1
|
Lin D, Fu Z, Liu J, Perrone-Bizzozero N, Hutchison KE, Bustillo J, Du Y, Pearlson G, Calhoun VD. Association between the oral microbiome and brain resting state connectivity in schizophrenia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573165. [PMID: 38234846 PMCID: PMC10793457 DOI: 10.1101/2023.12.22.573165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Recent microbiome-brain axis findings have shown evidence of the modulation of microbiome community as an environmental mediator in brain function and psychiatric illness. This work is focused on the role of the microbiome in understanding a rarely investigated environmental involvement in schizophrenia (SZ), especially in relation to brain circuit dysfunction. We leveraged high throughput microbial 16s rRNA sequencing and functional neuroimaging techniques to enable the delineation of microbiome-brain network links in SZ. N=213 SZ and healthy control (HC) subjects were assessed for the oral microbiome. Among them, 139 subjects were scanned by resting-state functional magnetic resonance imaging (rsfMRI) to derive brain functional connectivity. We found a significant microbiome compositional shift in SZ beta diversity (weighted UniFrac distance, p= 6×10 -3 ; Bray-Curtis distance p = 0.021). Fourteen microbial species involving pro-inflammatory and neurotransmitter signaling and H 2 S production, showed significant abundance alterations in SZ. Multivariate analysis revealed one pair of microbial and functional connectivity components showing a significant correlation of 0.46. Thirty five percent of microbial species and 87.8% of brain functional network connectivity from each component also showed significant differences between SZ and HC with strong performance in classifying SZ from HC, with an area under curve (AUC) = 0.84 and 0.87, respectively. The results suggest a potential link between oral microbiome dysbiosis and brain functional connectivity alteration in relation to SZ, possibly through immunological and neurotransmitter signaling pathways and the hypothalamic-pituitary-adrenal axis, supporting for future work in characterizing the role of oral microbiome in mediating effects on SZ brain functional activity.
Collapse
|
2
|
Understanding host-microbiota interactions in the commercial piglet around weaning. Sci Rep 2021; 11:23488. [PMID: 34873196 PMCID: PMC8648723 DOI: 10.1038/s41598-021-02754-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Weaning is a critical period in the life of pigs with repercussions on their health and welfare and on the economy of the swine industry. This study aimed to assess the effect of the commercial early weaning on gut microbiota, intestinal gene expression and serum metabolomic response via an integrated-omic approach combining 16S rRNA gene sequencing, the OpenArray gene expression technology and 1H-NMR spectroscopy. Fourteen piglets from different litters were sampled for blood, jejunum tissue and caecal content two days before (− 2d), and three days after (+ 3d) weaning. A clearly differential ordination of caecal microbiota was observed. Higher abundances of Roseburia, Ruminococcus, Coprococcus, Dorea and Lachnospira genera in weaned piglets compared to prior to weaning showed the quick microbial changes of the piglets’ gut microbiota. Downregulation of OCLN, CLDN4, MUC2, MUC13, SLC15A1 and SLC13A1 genes, also evidenced the negative impact of weaning on gut barrier and digestive functions. Metabolomic approach pinpointed significant decreases in choline, LDL, triglycerides, fatty acids, alanine and isoleucine and increases in 3-hydroxybutyrate after weaning. Moreover, the correlation between microbiota and metabolome datasets revealed the existence of metabolic clusters interrelated to different bacterial clusters. Our results demonstrate the impact of weaning stress on the piglet and give insights regarding the associations between gut microbiota and the animal gene activity and metabolic response.
Collapse
|
3
|
Ozkan H, Tuzun F, Taheri S, Korhan P, Akokay P, Yılmaz O, Duman N, Özer E, Tufan E, Kumral A, Özkul Y. Epigenetic Programming Through Breast Milk and Its Impact on Milk-Siblings Mating. Front Genet 2020; 11:569232. [PMID: 33133155 PMCID: PMC7565666 DOI: 10.3389/fgene.2020.569232] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Abstract
Background The epigenetic effects of transmission of certain regulatory molecules, such as miRNAs, through maternal milk on future generations, are still unknown and have not been fully understood yet. We hypothesized that breastfeeding regularly by adoptive-mother may cause transmission of miRNAs as epigenetic regulating factors to the infant, and the marriage of milk-siblings may cause various pathologies in the future generations. Results A cross-fostering model using a/a and Avy/a mice had been established. F2 milk-sibling and F2 control groups were obtained from mating of milk-siblings or unrelated mice. Randomized selected animals in the both F2 groups were sacrificed for miRNA expression studies and the remainings were followed for phenotypic changes (coat color, obesity, hyperglycemia, liver pathology, and life span). The lifespan in the F2 milk-sibling group was shorter than the control group (387 vs 590 days, p = 0.011) and they were more obese during the aging period. Histopathological examination of liver tissues revealed abnormal findings in F2 milk-sibling group. In order to understand the epigenetic mechanisms leading to these phenotypic changes, we analyzed miRNA expression differences between offspring of milk-sibling and control matings and focused on the signaling pathways regulating lifespan and metabolism. Bioinformatic analysis demonstrated that differentially expressed miRNAs were associated with pathways regulating metabolism, survival, and cancer development such as the PI3K-Akt, ErbB, mTOR, and MAPK, insulin signaling pathways. We further analyzed the expression patterns of miR-186-5p, miR-141-3p, miR-345-5p, and miR-34c-5p and their candidate target genes Mapk8, Gsk3b, and Ppargc1a in ovarian and liver tissues. Conclusion Our findings support for the first time that the factors modifying the epigenetic mechanisms may be transmitted by breast milk and these epigenetic interactions may be transferred transgenerationally. Results also suggested hereditary epigenetic effects of cross-fostering on future generations and the impact of mother-infant dyad on epigenetic programming.
Collapse
Affiliation(s)
- Hasan Ozkan
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Funda Tuzun
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Serpil Taheri
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.,Erciyes University's Betul-Ziya Eren Genome and Stem Cell (Genkok), Kayseri, Turkey
| | - Peyda Korhan
- İzmir Biomedicine and Genome Center, Dokuz Eylul University, İzmir, Turkey
| | - Pınar Akokay
- Department of Medical Laboratory Techniques, İzmir Kavram University, İzmir, Turkey
| | - Osman Yılmaz
- The Experimental Animal Laboratory, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Nuray Duman
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Erdener Özer
- Department of Pathology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Esra Tufan
- Erciyes University's Betul-Ziya Eren Genome and Stem Cell (Genkok), Kayseri, Turkey
| | - Abdullah Kumral
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Yusuf Özkul
- Erciyes University's Betul-Ziya Eren Genome and Stem Cell (Genkok), Kayseri, Turkey.,Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
4
|
Oleskin AV, Shenderov BA. Probiotics and Psychobiotics: the Role of Microbial Neurochemicals. Probiotics Antimicrob Proteins 2020; 11:1071-1085. [PMID: 31493127 DOI: 10.1007/s12602-019-09583-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In light of recent data, microorganisms should be construed as organisms that are capable of communication and collective behaviors. Microbial communication signals are involved both in interactions among microbial cells within microbial social systems, including the human body-inhabiting microconsortium, and the dialog between the microbiota and the host organism. The microbiota inhabits various niches of the host organism, especially the gastrointestinal (GI) tract. Microorganisms release diverse signal molecules and, in addition, specifically respond to host signals. This enables them to constantly interact with the nervous system including the brain and the immune system of the host organism. Evolutionarily conserved signals that are involved in the communication between microbiota and the host include neuroactive substances (neurochemicals) such as peptides, amino acids, biogenic amines, short-chain fatty acids, and gaseous substances. This ongoing dialog may either stabilize the host's physical and mental health state or, alternatively, cause serious health problems. Attempts are made to correct imbalances in the brain-gut-microbiota axis with probiotics including their subgroup called psychobiotics that release neuroactive substances directly influencing the human brain, psyche, and behavior. A number of recent review works address the microbiota-host system and its communication signals. Some of the publications focus on the involvement of neurochemicals in the bidirectional communication within the host-microbiota system. However, this work concentrates on the impact of bacterial cell components, metabolites, and signal molecules as promising alternatives to the currently widespread probiotics that have both advantages and disadvantages. Such biologically active agents of microbial origin are referred to as postbiotics or, alternatively, metabiotics (the term preferred in this work).
Collapse
Affiliation(s)
- Alexander V Oleskin
- General Ecology Department, School of Biology, Moscow State University, Vorobiev Hills, Moscow, Russia, 119991.
| | - Boris A Shenderov
- Laboratory of Bacteriology and Parasitology, Centre for Strategic Planning, Russian Ministry of Health; Moscow, Research Laboratory for Design & Implementation of Personalized Nutrition-Related Products & Diets, K. G. Razumovsky University of Technology & Management, Moscow, Russia
| |
Collapse
|
5
|
Lobzhanidze G, Japaridze N, Lordkipanidze T, Rzayev F, MacFabe D, Zhvania M. Behavioural and brain ultrastructural changes following the systemic administration of propionic acid in adolescent male rats. Further development of a rodent model of autism. Int J Dev Neurosci 2020; 80:139-156. [PMID: 31997401 DOI: 10.1002/jdn.10011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/08/2020] [Accepted: 01/19/2020] [Indexed: 12/18/2022] Open
Abstract
Short chain fatty acids, produced as gut microbiome metabolites but also present in the diet, exert broad effects in host physiology. Propionic acid (PPA), along with butyrate and acetate, plays a growing role in health, but also in neurological conditions. Increased PPA exposure in humans, animal models and cell lines elicit diverse behavioural and biochemical changes consistent with organic acidurias, mitochondrial disorders and autism spectrum disorders (ASD). ASD is considered a disorder of synaptic dysfunction and cell signalling, but also neuroinflammatory and neurometabolic components. We examined behaviour (Morris water and radial arm mazes) and the ultrastructure of the hippocampus and medial prefrontal cortex (electron microscopy) following a single intraperitoneal (i.p.) injection of PPA (175 mg/kg) in male adolescent rats. PPA treatment showed altered social and locomotor behaviour without changes in learning and memory. Both transient and enduring ultrastructural alterations in synapses, astro- and microglia were detected in the CA1 hippocampal area. Electron microscopic analysis showed the PPA treatment significantly decreased the total number of synaptic vesicles, presynaptic mitochondria and synapses with a symmetric active zone. Thus, brief systemic administration of this dietary and enteric short chain fatty acid produced behavioural and dynamic brain ultrastructural changes, providing further validation of the PPA model of ASD.
Collapse
Affiliation(s)
- Giorgi Lobzhanidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia.,Medical School, New Vision University, Tbilisi, Georgia
| | - Tamar Lordkipanidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Fuad Rzayev
- Laboratory of Electron Microscopy, Research Center of Azerbaijan Medical University, Baku, Azerbaijan
| | - Derrick MacFabe
- The Kilee Patchell-Evans Autism Research Group, London, ON, Canada.,Faculty of Medicine, Department of Microbiology, Center for Healthy Eating and Food Innovation, Maastricht University, Maastricht, the Netherlands
| | - Mzia Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| |
Collapse
|
6
|
Devaux CA, Raoult D. The Microbiological Memory, an Epigenetic Regulator Governing the Balance Between Good Health and Metabolic Disorders. Front Microbiol 2018; 9:1379. [PMID: 29997595 PMCID: PMC6028609 DOI: 10.3389/fmicb.2018.01379] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022] Open
Abstract
If the transmission of biological information from one generation to the next is based on DNA, most heritable phenotypic traits such as chronic metabolic diseases, are not linked to genetic variation in DNA sequences. Non-genetic heritability might have several causes including epigenetic, parental effect, adaptive social learning, and influence of the ecological environment. Distinguishing among these causes is crucial to resolve major phenotypic enigmas. Strong evidence indicates that changes in DNA expression through various epigenetic mechanisms can be linked to parent-offspring resemblance in terms of sensitivity to metabolic diseases. Among non-genetic heritable traits, early nutrition could account for a long term deviant programming of genes expression responsible for metabolic diseases in adulthood. Nutrition could shape an inadequate gut microbiota (dysbiosis), triggering epigenetic deregulation of transcription which can be observed in chronic metabolic diseases. We review herein the evidence that dysbiosis might be a major cause of heritable epigenetic patterns found to be associated with metabolic diseases. By taking into account the recent advances on the gut microbiome, we have aggregated together different observations supporting the hypothesis that the gut microbiota could promote the molecular crosstalk between bacteria and surrounding host cells which controls the pathological epigenetic signature. We introduce for the first time the concept of "microbiological memory" as the main regulator of the epigenetic signatures, thereby indicating that different causes of non-genetic heritability can interact in complex pathways to produce inheritance.
Collapse
Affiliation(s)
- Christian A. Devaux
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix-Marseille University, Marseille, France
- Centre National de la Recherche Scientifique, Marseille, France
| | - Didier Raoult
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix-Marseille University, Marseille, France
| |
Collapse
|
7
|
Wang HD, Liu L, Zhao HR, Hou QF, Yan JB, Shi WL, Guo QN, Wang L, Liao SX, Zhu BF. Detection of fetal epigenetic biomarkers through genome-wide DNA methylation study for non-invasive prenatal diagnosis. Mol Med Rep 2017; 15:3989-3998. [PMID: 28440505 PMCID: PMC5436219 DOI: 10.3892/mmr.2017.6506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/13/2017] [Indexed: 12/20/2022] Open
Abstract
The discovery of cell-free DNA fetal (cff DNA) in maternal plasma during pregnancy provides a novel perspective for the development of non-invasive prenatal diagnosis (NIPD). Against the background of maternal DNA, the use of the relatively low concentration of cff DNA is limited in NIPD. Therefore, in order to overcome the complication of the background of maternal DNA and expand the scope of cff DNA application in clinical practice, it is necessary to identify novel universal fetal-specific DNA markers. The GeneChip Human Promoter 1.0R Array set was used in the present study to analyze the methylation status of 12 placental tissue and maternal peripheral blood whole-genome DNA samples. In total, 5 fetus differential hypermethylation regions and 6 fetus differential hypomethylation regions were identified. In order to verify the 11 selected methylation regions and detect the differential CpG sites in these regions, a bisulfate direct sequencing strategy was used. In total, 87 fetal differential methylation CpG sites were identified from 123 CpG sites. The detection of fetal differential methylation DNA regions and CpG sites may be instrumental in the development of efficient NIPD and in the expansion of its application in other disorders.
Collapse
Affiliation(s)
- Hong-Dan Wang
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Lin Liu
- Department of Cardiovascular Ultrasound, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Hui-Ru Zhao
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Qiao-Fang Hou
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jing-Bin Yan
- Shanghai Children's Hospital, Shanghai Institute of Medical Genetics, Key Laboratory of Embryo Molecular Biology, Ministry of Health of China and Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, P.R. China
| | - Wei-Li Shi
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Qian-Nan Guo
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Li Wang
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Shi-Xiu Liao
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Bo-Feng Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
8
|
Mardo K, Visnapuu T, Vija H, Aasamets A, Viigand K, Alamäe T. A Highly Active Endo-Levanase BT1760 of a Dominant Mammalian Gut Commensal Bacteroides thetaiotaomicron Cleaves Not Only Various Bacterial Levans, but Also Levan of Timothy Grass. PLoS One 2017; 12:e0169989. [PMID: 28103254 PMCID: PMC5245892 DOI: 10.1371/journal.pone.0169989] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/27/2016] [Indexed: 12/11/2022] Open
Abstract
Bacteroides thetaiotaomicron, an abundant commensal of the human gut, degrades numerous complex carbohydrates. Recently, it was reported to grow on a β-2,6-linked polyfructan levan produced by Zymomonas mobilis degrading the polymer into fructooligosaccharides (FOS) with a cell surface bound endo-levanase BT1760. The FOS are consumed by B. thetaiotaomicron, but also by other gut bacteria, including health-promoting bifidobacteria and lactobacilli. Here we characterize biochemical properties of BT1760, including the activity of BT1760 on six bacterial levans synthesized by the levansucrase Lsc3 of Pseudomonas syringae pv. tomato, its mutant Asp300Asn, levansucrases of Zymomonas mobilis, Erwinia herbicola, Halomonas smyrnensis as well as on levan isolated from timothy grass. For the first time a plant levan is shown as a perfect substrate for an endo-fructanase of a human gut bacterium. BT1760 degraded levans to FOS with degree of polymerization from 2 to 13. At optimal reaction conditions up to 1 g of FOS were produced per 1 mg of BT1760 protein. Low molecular weight (<60 kDa) levans, including timothy grass levan and levan synthesized from sucrose by the Lsc3Asp300Asn, were degraded most rapidly whilst levan produced by Lsc3 from raffinose least rapidly. BT1760 catalyzed finely at human body temperature (37°C) and in moderately acidic environment (pH 5–6) that is typical for the gut lumen. According to differential scanning fluorimetry, the Tm of the endo-levanase was 51.5°C. All tested levans were sufficiently stable in acidic conditions (pH 2.0) simulating the gastric environment. Therefore, levans of both bacterial and plant origin may serve as a prebiotic fiber for B. thetaiotaomicron and contribute to short-chain fatty acids synthesis by gut microbiota. In the genome of Bacteroides xylanisolvens of human origin a putative levan degradation locus was disclosed.
Collapse
Affiliation(s)
- Karin Mardo
- Department of Genetics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Triinu Visnapuu
- Department of Genetics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heiki Vija
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Anneli Aasamets
- Department of Genetics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Katrin Viigand
- Department of Genetics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Tiina Alamäe
- Department of Genetics, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- * E-mail:
| |
Collapse
|
9
|
Davydov BN, Samoukina AM, Mikhailova ES, Gavrilova OA, Alekseeva YA. [Variations of oral fluid microbiota in healthy children and adolescents]. STOMATOLOGIIA 2017; 96:56-59. [PMID: 28317832 DOI: 10.17116/stomat201796156-59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The aim of the study was to establish the bacterial and viral associations of saliva and the immune resistance in 127 healthy people of different ages. The analysis sorted out three variants of oral fluid microbiota differing by certain combination of indigenous and facultative microflora. It was found that with age there is a significant increase in the number of adolescents with the third variant of the microbiota, characterized by a decrease in the number of indigenous microflora and increase in opportunistic microorganisms and viruses. With an increase in microecological shifts a tendency to decrease the level of lysozyme, an increase in the number of secretory IgA, accompanied by a decrease in antibody was revealed. It has been shown that bacterial and viral component of the microbiota of the oral fluid are in a dynamic relationship with each other, as well as immune resistance and can serve as an indicator of the health level and the selection criteria at the stages of clinical examination of children and adolescents, with the risk of dental disease.
Collapse
|
10
|
Slattery J, MacFabe DF, Frye RE. The Significance of the Enteric Microbiome on the Development of Childhood Disease: A Review of Prebiotic and Probiotic Therapies in Disorders of Childhood. Clin Med Insights Pediatr 2016; 10:91-107. [PMID: 27774001 PMCID: PMC5063840 DOI: 10.4137/cmped.s38338] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 02/07/2023] Open
Abstract
Recent studies have highlighted the fact that the enteric microbiome, the trillions of microbes that inhabit the human digestive tract, has a significant effect on health and disease. Methods for manipulating the enteric microbiome, particularly through probiotics and microbial ecosystem transplantation, have undergone some study in clinical trials. We review some of the evidence for microbiome alteration in relation to childhood disease and discuss the clinical trials that have examined the manipulation of the microbiome in an effort to prevent or treat childhood disease with a primary focus on probiotics, prebiotics, and/or synbiotics (ie, probiotics + prebiotics). Studies show that alterations in the microbiome may be a consequence of events occurring during infancy and/or childhood such as prematurity, C-sections, and nosocomial infections. In addition, certain childhood diseases have been associated with microbiome alterations, namely necrotizing enterocolitis, infantile colic, asthma, atopic disease, gastrointestinal disease, diabetes, malnutrition, mood/anxiety disorders, and autism spectrum disorders. Treatment studies suggest that probiotics are potentially protective against the development of some of these diseases. Timing and duration of treatment, the optimal probiotic strain(s), and factors that may alter the composition and function of the microbiome are still in need of further research. Other treatments such as prebiotics, fecal microbial transplantation, and antibiotics have limited evidence. Future translational work, in vitro models, long-term and follow-up studies, and guidelines for the composition and viability of probiotic and microbial therapies need to be developed. Overall, there is promising evidence that manipulating the microbiome with probiotics early in life can help prevent or reduce the severity of some childhood diseases, but further research is needed to elucidate biological mechanisms and determine optimal treatments.
Collapse
Affiliation(s)
- John Slattery
- Arkansas Children’s Research Institute, Little Rock, AR, USA
- Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Derrick F. MacFabe
- The Kilee Patchell-Evans Autism Research Group, Departments of Psychology (Neuroscience) and Psychiatry, Division of Developmental Disabilities, University of Western Ontario, London, ON, Canada
| | - Richard E. Frye
- Arkansas Children’s Research Institute, Little Rock, AR, USA
- Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
11
|
Frye RE, Rose S, Chacko J, Wynne R, Bennuri SC, Slattery JC, Tippett M, Delhey L, Melnyk S, Kahler SG, MacFabe DF. Modulation of mitochondrial function by the microbiome metabolite propionic acid in autism and control cell lines. Transl Psychiatry 2016; 6:e927. [PMID: 27779624 PMCID: PMC5290345 DOI: 10.1038/tp.2016.189] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/26/2016] [Accepted: 08/02/2016] [Indexed: 12/12/2022] Open
Abstract
Propionic acid (PPA) is a ubiquitous short-chain fatty acid, which is a major fermentation product of the enteric microbiome. PPA is a normal intermediate of metabolism and is found in foods, either naturally or as a preservative. PPA and its derivatives have been implicated in both health and disease. Whereas PPA is an energy substrate and has many proposed beneficial effects, it is also associated with human disorders involving mitochondrial dysfunction, including propionic acidemia and autism spectrum disorders (ASDs). We aimed to investigate the dichotomy between the health and disease effects of PPA by measuring mitochondrial function in ASD and age- and gender-matched control lymphoblastoid cell lines (LCLs) following incubation with PPA at several concentrations and durations both with and without an in vitro increase in reactive oxygen species (ROS). Mitochondrial function was optimally increased at particular exposure durations and concentrations of PPA with ASD LCLs, demonstrating a greater enhancement. In contrast, increasing ROS negated the positive PPA effect with the ASD LCLs, showing a greater detriment. These data demonstrate that enteric microbiome metabolites such as PPA can have both beneficial and toxic effects on mitochondrial function, depending on concentration, exposure duration and microenvironment redox state with these effects amplified in LCLs derived from individuals with ASD. As PPA, as well as enteric bacteria, which produce PPA, have been implicated in a wide variety of diseases, including ASD, diabetes, obesity and inflammatory diseases, insight into this metabolic modulator from the host microbiome may have wide applications for both health and disease.
Collapse
Affiliation(s)
- R E Frye
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA,Arkansas Children's Research Institute, Slot 512-41B, 13 Children's Way, Little Rock, AR 72202, USA. E-mail:
| | - S Rose
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - J Chacko
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - R Wynne
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - S C Bennuri
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - J C Slattery
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - M Tippett
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - L Delhey
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - S Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - S G Kahler
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - D F MacFabe
- Kilee Patchell-Evans Autism Research Group, Division of Developmental Disabilities, Department of Psychology/Psychiatry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
12
|
Oleskin AV, Shenderov BA. Neuromodulatory effects and targets of the SCFAs and gasotransmitters produced by the human symbiotic microbiota. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2016; 27:30971. [PMID: 27389418 PMCID: PMC4937721 DOI: 10.3402/mehd.v27.30971] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/10/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022]
Abstract
The symbiotic gut microbiota plays an important role in the development and homeostasis of the host organism. Its physiological, biochemical, behavioral, and communicative effects are mediated by multiple low molecular weight compounds. Recent data on small molecules produced by gut microbiota in mammalian organisms demonstrate the paramount importance of these biologically active molecules in terms of biology and medicine. Many of these molecules are pleiotropic mediators exerting effects on various tissues and organs. This review is focused on the functional roles of gaseous molecules that perform neuromediator and/or endocrine functions. The molecular mechanisms that underlie the effects of microbial fermentation-derived gaseous metabolites are not well understood. It is possible that these metabolites produce their effects via immunological, biochemical, and neuroendocrine mechanisms that involve endogenous and microbial modulators and transmitters; of considerable importance are also changes in epigenetic transcriptional factors, protein post-translational modification, lipid and mitochondrial metabolism, redox signaling, and ion channel/gap junction/transporter regulation. Recent findings have revealed that interactivity among such modulators/transmitters is a prerequisite for the ongoing dialog between microbial cells and host cells, including neurons. Using simple reliable methods for the detection and measurement of short-chain fatty acids (SCFAs) and small gaseous molecules in eukaryotic tissues and prokaryotic cells, selective inhibitors of enzymes that participate in their synthesis, as well as safe chemical and microbial donors of pleiotropic mediators and modulators of host intestinal microbial ecology, should enable us to apply these chemicals as novel therapeutics and medical research tools.
Collapse
Affiliation(s)
- Alexander V Oleskin
- General Ecology Department, Biology Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Boris A Shenderov
- Moscow Research Institute of Epidemiology and Microbiology after G.N. Gabrichevsky, Moscow, Russia; ;
| |
Collapse
|
13
|
Frye RE, Rossignol DA. Identification and Treatment of Pathophysiological Comorbidities of Autism Spectrum Disorder to Achieve Optimal Outcomes. CLINICAL MEDICINE INSIGHTS-PEDIATRICS 2016; 10:43-56. [PMID: 27330338 PMCID: PMC4910649 DOI: 10.4137/cmped.s38337] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/15/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023]
Abstract
Despite the fact that the prevalence of autism spectrum disorder (ASD) continues to rise, no effective medical treatments have become standard of care. In this paper we review some of the pathophysiological abnormalities associated with ASD and their potential associated treatments. Overall, there is evidence for some children with ASD being affected by seizure and epilepsy, neurotransmitter dysfunction, sleep disorders, metabolic abnormalities, including abnormalities in folate, cobalamin, tetrahydrobiopterin, carnitine, redox and mitochondrial metabolism, and immune and gastrointestinal disorders. Although evidence for an association between these pathophysiological abnormalities and ASD exists, the exact relationship to the etiology of ASD and its associated symptoms remains to be further defined in many cases. Despite these limitations, treatments targeting some of these pathophysiological abnormalities have been studied in some cases with high-quality studies, whereas treatments for other pathophysiological abnormalities have not been well studied in many cases. There are some areas of more promising treatments specific for ASD including neurotransmitter abnormalities, particularly imbalances in glutamate and acetylcholine, sleep onset disorder (with behavioral therapy and melatonin), and metabolic abnormalities in folate, cobalamin, tetrahydrobiopterin, carnitine, and redox pathways. There is some evidence for treatments of epilepsy and seizures, mitochondrial and immune disorders, and gastrointestinal abnormalities, particularly imbalances in the enteric microbiome, but further clinical studies are needed in these areas to better define treatments specific to children with ASD. Clearly, there are some promising areas of ASD research that could lead to novel treatments that could become standard of care in the future, but more research is needed to better define subgroups of children with ASD who are affected by specific pathophysiological abnormalities and the optimal treatments for these abnormalities.
Collapse
Affiliation(s)
- Richard E Frye
- Arkansas Children's Research Institute, Little Rock, AR, USA.; Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | |
Collapse
|
14
|
El Aidy S, Stilling R, Dinan TG, Cryan JF. Microbiome to Brain: Unravelling the Multidirectional Axes of Communication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:301-36. [PMID: 26589226 DOI: 10.1007/978-3-319-20215-0_15] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gut microbiome plays a crucial role in host physiology. Disruption of its community structure and function can have wide-ranging effects making it critical to understand exactly how the interactive dialogue between the host and its microbiota is regulated to maintain homeostasis. An array of multidirectional signalling molecules is clearly involved in the host-microbiome communication. This interactive signalling not only impacts the gastrointestinal tract, where the majority of microbiota resides, but also extends to affect other host systems including the brain and liver as well as the microbiome itself. Understanding the mechanistic principles of this inter-kingdom signalling is fundamental to unravelling how our supraorganism function to maintain wellbeing, subsequently opening up new avenues for microbiome manipulation to favour desirable mental health outcome.
Collapse
Affiliation(s)
- Sahar El Aidy
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Roman Stilling
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.,Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland. .,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
15
|
Moos WH, Faller DV, Harpp DN, Kanara I, Pernokas J, Powers WR, Steliou K. Microbiota and Neurological Disorders: A Gut Feeling. Biores Open Access 2016; 5:137-45. [PMID: 27274912 PMCID: PMC4892191 DOI: 10.1089/biores.2016.0010] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the past century, noncommunicable diseases have surpassed infectious diseases as the principal cause of sickness and death, worldwide. Trillions of commensal microbes live in and on our body, and constitute the human microbiome. The vast majority of these microorganisms are maternally derived and live in the gut, where they perform functions essential to our health and survival, including: digesting food, activating certain drugs, producing short-chain fatty acids (which help to modulate gene expression by inhibiting the deacetylation of histone proteins), generating anti-inflammatory substances, and playing a fundamental role in the induction, training, and function of our immune system. Among the many roles the microbiome ultimately plays, it mitigates against untoward effects from our exposure to the environment by forming a biotic shield between us and the outside world. The importance of physical activity coupled with a balanced and healthy diet in the maintenance of our well-being has been recognized since antiquity. However, it is only recently that characterization of the host-microbiome intermetabolic and crosstalk pathways has come to the forefront in studying therapeutic design. As reviewed in this report, synthetic biology shows potential in developing microorganisms for correcting pathogenic dysbiosis (gut microbiota-host maladaptation), although this has yet to be proven. However, the development and use of small molecule drugs have a long and successful history in the clinic, with small molecule histone deacetylase inhibitors representing one relevant example already approved to treat cancer and other disorders. Moreover, preclinical research suggests that epigenetic treatment of neurological conditions holds significant promise. With the mouth being an extension of the digestive tract, it presents a readily accessible diagnostic site for the early detection of potential unhealthy pathogens resident in the gut. Taken together, the data outlined herein provide an encouraging roadmap toward important new medicines and companion diagnostic platforms in a wide range of therapeutic indications.
Collapse
Affiliation(s)
- Walter H. Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, California
- Address correspondence to: Walter H. Moos, PhD, Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, 600 16th Street, Mail Code 2280, Genentech Hall S512D, Mission Bay Campus, San Francisco, CA 94158, E-mail: , ; or Kosta Steliou, PhD, PhenoMatriX, Inc., 9 Hawthorne Place Suite 4R, Boston, MA 02114, E-mail: ,
| | - Douglas V. Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
| | - David N. Harpp
- Department of Chemistry, McGill University, Montreal, Canada
| | - Iphigenia Kanara
- Weatherhead Center for International Affairs, Harvard University, Cambridge, Massachusetts
- Consulate General of Greece in Boston, Boston, Massachusetts
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Whitney R. Powers
- Department of Health Sciences, Boston University, Boston, Massachusetts
- Department of Anatomy, Boston University School of Medicine, Boston, Massachusetts
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
- PhenoMatriX, Inc., Boston, Massachusetts
- Address correspondence to: Walter H. Moos, PhD, Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, 600 16th Street, Mail Code 2280, Genentech Hall S512D, Mission Bay Campus, San Francisco, CA 94158, E-mail: , ; or Kosta Steliou, PhD, PhenoMatriX, Inc., 9 Hawthorne Place Suite 4R, Boston, MA 02114, E-mail: ,
| |
Collapse
|
16
|
Langley G, Austin CP, Balapure AK, Birnbaum LS, Bucher JR, Fentem J, Fitzpatrick SC, Fowle JR, Kavlock RJ, Kitano H, Lidbury BA, Muotri AR, Peng SQ, Sakharov D, Seidle T, Trez T, Tonevitsky A, van de Stolpe A, Whelan M, Willett C. Lessons from Toxicology: Developing a 21st-Century Paradigm for Medical Research. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:A268-72. [PMID: 26523530 PMCID: PMC4629751 DOI: 10.1289/ehp.1510345] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Biomedical developments in the 21st century provide an unprecedented opportunity to gain a dynamic systems-level and human-specific understanding of the causes and pathophysiologies of disease. This understanding is a vital need, in view of continuing failures in health research, drug discovery, and clinical translation. The full potential of advanced approaches may not be achieved within a 20th-century conceptual framework dominated by animal models. Novel technologies are being integrated into environmental health research and are also applicable to disease research, but these advances need a new medical research and drug discovery paradigm to gain maximal benefits. We suggest a new conceptual framework that repurposes the 21st-century transition underway in toxicology. Human disease should be conceived as resulting from integrated extrinsic and intrinsic causes, with research focused on modern human-specific models to understand disease pathways at multiple biological levels that are analogous to adverse outcome pathways in toxicology. Systems biology tools should be used to integrate and interpret data about disease causation and pathophysiology. Such an approach promises progress in overcoming the current roadblocks to understanding human disease and successful drug discovery and translation. A discourse should begin now to identify and consider the many challenges and questions that need to be solved.
Collapse
Affiliation(s)
- Gill Langley
- Research and Toxicology Department, Humane Society International, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Paul B, Barnes S, Demark-Wahnefried W, Morrow C, Salvador C, Skibola C, Tollefsbol TO. Influences of diet and the gut microbiome on epigenetic modulation in cancer and other diseases. Clin Epigenetics 2015; 7:112. [PMID: 26478753 PMCID: PMC4609101 DOI: 10.1186/s13148-015-0144-7] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/22/2015] [Indexed: 02/06/2023] Open
Abstract
Epigenetic modulation of gene activity occurs in response to non-genetic factors such as body weight status, physical activity, dietary factors, and environmental toxins. In addition, each of these factors is thought to affect and be affected by the gut microbiome. A primary mechanism that links these various factors together in mediating control of gene expression is the production of metabolites that serve as critical cofactors and allosteric regulators of epigenetic processes. Here, we review the involvement of the gut microbiota and its interactions with dietary factors, many of which have known cellular bioactivity, focusing on particular epigenetic processes affected and the influence they have on human health and disease, particularly cancer and response to treatment. Advances in DNA sequencing have expanded the capacity for studying the microbiome. Combining this with rapidly improving techniques to measure the metabolome provides opportunities to understand complex relationships that may underlie the development and progression of cancer as well as treatment-related sequelae. Given broad reaching and fundamental biology, both at the cellular and organismal levels, we propose that interactive research programs, which utilize a wide range of mutually informative experimental model systems—each one optimally suited for answering particular questions—provide the best path forward for breaking ground on new knowledge and ultimately understanding the epigenetic significance of the gut microbiome and its response to dietary factors in cancer prevention and therapy.
Collapse
Affiliation(s)
- Bidisha Paul
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170 USA
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL USA ; Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL USA
| | - Wendy Demark-Wahnefried
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL USA ; Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL USA
| | - Casey Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL USA
| | - Carolina Salvador
- Division of Medical Oncology/Hematology, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL USA
| | - Christine Skibola
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL USA ; Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170 USA ; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL USA ; Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL USA ; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
18
|
Zakharevich NV, Averina OV, Klimina KM, Kudryavtseva AV, Kasianov AS, Makeev VJ, Danilenko VN. Complete Genome Sequence of Bifidobacterium longum GT15: Identification and Characterization of Unique and Global Regulatory Genes. MICROBIAL ECOLOGY 2015; 70:819-834. [PMID: 25894918 DOI: 10.1007/s00248-015-0603-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 03/23/2015] [Indexed: 06/04/2023]
Abstract
In this study, we report the first completely annotated genome sequence of the Russia origin Bifidobacterium longum subsp. longum strain GT15. Comparative genomic analysis of this genome with other available completely annotated genome sequences of B. longum strains isolated from other countries has revealed a high degree of conservation and synteny across the entire genomes. However, it was discovered that the open reading frames to 35 genes were detected only from the B. longum GT15 genome and absent from other genomes B. longum strains (not of Russian origin). These so-called unique genes (UGs) represent a total length of 39,066 bp, with G + C content ranging from 37 to 65 %. Interestingly, certain genes were detected in other B. longum strains of Russian origin. In our analysis, we examined genes for global regulatory systems: proteins of toxin-antitoxin (TA) systems type II, serine/threonine protein kinases (STPKs) of eukaryotic type, and genes of the WhiB-like family proteins. In addition, we have made in silico analysis of all the most significant probiotic genes and considered genes involved in epigenetic regulation and genes responsible for producing various neuromediators. This genome sequence may elucidate the biology of this probiotic strain as a promising candidate for practical (pharmaceutical) applications.
Collapse
Affiliation(s)
| | - Olga V Averina
- Vavilov Institute of General Genetics, Gubkina str. 3, 119991, Moscow, Russia
| | - Ksenia M Klimina
- Vavilov Institute of General Genetics, Gubkina str. 3, 119991, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Vavilova str. 32, 119991, Moscow, Russia
| | - Artem S Kasianov
- Vavilov Institute of General Genetics, Gubkina str. 3, 119991, Moscow, Russia
| | - Vsevolod J Makeev
- Vavilov Institute of General Genetics, Gubkina str. 3, 119991, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Vavilova str. 32, 119991, Moscow, Russia
| | - Valery N Danilenko
- Vavilov Institute of General Genetics, Gubkina str. 3, 119991, Moscow, Russia
| |
Collapse
|
19
|
Boyce WT, Kobor MS. Development and the epigenome: the 'synapse' of gene-environment interplay. Dev Sci 2015; 18:1-23. [PMID: 25546559 DOI: 10.1111/desc.12282] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/14/2014] [Indexed: 12/21/2022]
Abstract
This paper argues that there is a revolution afoot in the developmental science of gene-environment interplay. We summarize, for an audience of developmental researchers and clinicians, how epigenetic processes - chromatin structural modifications that regulate gene expression without changing DNA sequences - may offer a strong, parsimonious account for the convergence of genetic and contextual variation in the genesis of adaptive and maladaptive development. Epigenetic processes may play a plausible explanatory role in understanding: divergent trajectories and sexual dimorphisms in brain development; statistical interactions between genes and environments; the biological embedding of early psychosocial adversities; the linkages of such adversities to disorders of mental health; the striking individual variation in the strength of those linkages; the molecular origins of critical and sensitive periods; and the transgenerational inheritance of risk and protection. Taken together, these arguments converge in a claim that epigenetic processes constitute a promising and illuminating point of connection - a 'synapse' - between genes and environments.
Collapse
Affiliation(s)
- W Thomas Boyce
- Departments of Pediatrics and Psychiatry, University of California, San Francisco, USA; Child and Brain Development Program, Canadian Institute for Advanced Research, Canada
| | | |
Collapse
|
20
|
Frye RE, Slattery J, MacFabe DF, Allen-Vercoe E, Parker W, Rodakis J, Adams JB, Krajmalnik-Brown R, Bolte E, Kahler S, Jennings J, James J, Cerniglia CE, Midtvedt T. Approaches to studying and manipulating the enteric microbiome to improve autism symptoms. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2015; 26:26878. [PMID: 25956237 PMCID: PMC4425814 DOI: 10.3402/mehd.v26.26878] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/05/2015] [Accepted: 04/06/2015] [Indexed: 02/07/2023]
Abstract
There is a growing body of scientific evidence that the health of the microbiome (the trillions of microbes that inhabit the human host) plays an important role in maintaining the health of the host and that disruptions in the microbiome may play a role in certain disease processes. An increasing number of research studies have provided evidence that the composition of the gut (enteric) microbiome (GM) in at least a subset of individuals with autism spectrum disorder (ASD) deviates from what is usually observed in typically developing individuals. There are several lines of research that suggest that specific changes in the GM could be causative or highly associated with driving core and associated ASD symptoms, pathology, and comorbidities which include gastrointestinal symptoms, although it is also a possibility that these changes, in whole or in part, could be a consequence of underlying pathophysiological features associated with ASD. However, if the GM truly plays a causative role in ASD, then the manipulation of the GM could potentially be leveraged as a therapeutic approach to improve ASD symptoms and/or comorbidities, including gastrointestinal symptoms. One approach to investigating this possibility in greater detail includes a highly controlled clinical trial in which the GM is systematically manipulated to determine its significance in individuals with ASD. To outline the important issues that would be required to design such a study, a group of clinicians, research scientists, and parents of children with ASD participated in an interdisciplinary daylong workshop as an extension of the 1st International Symposium on the Microbiome in Health and Disease with a Special Focus on Autism (www.microbiome-autism.com). The group considered several aspects of designing clinical studies, including clinical trial design, treatments that could potentially be used in a clinical trial, appropriate ASD participants for the clinical trial, behavioral and cognitive assessments, important biomarkers, safety concerns, and ethical considerations. Overall, the group not only felt that this was a promising area of research for the ASD population and a promising avenue for potential treatment but also felt that further basic and translational research was needed to clarify the clinical utility of such treatments and to elucidate possible mechanisms responsible for a clinical response, so that new treatments and approaches may be discovered and/or fostered in the future.
Collapse
Affiliation(s)
- Richard E Frye
- Division of Neurology, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA;
| | - John Slattery
- Division of Neurology, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Derrick F MacFabe
- Department of Psychology and Psychiatry, Western University, London, ON, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | - John Rodakis
- N of One: Autism Research Foundation, Dallas, TX, USA
| | - James B Adams
- School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA
| | - Rosa Krajmalnik-Brown
- Swette Center for Environmental Biotechnology, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Ellen Bolte
- N of One: Autism Research Foundation, Dallas, TX, USA
| | - Stephen Kahler
- Division of Neurology, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Jill James
- Department of Developmental Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | | |
Collapse
|
21
|
Chango A, Pogribny IP. Considering maternal dietary modulators for epigenetic regulation and programming of the fetal epigenome. Nutrients 2015; 7:2748-70. [PMID: 25875118 PMCID: PMC4425171 DOI: 10.3390/nu7042748] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/16/2015] [Accepted: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Fetal life is characterized by a tremendous plasticity and ability to respond to various environmental and lifestyle factors, including maternal nutrition. Identification of the role of dietary factors that can modulate and reshape the cellular epigenome during development, including methyl group donors (e.g., folate, choline) and bioactive compounds (e.g., polyphenols) is of great importance; however, there is insufficient knowledge of a particular effect of each type of modulator and/or their combination on fetal life. To enhance the quality and safety of food products for proper fetal health and disease prevention in later life, a better understanding of the underlying mechanisms of dietary epigenetic modulators during the critical prenatal period is necessary. This review focuses on the influence of maternal dietary components on DNA methylation, histone modification, and microRNAs, and summarizes current knowledge of the effect and importance of dietary components on epigenetic mechanisms that control the proper expression of genetic information. Evidence reveals that some components in the maternal diet can directly or indirectly affect epigenetic mechanisms. Understanding the underlying mechanisms of how early-life nutritional environment affects the epigenome during development is of great importance for the successful prevention of adult chronic diseases through optimal maternal nutrition.
Collapse
Affiliation(s)
- Abalo Chango
- Polytechnic Institute LaSalle Beauvais, Department of Nutrition and Health Sciences, EGEAL UP:2012.10.101, F-60026 Beauvais Cedex, France.
| | - Igor P Pogribny
- Division of Biochemical Toxicology, Food and Drug Administration National Center for Toxicological Research, Jefferson, AR 72079, USA.
| |
Collapse
|
22
|
Dietert RR, Dietert JM. The Microbiome and Sustainable Healthcare. Healthcare (Basel) 2015; 3:100-29. [PMID: 27417751 PMCID: PMC4934527 DOI: 10.3390/healthcare3010100] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/09/2015] [Accepted: 02/16/2015] [Indexed: 12/19/2022] Open
Abstract
Increasing prevalences, morbidity, premature mortality and medical needs associated with non-communicable diseases and conditions (NCDs) have reached epidemic proportions and placed a major drain on healthcare systems and global economies. Added to this are the challenges presented by overuse of antibiotics and increased antibiotic resistance. Solutions are needed that can address the challenges of NCDs and increasing antibiotic resistance, maximize preventative measures, and balance healthcare needs with available services and economic realities. Microbiome management including microbiota seeding, feeding, and rebiosis appears likely to be a core component of a path toward sustainable healthcare. Recent findings indicate that: (1) humans are mostly microbial (in terms of numbers of cells and genes); (2) immune dysfunction and misregulated inflammation are pivotal in the majority of NCDs; (3) microbiome status affects early immune education and risk of NCDs, and (4) microbiome status affects the risk of certain infections. Management of the microbiome to reduce later-life health risk and/or to treat emerging NCDs, to spare antibiotic use and to reduce the risk of recurrent infections may provide a more effective healthcare strategy across the life course particularly when a personalized medicine approach is considered. This review will examine the potential for microbiome management to contribute to sustainable healthcare.
Collapse
Affiliation(s)
- Rodney R Dietert
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | |
Collapse
|
23
|
Scott KP, Antoine JM, Midtvedt T, van Hemert S. Manipulating the gut microbiota to maintain health and treat disease. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2015; 26:25877. [PMID: 25651995 PMCID: PMC4315778 DOI: 10.3402/mehd.v26.25877] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The intestinal microbiota composition varies between healthy and diseased individuals for numerous diseases. Although any cause or effect relationship between the alterations in the gut microbiota and disease is not always clear, targeting the intestinal microbiota might offer new possibilities for prevention and/or treatment of disease. OBJECTIVE Here we review some examples of manipulating the intestinal microbiota by prebiotics, probiotics, and fecal microbial transplants. RESULTS Prebiotics are best known for their ability to increase the number of bifidobacteria. However, specific prebiotics could potentially also stimulate other species they can also stimulate other species associated with health, like Akkermansia muciniphila, Ruminococcus bromii, the Roseburia/Enterococcus rectale group, and Faecalibacterium prausnitzii. Probiotics have beneficial health effects for different diseases and digestive symptoms. These effects can be due to the direct effect of the probiotic bacterium or its products itself, as well as effects of the probiotic on the resident microbiota. Probiotics can influence the microbiota composition as well as the activity of the resident microbiota. Fecal microbial transplants are a drastic intervention in the gut microbiota, aiming for total replacement of one microbiota by another. With numerous successful studies related to antibiotic-associated diarrhea and Clostridium difficile infection, the potential of fecal microbial transplants to treat other diseases like inflammatory bowel disease, irritable bowel syndrome, and metabolic and cardiovascular disorders is under investigation. CONCLUSIONS Improved knowledge on the specific role of gut microbiota in prevention and treatment of disease will help more targeted manipulation of the intestinal microbiota. Further studies are necessary to see the (long term) effects for health of these interventions.
Collapse
Affiliation(s)
- Karen P Scott
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK
| | | | - Tore Midtvedt
- Department of Microbiology, Tumor and Cell Biology (MTC) Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
24
|
Yuan L, Ma W, Liu J, Meng L, Liu J, Li S, Han J, Liu Q, Feng L, Wang C, Xiao R. Effects of GSTM1/GSTT1 gene polymorphism and fruit & vegetable consumption on antioxidant biomarkers and cognitive function in the elderly: a community based cross-sectional study. PLoS One 2014; 9:e113588. [PMID: 25420021 PMCID: PMC4242636 DOI: 10.1371/journal.pone.0113588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/27/2014] [Indexed: 01/01/2023] Open
Abstract
Background It was reported that Glutathione S-transferase (GST) gene polymorphism and fruit and vegetable (FV) intake were associated with body antioxidant capacity. The oxidative/anti-oxidative imbalance played an important role in the pathogenesis of AD. However, the association of GST genotype, dietary FV consumption with body antioxidant biomarkers and cognitive function in the elderly is not clear. Objective The aim of the present study was to determine the association of GST genotype, and dietary FV intake, with antioxidant biomarkers and cognitive function in the elderly. Methods Food frequency questionnaire was used to collect data of dietary FV intakes in 504 community dwelling elderly aged from 55 to 75 years old. GSTM1 and GSTT1 genotypes were determined by using multiple-PCR method. Plasma and erythrocyte antioxidant biomarkers were measured. Cognitive function was measured by using Montreal Cognitive Assessment. Statistical analysis was applied for exploring the association of GST genotype and FV intake with antioxidant biomarkers level and cognitive function in the elderly. Results Individual GSTM1 or GSTT1 gene deletion affects body antioxidant biomarkers levels, including erythrocyte GST activity, plasma total antioxidant capacity, and glutathione levels. GSTM1and/or GSTT1 gene deletion have no effects on cognitive function in the surveyed participants. The effect of GST genotype on antioxidant biomarkers are FV intake dependent. There is interaction of FV intake and GST genotype on cognitive function in the elderly. Conclusion GST genotype or daily FV consumption impact body antioxidant biomarkers, but not cognitive function in the elderly. There were combined effects of GST genotype and FV consumption on cognitive function in the elderly population. Large scale perspective population study is required to explore the association of GST genetic polymorphism, FV consumption and antioxidant biomarkers and cognitive function in the elderly.
Collapse
Affiliation(s)
- Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, P.R.China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, P.R. China
| | - Weiwei Ma
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Jinmeng Liu
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Liping Meng
- Institute of Nutrition and Food Safety, Chinese Center for Disease Control and Prevention, Beijing, P.R. China
| | - Jixia Liu
- Nanyuan Community Health Service Center of Fengtai District, Beijing, P.R. China
| | - Shuang Li
- Nanyuan Community Health Service Center of Fengtai District, Beijing, P.R. China
| | - Jing Han
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Quanri Liu
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Lingli Feng
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Chao Wang
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Rong Xiao
- School of Public Health, Capital Medical University, Beijing, P.R.China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, P.R. China
- * E-mail:
| |
Collapse
|
25
|
Stilling RM, Bordenstein SR, Dinan TG, Cryan JF. Friends with social benefits: host-microbe interactions as a driver of brain evolution and development? Front Cell Infect Microbiol 2014; 4:147. [PMID: 25401092 PMCID: PMC4212686 DOI: 10.3389/fcimb.2014.00147] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/03/2014] [Indexed: 12/21/2022] Open
Abstract
The tight association of the human body with trillions of colonizing microbes that we observe today is the result of a long evolutionary history. Only very recently have we started to understand how this symbiosis also affects brain function and behavior. In this hypothesis and theory article, we propose how host-microbe associations potentially influenced mammalian brain evolution and development. In particular, we explore the integration of human brain development with evolution, symbiosis, and RNA biology, which together represent a “social triangle” that drives human social behavior and cognition. We argue that, in order to understand how inter-kingdom communication can affect brain adaptation and plasticity, it is inevitable to consider epigenetic mechanisms as important mediators of genome-microbiome interactions on an individual as well as a transgenerational time scale. Finally, we unite these interpretations with the hologenome theory of evolution. Taken together, we propose a tighter integration of neuroscience fields with host-associated microbiology by taking an evolutionary perspective.
Collapse
Affiliation(s)
- Roman M Stilling
- Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department Anatomy and Neuroscience, University College Cork Cork, Ireland
| | - Seth R Bordenstein
- Departments of Biological Sciences and Pathology, Microbiology, and Immunology, Vanderbilt University Nashville, TN, USA
| | - Timothy G Dinan
- Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department of Psychiatry, University College Cork Cork, Ireland
| | - John F Cryan
- Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department Anatomy and Neuroscience, University College Cork Cork, Ireland
| |
Collapse
|
26
|
Kanherkar RR, Bhatia-Dey N, Csoka AB. Epigenetics across the human lifespan. Front Cell Dev Biol 2014; 2:49. [PMID: 25364756 PMCID: PMC4207041 DOI: 10.3389/fcell.2014.00049] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/22/2014] [Indexed: 12/17/2022] Open
Abstract
Epigenetics has the potential to explain various biological phenomena that have heretofore defied complete explication. This review describes the various types of endogenous human developmental milestones such as birth, puberty, and menopause, as well as the diverse exogenous environmental factors that influence human health, in a chronological epigenetic context. We describe the entire course of human life from periconception to death and chronologically note all of the potential internal timepoints and external factors that influence the human epigenome. Ultimately, the environment presents these various factors to the individual that influence the epigenome, and the unique epigenetic and genetic profile of each individual also modulates the specific response to these factors. During the course of human life, we are exposed to an environment that abounds with a potent and dynamic milieu capable of triggering chemical changes that activate or silence genes. There is constant interaction between the external and internal environments that is required for normal development and health maintenance as well as for influencing disease load and resistance. For example, exposure to pharmaceutical and toxic chemicals, diet, stress, exercise, and other environmental factors are capable of eliciting positive or negative epigenetic modifications with lasting effects on development, metabolism and health. These can impact the body so profoundly as to permanently alter the epigenetic profile of an individual. We also present a comprehensive new hypothesis of how these diverse environmental factors cause both direct and indirect epigenetic changes and how this knowledge can ultimately be used to improve personalized medicine.
Collapse
Affiliation(s)
- Riya R Kanherkar
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Naina Bhatia-Dey
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Antonei B Csoka
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| |
Collapse
|
27
|
Sun GD, Cui WP, Guo QY, Miao LN. Histone lysine methylation in diabetic nephropathy. J Diabetes Res 2014; 2014:654148. [PMID: 25215303 PMCID: PMC4158558 DOI: 10.1155/2014/654148] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/14/2014] [Indexed: 01/21/2023] Open
Abstract
Diabetic nephropathy (DN) belongs to debilitating microvascular complications of diabetes and is the leading cause of end-stage renal diseases worldwide. Furthermore, outcomes from the DCCT/EDIC study showed that DN often persists and progresses despite intensive glucose control in many diabetes patients, possibly as a result of prior episode of hyperglycemia, which is called "metabolic memory." The underlying mechanisms responsible for the development and progression of DN remain poorly understood. Activation of multiple signaling pathways and key transcription factors can lead to aberrant expression of DN-related pathologic genes in target renal cells. Increasing evidence suggests that epigenetic mechanisms in chromatin such as DNA methylation, histone acetylation, and methylation can influence the pathophysiology of DN and metabolic memory. Exciting researches from cell culture and experimental animals have shown that key histone methylation patterns and the related histone methyltransferases and histone demethylases can play important roles in the regulation of inflammatory and profibrotic genes in renal cells under diabetic conditions. Because histone methylation is dynamic and potentially reversible, it can provide a window of opportunity for the development of much-needed novel therapeutic potential for DN in the future. In this minireview, we discuss recent advances in the field of histone methylation and its roles in the pathogenesis and progression of DN.
Collapse
Affiliation(s)
- Guang-dong Sun
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
- *Guang-dong Sun: and
| | - Wen-peng Cui
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Qiao-yan Guo
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Li-ning Miao
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
- *Li-ning Miao:
| |
Collapse
|