A proof of concept study towards multi‐omics‐based computational drug repositioning in Alzheimer’s disease

Background Alzheimer’s disease (AD) is a devastating neurodegenerative disorder for which there currently are no disease‐modifying treatments available. To accelerate the path to effective intervention strategies, drug repositioning – the application of available compounds in a novel disease context...

Full description

Saved in:
Bibliographic Details
Published inAlzheimer's & dementia Vol. 17; pp. e056673 - n/a
Main Authors Wörheide, Maria A., Krumsiek, Jan, Kastenmüller, Gabi, Kaddurah‐Daouk, Rima F., Arnold, Matthias
Format Journal Article
LanguageEnglish
Published United States 01.12.2021
Online AccessGet full text

Cover

Loading…
Abstract Background Alzheimer’s disease (AD) is a devastating neurodegenerative disorder for which there currently are no disease‐modifying treatments available. To accelerate the path to effective intervention strategies, drug repositioning – the application of available compounds in a novel disease context – has gained increasing attention as a promising alternative to de novo drug development. Rich multi‐omics data, generated by large international and interdisciplinary AD consortia, is now enabling the implementation of novel methods that have the potential to drive the computational identification and prioritization of promising repositioning candidates. Method We recently developed the AD atlas, a web‐based multi‐omics resource that integrates multiple layers of heterogenous data from different studies and cohorts, including omics QTLs, transcriptomic, proteomic and metabolomic correlation networks, as well as genetic and multi‐omics associations with AD and associated biomarkers/endophenotypes. Using this atlas, we generated and analyzed molecular context networks surrounding AD‐associated genes as well as those targeted by drug repositioning candidates proposed in the literature. Subsequent enrichment analysis on AD subnetworks was used to identify drugs with overlapping molecular signatures on the gene expression level, while target networks of repositioning candidates were investigated for their potential involvement in AD pathogenesis. Result We found ample evidence for the potential of integrative multi‐omics approaches for drug repositioning in AD. For instance, enrichment analysis of the context network surrounding the AD‐associated genes APOE and CLU identified multiple repositioning candidates, where the top hits were drugs that were either previously proposed as promising or already subjected to clinical trials, such as fluoxetine, rosiglitazone, and valproate. Investigating candidate drugs, the exploration of the context network targeted by statins revealed functional links to TYROBP/TREM2 signaling, suggesting a potential protective effect of this drug class through modulation of neuroinflammatory pathways. Conclusion Our results highlight multiple opportunities to advance drug repositioning efforts in AD by integrative analysis of comprehensive multi‐omics data. Automation of our analyses using network‐based machine learning approaches and extension of the AD atlas with multi‐omics data from drug screens to resolve directionalities will allow us to globally identify molecular pathways disturbed in AD that are targetable by drug repositioning candidates.
AbstractList Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which there currently are no disease-modifying treatments available. To accelerate the path to effective intervention strategies, drug repositioning - the application of available compounds in a novel disease context - has gained increasing attention as a promising alternative to de novo drug development. Rich multi-omics data, generated by large international and interdisciplinary AD consortia, is now enabling the implementation of novel methods that have the potential to drive the computational identification and prioritization of promising repositioning candidates. We recently developed the AD atlas, a web-based multi-omics resource that integrates multiple layers of heterogenous data from different studies and cohorts, including omics QTLs, transcriptomic, proteomic and metabolomic correlation networks, as well as genetic and multi-omics associations with AD and associated biomarkers/endophenotypes. Using this atlas, we generated and analyzed molecular context networks surrounding AD-associated genes as well as those targeted by drug repositioning candidates proposed in the literature. Subsequent enrichment analysis on AD subnetworks was used to identify drugs with overlapping molecular signatures on the gene expression level, while target networks of repositioning candidates were investigated for their potential involvement in AD pathogenesis. We found ample evidence for the potential of integrative multi-omics approaches for drug repositioning in AD. For instance, enrichment analysis of the context network surrounding the AD-associated genes APOE and CLU identified multiple repositioning candidates, where the top hits were drugs that were either previously proposed as promising or already subjected to clinical trials, such as fluoxetine, rosiglitazone, and valproate. Investigating candidate drugs, the exploration of the context network targeted by statins revealed functional links to TYROBP/TREM2 signaling, suggesting a potential protective effect of this drug class through modulation of neuroinflammatory pathways. Our results highlight multiple opportunities to advance drug repositioning efforts in AD by integrative analysis of comprehensive multi-omics data. Automation of our analyses using network-based machine learning approaches and extension of the AD atlas with multi-omics data from drug screens to resolve directionalities will allow us to globally identify molecular pathways disturbed in AD that are targetable by drug repositioning candidates.
Background Alzheimer’s disease (AD) is a devastating neurodegenerative disorder for which there currently are no disease‐modifying treatments available. To accelerate the path to effective intervention strategies, drug repositioning – the application of available compounds in a novel disease context – has gained increasing attention as a promising alternative to de novo drug development. Rich multi‐omics data, generated by large international and interdisciplinary AD consortia, is now enabling the implementation of novel methods that have the potential to drive the computational identification and prioritization of promising repositioning candidates. Method We recently developed the AD atlas, a web‐based multi‐omics resource that integrates multiple layers of heterogenous data from different studies and cohorts, including omics QTLs, transcriptomic, proteomic and metabolomic correlation networks, as well as genetic and multi‐omics associations with AD and associated biomarkers/endophenotypes. Using this atlas, we generated and analyzed molecular context networks surrounding AD‐associated genes as well as those targeted by drug repositioning candidates proposed in the literature. Subsequent enrichment analysis on AD subnetworks was used to identify drugs with overlapping molecular signatures on the gene expression level, while target networks of repositioning candidates were investigated for their potential involvement in AD pathogenesis. Result We found ample evidence for the potential of integrative multi‐omics approaches for drug repositioning in AD. For instance, enrichment analysis of the context network surrounding the AD‐associated genes APOE and CLU identified multiple repositioning candidates, where the top hits were drugs that were either previously proposed as promising or already subjected to clinical trials, such as fluoxetine, rosiglitazone, and valproate. Investigating candidate drugs, the exploration of the context network targeted by statins revealed functional links to TYROBP/TREM2 signaling, suggesting a potential protective effect of this drug class through modulation of neuroinflammatory pathways. Conclusion Our results highlight multiple opportunities to advance drug repositioning efforts in AD by integrative analysis of comprehensive multi‐omics data. Automation of our analyses using network‐based machine learning approaches and extension of the AD atlas with multi‐omics data from drug screens to resolve directionalities will allow us to globally identify molecular pathways disturbed in AD that are targetable by drug repositioning candidates.
Author Wörheide, Maria A.
Arnold, Matthias
Krumsiek, Jan
Kaddurah‐Daouk, Rima F.
Kastenmüller, Gabi
Author_xml – sequence: 1
  givenname: Maria A.
  surname: Wörheide
  fullname: Wörheide, Maria A.
  email: maria.woerheide@helmholtz-muenchen.de
  organization: Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health
– sequence: 2
  givenname: Jan
  surname: Krumsiek
  fullname: Krumsiek, Jan
  organization: Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine
– sequence: 3
  givenname: Gabi
  surname: Kastenmüller
  fullname: Kastenmüller, Gabi
  organization: German Center for Diabetes Research (DZD)
– sequence: 4
  givenname: Rima F.
  surname: Kaddurah‐Daouk
  fullname: Kaddurah‐Daouk, Rima F.
  organization: Duke University
– sequence: 5
  givenname: Matthias
  surname: Arnold
  fullname: Arnold, Matthias
  organization: Duke University
BackLink https://www.ncbi.nlm.nih.gov/pubmed/35109201$$D View this record in MEDLINE/PubMed
BookMark eNo9kE1qwzAQhUVJaX7aTQ9QdAGnkmXZ1tKE_kGgm3bTjZGtUapiW8KyCckqR-i218tJ6uA2MPAeM28G5pujSWMbQOiWkiUlJLyX1X5JeBwn7ALNKOdhwMNETM4-JlM09_6LkIiklF-hKeOUiJDQGXIZdq21Gg9V2qYE12Hf9WqHO7uVrfK47qvOHA_ftjalH7SQHtSQrV3fyc7YRlZYtf0Gt-CsN6eOaTbYNDir9p9gamiPhx-PlfEwrF6jSy0rDzd_ukDvjw9vq-dg_fr0ssrWgaOMs6BQKUSxLkIJnBU8DnVEmCgEFBp0JKUMY6m1SBNBSpGAYjFLRZkIVUqesiRlC3Q33nV9UYPKXWtq2e7y_9eHAB0DW1PB7jynJD9BzQeo-Qg1z9Yfo2O_aiNxfw
ContentType Journal Article
Copyright 2021 the Alzheimer's Association
2021 the Alzheimer's Association.
Copyright_xml – notice: 2021 the Alzheimer's Association
– notice: 2021 the Alzheimer's Association.
DBID NPM
DOI 10.1002/alz.056673
DatabaseName PubMed
DatabaseTitle PubMed
DatabaseTitleList PubMed

Database_xml – sequence: 1
  dbid: NPM
  name: PubMed
  url: https://proxy.k.utb.cz/login?url=http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=PubMed
  sourceTypes: Index Database
DeliveryMethod fulltext_linktorsrc
EISSN 1552-5279
EndPage n/a
ExternalDocumentID 35109201
ALZ056673
Genre article
Journal Article
GroupedDBID ---
--K
--M
.~1
0R~
1B1
1OC
1~.
1~5
24P
33P
4.4
457
4G.
53G
5VS
7-5
71M
7RV
7X7
8FI
8FJ
8P~
AACTN
AAEDT
AAHHS
AAIKJ
AAKOC
AALRI
AANLZ
AAOAW
AAXLA
AAXUO
ABBQC
ABCQJ
ABCUV
ABIVO
ABJNI
ABMAC
ABMZM
ABUWG
ACCFJ
ACCZN
ACGFS
ACGOF
ACPOU
ACXQS
ADBBV
ADBTR
ADEZE
ADHUB
ADKYN
ADMUD
ADPDF
ADVLN
ADZMN
ADZOD
AEEZP
AEIGN
AEKER
AENEX
AEQDE
AEUYR
AEVXI
AFKRA
AFTJW
AGHFR
AGUBO
AGWIK
AGYEJ
AITUG
AIURR
AIWBW
AJBDE
AJOXV
AJRQY
AKRWK
ALMA_UNASSIGNED_HOLDINGS
ALUQN
AMFUW
AMRAJ
AMYDB
ANZVX
AZQEC
BENPR
BFHJK
BLXMC
C45
CCPQU
DCZOG
EBS
EJD
EMOBN
EO8
EO9
EP2
EP3
F5P
FDB
FEDTE
FIRID
FNPLU
FYUFA
G-Q
GBLVA
HMCUK
HVGLF
HX~
HZ~
IHE
J1W
K9-
LATKE
LEEKS
M0R
M41
MO0
MOBAO
N9A
NAPCQ
O-L
O9-
OAUVE
OVD
OVEED
OZT
P-8
P-9
P2P
PC.
PGMZT
PIMPY
PSYQQ
Q38
QTD
RIG
ROL
RPM
RPZ
SDF
SDG
SEL
SES
SSZ
SUPJJ
T5K
TEORI
UKHRP
~G-
NPM
ID FETCH-LOGICAL-p1353-bd8e46fb2ae53b562f4039b9ebfef4aaa26aff98790c97ed36389c79dca583783
ISSN 1552-5260
IngestDate Sat Sep 28 08:27:41 EDT 2024
Sat Aug 24 00:57:39 EDT 2024
IsPeerReviewed true
IsScholarly true
Language English
License 2021 the Alzheimer's Association.
LinkModel OpenURL
MergedId FETCHMERGED-LOGICAL-p1353-bd8e46fb2ae53b562f4039b9ebfef4aaa26aff98790c97ed36389c79dca583783
PMID 35109201
PageCount 1
ParticipantIDs pubmed_primary_35109201
wiley_primary_10_1002_alz_056673_ALZ056673
PublicationCentury 2000
PublicationDate December 2021
2021-Dec
PublicationDateYYYYMMDD 2021-12-01
PublicationDate_xml – month: 12
  year: 2021
  text: December 2021
PublicationDecade 2020
PublicationPlace United States
PublicationPlace_xml – name: United States
PublicationTitle Alzheimer's & dementia
PublicationTitleAlternate Alzheimers Dement
PublicationYear 2021
SSID ssj0040815
Score 2.3646595
Snippet Background Alzheimer’s disease (AD) is a devastating neurodegenerative disorder for which there currently are no disease‐modifying treatments available. To...
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which there currently are no disease-modifying treatments available. To accelerate the...
SourceID pubmed
wiley
SourceType Index Database
Publisher
StartPage e056673
Title A proof of concept study towards multi‐omics‐based computational drug repositioning in Alzheimer’s disease
URI https://onlinelibrary.wiley.com/doi/abs/10.1002%2Falz.056673
https://www.ncbi.nlm.nih.gov/pubmed/35109201
Volume 17
hasFullText 1
inHoldings 1
isFullTextHit
isPrint
link http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1La9wwEBbbBEouJaWvtGnRoVBoULqR_Dy6ybahz0tSQi9GsqTETdZZ1uvL_oH-7Y4efrEttIXFGFuWF83H-JM08w1CLxM91RKYOEngY0UCoSnhMeMkSEJ-FBZUcivH8PlLdHoefLgILyaTn4OopWYlDov1b_NK_seqcA3sarJk_8GyXadwAc7BvnAEC8Pxr2ycmfAqYHs2NNymHzq5WCCUJhi2duGCxCQe18R8r2wK26JZtSuActlc2m0DF7nl81uym_WVKm1ZlbgebeG0crWjBgY80q4ylr2PNxvwb6PllZHR8jlBJe8XTj8um3ldqmsXqNtHAnAAXTW3Dx-3SYrvuSj7BlI24B_JCb9trp0ywJz78GS_eEGPBoEgyjvc0EyGXUGZziPHB7am6QEbuFYFVC1yZU823L6TkeU368PNRmCyxdwCgIH_SWn7-pHIdnvrDtqG_xLCLH7767fZ7KT9qAfAnMJO3Za-6V-1g-62Dw9IzHCiY5nK2S6656cYOHN4uY8mqnqAfmTYYgXDz2MFW6xgjxW8gRU8wgo2WMEjrOCywh0UXtXYI-UhOn83Ozs-Jb7OBlmYqidEyEQFkRaUq5AJIMQ6mLJUpEpopQPOOY241mkSp9MijZVkhuUWcSoLHpp6BOwR2qpuK_UEYRnJhCkgRYxT6JKmSaSKUCVGZY7ySO-hx26I8oUTU8nbwdtDr-2YdTecnjbNYahzN9R59um7O3v6x26eoZ0eZ_toa7Vs1HOgjSvxwpv0F4UGbqQ
link.rule.ids 315,786,790,27957,27958
linkProvider Ovid
openUrl ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=A+proof+of+concept+study+towards+multi-omics-based+computational+drug+repositioning+in+Alzheimer%27s+disease&rft.jtitle=Alzheimer%27s+%26+dementia&rft.au=W%C3%B6rheide%2C+Maria+A&rft.au=Krumsiek%2C+Jan&rft.au=Kastenm%C3%BCller%2C+Gabi&rft.au=Kaddurah-Daouk%2C+Rima+F&rft.date=2021-12-01&rft.eissn=1552-5279&rft.volume=17+Suppl+3&rft.spage=e056673&rft_id=info:doi/10.1002%2Falz.056673&rft_id=info%3Apmid%2F35109201&rft.externalDocID=35109201
thumbnail_l http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=1552-5260&client=summon
thumbnail_m http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=1552-5260&client=summon
thumbnail_s http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=1552-5260&client=summon