The nasal and gut microbiome in Parkinson's disease and idiopathic rapid eye movement sleep behavior disorder

Anna Heintz-Buschart, Urvashi Pandey, Tamara Wicke, Friederike Sixel-Döring, Annette Janzen, Elisabeth Sittig-Wiegand, Claudia Trenkwalder, Wolfgang H Oertel, Brit Mollenhauer, Paul Wilmes, Anna Heintz-Buschart, Urvashi Pandey, Tamara Wicke, Friederike Sixel-Döring, Annette Janzen, Elisabeth Sittig-Wiegand, Claudia Trenkwalder, Wolfgang H Oertel, Brit Mollenhauer, Paul Wilmes

Abstract

Background: Increasing evidence connects the gut microbiota and the onset and/or phenotype of Parkinson's disease (PD). Differences in the abundances of specific bacterial taxa have been reported in PD patients. It is, however, unknown whether these differences can be observed in individuals at high risk, for example, with idiopathic rapid eye movement sleep behavior disorder, a prodromal condition of α-synuclein aggregation disorders including PD.

Objectives: To compare microbiota in carefully preserved nasal wash and stool samples of subjects with idiopathic rapid eye movement sleep behavior disorder, manifest PD, and healthy individuals.

Methods: Microbiota of flash-frozen stool and nasal wash samples from 76 PD patients, 21 idiopathic rapid eye movement sleep behavior disorder patients, and 78 healthy controls were assessed by 16S and 18S ribosomal RNA amplicon sequencing. Seventy variables, related to demographics, clinical parameters including nonmotor symptoms, and sample processing, were analyzed in relation to microbiome variability and controlled differential analyses were performed.

Results: Differentially abundant gut microbes, such as Akkermansia, were observed in PD, but no strong differences in nasal microbiota. Eighty percent of the differential gut microbes in PD versus healthy controls showed similar trends in idiopathic rapid eye movement sleep behavior disorder, for example, Anaerotruncus and several Bacteroides spp., and correlated with nonmotor symptoms. Metagenomic sequencing of select samples enabled the reconstruction of genomes of so far uncharacterized differentially abundant organisms.

Conclusion: Our study reveals differential abundances of gut microbial taxa in PD and its prodrome idiopathic rapid eye movement sleep behavior disorder in comparison to the healthy controls, and highlights the potential of metagenomics to identify and characterize microbial taxa, which are enriched or depleted in PD and/or idiopathic rapid eye movement sleep behavior disorder. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.

Keywords: 16S rRNA gene amplicon sequencing; PD; RBD; genome reconstructions; nonmotor phenotype.

© 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.

Figures

Figure 1
Figure 1
Overview of the observed community structures. (A) DNA yield per mL nasal wash or g of stool. (B) Estimates of OTU richness in nasal and gut samples. (C) The most common bacterial families and eukaryotic divisions or subdivisions in nasal and gut samples. Principal coordinate analyses of Jensen‐Shannon diversities between (D) OTU profiles of nasal and gut samples, (E) prokaryotic family profiles of nasal samples, and (F) OTU profiles of gut samples with circles around 70% confidence intervals for (E) sex (red, female; blue, male) and (F) study group (blue, healthy; red, PD; purple, iRBD). Colors and symbols used for the study participants throughout the articles are represented below. PC, principal coordinate.
Figure 2
Figure 2
Heatmaps of most differentially abundant taxa in PD patients and individuals with RBD. Relative abundances of prokaryotic OTUs and higher‐level taxa of the gut microbiome that were found to be differentially abundant in (A) PD patients, or (B) iRBD patients or both compared to the HCs (FDR‐adjusted DESeq2 P values < 0.001 and/or confirmation by ANCOM). Legends for the cohort‐related indications at the top of the heatmaps, for the summarizing heatmaps to the left, and for the central heatmaps are given to the left and below the heatmaps (for the complete set of differentially abundant taxa, detailed histograms, summary data, and further explanations, see Supplementary Information). For the OTUs, the lowest confident classifications are displayed; i.s.: incertae sedis; *the Bacteroides OTU_184 was found in both displayed sets. FC, fold change; n.a., not applicable.
Figure 3
Figure 3
Overviews of genomic reconstructions of two novel microbial populations depleted in patients with PD. (A) OTU_171, classified as Melainabacterium, (B) OTU_469, classified as alpha‐proteobacterium. 1) Contiguous sequences with lengths, 2) metagenomic depth of coverage, 3) % similarity to MelB1,57 4 and 5) predicted proteins colored according to functional categories, 6 and 7) tRNA and rRNA loci, and E) endoglucanase with synuclein‐like domain; pink spokes highlight unique essential genes, golden spokes highlight phylogenetic marker genes. kb = kilobase; tRNA, transfer RNA.

References

    1. Forsyth CB, Shannon KM, Kordower JH, et al. Increased intestinal permeability correlates with sigmoid mucosa alpha‐synuclein staining and endotoxin exposure markers in early Parkinson's disease. PLoS One 2011;6:e28032.
    1. Beach TG, White CL III, Hladik CL, et al. Olfactory bulb α‐synucleinopathy has high specificity and sensitivity for Lewy body disorders. Acta Neuropathol 2008;117:169‐174.
    1. Arizona Parkinson's Disease Consortium , Beach TG, Adler CH, et al. Multi‐organ distribution of phosphorylated α‐synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol 2010;119:689‐702.
    1. Mu L, Sobotka S, Chen J, et al. Alpha‐synuclein pathology and axonal degeneration of the peripheral motor nerves innervating pharyngeal muscles in Parkinson disease. J Neuropathol Exp Neurol 2013;72:119‐129.
    1. Doppler K, Jentschke HM, Schulmeyer L, et al. Dermal phospho‐alpha‐synuclein deposits confirm REM sleep behaviour disorder as prodromal Parkinson's disease. Acta Neuropathol 2017;133:535‐545.
    1. Braak H, de Vos RAI, Bohl J, Del Tredici K. Gastric α‐synuclein immunoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Parkinson's disease‐related brain pathology. Neurosci Lett 2006;396:67‐72.
    1. Svensson E, Horváth‐Puhó E, Thomsen RW, et al. Vagotomy and subsequent risk of Parkinson's disease. Ann Neurol 2015;78:522‐529.
    1. Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging 2003;24:197‐211.
    1. Devos D, Lebouvier T, Lardeux B, et al. Colonic inflammation in Parkinson's disease. Neurobiol Dis 2013;50:42‐48.
    1. Dobbs SM, Dobbs RJ, Weller C, et al. Peripheral aetiopathogenic drivers and mediators of Parkinson's disease and co‐morbidities: role of gastrointestinal microbiota. J Neurovirol 2015;22:22‐32.
    1. Hawkes CH, Del Tredici K, Braak H. Parkinson's disease: a dual‐hit hypothesis. Neuropathol Appl Neurobiol 2007;33:599‐614.
    1. Hoglinger GU, Alvarez‐Fischer D, Arias‐Carrion O, et al. A new dopaminergic nigro‐olfactory projection. Acta Neuropathol 2015;130:333‐348.
    1. Pan‐Montojo F, Schwarz M, Winkler C, et al. Environmental toxins trigger PD‐like progression via increased alpha‐synuclein release from enteric neurons in mice. Sci Rep 2012;2:898.
    1. Chen SG, Stribinskis V, Rane MJ, et al. Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha‐Synuclein Aggregation in Aged Fischer 344 Rats and Caenorhabditis elegans . Sci Rep 2016;6:34477.
    1. Scheperjans F, Aho V, Pereira PAB, et al. Gut microbiota are related to Parkinson's disease and clinical phenotype. Mov Disord 2015;30:350‐358.
    1. Keshavarzian A, Green SJ, Engen PA, et al. Colonic bacterial composition in Parkinson's disease. Mov Disord 2015;30:1351‐1360.
    1. Unger MM, Spiegel J, Dillmann KU, et al. Short chain fatty acids and gut microbiota differ between patients with Parkinson's disease and age‐matched controls. Parkinsonism Relat Disord 2016;32:66‐72.
    1. Hasegawa S, Goto S, Tsuji H, et al. Intestinal dysbiosis and lowered serum lipopolysaccharide‐binding protein in Parkinson's disease. PLoS One 2015;10:e0142164.
    1. Hill‐Burns EM, Debelius JW, Morton JT, et al. Parkinson's disease and Parkinson's disease medications have distinct signatures of the gut microbiome. Mov Disord 2017;32:739‐749.
    1. Bedarf JR, Hildebrand F, Coelho LP, et al. Functional implications of microbial and viral gut metagenome changes in early stage L‐DOPA‐naïve Parkinson's disease patients. Genome Med 2017;9:39.
    1. Hopfner F, Künstner A, Müller SH, et al. Gut microbiota in Parkinson disease in a northern German cohort. Brain Res 2017;1667:41‐45.
    1. Scheperjans F. Gut microbiota, 1013 new pieces in the Parkinson's disease puzzle. Curr Opin Neurol 2016;29:773‐780.
    1. Pereira PA, Aho VT, Paulin L, Pekkonen E, Auvinen P, Scheperjans F. Oral and nasal microbiota in Parkinson's disease. Parkinsonism Relat Disord 2017;38:61‐67.
    1. Schrag A, Horsfall L, Walters K, Noyce A. Prediagnostic presentations of Parkinson's disease in primary care: a case‐control study. Lancet Neurol 2015;14:57‐64.
    1. Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder: a 16‐year update on a previously reported series. Sleep Med 2013;14:744‐748.
    1. Chaudhuri KR, Schapira AH. Non‐motor symptoms of Parkinson's disease: dopaminergic pathophysiology and treatment. Lancet Neurol 2009;8:464‐474.
    1. Mollenhauer B, Zimmermann J, Sixel‐Doring F, et al. Monitoring of 30 marker candidates in early Parkinson disease as progression markers. Neurology 2016;87:168‐177.
    1. Mollenhauer B, Trautmann E, Sixel‐Doring F, et al. Nonmotor and diagnostic findings in subjects with de novo Parkinson disease of the DeNoPa cohort. Neurology 2013;81:1226‐1234.
    1. Schenck CH, Montplaisir JY, Frauscher B, et al. Rapid eye movement sleep behavior disorder: devising controlled active treatment studies for symptomatic and neuroprotective therapy—a consensus statement from the International Rapid Eye Movement Sleep Behavior Disorder Study Group. Sleep Med 2013;14:795‐806.
    1. Goetz CG, Tilley BC, Shaftman SR, et al. Movement Disorder Society‐sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS‐UPDRS): scale presentation and clinimetric testing results. Mov Disord 2008;23:2129‐2170.
    1. Visser M, Marinus J, Stiggelbout AM, van Hilten JJ. Assessment of autonomic dysfunction in Parkinson's disease: the SCOPA‐AUT. Mov Disord 2004;19:1306‐1312.
    1. Gauggel S, Birkner B. [Validity and reliability of a German version of the Geriatric Depression Scale (GDS)]. [Article in German]. Z Klin Psychol Psych 1999;28:18‐27.
    1. Schmidtke A, Fleckenstein P, Moises W, Beckmann H. [Studies of the reliability and validity of the German version of the Montgomery‐Asberg Depression Rating Scale (MADRS)]. [Article in German]. Schweiz Arch Neurol Psychiatr (1985) 1988;139:51‐65.
    1. Folstein MF, Folstein SE, McHugh PR. “Mini‐mental state.” A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975;12:189‐198.
    1. Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc 2005;53:695‐699.
    1. Bloch KE, Schoch OD, Zhang JN, Russi EW. German version of the Epworth Sleepiness Scale. Respiration 1999;66:440‐447.
    1. Schoenebeck B, May C, Güldner C, et al. Improved preparation of nasal lavage fluid (NLF) as a noninvasive sample for proteomic biomarker discovery. Biochim Biophys Acta 2015;1854:741‐745.
    1. Heintz‐Buschart A, May P, Laczny CC, et al. Integrated multi‐omics of the human gut microbiome in a case study of familial type 1 diabetes. Nat Microbiol 2016;2:16180.
    1. Roume H, Muller EE, Cordes T, Renaut J, Hiller K, Wilmes P. A biomolecular isolation framework for eco‐systems biology. ISME J 2013;7:110‐121.
    1. Mathay C, Hamot G, Henry E, et al. Method optimization for fecal sample collection and fecal DNA extraction. Biopreserv Biobank 2015;13:79‐93.
    1. Wampach L, Heintz‐Buschart A, Hogan A, et al. Colonization and succession within the human gut microbiome by archaea, bacteria and microeukaryotes during the first year of life. Front Microbiol 2017;8:738.
    1. Hildebrand F, Tadeo R, Voigt AY, Bork P, Raes J. LotuS: an efficient and user‐friendly OTU processing pipeline. Microbiome 2014;2:30.
    1. Hugerth LW, Muller EE, Hu YO, et al. Systematic design of 18S rRNA gene primers for determining eukaryotic diversity in microbial consortia. PLoS One 2014;9:e95567.
    1. R Core Team . R: A Language and Environment for Statistical Computing. Vienna: R Foundation for Statistical Computing; 2014.
    1. Narayanasamy S, Jarosz Y, Muller EE, et al. IMP: a pipeline for reproducible metagenomic and metatranscriptomic analyses. Genome Biol 2016;17:260.
    1. Laczny CC, Muller EE, Heintz‐Buschart A, et al. Identification, recovery, and refinement of hitherto undescribed population‐level genomes from the human gastrointestinal tract. Front Microbiol 2016;7:533.
    1. Aziz RK, Bartels D, Best AA, et al. The RAST Server: rapid annotations using subsystems technology. BMC Genomics 2008;9:75.
    1. Oksanen J, Blanchet FG, Kindt R, et al. The vegan package. Community ecology package 2007;10:631‐637.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One 2013;8:e61217.
    1. Paradis E, Claude J, Strimmer K. APE: Analyses of phylogenetics and evolution in R language. Bioinformatics 2004;20:289‐290.
    1. Sievers F, Wilm A, Dineen D, et al. Fast, scalable generation of high‐quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol 2011;7:1‐6.
    1. Price MN, Dehal PS, Arkin AP. FastTree 2—approximately maximum‐likelihood trees for large alignments. PLoS One 2010;5:e9490.
    1. Zhernakova A, Kurilshikov A, Bonder MJ, et al. Population‐based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 2016;352:565‐569.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA‐seq data with DESeq2. Genome Biol 2014;15:31.
    1. Mandal S, Van Treuren W, White RA, Eggesbø M, Knight R, Peddada SD. Analysis of composition of microbiomes: a novel method for studying microbial composition. Microb Ecol Health Dis 2015;26:27663.
    1. Jun SR, Robeson MS, Hauser LJ, Schadt CW, Gorin AA. PanFP: pangenome‐based functional profiles for microbial communities. BMC Res Notes 2015;8:479.
    1. Di Rienzi SC, Sharon I, Wrighton KC, et al. The human gut and groundwater harbor non‐photosynthetic bacteria belonging to a new candidate phylum sibling to Cyanobacteria. eLife 2013;2:e01102.
    1. Rantala‐Ylinen A, Kana S, Wang H, et al. Anatoxin‐a Synthetase Gene Cluster of the Cyanobacterium Anabaena sp. Strain 37 and Molecular Methods To Detect Potential Producers. Appl Environ Microbiol 2011;77:7271‐7278.
    1. Méjean A, Paci G, Gautier V, Ploux O. Biosynthesis of anatoxin‐a and analogues (anatoxins) in cyanobacteria. Toxicon 2014;91:15‐22.
    1. Kellmann R, Mihali TK, Jeon YJ, Pickford R, Pomati F, Neilan BA. Biosynthetic Intermediate Analysis and Functional Homology Reveal a Saxitoxin Gene Cluster in Cyanobacteria. Appl Environ Microbiol 2008;74:4044‐4053.
    1. Gorzelak MA, Gill SK, Tasnim N, Ahmadi‐Vand Z, Jay M, Gibson DL. Methods for improving human gut microbiome data by reducing variability through sample processing and storage of stool. PLoS One 2015;10:e0134802.
    1. Bassis CM, Erb‐Downward JR, Dickson RP, et al. Analysis of the upper respiratory tract microbiotas as the source of the lung and gastric microbiotas in healthy individuals. MBio 2015;6:e00037.
    1. Huttenhower C, Gevers D, Knight R, et al. Structure, function and diversity of the healthy human microbiome. Nature 2012;486:207‐214.
    1. Biswas K, Hoggard M, Jain R, Taylor MW, Douglas RG. The nasal microbiota in health and disease: variation within and between subjects. Front Microbiol 2015;9:134.
    1. Bassis CM, Tang AL, Young VB, Pynnonen MA. The nasal cavity microbiota of healthy adults. Microbiome 2014;2:27.
    1. Forslund K, Hildebrand F, Nielsen T, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015;528:262‐266.
    1. Vandeputte D, Falony G, Vieira‐Silva S, Tito RY, Joossens M, Raes J. Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut 2015;65:57‐62.
    1. Gobert AP, Sagrestani G, Delmas E, et al. The human intestinal microbiota of constipated‐predominant irritable bowel syndrome patients exhibits anti‐inflammatory properties. Sci Rep 2016;6:39399.
    1. Kong F, Hua Y, Zeng B, Ning R, Li Y, Zhao J. Gut microbiota signatures of longevity. Curr Biol 2016;26:R832‐R833.
    1. Reunanen J, Kainulainen V, Huuskonen L, et al. Akkermansia muciniphila. adheres to enterocytes and strengthens the integrity of the epithelial cell layer. Appl Environ Microbiol 2015;81:3655‐3662.
    1. Plovier H, Everard A, Druart C, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med 2017;23:107‐113.
    1. Ottman N, Reunanen J, Meijerink M, et al. Pili‐like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS One 2017;12:e0173004.
    1. Desai MS, Seekatz AM, Koropatkin NM, et al. A dietary fiber‐deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 2016;167:1339‐1353.e21.
    1. Sampson TR, Debelius JW, Thron T, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell 2016;167:1469‐1480.e12.
    1. Felice VD, Quigley EM, Sullivan AM, O'Keeffe GW, O'Mahony SM. Microbiota‐gut‐brain signalling in Parkinson's disease: implications for non‐motor symptoms. Parkinsonism Relat Disord 2016;27:1‐8.
    1. Jiang H, Ling Z, Zhang Y, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun 2015;48:186‐194.
    1. Aizawa E, Tsuji H, Asahara T, et al. Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J Affect Disord 2016;202:254‐257.
    1. Kelly JR, Borre Y, O'Brien C, et al. Transferring the blues: depression‐associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res 2016;82:109‐118.
    1. Zheng P, Zeng B, Zhou C, et al. Gut microbiome remodeling induces depressive‐like behaviors through a pathway mediated by the host. Mol Psychiatry 2016;21:786‐796.
    1. Sunagawa S, Mende DR, Zeller G, et al. Metagenomic species profiling using universal phylogenetic marker genes. Nat Methods 2013;10:1196‐1199.
    1. Cox PA, Banack SA, Murch SJ, et al. Diverse taxa of cyanobacteria produce beta‐N‐methylamino‐L‐alanine, a neurotoxic amino acid. Proc Natl Acad Sci U S A 2005;102:5074‐5078.
    1. Dunlop RA, Cox PA, Banack SA, Rodgers KJ. The non‐protein amino acid BMAA is misincorporated into human proteins in place of l‐serine causing protein misfolding and aggregation. PLoS One 2013;8:e75376.
    1. Murch SJ, Cox PA, Banack SA, Steele JC, Sacks OW. Occurrence of beta‐methylamino‐l‐alanine (BMAA) in ALS/PDC patients from Guam. Acta Neurol Scand 2004;110:267‐269.
    1. Pablo J, Banack SA, Cox PA, et al. Cyanobacterial neurotoxin BMAA in ALS and Alzheimer's disease. Acta Neurol Scand 2009;120:216‐225.
    1. Downing S, Downing TG. The metabolism of the non‐proteinogenic amino acid β‐N‐methylamino‐L‐alanine (BMAA) in the cyanobacterium Synechocystis PCC6803. Toxicon 2016;115:41‐48.
    1. Durso LM, Smith D, Hutkins RW. Measurements of fitness and competition in commensal Escherichia coli and E. coli O157:H7 strains. Appl Environ Microbiol 2004;70:6466‐6472.

Source: PubMed

3
Iratkozz fel