Mitochondrial aspartate-glutamate carrier isoform 1 (AGC1) deficiency is an ultra-rare genetic disease characterized by global hypomyelination and brain atrophy, caused by mutations in the SLC25A12 gene leading to a reduction in AGC1 activity. In both neuronal precursor cells and oligodendrocytes precursor cells (NPCs and OPCs), the AGC1 determines reduced proliferation with an accelerated differentiation of OPCs, both associated with gene expression dysregulation. Epigenetic regulation of gene expression through histone acetylation plays a crucial role in the proliferation/differentiation of both NPCs and OPCs and is modulated by mitochondrial metabolism. In AGC1 deficiency models, both OPCs and NPCs show an altered expression of transcription factors involved in the proliferation/differentiation of brain precursor cells (BPCs) as well as a reduction in histone acetylation with a parallel alteration in the expression and activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs). In this study, histone acetylation dysfunctions have been dissected in in vitro models of AGC1 deficiency OPCs (Oli-Neu cells) and NPCs (neurospheres), in physiological conditions and following pharmacological treatments. The inhibition of HATs by curcumin arrests the proliferation of OPCs leading to their differentiation, while the inhibition of HDACs by suberanilohydroxamic acid (SAHA) has only a limited effect on proliferation, but it significantly stimulates the differentiation of OPCs. In NPCs, both treatments determine an alteration in the commitment toward glial cells. These data contribute to clarifying the molecular and epigenetic mechanisms regulating the proliferation/differentiation of OPCs and NPCs. This will help to identify potential targets for new therapeutic approaches that are able to increase the OPCs pool and to sustain their differentiation toward oligodendrocytes and to myelination/remyelination processes in AGC1 deficiency, as well as in other white matter neuropathologies.

Histone Acetylation Defects in Brain Precursor Cells: A Potential Pathogenic Mechanism Causing Proliferation and Differentiation Dysfunctions in Mitochondrial Aspartate-Glutamate Carrier Isoform 1 Deficiency / Poeta E.; Petralla S.; Babini G.; Renzi B.; Celauro L.; Magnifico M.C.; Barile S.N.; Masotti M.; De Chirico F.; Massenzio F.; Viggiano L.; Palmieri L.; Virgili M.; Lasorsa F.M.; Monti B.. - In: FRONTIERS IN CELLULAR NEUROSCIENCE. - ISSN 1662-5102. - ELETTRONICO. - 15:(2022), pp. 773709.1-773709.23. [10.3389/fncel.2021.773709]

Histone Acetylation Defects in Brain Precursor Cells: A Potential Pathogenic Mechanism Causing Proliferation and Differentiation Dysfunctions in Mitochondrial Aspartate-Glutamate Carrier Isoform 1 Deficiency

Poeta E.
Primo
Writing – Original Draft Preparation
;
Petralla S.
Secondo
Investigation
;
Babini G.
Membro del Collaboration Group
;
Renzi B.
Membro del Collaboration Group
;
Masotti M.
Membro del Collaboration Group
;
De Chirico F.
Membro del Collaboration Group
;
Massenzio F.
Membro del Collaboration Group
;
Virgili M.
Membro del Collaboration Group
;
Monti B.
Ultimo
Conceptualization
2022

Abstract

Mitochondrial aspartate-glutamate carrier isoform 1 (AGC1) deficiency is an ultra-rare genetic disease characterized by global hypomyelination and brain atrophy, caused by mutations in the SLC25A12 gene leading to a reduction in AGC1 activity. In both neuronal precursor cells and oligodendrocytes precursor cells (NPCs and OPCs), the AGC1 determines reduced proliferation with an accelerated differentiation of OPCs, both associated with gene expression dysregulation. Epigenetic regulation of gene expression through histone acetylation plays a crucial role in the proliferation/differentiation of both NPCs and OPCs and is modulated by mitochondrial metabolism. In AGC1 deficiency models, both OPCs and NPCs show an altered expression of transcription factors involved in the proliferation/differentiation of brain precursor cells (BPCs) as well as a reduction in histone acetylation with a parallel alteration in the expression and activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs). In this study, histone acetylation dysfunctions have been dissected in in vitro models of AGC1 deficiency OPCs (Oli-Neu cells) and NPCs (neurospheres), in physiological conditions and following pharmacological treatments. The inhibition of HATs by curcumin arrests the proliferation of OPCs leading to their differentiation, while the inhibition of HDACs by suberanilohydroxamic acid (SAHA) has only a limited effect on proliferation, but it significantly stimulates the differentiation of OPCs. In NPCs, both treatments determine an alteration in the commitment toward glial cells. These data contribute to clarifying the molecular and epigenetic mechanisms regulating the proliferation/differentiation of OPCs and NPCs. This will help to identify potential targets for new therapeutic approaches that are able to increase the OPCs pool and to sustain their differentiation toward oligodendrocytes and to myelination/remyelination processes in AGC1 deficiency, as well as in other white matter neuropathologies.
2022
Histone Acetylation Defects in Brain Precursor Cells: A Potential Pathogenic Mechanism Causing Proliferation and Differentiation Dysfunctions in Mitochondrial Aspartate-Glutamate Carrier Isoform 1 Deficiency / Poeta E.; Petralla S.; Babini G.; Renzi B.; Celauro L.; Magnifico M.C.; Barile S.N.; Masotti M.; De Chirico F.; Massenzio F.; Viggiano L.; Palmieri L.; Virgili M.; Lasorsa F.M.; Monti B.. - In: FRONTIERS IN CELLULAR NEUROSCIENCE. - ISSN 1662-5102. - ELETTRONICO. - 15:(2022), pp. 773709.1-773709.23. [10.3389/fncel.2021.773709]
Poeta E.; Petralla S.; Babini G.; Renzi B.; Celauro L.; Magnifico M.C.; Barile S.N.; Masotti M.; De Chirico F.; Massenzio F.; Viggiano L.; Palmieri L.; Virgili M.; Lasorsa F.M.; Monti B.
File in questo prodotto:
File Dimensione Formato  
fncel-15-773709.pdf

accesso aperto

Tipo: Versione (PDF) editoriale
Licenza: Licenza per accesso libero gratuito
Dimensione 9.1 MB
Formato Adobe PDF
9.1 MB Adobe PDF Visualizza/Apri

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11585/859911
Citazioni
  • ???jsp.display-item.citation.pmc??? 0
  • Scopus 0
  • ???jsp.display-item.citation.isi??? 0
social impact