2022
DOI: 10.1002/glia.24153
|View full text |Cite
|
Sign up to set email alerts
|

Viral mediated knockdown of GATA6 in SMA iPSC‐derived astrocytes prevents motor neuron loss and microglial activation

Abstract: Spinal muscular atrophy (SMA), a pediatric genetic disorder, is characterized by the profound loss of spinal cord motor neurons and subsequent muscle atrophy and death. Although the mechanisms underlying motor neuron loss are not entirely clear, data from our work and others support the idea that glial cells contribute to disease pathology. GATA6, a transcription factor that we have previously shown to be upregulated in SMA astrocytes, is negatively regulated by SMN (survival motor neuron) and can increase the… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

0
5
0

Year Published

2022
2022
2024
2024

Publication Types

Select...
7

Relationship

2
5

Authors

Journals

citations
Cited by 18 publications
(5 citation statements)
references
References 101 publications
0
5
0
Order By: Relevance
“…Using a microelectrode array (MEA) approach, we first wanted to determine if SMA patient astrocytes possess intrinsic defects in synaptic neuromodulation that could drive motor neuron dysfunction. The initial, independent derivation of iPSC‐derived spinal cord‐patterned motor neurons (Maury et al, 2015) and astrocytes (Allison et al, 2022) allowed for the assessment of different healthy and SMA patient co‐culture combinations to determine if changes in neural activity are motor neuron‐intrinsic or driven by diseased astrocytes. Specifically, we assessed if motor neuron activity phenotypes could be attributed to astrocytic cellular interactions (direct contact motor neuron‐astrocyte co‐cultures) and/or secreted factors (astrocyte conditioned media [ACM] treated motor neurons).…”
Section: Resultsmentioning
confidence: 99%
See 2 more Smart Citations
“…Using a microelectrode array (MEA) approach, we first wanted to determine if SMA patient astrocytes possess intrinsic defects in synaptic neuromodulation that could drive motor neuron dysfunction. The initial, independent derivation of iPSC‐derived spinal cord‐patterned motor neurons (Maury et al, 2015) and astrocytes (Allison et al, 2022) allowed for the assessment of different healthy and SMA patient co‐culture combinations to determine if changes in neural activity are motor neuron‐intrinsic or driven by diseased astrocytes. Specifically, we assessed if motor neuron activity phenotypes could be attributed to astrocytic cellular interactions (direct contact motor neuron‐astrocyte co‐cultures) and/or secreted factors (astrocyte conditioned media [ACM] treated motor neurons).…”
Section: Resultsmentioning
confidence: 99%
“…Overall, our work defines a novel SMN‐associated disease mechanism involving abnormal astrocyte glutamate transporter activity that diminishes motor neuron activity in SMA. In vivo, astrocytic‐driven dysfunction is likely to occur in combination with intrinsic motor neuron defects and sensory neuron‐mediated dysfunction, with further perturbation likely happening in other glial cell types including microglia (Allison et al, 2022; Khayrullina et al, 2022; Tarabal et al, 2014). Studies have already begun to test the efficacy of therapeutic compounds in order to increase motor neuron activity at the central afferent synapse (Kovalchuk et al, 2018; Simon et al, 2021).…”
Section: Discussionmentioning
confidence: 99%
See 1 more Smart Citation
“…LPS systemic administration is known to rapidly promote pro-inflammatory microglia phenotypes in the CNS [91], and the bidirectional communication of these populations is a crucial mediator in terms of delaying or accelerating the establishment of neuroinflammation [92]. In SMA, aberrant communication of these populations may contribute to disease pathology [93]. In our study, LPS systemic administration was strongly correlated with an increase in CNS pro-inflammatory microglia and astrocytes (defined as Iba-1 + iNOS + and GFAP + C3 + cells, respectively), and this effect, although present in both SMN∆7 and control mice, was more prominent for mice with experimental SMA compared to control mice.…”
Section: Discussionmentioning
confidence: 99%
“…In the nervous system, neuronal activation of NF-κB is critical for cell survival and differentiation, whereas non-neuronal cell activation of NF-κB has a detrimental effect on neurons, triggering inflammatory and cell death mechanisms ( Mincheva-Tasheva and Soler, 2013 ). For instance, increased phosphorylation and upregulation of NF-κB was described in SMN-depleted macrophages ( Ando et al, 2020 ) and human iPSC-derived SMA astrocytes ( Allison et al, 2022 ), suggesting that SMN protein may regulate the activation of these cells and the inflammatory response in SMA. In cultured MNs, pharmacological inhibition of NF-κB pathway or endogenous RelA knockdown reduces SMN level ( Arumugam et al, 2018 ), suggesting an active role of NF-κB in SMN expression in these cells.…”
Section: Discussionmentioning
confidence: 99%