Your browser doesn't support javascript.
loading
TREM2 Alzheimer's variant R47H causes similar transcriptional dysregulation to knockout, yet only subtle functional phenotypes in human iPSC-derived macrophages.
Hall-Roberts, Hazel; Agarwal, Devika; Obst, Juliane; Smith, Thomas B; Monzón-Sandoval, Jimena; Di Daniel, Elena; Webber, Caleb; James, William S; Mead, Emma; Davis, John B; Cowley, Sally A.
Afiliação
  • Hall-Roberts H; James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK. hazel.hall-roberts@path.ox.ac.uk.
  • Agarwal D; Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK. hazel.hall-roberts@path.ox.ac.uk.
  • Obst J; Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK.
  • Smith TB; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
  • Monzón-Sandoval J; Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK.
  • Di Daniel E; Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK.
  • Webber C; UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
  • James WS; Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK.
  • Mead E; UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
  • Davis JB; James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
  • Cowley SA; Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK.
Alzheimers Res Ther ; 12(1): 151, 2020 11 16.
Article em En | MEDLINE | ID: mdl-33198789
ABSTRACT

BACKGROUND:

TREM2 is a microglial cell surface receptor, with risk mutations linked to Alzheimer's disease (AD), including R47H. TREM2 signalling via SYK aids phagocytosis, chemotaxis, survival, and changes to microglial activation state. In AD mouse models, knockout (KO) of TREM2 impairs microglial clustering around amyloid and prevents microglial activation. The R47H mutation is proposed to reduce TREM2 ligand binding. We investigated cell phenotypes of the R47H mutant and TREM2 KO in a model of human microglia, and compared their transcriptional signatures, to determine the mechanism by which R47H TREM2 disrupts function.

METHODS:

We generated human microglia-like iPSC-macrophages (pMac) from isogenic induced pluripotent stem cell (iPSC) lines, with homozygous R47H mutation or TREM2 knockout (KO). We firstly validated the effect of the R47H mutant on TREM2 surface and subcellular localization in pMac. To assess microglial phenotypic function, we measured phagocytosis of dead neurons, cell morphology, directed migration, survival, and LPS-induced inflammation. We performed bulk RNA-seq, comparing significant differentially expressed genes (DEGs; p < 0.05) between the R47H and KO versus WT, and bioinformatically predicted potential upstream regulators of TREM2-mediated gene expression.

RESULTS:

R47H modified surface expression and shedding of TREM2, but did not impair TREM2-mediated signalling, or gross phenotypes that were dysregulated in the TREM2 KO (phagocytosis, motility, survival). However, altered gene expression in the R47H TREM2 pMac overlapped by 90% with the TREM2 KO and was characterised by dysregulation of genes involved with immunity, proliferation, activation, chemotaxis, and adhesion. Downregulated mediators of ECM adhesion included the vitronectin receptor αVß3, and consequently, R47H TREM2 pMac adhered weakly to vitronectin compared with WT pMac. To counteract these transcriptional defects, we investigated TGFß1, as a candidate upstream regulator. TGFß1 failed to rescue vitronectin adhesion of pMac, although it improved αVß3 expression.

CONCLUSIONS:

The R47H mutation is not sufficient to cause gross phenotypic defects of human pMac under standard culture conditions. However, overlapping transcriptional defects with TREM2 KO supports the hypothesised partial loss-of-function effects of the R47H mutation. Furthermore, transcriptomics can guide us to more subtle phenotypic defects in the R47H cells, such as reduced cell adhesion, and can be used to predict targets for therapeutic intervention.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Células-Tronco Pluripotentes Induzidas / Doença de Alzheimer Tipo de estudo: Etiology_studies / Prognostic_studies Limite: Humans Idioma: En Revista: Alzheimers Res Ther Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Reino Unido

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Células-Tronco Pluripotentes Induzidas / Doença de Alzheimer Tipo de estudo: Etiology_studies / Prognostic_studies Limite: Humans Idioma: En Revista: Alzheimers Res Ther Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Reino Unido