Equivalencies: 0 | Classes: 0 | Children: 0 | Explore

Entity

Name
PPP1CA
Namespace
HGNC
Namespace Version
20180215
Namespace URL
https://arty.scai.fraunhofer.de/artifactory/bel/namespace/hgnc/hgnc-20180215.belns

Appears in Networks 2

Tau oligomers and tau toxicity in neurodegenerative disease v1.0.0

Tau oligomers and tau toxicity in neurodegenerative disease by Ward et al., 2012

Tau Modifications v1.9.5

Tau Modifications Sections of NESTOR

In-Edges 6

a(HBP:"Tau aggregates") increases act(p(HGNC:PPP1CA)) View Subject | View Object

However, when tau aggregates, this conformation is altered, exposing PAD and allowing activation of the PP1/GSK3 signalling pathway facilitating FAT inhibition [21] PubMed:22817713

a(CHEBI:"quinolinic acid") decreases p(HGNC:PPP1CA) View Subject | View Object

Kynurenic pathway is overactive in AD. QA is co-localized with hyperphosphorylated tau within cortical neurons in AD brain. In primary cultures of human neurons, QA treatment increased tau phosphorylation at serine 199/202, threonine 231 and serine 396/404 in a dose dependent manner. This increase in tau phosphorylation was paralleled by a substantial decrease in the total protein phosphatase activity, mostly in PP2A expression and modest in PP1. PubMed:19623258

Appears in Networks:

bp(GO:"axonal transport") negativeCorrelation act(p(HGNC:PPP1CA)) View Subject | View Object

Filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT PubMed:21734277

Appears in Networks:

m(MIRBASE:"hsa-mir-125b-1") directlyDecreases act(p(HGNC:PPP1CA)) View Subject | View Object

In primary neurons, overexpression of miR-125b causes tau hyperphosphorylation and an upregulation of p35, cdk5, and p44/42-MAPK signaling. In parallel, the phosphatases DUSP6 and PPP1CA and the anti-apoptotic factor Bcl-W are downregulated as direct targets of miR-125b. Knockdown of these phosphatases induces tau hyperphosphorylation, and overexpression of PPP1CA and Bcl-W prevents miR-125b-induced tau phosphorylation, suggesting that they mediate the effects of miR-125b on tau. Conversely, suppression of miR-125b in neurons by tough decoys reduces tau phosphorylation and kinase expression/activity. Injecting miR-125b into the hippocampus of mice impairs associative learning and is accompanied by downregulation of Bcl-W, DUSP6, and PPP1CA, resulting in increased tau phosphorylation in vivo. Importantly, DUSP6 and PPP1CA are also reduced in AD brains. PubMed:25001178

Appears in Networks:

p(HBP:"phosphatase-activating domain") directlyIncreases act(p(HGNC:PPP1CA)) View Subject | View Object

Filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT PubMed:21734277

Appears in Networks:

p(HGNC:PPP1R15A) regulates act(p(HGNC:PPP1CA)) View Subject | View Object

The growth arrest and DNA damage protein (GADD) 34 is up-regulated in response to ER stress and regulates subunit of protein phosphatase 1 (PP1) complex that dephosphorylates eukaryotic translation initiator factor 2α (elF2α). PubMed:26142647

Appears in Networks:

Out-Edges 11

p(HGNC:PPP1CA) decreases bp(GO:"anterograde axonal protein transport") View Subject | View Object

Further investigation illustrated that this inhibition occurs via activation of a signalling cascade involving PP1 (protein phosphatase 1) and GSK3 (glycogen synthase kinase 3) [24] PubMed:22817713

p(HGNC:PPP1CA) directlyDecreases p(HGNC:TARDBP, pmod(Ph, Ser, 397)) View Subject | View Object

We recently found that overexpression of TDP-43 enhances tau exon 10 inclusion and tau mRNA instability. Overexpression of PP1α and PP1γ, but not PP1β, suppressed TDP-43 phosphorylation at Ser403/404 and Ser409/410 and TDP-43-induced tau exon 10 inclusion. PubMed:29334120

Appears in Networks:

p(HGNC:PPP1CA) directlyDecreases p(HGNC:TARDBP, pmod(Ph, Ser, 403)) View Subject | View Object

We recently found that overexpression of TDP-43 enhances tau exon 10 inclusion and tau mRNA instability. Overexpression of PP1α and PP1γ, but not PP1β, suppressed TDP-43 phosphorylation at Ser403/404 and Ser409/410 and TDP-43-induced tau exon 10 inclusion. PubMed:29334120

Appears in Networks:

p(HGNC:PPP1CA) directlyDecreases p(HGNC:TARDBP, pmod(Ph, Ser, 404)) View Subject | View Object

We recently found that overexpression of TDP-43 enhances tau exon 10 inclusion and tau mRNA instability. Overexpression of PP1α and PP1γ, but not PP1β, suppressed TDP-43 phosphorylation at Ser403/404 and Ser409/410 and TDP-43-induced tau exon 10 inclusion. PubMed:29334120

Appears in Networks:

p(HGNC:PPP1CA) directlyDecreases p(HGNC:TARDBP, pmod(Ph, Ser, 409)) View Subject | View Object

We recently found that overexpression of TDP-43 enhances tau exon 10 inclusion and tau mRNA instability. Overexpression of PP1α and PP1γ, but not PP1β, suppressed TDP-43 phosphorylation at Ser403/404 and Ser409/410 and TDP-43-induced tau exon 10 inclusion. PubMed:29334120

Appears in Networks:

p(HGNC:PPP1CA) directlyDecreases p(HGNC:TARDBP, pmod(Ph, Ser, 410)) View Subject | View Object

We recently found that overexpression of TDP-43 enhances tau exon 10 inclusion and tau mRNA instability. Overexpression of PP1α and PP1γ, but not PP1β, suppressed TDP-43 phosphorylation at Ser403/404 and Ser409/410 and TDP-43-induced tau exon 10 inclusion. PubMed:29334120

Appears in Networks:

act(p(HGNC:PPP1CA)) directlyDecreases p(HGNC:EIF2S1, pmod(Ph)) View Subject | View Object

The growth arrest and DNA damage protein (GADD) 34 is up-regulated in response to ER stress and regulates subunit of protein phosphatase 1 (PP1) complex that dephosphorylates eukaryotic translation initiator factor 2α (elF2α). PubMed:26142647

Appears in Networks:

act(p(HGNC:PPP1CA)) directlyDecreases p(HGNC:GSK3B, pmod(Ph, Ser, 9)) View Subject | View Object

Filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT PubMed:21734277

Appears in Networks:

act(p(HGNC:PPP1CA)) increases act(p(HGNC:GSK3B)) View Subject | View Object

Filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT PubMed:21734277

Appears in Networks:

act(p(HGNC:PPP1CA)) negativeCorrelation bp(GO:"axonal transport") View Subject | View Object

Filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT PubMed:21734277

Appears in Networks:

act(p(HGNC:PPP1CA)) directlyDecreases p(HGNC:MAPT, pmod(HBP:hyperphosphorylation)) View Subject | View Object

In primary neurons, overexpression of miR-125b causes tau hyperphosphorylation and an upregulation of p35, cdk5, and p44/42-MAPK signaling. In parallel, the phosphatases DUSP6 and PPP1CA and the anti-apoptotic factor Bcl-W are downregulated as direct targets of miR-125b. Knockdown of these phosphatases induces tau hyperphosphorylation, and overexpression of PPP1CA and Bcl-W prevents miR-125b-induced tau phosphorylation, suggesting that they mediate the effects of miR-125b on tau. Conversely, suppression of miR-125b in neurons by tough decoys reduces tau phosphorylation and kinase expression/activity. Injecting miR-125b into the hippocampus of mice impairs associative learning and is accompanied by downregulation of Bcl-W, DUSP6, and PPP1CA, resulting in increased tau phosphorylation in vivo. Importantly, DUSP6 and PPP1CA are also reduced in AD brains. PubMed:25001178

Appears in Networks:

About

BEL Commons is developed and maintained in an academic capacity by Charles Tapley Hoyt and Daniel Domingo-Fernández at the Fraunhofer SCAI Department of Bioinformatics with support from the IMI project, AETIONOMY. It is built on top of PyBEL, an open source project. Please feel free to contact us here to give us feedback or report any issues. Also, see our Publishing Notes and Data Protection information.

If you find BEL Commons useful in your work, please consider citing: Hoyt, C. T., Domingo-Fernández, D., & Hofmann-Apitius, M. (2018). BEL Commons: an environment for exploration and analysis of networks encoded in Biological Expression Language. Database, 2018(3), 1–11.