Wie advies wilt over hoe het microbioom te verbeteren zou contact op kunnen nemen met deze website: Www.microbiome-Center.nl Voor zowel artsen als individuele burgers staat een groep van artsen en wetenschappers klaar om u een persoonlijk advies te geven.

Gerelateerd aan dit onderwerp is het wellicht interessant dit artikel in Kennislink  ook te lezen.

22 februari 2022: lees ook dit artikel: https://kanker-actueel.nl/NL/darmflora-beinvloed-door-voeding-en-leefstijl-speelt-cruciale-rol-in-ontstaan-van-ziektes-ook-bij-ontstaan-van-kanker-en-behandelingen-daarvan.html

Lees ook dit artikel over het darmmicrobioom en hoe je die kan testen: https://kanker-actueel.nl/NL/wat-is-een-darmmicrobioomtest-en-wat-zegt-een-uitslag-over-de-status-van-je-darmen-en-gezondheid-in-het-algemeen.html

22 februari 2022: VUmc Amsterdam

Uit recent Nederlands onderzoek blijkt dat patiënten met Alzheimer minder van een bepaald type bacteriën in hun darm hebben dan gezonde mensen. Die goede bacteriën worden vaak gezien bij mensen die veel vezels eten. Al eerder is aangetoond dat veel groente en fruit eten helpt tegen het ontstaan van dementie. Nu hebben onderzoekers van het VUmc ook daadwerkelijk aangetoond dat de darmflora - darmmicrobioom is gekoppeld aan Alzheimer. Al is nog niet duidelijk of Alzheimer ontstaat door een bepaalde darmflora - microbioom of andersom dat Alzheimer invloed heeft op de darmflora. 

Uit een AD artikel: Alzheimerpatiënten hebben vooral minder bacteriën van het soort dat zogenoemde korteketenvetzuren produceert. Van die vetzuren zoals boterzuur is al langer bekend dat ze een belangrijke rol spelen in het afweersysteem.

Wat we tot nu toe wisten over de relatie tussen het microbioom en alzheimer, kwam vooral uit dierproeven. Maar mensen zijn geen muizen, zegt Wiesje van der Flier, wetenschappelijk directeur van Alzheimercentrum Amsterdam van Amsterdam UMC. in een artikel in het Algemeen Dagblad. "Wel is er bij mensen eerder onderzoek gedaan naar het microbioom en de hersenen. Maar dat gaf nog geen inzicht in de relatie tussen darmbacteriën en alzheimerschade.”

"Bacteriën die stoffen als boterzuur produceren spelen ook een rol bij depressie”, zegt Robert Jan Brummer, MDL-arts en hoogleraar maag- darm- en leverziekten en klinische voeding. Duidelijk is alleen niet waar een depressie begint: in de darm of in de hersenen. Dat is voor Brummer ook niet interessant. ,,Als je de cirkel maar doorbreekt.”

Dat ligt voor alzheimer anders, zegt Van der Flier. ,,Een depressie kan weggaan en terugkomen. Alzheimer is een progressieve ziekte die nooit meer weggaat. ,,Daarom willen we weten of veranderingen in het microbioom tot alzheimer hersenschade leiden. Of veroorzaakt alzheimerhersenschade veranderingen in het microbioom?”

Het Algemeen Dagblad heeft dit artikel over de studie van het VUmc in de rubriek Koken en Weten: 

Alzheimerpatiënten missen goede bacterie in hun darmen, die gezonde mensen wel hebben

KOKEN & WETEN

In de rubriek Koken & Weten duikt gezondheidsjournalist Tijn Elferink in een voedingsonderwerp waar verwarring over bestaat. Dit keer: is er een dieet dat alzheimer kan voorkomen? 

Het is nog te kort door de bocht om te zeggen: ons eten heeft invloed op het ontstaan van de ziekte alzheimer. Maar uit nieuw Nederlands onderzoek blijkt wel dat patiënten met alzheimer minder van een bepaald type bacteriën in hun darm hebben dan gezonde mensen. Die bacteriën worden vaak gezien bij mensen die veel vezels eten. Niet voor niets dat al wel duidelijk is dat veel groente en fruit eten helpt tegen het ontstaan van dementie.>>>>>>>lees verder

Het studierapport van de onderzoekrs uit het VUmc is dit. Klik op de titel van het abstract voor het volledige verslag. 

ORIGINAL RESEARCH article

Front. Immunol., 31 January 2022 | https://doi.org/10.3389/fimmu.2021.794519

Gut Microbiota Composition Is Related to AD Pathology

Barbara J. H. Verhaar1,2,3*, Heleen M. A. Hendriksen3, Francisca A. de Leeuw3, Astrid S. Doorduijn3, Mardou van Leeuwenstijn3, Charlotte E. Teunissen4, Frederik Barkhof5,6, Philip Scheltens3, Robert Kraaij7, Cornelia M. van Duijn8,9, Max Nieuwdorp2, Majon Muller1 and Wiesje M. van der Flier3,10
  • 1Department of Internal Medicine - Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
  • 2Department of Internal and Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
  • 3Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
  • 4Department of Clinical Chemistry, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
  • 5Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
  • 6University College London (UCL) Institutes of Neurology, Faculty of Brain Sciences, London, United Kingdom
  • 7Department of Internal Medicine, Erasmus Medical Center (MC), Rotterdam, Netherlands
  • 8Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam, Netherlands
  • 9Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
  • 10Department of Epidemiology and Data Science, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands

Introduction: Several studies have reported alterations in gut microbiota composition of Alzheimer’s disease (AD) patients. However, the observed differences are not consistent across studies. We aimed to investigate associations between gut microbiota composition and AD biomarkers using machine learning models in patients with AD dementia, mild cognitive impairment (MCI) and subjective cognitive decline (SCD).

Materials and Methods: We included 170 patients from the Amsterdam Dementia Cohort, comprising 33 with AD dementia (66 ± 8 years, 46%F, mini-mental state examination (MMSE) 21[19-24]), 21 with MCI (64 ± 8 years, 43%F, MMSE 27[25-29]) and 116 with SCD (62 ± 8 years, 44%F, MMSE 29[28-30]). Fecal samples were collected and gut microbiome composition was determined using 16S rRNA sequencing. Biomarkers of AD included cerebrospinal fluid (CSF) amyloid-beta 1-42 (amyloid) and phosphorylated tau (p-tau), and MRI visual scores (medial temporal atrophy, global cortical atrophy, white matter hyperintensities). Associations between gut microbiota composition and dichotomized AD biomarkers were assessed with machine learning classification models. The two models with the highest area under the curve (AUC) were selected for logistic regression, to assess associations between the 20 best predicting microbes and the outcome measures from these machine learning models while adjusting for age, sex, BMI, diabetes, medication use, and MMSE.

Results: The machine learning prediction for amyloid and p-tau from microbiota composition performed best with AUCs of 0.64 and 0.63. Highest ranked microbes included several short chain fatty acid (SCFA)-producing species. Higher abundance of  leptum and lower abundance of  ventriosum group spp., Lachnospiraceae spp., Marvinbryantia spp., Monoglobus spp.,  torques group spp., Roseburia hominis, and Christensenellaceae R-7 spp., was associated with higher odds of amyloid positivity. We found associations between lower abundance of Lachnospiraceae spp., Lachnoclostridium spp., Roseburia hominis and Bilophila wadsworthia and higher odds of positive p-tau status.

Conclusions: Gut microbiota composition was associated with amyloid and p-tau status. We extend on recent studies that observed associations between SCFA levels and AD CSF biomarkers by showing that lower abundances of SCFA-producing microbes were associated with higher odds of positive amyloid and p-tau status.

Data Availability Statement

The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found below: https://www.ebi.ac.uk/ena/browser/home, accession ID: PRJEB49329.

Ethics Statement

The studies involving human participants were reviewed and approved by Medisch Ethische Toetsingscommissie VUmc. The patients/participants provided their written informed consent to participate in this study.

Author Contributions

WF, CT, FB, PS, and CD contributed to conception and design of the study. HH, FL, AD, ML, and BV collected the data. RK was responsible for the sequencing of the samples. BV performed the statistical analyses. WF, MN, and MM contributed to the interpretation of the results. BV wrote the first draft of the manuscript. All authors contributed to manuscript revision, read, and approved the submitted version.

Funding

Research of Alzheimer Center Amsterdam is part of the neurodegeneration research program of Amsterdam Neuroscience. Alzheimer Center Amsterdam is supported by Stichting Alzheimer Nederland and Stichting VUmc fonds. The chair of WF is supported by the Pasman stichting. WF is recipient of a grant by Stichting Equilibrio and of ZonMW-Memorabel funded #733050814. The SCIENCe project is supported by a research grant of stichting Dioraphte. BV is appointed on an Amsterdam Cardiovascular Sciences (ACSPhD2019P003) and an Alzheimer Nederland grant (WE.03-2017-12). FB is supported by the NIHR biomedical research centre at UCLH. MN is supported by a personal ZONMW-VICI grant 2020 (09150182010020).

Conflict of Interest

CT received grants from the European Commission, the Dutch Research Council (ZonMW), Association of Frontotemporal Dementia/Alzheimer’s Drug Discovery Foundation, The Weston Brain Institute, Alzheimer Netherlands. CT has a collaboration contract with ADx Neurosciences, performed contract research or received grants from Probiodrug, Biogen, Esai, Toyama, Janssen prevention center, Boehringer, AxonNeurosciences, Fujirebio, EIP farma, PeopleBio, and Roche. FB is a consultant for Biogen-Idec, Bayer-Schering, Merck-Serono, Roche, Combinostics and IXICO; has received sponsorship from European Commission–Horizon 2020, National Institute for Health Research–University College London Hospitals Biomedical Research Centre, Novartis, and Merck; and serves on the editorial boards of Radiology, Neuroradiology, Multiple Sclerosis Journal, and Neurology. PS has received consultancy/speaker fees from Lilly, GE Healthcare, Novartis, Sanofi, Nutricia, Probiodrug, Biogen, Roche, Avraham, and EIP Pharma. PS has acquired grant support from GE Healthcare, Danone Research, Piramal, and MERCK. All funding was paid to the institution. MN is part of the Scientific Advisory Board of Caelus Health, The Netherlands and Kaleido Biosciences, USA. However, none of these are directly relevant to the current paper. WF received research funding from ZonMW, NWO, EU-FP7, EU-JPND, Alzheimer Nederland, CardioVascular Onderzoek Nederland, Health~Holland, Topsector Life Sciences & Health, stichting Dioraphte, Gieskes-Strijbis fonds, stichting Equilibrio, Pasman stichting, Biogen MA Inc, Boehringer Ingelheim, Life-MI, AVID, Roche BV, Fujifilm, Combinostics. WF holds the Pasman chair. WF is recipient of ABOARD, which is a public-private partnership receiving funding from ZonMW (#73305095007) and Health~Holland, Topsector Life Sciences & Health (PPP-allowance; #LSHM20106). She has performed contract research for Biogen MA Inc, and Boehringer Ingelheim. She has been an invited speaker at Boehringer Ingelheim, Biogen MA Inc, Danone, Eisai, WebMD Neurology (Medscape). WF is consultant to Oxford Health Policy Forum CIC, Roche, and Biogen MA Inc. WF participated in an advisory board of Biogen MA Inc and Roche. WF was associate editor of Alzheimer, Research & Therapy in 2020/2021. WF is associate editor at Brain. All funding was paid to the institution.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fimmu.2021.794519/full#supplementary-material

References

1. Heneka MT, Carson MJ, Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s Disease. Lancet Neurol (2015) 14:388–405. doi: 10.1016/S1474-4422(15)70016-5

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the Immune System. Nat Rev Immunol (2017) 17(4):219–32. doi: 10.1038/nri.2017.7

PubMed Abstract | CrossRef Full Text | Google Scholar

3. van Olst L, Roks SJM, Kamermans A, Verhaar BJH, van der Geest AM, Muller M, et al. Contribution of Gut Microbiota to Immunological Changes in Alzheimer’s Disease. Front Immunol (2021) 12:683068. doi: 10.3389/fimmu.2021.683068

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Saresella M, Marventano I, Barone M, La Rosa F, Piancone F, Mendozzi L, et al. Alterations in Circulating Fatty Acid Are Associated With Gut Microbiota Dysbiosis and Inflammation in Multiple Sclerosis. Front Immunol (2020) 11:1390. doi: 10.3389/fimmu.2020.01390

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Moccia M, Clerici M, Keshavarzian A, Engen PA, Zaferiou A, Rasmussen H, et al. Single-Arm, Non-Randomized, Time Series, Single-Subject Study of Fecal Microbiota Transplantation in Multiple Sclerosis. Front Neurol (2020) 11:978. doi: 10.3389/fneur.2020.00978

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Ley RE, Peterson DA, Gordon JI. Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human Intestine. Cell (2006) 124:837–48. doi: 10.1016/j.cell.2006.02.017

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH. The Influence of Diet on the Gut Microbiota. Pharmacol Res (2013) 69(1):52–60. doi: 10.1016/j.phrs.2012.10.020

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Gentile CL, Weir TL. The Gut Microbiota at the Intersection of Diet and Human Health. Science (2018) 362(6416):776–80. doi: 10.1126/science.aau5812

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Cerf-Bensussan N, Gaboriau-Routhiau V. The Immune System and the Gut Microbiota: Friends or Foes? Nat Rev Immunol (2010) 10:735–44. doi: 10.1038/nri2850

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Deleu S, Machiels K, Raes J, Verbeke K, Vermeire S. Short Chain Fatty Acids and its Producing Organisms: An Overlooked Therapy for IBD? EBioMedicine (2021) 66:103293. doi: 10.1016/j.ebiom.2021.103293

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Zhang L, Wang Y, Xiayu X, Shi C, Chen W, Song N, et al. Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease. J Alzheimer’s Dis (2017) 60(4):1241–57. doi: 10.3233/JAD-170020

CrossRef Full Text | Google Scholar

12. Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, et al. Fecal Microbiota Transplantation Alleviated Alzheimer’s Disease-Like Pathogenesis in APP/PS1 Transgenic Mice. Transl Psychiatry (2019) 9(1):1–13. doi: 10.1038/s41398-019-0525-3

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, et al. Transfer of a Healthy Microbiota Reduces Amyloid and Tau Pathology in an Alzheimer’s Disease Animal Model. Gut (2020) 69:283–94. doi: 10.1136/gutjnl-2018-317431

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Cox LM, Schafer MJ, Sohn J, Vincentini J, Weiner HL, Ginsberg SD, et al. Calorie Restriction Slows Age-Related Microbiota Changes in an Alzheimer’s Disease Model in Female Mice. Sci Rep (2019) 9(1):1–14. doi: 10.1038/s41598-019-54187-x

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Fernando WMADB, Martins IJ, Morici M, Bharadwaj P, Rainey-Smith SR, Lim WLF, et al. Sodium Butyrate Reduces Brain Amyloid-β Levels and Improves Cognitive Memory Performance in an Alzheimer’s Disease Transgenic Mouse Model at an Early Disease Stage. J Alzheimer’s Dis (2020) 74(1):91–9. doi: 10.3233/JAD-190120

CrossRef Full Text | Google Scholar

16. Vogt NM, Kerby RL, Dill-Mcfarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut Microbiome Alterations in Alzheimer’s Disease. Sci Rep (2017) 7):13537. doi: 10.1038/s41598-017-13601-y

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Zhuang ZQ, Shen LL, Li WW, Fu X, Zeng F, Gui L, et al. Gut Microbiota Is Altered in Patients With Alzheimer’s Disease. J Alzheimer’s Dis (2018) 63:1337–46. doi: 10.3233/JAD-180176

CrossRef Full Text | Google Scholar

18. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, et al. Alzheimer’s Disease Microbiome Is Associated With Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway. MBio (2019) 10:e00632-19. doi: 10.1128/mBio.00632-19

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Liu P, Wu L, Peng G, Han Y, Tang R, Ge J, et al. Altered Microbiomes Distinguish Alzheimer’s Disease From Amnestic Mild Cognitive Impairment and Health in a Chinese Cohort. Brain Behav Immun (2019) 80:633–43. doi: 10.1016/j.bbi.2019.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Ueda A, Shinkai S, Shiroma H, Taniguchi Y, Tsuchida S, Kariya T, et al. Identification of Faecalibacterium Prausnitzii Strains for Gut Microbiome-Based Intervention in Alzheimer’s-Type Dementia. Cell Rep Med (2021) 2(9):100398. doi: 10.1016/j.xcrm.2021.100398

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, et al. Association of Brain Amyloidosis With Pro-Inflammatory Gut Bacterial Taxa and Peripheral Inflammation Markers in Cognitively Impaired Elderly. Neurobiol Aging (2017) 49:60–8. doi: 10.1016/j.neurobiolaging.2016.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Mirzayi C, Renson A, Furlanello C, Sansone S-A, Zohra F, Elsafoury S, et al. Reporting Guidelines for Human Microbiome Research: The STORMS Checklist. Nat Med (2021) 27(11):1885–92. doi: 10.1038/s41591-021-01552-x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. van der Flier WM, Pijnenburg YAL, Prins N, Lemstra AW, Bouwman FH, Teunissen CE, et al. Optimizing Patient Care and Research: The Amsterdam Dementia Cohort. J Alzheimer’s Dis (2014) 41(1):313–27. doi: 10.3233/JAD-132306

CrossRef Full Text | Google Scholar

24. van der Flier WM, Scheltens P. Amsterdam Dementia Cohort: Performing Research to Optimize Care. J Alzheimer’s Dis (2018) 62(3):1091–111. doi: 10.3233/JAD-170850

CrossRef Full Text | Google Scholar

25. Slot RER, Verfaillie SCJ, Overbeek JM, Timmers T, Wesselman LMP, Teunissen CE, et al. Subjective Cognitive Impairment Cohort (SCIENCe): Study Design and First Results. Alzheimer’s Res Ther (2018) 10(1):1–13. doi: 10.1186/s13195-018-0390-y

CrossRef Full Text | Google Scholar

26. Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, et al. The Diagnosis of Mild Cognitive Impairment Due to Alzheimer’s Disease: Recommendations From the National Institute on Aging-Alzheimer’s Association Workgroups on Diagnostic Guidelines for Alzheimer’s Disease. Alzheimer’s Dement (2011) 7(3):270–9. doi: 10.1016/j.jalz.2011.03.008

CrossRef Full Text | Google Scholar

27. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical Diagnosis of Alzheimer’s Disease. Neurology (1984) 34(7):939–9. doi: 10.1212/WNL.34.7.939

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Tombaugh TN, McIntyre NJ. The Mini-Mental State Examination: A Comprehensive Review. J Am Geriatr Soc (1992) 40(9):922–35. doi: 10.1111/j.1532-5415.1992.tb01992.x

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Fadrosh DW, Ma B, Gajer P, Sengamalay N, Ott S, Brotman RM, et al. An Improved Dual-Indexing Approach for Multiplexed 16S rRNA Gene Sequencing on the Illumina MiSeq Platform. Microbiome (2014) 2:1–7. doi: 10.1186/2049-2618-2-6

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Mulder C, Verwey NA, van der Flier WM, Bouwman FH, Kok A, van Elk EJ, et al. Amyloid-β(1–42), Total Tau, and Phosphorylated Tau as Cerebrospinal Fluid Biomarkers for the Diagnosis of Alzheimer Disease. Clin Chem (2010) 56(2):248–53. doi: 10.1373/clinchem.2009.130518

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Tijms BM, Willemse EAJ, Zwan MD, Mulder SD, Visser PJ, van Berckel BNM, et al. Unbiased Approach to Counteract Upward Drift in Cerebrospinal Fluid Amyloid-β 1–42 Analysis Results. Clin Chem (2018) 64(3):576–85. doi: 10.1373/clinchem.2017.281055

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Willemse EAJ, Maurik ISv, Tijms BM, Bouwman FH, Franke A, Hubeek I, et al. Diagnostic Performance of Elecsys Immunoassays for Cerebrospinal Fluid Alzheimer’s Disease Biomarkers in a Nonacademic, Multicenter Memory Clinic Cohort: The ABIDE Project. Alzheimer’s Dement Diagn Assess Dis Monit (2018) 10:563. doi: 10.1016/J.DADM.2018.08.006

CrossRef Full Text | Google Scholar

33. Scheltens P, Kuiper M, Ch Wolters E, Barkhof F, Valk J, Weinsten HC, et al. Atrophy of Medial Temporal Lobes on MRI in “Probable” Alzheimer’s Disease and Normal Ageing: Diagnostic Value and Neuropsychological Correlates. J Neurol Neurosurg Psychiatry (1992) 55:967–72. doi: 10.1136/jnnp.55.10.967

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Rhodius-Meester HFM, Benedictus MR, Wattjes MP, Barkhof F, Scheltens P, Muller M, et al. MRI Visual Ratings of Brain Atrophy and White Matter Hyperintensities Across the Spectrum of Cognitive Decline Are Differently Affected by Age and Diagnosis. Front Aging Neurosci (2017) 9:117. doi: 10.3389/fnagi.2017.00117

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Pasquier F, Leys D, Weerts JGE, Mounier-Vehier F, Barkhof F, Scheltens P. Inter-And Intraobserver Reproducibility of Cerebral Atrophy Assessment on Mri Scans With Hemispheric Infarcts. Eur Neurol (1996) 36:268–72. doi: 10.1159/000117270

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Fazekas F, Chawluk JB, Alavi A, Hurtig HI, Zimmerman RA. MR Signal Abnormalities at 1.5 T in Alzheimer’s Dementia and Normal Aging. Am J Roentgenol (1987) 8:421–426. doi: 10.2214/ajr.149.2.351

CrossRef Full Text | Google Scholar

37. Wardlaw JM, Smith EE, Biessels GJ, Cordonnier C, Fazekas F, Frayne R, et al. Neuroimaging Standards for Research Into Small Vessel Disease and its Contribution to Ageing and Neurodegeneration. Lancet Neurol (2013) 12:822–38. doi: 10.1016/S1474-4422(13)70124-8

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Faith DP. Conservation Evaluation and Phylogenetic Diversity. Biol Conserv (1992) 61(1):1–10. doi: 10.1016/0006-3207(92)91201-3

CrossRef Full Text | Google Scholar

39. Hill MO. Diversity and Evenness: A Unifying Notation and Its Consequences. Ecology (1973) 54(2):427–32. doi: 10.2307/1934352

CrossRef Full Text | Google Scholar

40. Chen T, Guestrin C. XGBoost: A Scalable Tree Boosting System. In: Proceedings of the ACM SIGKDD International Conference on Knowledge Discovery and Data Mining (2016) 785–94. doi: 10.1145/2939672.2939785

CrossRef Full Text | Google Scholar

41. Wang X-W, Liu Y-Y. Comparative Study of Classifiers for Human Microbiome Data. Med Microecol (2020) 4:100013. doi: 10.1016/j.medmic.2020.100013

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, et al. The Gut Microbiota Influences Blood-Brain Barrier Permeability in Mice. Sci Transl Med (2014) 6(263):263ra158. doi: 10.1126/scitranslmed.3009759

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Xaio H, Banks WA, Niehoff ML, Morley JE. Effect of LPS on the Permeability of the Blood–Brain Barrier to Insulin. Brain Res (2001) 896(1–2):36–42. doi: 10.1016/S0006-8993(00)03247-9

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Moissl-Eichinger C, Willing BP, Burke CM, Lukiw WJ. Bacteroides Fragilis Lipopolysaccharide and Inflammatory Signaling in Alzheimer’s Disease. Front Microbiol (2016) 7:1544. doi: 10.3389/fmicb.2016.01544

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Shen Y, Torchia MLG, Lawson GW, Karp CL, Ashwell JD, Mazmanian SK. Outer Membrane Vesicles of a Human Commensal Mediate Immune Regulation and Disease Protection. Cell Host Microbe (2012) 12(4):509–20. doi: 10.1016/j.chom.2012.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Wang Y, Begum-Haque S, Telesford KM, Ochoa-Repáraz J, Christy M, Kasper EJ, et al. A Commensal Bacterial Product Elicits and Modulates Migratory Capacity of CD39+ CD4 T Regulatory Subsets in the Suppression of Neuroinflammation. Gut Microbes (2014) 5(4):552–61. doi: 10.4161/gmic29797

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Venegas DP, de la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, et al. Short Chain Fatty Acids (SCFAs) Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol (2019) 10:277. doi: 10.3389/fimmu.2019.00277

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Wenzel TJ, Gates EJ, Ranger AL, Klegeris A. Short-Chain Fatty Acids (SCFAs) Alone or in Combination Regulate Select Immune Functions of Microglia-Like Cells. Mol Cell Neurosci (2020) 105:103493. doi: 10.1016/j.mcn.2020.103493

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Verberk IMW, Laarhuis MB, van den Bosch KA, Ebenau JL, van Leeuwenstijn M, Prins ND, et al. Serum Markers Glial Fibrillary Acidic Protein and Neurofilament Light for Prognosis and Monitoring in Cognitively Normal Older People: A Prospective Memory Clinic-Based Cohort Study. Lancet Heal Longev (2021) 2(2):e87–95. doi: 10.1016/S2666-7568(20)30061-1

CrossRef Full Text | Google Scholar

50. Claesson MJ, Jeffery IB, Conde S, Power SE, O’connor EM, Cusack S, et al. Gut Microbiota Composition Correlates With Diet and Health in the Elderly. Nature (2012) 488(7410):178–84. doi: 10.1038/nature11319

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Poehlman ET, Dvorak RV. Energy Expenditure, Energy Intake, and Weight Loss in Alzheimer Disease. Am J Clin Nutr (2000) 71(2):650S–5S. doi: 10.1093/ajcn/71.2.650s

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Doorduijn AS, Schueren MAE devd, Rest Ov de, Leeuw FA de, Hendriksen HMA, Teunissen CE, et al. Energy Intake and Expenditure in Patients With Alzheimer’s Disease and Mild Cognitive Impairment: The NUDAD Project. Alzheimer’s Res Ther (2020) 12(1):1–8. doi: 10.1186/s13195-020-00687-2

CrossRef Full Text | Google Scholar

53. Vieira AT, Fukumori C, Ferreira CM. New Insights Into Therapeutic Strategies for Gut Microbiota Modulation in Inflammatory Diseases. Clin Transl Immunol (2016) 5(6):e87. doi: 10.1038/cti.2016.38

CrossRef Full Text | Google Scholar

54. Meijnikman AS, Gerdes VE, Nieuwdorp M, Herrema H. Evaluating Causality of Gut Microbiota in Obesity and Diabetes in Humans. Endocr Rev (2018) 39(2):133–53. doi: 10.1210/er.2017-00192

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Groot P, Nikolic T, Pellegrini S, Sordi V, Imangaliyev S, Rampanelli E, et al. Faecal Microbiota Transplantation Halts Progression of Human New-Onset Type 1 Diabetes in a Randomised Controlled Trial. Gut (2021) 70(1):92–105. doi: 10.1136/gutjnl-2020-322630

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Hartstra AV, Schüppel V, Imangaliyev S, Schrantee A, Prodan A, Collard D, et al. Infusion of Donor Feces Affects the Gut–Brain Axis in Humans With Metabolic Syndrome. Mol Metab (2020) 42:101076. doi: 10.1016/j.molmet.2020.101076

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Kootte RS, Levin E, Salojärvi J, Smits LP, Hartstra AV, Udayappan SD, et al. Improvement of Insulin Sensitivity After Lean Donor Feces in Metabolic Syndrome Is Driven by Baseline Intestinal Microbiota Composition. Cell Metab (2017) 26(4):611–619.e6. doi: 10.1016/j.cmet.2017.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Chen J, Zaman A, Ramakrishna B, Olesen SW. Stool Banking for Fecal Microbiota Transplantation: Methods and Operations at a Large Stool Bank. Front Cell Infect Microbiol (2021) 0:281. doi: 10.3389/fcimb.2021.622949

CrossRef Full Text | Google Scholar

59. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and Prebiotics in Intestinal Health and Disease: From Biology to the Clinic. Nat Rev Gastroenterol Hepatol (2019) 16(10):605–16. doi: 10.1038/s41575-019-0173-3

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Zhu G, Zhao J, Zhang H, Chen W, Wang G. Probiotics for Mild Cognitive Impairment and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Foods (2021) 10(7):1672. doi: 10.3390/foods10071672

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Andrade JC, Almeida D, Domingos M, Seabra CL, Machado D, Freitas AC, et al. Commensal Obligate Anaerobic Bacteria and Health: Production, Storage, and Delivery Strategies. Front Bioeng Biotechnol (2020) 8:1–23. doi: 10.3389/fbioe.2020.00550

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Wijdeveld M, Nieuwdorp M, IJzerman R. The Interaction Between Microbiome and Host Central Nervous System: The Gut-Brain Axis as a Potential New Therapeutic Target in the Treatment of Obesity and Cardiometabolic Disease. Expert Opin Ther Targets (2020) 24(7):639–53. doi: 10.1080/14728222.2020.1761958

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Bouter KEC, Bakker GJ, Levin E, Hartstra AV, Kootte RS, Udayappan SD, et al. Differential Metabolic Effects of Oral Butyrate Treatment in Lean Versus Metabolic Syndrome Subjects Article. Clin Transl Gastroenterol (2018) 9(5):155. doi: 10.1038/s41424-018-0025-4

PubMed Abstract | CrossRef Full Text | Google Scholar


Plaats een reactie ...

Reageer op "Darmflora: Alzheimerpatiënten missen goede bacterie in hun darmen, die gezonde mensen wel hebben. Veel vezels en gevarieerd eten zou Alzheimer kunnen voorkomen"


Gerelateerde artikelen
 

Gerelateerde artikelen

Alcohol tast de darmbiotica >> Algemeen: Aangepast voedingspatroon >> Algemeen: Wat is er met ons >> Bloedverdunners die vitamine >> Caffeine: mensen met een dieet >> Darmflora: Alzheimerpatiënten >> Darmflora beinvloed door voeding >> Deze 6 wetenschappelijke onderzoeken >> Dierlijke vetten, roken en >> Een nieuw drinkvoedingsproduct, >>