ASD often develop comorbid medical conditions including intestinal problems, which may reflect aberrations in the bidirectional communication between the brain and the gut. The impact of faecal microbial composition in brain development and behavioural functions has been repeatedly linked to ASD, as well as changes in the metabolic profile of individuals affected by ASD. Since metabolism is one of the major drivers of microbiome-host interactions, this review aims to report emerging literature showing shifts in gut microbiota metabolic function in ASD. Additionally, we discuss how these changes may be involved in and/or perpetuate ASD pathology. These valuable insights can help us to better comprehend ASD pathogenesis and may provide relevant biomarkers for improving diagnosis and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Lucía N. Peralta-Marzal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.P.); (J.G.); (A.D.K.)
| | - Naika Prince
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.P.); (J.G.); (A.D.K.)
| | - Djordje Bajic
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA;
- Microbial Sciences Institute, Yale University, West Haven, CT 06516, USA
| | - Léa Roussin
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (L.R.); (S.R.)
| | - Laurent Naudon
- CNRS, Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France;
| | - Sylvie Rabot
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (L.R.); (S.R.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.P.); (J.G.); (A.D.K.)
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.P.); (J.G.); (A.D.K.)
| | - Paula Perez-Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.P.); (J.G.); (A.D.K.)
| |
Collapse
|
36
|
P-Doped Carbon Quantum Dots with Antibacterial Activity. MICROMACHINES 2021; 12:mi12091116. [PMID: 34577758 PMCID: PMC8466419 DOI: 10.3390/mi12091116] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 02/01/2023]
Abstract
It is a major challenge to effectively inhibit microbial pathogens in the treatment of infectious diseases. Research on the application of nanomaterials as antibacterial agents has evidenced their great potential for the remedy of infectious disease. Among these nanomaterials, carbon quantum dots (CQDs) have attracted much attention owing to their unique optical properties and high biosafety. In this work, P-doped CQDs were prepared by simple hydrothermal treatment of m-aminophenol and phosphoric acid with fluorescence emission at 501 nm when excited at 429 nm. The P-doped CQDs showed effective antibacterial activity against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). The minimal inhibitory concentrations (MICs) of P-doped CQD were 1.23 mg/mL for E. coli and 1.44 mg/mL for S. aureus. Furthermore, the morphologies of E. coli cells were damaged and S. aureus became irregular when treated with the P-doped CQDs. The results of zeta potential analysis demonstrated that the P-doped CQDs inhibit antibacterial activity and destroy the structure of bacteria by electronic interaction. In combination, the results of this study indicate that the as-prepared P-doped CQDs can be a promising candidate for the treatment of bacterial infections.
Collapse
|
37
|
Henry LP, Bruijning M, Forsberg SKG, Ayroles JF. The microbiome extends host evolutionary potential. Nat Commun 2021; 12:5141. [PMID: 34446709 PMCID: PMC8390463 DOI: 10.1038/s41467-021-25315-x] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
The microbiome shapes many host traits, yet the biology of microbiomes challenges traditional evolutionary models. Here, we illustrate how integrating the microbiome into quantitative genetics can help untangle complexities of host-microbiome evolution. We describe two general ways in which the microbiome may affect host evolutionary potential: by shifting the mean host phenotype and by changing the variance in host phenotype in the population. We synthesize the literature across diverse taxa and discuss how these scenarios could shape the host response to selection. We conclude by outlining key avenues of research to improve our understanding of the complex interplay between hosts and microbiomes.
Collapse
Affiliation(s)
- Lucas P. Henry
- grid.16750.350000 0001 2097 5006Dept. of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ USA ,grid.16750.350000 0001 2097 5006Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ USA
| | - Marjolein Bruijning
- grid.16750.350000 0001 2097 5006Dept. of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ USA
| | - Simon K. G. Forsberg
- grid.16750.350000 0001 2097 5006Dept. of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ USA ,grid.16750.350000 0001 2097 5006Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ USA ,grid.8993.b0000 0004 1936 9457Dept. of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Julien F. Ayroles
- grid.16750.350000 0001 2097 5006Dept. of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ USA ,grid.16750.350000 0001 2097 5006Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ USA
| |
Collapse
|
38
|
Hu Y, He J, Zheng P, Mao X, Huang Z, Yan H, Luo Y, Yu J, Luo J, Yu B, Chen D. Prebiotic inulin as a treatment of obesity related nonalcoholic fatty liver disease through gut microbiota: a critical review. Crit Rev Food Sci Nutr 2021; 63:862-872. [PMID: 34292103 DOI: 10.1080/10408398.2021.1955654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The microbial-derived products, including short chain fatty acids, lipopolysaccharide and secondary bile acids, have been shown to participate in the regulation of hepatic lipid metabolism. Previous studies have demonstrated that prebiotics, such as oligosaccharide and inulin, have abilities to change the concentration of microbial-derived products through modulating the microbial community structure, thus controlling body weight and alleviating hepatic fat accumulation. However, recent evidence indicates that there are individual differences in host response upon inulin treatment due to the differences in host microbial composition before dietary intervention. Probably it is because of the multiple relationships among bacterial species (e.g., competition and mutualism), which play key roles in the degradation of inulin and the regulation of microbial structure. Thereby, analyzing the composition and function of initial gut microbiota is essential for improving the efficacy of prebiotics supplementation. Furthermore, considering that different structures of polysaccharides can be used by different microorganisms, the chemical structure of processed inulin should be tested before using prebiotic inulin to treat obesity related nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Yaolian Hu
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Jun He
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Ping Zheng
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Xiangbing Mao
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Zhiqing Huang
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Hui Yan
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Yuheng Luo
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Jie Yu
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Junqiu Luo
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Bing Yu
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| | - Daiwen Chen
- Key laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, People's Republic of China
| |
Collapse
|
39
|
Josephs-Spaulding J, Krogh TJ, Rettig HC, Lyng M, Chkonia M, Waschina S, Graspeuntner S, Rupp J, Møller-Jensen J, Kaleta C. Recurrent Urinary Tract Infections: Unraveling the Complicated Environment of Uncomplicated rUTIs. Front Cell Infect Microbiol 2021; 11:562525. [PMID: 34368008 PMCID: PMC8340884 DOI: 10.3389/fcimb.2021.562525] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
Urinary tract infections (UTIs) are frequent in humans, affecting the upper and lower urinary tract. Present diagnosis relies on the positive culture of uropathogenic bacteria from urine and clinical markers of inflammation of the urinary tract. The bladder is constantly challenged by adverse environmental stimuli which influence urinary tract physiology, contributing to a dysbiotic environment. Simultaneously, pathogens are primed by environmental stressors such as antibiotics, favoring recurrent UTIs (rUTIs), resulting in chronic illness. Due to different confounders for UTI onset, a greater understanding of the fundamental environmental mechanisms and microbial ecology of the human urinary tract is required. Such advancements could promote the tandem translation of bench and computational studies for precision treatments and clinical management of UTIs. Therefore, there is an urgent need to understand the ecological interactions of the human urogenital microbial communities which precede rUTIs. This review aims to outline the mechanistic aspects of rUTI ecology underlying dysbiosis between both the human microbiome and host physiology which predisposes humans to rUTIs. By assessing the applications of next generation and systems level methods, we also recommend novel approaches to elucidate the systemic consequences of rUTIs which requires an integrated approach for successful treatment. To this end, we will provide an outlook towards the so-called 'uncomplicated environment of UTIs', a holistic and systems view that applies ecological principles to define patient-specific UTIs. This perspective illustrates the need to withdraw from traditional reductionist perspectives in infection biology and instead, a move towards a systems-view revolving around patient-specific pathophysiology during UTIs.
Collapse
Affiliation(s)
- Jonathan Josephs-Spaulding
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-Universität, Kiel, Germany
| | - Thøger Jensen Krogh
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Hannah Clara Rettig
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Mark Lyng
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Mariam Chkonia
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Silvio Waschina
- Research Group Nutriinformatics, Institute of Human Nutrition and Food Science, Christian-Albrechts-Universität, Kiel, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Jakob Møller-Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-Universität, Kiel, Germany
| |
Collapse
|
40
|
Wang T, Weiss A, Ha Y, You L. Predicting plasmid persistence in microbial communities by coarse-grained modeling. Bioessays 2021; 43:e2100084. [PMID: 34278591 DOI: 10.1002/bies.202100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 11/08/2022]
Abstract
Plasmids are a major type of mobile genetic elements (MGEs) that mediate horizontal gene transfer. The stable maintenance of plasmids plays a critical role in the functions and survival for microbial populations. However, predicting and controlling plasmid persistence and abundance in complex microbial communities remain challenging. Computationally, this challenge arises from the combinatorial explosion associated with the conventional modeling framework. Recently, a plasmid-centric framework (PCF) has been developed to overcome this computational bottleneck. This framework enables the derivation of a simple metric, the persistence potential, to predict plasmid persistence and abundance. Here, we discuss how PCF can be extended to account for plasmid interactions. We also discuss how such model-guided predictions of plasmid fates can benefit from the development of new experimental tools and data-driven computational methods.
Collapse
Affiliation(s)
- Teng Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Andrea Weiss
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Yuanchi Ha
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA.,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
41
|
Huang S, Jiang S, Huo D, Allaband C, Estaki M, Cantu V, Belda-Ferre P, Vázquez-Baeza Y, Zhu Q, Ma C, Li C, Zarrinpar A, Liu YY, Knight R, Zhang J. Candidate probiotic Lactiplantibacillus plantarum HNU082 rapidly and convergently evolves within human, mice, and zebrafish gut but differentially influences the resident microbiome. MICROBIOME 2021; 9:151. [PMID: 34193290 PMCID: PMC8247228 DOI: 10.1186/s40168-021-01102-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/24/2021] [Indexed: 05/07/2023]
Abstract
BACKGROUND Improving probiotic engraftment in the human gut requires a thorough understanding of the in vivo adaptive strategies of probiotics in diverse contexts. However, for most probiotic strains, these in vivo genetic processes are still poorly characterized. Here, we investigated the effects of gut selection pressures from human, mice, and zebrafish on the genetic stability of a candidate probiotic Lactiplantibacillus plantarum HNU082 (Lp082) as well as its ecological and evolutionary impacts on the indigenous gut microbiota using shotgun metagenomic sequencing in combination with isolate resequencing methods. RESULTS We combined both metagenomics and isolate whole genome sequencing approaches to systematically study the gut-adaptive evolution of probiotic L. plantarum and the ecological and evolutionary changes of resident gut microbiomes in response to probiotic ingestion in multiple host species. Independent of host model, Lp082 colonized and adapted to the gut by acquiring highly consistent single-nucleotide mutations, which primarily modulated carbohydrate utilization and acid tolerance. We cultivated the probiotic mutants and validated that these gut-adapted mutations were genetically stable for at least 3 months and improved their fitness in vitro. In turn, resident gut microbial strains, especially competing strains with Lp082 (e.g., Bacteroides spp. and Bifidobacterium spp.), actively responded to Lp082 engraftment by accumulating 10-70 times more evolutionary changes than usual. Human gut microbiota exhibited a higher ecological and genetic stability than that of mice. CONCLUSIONS Collectively, our results suggest a highly convergent adaptation strategy of Lp082 across three different host environments. In contrast, the evolutionary changes within the resident gut microbes in response to Lp082 were more divergent and host-specific; however, these changes were not associated with any adverse outcomes. This work lays a theoretical foundation for leveraging animal models for ex vivo engineering of probiotics to improve engraftment outcomes in humans. Video abstract.
Collapse
Affiliation(s)
- Shi Huang
- School of Food Science and Engineering, Hainan University, Haikou, China
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Shuaiming Jiang
- School of Food Science and Engineering, Hainan University, Haikou, China
| | - Dongxue Huo
- School of Food Science and Engineering, Hainan University, Haikou, China
| | - Celeste Allaband
- Biomedical Sciences Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Mehrbod Estaki
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Victor Cantu
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Pedro Belda-Ferre
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Yoshiki Vázquez-Baeza
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Qiyun Zhu
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Chenchen Ma
- School of Food Science and Engineering, Hainan University, Haikou, China
| | - Congfa Li
- School of Food Science and Engineering, Hainan University, Haikou, China
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China
| | - Amir Zarrinpar
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- UCSD Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- VA San Diego Healthcare, 3350 La Jolla Village Dr, San Diego, CA, 92161, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Rob Knight
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Biomedical Sciences Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Department of Computer Science and Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, China.
- UCSD Health Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China.
| |
Collapse
|
42
|
Westerman EL, Bowman SEJ, Davidson B, Davis MC, Larson ER, Sanford CPJ. Deploying Big Data to Crack the Genotype to Phenotype Code. Integr Comp Biol 2021; 60:385-396. [PMID: 32492136 DOI: 10.1093/icb/icaa055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mechanistically connecting genotypes to phenotypes is a longstanding and central mission of biology. Deciphering these connections will unite questions and datasets across all scales from molecules to ecosystems. Although high-throughput sequencing has provided a rich platform on which to launch this effort, tools for deciphering mechanisms further along the genome to phenome pipeline remain limited. Machine learning approaches and other emerging computational tools hold the promise of augmenting human efforts to overcome these obstacles. This vision paper is the result of a Reintegrating Biology Workshop, bringing together the perspectives of integrative and comparative biologists to survey challenges and opportunities in cracking the genotype to phenotype code and thereby generating predictive frameworks across biological scales. Key recommendations include promoting the development of minimum "best practices" for the experimental design and collection of data; fostering sustained and long-term data repositories; promoting programs that recruit, train, and retain a diversity of talent; and providing funding to effectively support these highly cross-disciplinary efforts. We follow this discussion by highlighting a few specific transformative research opportunities that will be advanced by these efforts.
Collapse
Affiliation(s)
- Erica L Westerman
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Sarah E J Bowman
- High-Throughput Crystallization Screening Center, Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA.,Department of Biochemistry, Jacobs School of Medicine & Biomedical Sciences at the University at Buffalo, Buffalo, NY 14203, USA
| | - Bradley Davidson
- Department of Biology, Swarthmore College, Swarthmore, PA 19081, USA
| | - Marcus C Davis
- Department of Biology, James Madison University, Harrisonburg, VA 22807, USA
| | - Eric R Larson
- Department of Natural Resources and Environmental Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Christopher P J Sanford
- Department of Ecology, Evolution and Organismal Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| |
Collapse
|
43
|
Al'Abri IS, Durmusoglu D, Crook N. What E. coli knows about your 1-year-old infant: Antibiotic use, lifestyle, birth mode, and siblings. Cell Host Microbe 2021; 29:854-855. [PMID: 34111392 DOI: 10.1016/j.chom.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The infant gut microbiota is shaped by diverse environmental exposures that alter its composition and can enrich antimicrobial resistance genes (ARGs). In this issue of Cell Host & Microbe, Li et al. (2021) studied the causes, spread, and dynamics of ARGs and their relationship with asthma-associated microbiota in Danish children.
Collapse
Affiliation(s)
- Ibrahim S Al'Abri
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - Deniz Durmusoglu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
44
|
Danehower S. Targeting gut dysbiosis as a means to enhance recovery from surgical brain injury. Surg Neurol Int 2021; 12:210. [PMID: 34084637 PMCID: PMC8168676 DOI: 10.25259/sni_72_2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/11/2021] [Indexed: 11/04/2022] Open
Abstract
Background Surgical brain injury (SBI) impacts roughly 800,000 people who undergo neurosurgical procedures each year. SBI is the result of unavoidable parenchymal damage, vessel disruption, and thermal injury that is an inherent part of all neurosurgical procedures. Clinically, SBI has been associated with postoperative seizures and long-term neurobehavioral deficits. Current therapies are aimed at providing symptom relief by reducing swelling and preventing seizures. However, there are no therapies aimed at reducing the extent of SBI preoperatively. The microbiome-gut-brain axis may serve as a potential target for the development of new preventative therapies due to its extensive involvement in central nervous system function. Methods An extensive literature review was conducted to determine whether there is a potential role for dysbiosis treatment in reducing the extent of SBI. Results Treatment of gut dysbiosis deserves further exploration as a potential means of reducing the extent of unavoidable SBI. Dysbiosis has been correlated with increased neuroinflammation through impaired immune regulation, increased blood-brain barrier permeability, and increased production of reactive metabolites. Recently, dysbiosis has also been linked to acute neurological dysfunction in the postoperative state. Importantly, treatment of dysbiosis has been correlated with better patient outcomes and decreased length of stay in surgical patients. Conclusion Current literature supports the role of dysbiosis treatment in the preoperative setting as a means of optimizing neurological recovery following unavoidable SBI that results from all neurosurgical procedures.
Collapse
Affiliation(s)
- Sarah Danehower
- Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| |
Collapse
|
45
|
D’Alessandro G, Lauro C, Quaglio D, Ghirga F, Botta B, Trettel F, Limatola C. Neuro-Signals from Gut Microbiota: Perspectives for Brain Glioma. Cancers (Basel) 2021; 13:2810. [PMID: 34199968 PMCID: PMC8200200 DOI: 10.3390/cancers13112810] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of glioma tumor in adult brain. Among the numerous factors responsible for GBM cell proliferation and invasion, neurotransmitters such as dopamine, serotonin and glutamate can play key roles. Studies performed in mice housed in germ-free (GF) conditions demonstrated the relevance of the gut-brain axis in a number of physiological and pathological conditions. The gut-brain communication is made possible by vagal/nervous and blood/lymphatic routes and pave the way for reciprocal modulation of functions. The gut microbiota produces and consumes a wide range of molecules, including neurotransmitters (dopamine, norepinephrine, serotonin, gamma-aminobutyric acid [GABA], and glutamate) that reach their cellular targets through the bloodstream. Growing evidence in animals suggests that modulation of these neurotransmitters by the microbiota impacts host neurophysiology and behavior, and affects neural cell progenitors and glial cells, along with having effects on tumor cell growth. In this review we propose a new perspective connecting neurotransmitter modulation by gut microbiota to glioma progression.
Collapse
Affiliation(s)
- Giuseppina D’Alessandro
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; (G.D.); (C.L.); (F.T.)
- IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; (G.D.); (C.L.); (F.T.)
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.Q.); (F.G.); (B.B.)
| | - Francesca Ghirga
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.Q.); (F.G.); (B.B.)
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.Q.); (F.G.); (B.B.)
| | - Flavia Trettel
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; (G.D.); (C.L.); (F.T.)
| | - Cristina Limatola
- IRCCS Neuromed, 86077 Pozzilli, IS, Italy
- Department of Physiology and Pharmacology, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia, 00185 Rome, Italy
| |
Collapse
|
46
|
Durmusoglu D, Al’Abri IS, Collins SP, Cheng J, Eroglu A, Beisel CL, Crook N. In Situ Biomanufacturing of Small Molecules in the Mammalian Gut by Probiotic Saccharomyces boulardii. ACS Synth Biol 2021; 10:1039-1052. [PMID: 33843197 DOI: 10.1021/acssynbio.0c00562] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Saccharomyces boulardii is a probiotic yeast that exhibits rapid growth at 37 °C, is easy to transform, and can produce therapeutic proteins in the gut. To establish its ability to produce small molecules encoded by multigene pathways, we measured the amount and variance in protein expression enabled by promoters, terminators, selective markers, and copy number control elements. We next demonstrated efficient (>95%) CRISPR-mediated genome editing in this strain, allowing us to probe engineered gene expression across different genomic sites. We leveraged these strategies to assemble pathways enabling a wide range of vitamin precursor (β-carotene) and drug (violacein) titers. We found that S. boulardii colonizes germ-free mice stably for over 30 days and competes for niche space with commensal microbes, exhibiting short (1-2 day) gut residence times in conventional and antibiotic-treated mice. Using these tools, we enabled β-carotene synthesis (194 μg total) in the germ-free mouse gut over 14 days, estimating that the total mass of additional β-carotene recovered in feces was 56-fold higher than the β-carotene present in the initial probiotic dose. This work quantifies heterologous small molecule production titers by S. boulardii living in the mammalian gut and provides a set of tools for modulating these titers.
Collapse
Affiliation(s)
- Deniz Durmusoglu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Ibrahim S. Al’Abri
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Scott P. Collins
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Junrui Cheng
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Room 3204, Kannapolis, North Carolina 28081, United States
| | - Abdulkerim Eroglu
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Room 3204, Kannapolis, North Carolina 28081, United States
- Department of Molecular and Structural Biochemistry, College of Agriculture and Life Sciences, North Carolina State University, 120 Broughton Drive, Room 351, Raleigh, North Carolina 27695-7622, United States
| | - Chase L. Beisel
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg 97080, Germany
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
47
|
Erturk-Hasdemir D, Ochoa-Repáraz J, Kasper DL, Kasper LH. Exploring the Gut-Brain Axis for the Control of CNS Inflammatory Demyelination: Immunomodulation by Bacteroides fragilis' Polysaccharide A. Front Immunol 2021; 12:662807. [PMID: 34025663 PMCID: PMC8131524 DOI: 10.3389/fimmu.2021.662807] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
The symbiotic relationship between animals and their resident microorganisms has profound effects on host immunity. The human microbiota comprises bacteria that reside in the gastrointestinal tract and are involved in a range of inflammatory and autoimmune diseases. The gut microbiota's immunomodulatory effects extend to extraintestinal tissues, including the central nervous system (CNS). Specific symbiotic antigens responsible for inducing immunoregulation have been isolated from different bacterial species. Polysaccharide A (PSA) of Bacteroides fragilis is an archetypical molecule for host-microbiota interactions. Studies have shown that PSA has beneficial effects in experimental disease models, including experimental autoimmune encephalomyelitis (EAE), the most widely used animal model for multiple sclerosis (MS). Furthermore, in vitro stimulation with PSA promotes an immunomodulatory phenotype in human T cells isolated from healthy and MS donors. In this review, we discuss the current understanding of the interactions between gut microbiota and the host in the context of CNS inflammatory demyelination, the immunomodulatory roles of gut symbionts. More specifically, we also discuss the immunomodulatory effects of B. fragilis PSA in the gut-brain axis and its therapeutic potential in MS. Elucidation of the molecular mechanisms responsible for the microbiota's impact on host physiology offers tremendous promise for discovering new therapies.
Collapse
Affiliation(s)
| | | | - Dennis L. Kasper
- Department of Immunology, Harvard Medical School, Boston, MA, United States
| | - Lloyd H. Kasper
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States
| |
Collapse
|
48
|
Henry LP, Ayroles JF. Meta-analysis suggests the microbiome responds to Evolve and Resequence experiments in Drosophila melanogaster. BMC Microbiol 2021; 21:108. [PMID: 33836662 PMCID: PMC8034159 DOI: 10.1186/s12866-021-02168-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 03/31/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Experimental evolution has a long history of uncovering fundamental insights into evolutionary processes, but has largely neglected one underappreciated component--the microbiome. As eukaryotic hosts evolve, the microbiome may also respond to selection. However, the microbial contribution to host evolution remains poorly understood. Here, we re-analyzed genomic data to characterize the metagenomes from ten Evolve and Resequence (E&R) experiments in Drosophila melanogaster to determine how the microbiome changed in response to host selection. RESULTS Bacterial diversity was significantly different in 5/10 studies, primarily in traits associated with metabolism or immunity. Duration of selection did not significantly influence bacterial diversity, highlighting the importance of associations with specific host traits. CONCLUSIONS Our genomic re-analysis suggests the microbiome often responds to host selection; thus, the microbiome may contribute to the response of Drosophila in E&R experiments. We outline important considerations for incorporating the microbiome into E&R experiments. The E&R approach may provide critical insights into host-microbiome interactions and fundamental insight into the genomic basis of adaptation.
Collapse
Affiliation(s)
- Lucas P Henry
- Department of Ecology & Evolutionary Biology, 150 Carl Icahn Laboratory, Princeton University, Princeton, NJ, 08544, USA.
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA.
| | - Julien F Ayroles
- Department of Ecology & Evolutionary Biology, 150 Carl Icahn Laboratory, Princeton University, Princeton, NJ, 08544, USA.
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
49
|
Ruuskanen MO, Sommeria-Klein G, Havulinna AS, Niiranen TJ, Lahti L. Modelling spatial patterns in host-associated microbial communities. Environ Microbiol 2021; 23:2374-2388. [PMID: 33734553 DOI: 10.1111/1462-2920.15462] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Microbial communities exhibit spatial structure at different scales, due to constant interactions with their environment and dispersal limitation. While this spatial structure is often considered in studies focusing on free-living environmental communities, it has received less attention in the context of host-associated microbial communities or microbiota. The wider adoption of methods accounting for spatial variation in these communities will help to address open questions in basic microbial ecology as well as realize the full potential of microbiome-aided medicine. Here, we first overview known factors affecting the composition of microbiota across diverse host types and at different scales, with a focus on the human gut as one of the most actively studied microbiota. We outline a number of topical open questions in the field related to spatial variation and patterns. We then review the existing methodology for the spatial modelling of microbiota. We suggest that methodology from related fields, such as systems biology and macro-organismal ecology, could be adapted to obtain more accurate models of spatial structure. We further posit that methodological developments in the spatial modelling and analysis of microbiota could in turn broadly benefit theoretical and applied ecology and contribute to the development of novel industrial and clinical applications.
Collapse
Affiliation(s)
- Matti O Ruuskanen
- Department of Internal Medicine, University of Turku, Turku, Finland.,Finnish Institute for Health and Welfare, Helsinki, Finland
| | | | - Aki S Havulinna
- Finnish Institute for Health and Welfare, Helsinki, Finland.,Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Teemu J Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland.,Finnish Institute for Health and Welfare, Helsinki, Finland.,Division of Medicine, Turku University Hospital, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| |
Collapse
|
50
|
Ahmad SY, Friel JK, Mackay DS. Effect of sucralose and aspartame on glucose metabolism and gut hormones. Nutr Rev 2021; 78:725-746. [PMID: 32065635 DOI: 10.1093/nutrit/nuz099] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Non-nutritive sweeteners are thought to be useful replacements for caloric sweeteners in sweet food and beverages, since the reduction in energy and carbohydrate intake may lead to health benefits stemming from weight management and glycemic control. However, the potential effects of non-nutritive sweeteners on glucose metabolism and gut hormones have not been determined definitively. Here, the available evidence of the effects of aspartame and sucralose consumption on glucose metabolism and gut hormones is reviewed. A majority of studies have found that consumption of aspartame or sucralose has no effect on concentrations of blood glucose, insulin, or gut hormones; however, 2 trials have shown that aspartame consumption affects glucose, insulin, and glucagon-like peptide 1 concentrations, while only a few trials have shown that sucralose consumption affects glucose, insulin, and glucagon-like peptide 1 concentrations. One study found higher glucose concentrations after sucralose consumption, while 3 studies found lower concentrations and 33 studies found no change in glucose concentrations. Moreover, only 4 studies reported increased concentrations of glucagon-like peptide 1. Three studies reported decreased insulin sensitivity following sucralose consumption, while 1 trial reported an increase in insulin sensitivity. In summary, the evidence from the clinical trials conducted to date is contradictory because of the different protocols used.
Collapse
Affiliation(s)
- Samar Y Ahmad
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James K Friel
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dylan S Mackay
- Department of Community Health Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|