Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation

Phosphoglycerate mutase 2 (PGAM2) is a critical glycolytic enzyme that is highly expressed in skeletal muscle. In humans, naturally occurring mutations in Phosphoglycerate mutase 2 have been etiologically linked to glycogen storage disease X (GSDX). Phosphoglycerate mutase 2 activity is regulated by...

Full description

Bibliographic Details
Main Authors: Yi Zhang, Ilimbek Beketaev, Yanlin Ma, Jun Wang
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-12-01
Series:Frontiers in Cell and Developmental Biology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fcell.2022.1052363/full
_version_ 1811200169134260224
author Yi Zhang
Yi Zhang
Ilimbek Beketaev
Yanlin Ma
Yanlin Ma
Jun Wang
author_facet Yi Zhang
Yi Zhang
Ilimbek Beketaev
Yanlin Ma
Yanlin Ma
Jun Wang
author_sort Yi Zhang
collection DOAJ
description Phosphoglycerate mutase 2 (PGAM2) is a critical glycolytic enzyme that is highly expressed in skeletal muscle. In humans, naturally occurring mutations in Phosphoglycerate mutase 2 have been etiologically linked to glycogen storage disease X (GSDX). Phosphoglycerate mutase 2 activity is regulated by several posttranslational modifications such as ubiquitination and acetylation. Here, we report that Phosphoglycerate mutase 2 activity is regulated by sumoylation—a covalent conjugation involved in a wide spectrum of cellular events. We found that Phosphoglycerate mutase 2 contains two primary SUMO acceptor sites, lysine (K)49 and K176, and that the mutation of either K to arginine (R) abolished Phosphoglycerate mutase 2 sumoylation. Given that K176 is more highly evolutionarily conserved across paralogs and orthologs than K49 is, we used the CRISPR-mediated homologous recombination technique in myogenic C2C12 cells to generate homozygous K176R knock-in cells (PGAM2K176R/K176R). Compared with wild-type (WT) C2C12 cells, PGAM2K176R/K176R C2C12 cells exhibited impaired myogenic differentiation, as indicated by decreased differentiation and fusion indexes. Furthermore, the results of glycolytic and mitochondrial stress assays with the XF96 Extracellular Flux analyzer revealed a reduced proton efflux rate (PER), glycolytic PER (glycoPER), extracellular acidification rate (ECAR), and oxygen consumption rate (OCR) in PGAM2K176R/K176R C2C12 cells, both at baseline and in response to stress. Impaired mitochondrial function was also observed in PGAM2K176R/K176R P19 cells, a carcinoma cell line. These findings indicate that the PGAM2-K176R mutation impaired glycolysis and mitochondrial function. Gene ontology term analysis of RNA sequencing data further revealed that several downregulated genes in PGAM2K176R/K176R C2C12 cells were associated with muscle differentiation/development/contraction programs. Finally, PGAM2 with either of two naturally occurring missense mutations linked to GSDX, E89A (conversion of glutamic acid 89 to alanine) or R90W (conversion of arginine 90 to tryptophan), exhibited reduced Phosphoglycerate mutase 2 sumoylation. Thus, sumoylation is an important mechanism that mediates Phosphoglycerate mutase 2 activity and is potentially implicated in Phosphoglycerate mutase 2 mutation-linked disease in humans.
first_indexed 2024-04-12T02:00:19Z
format Article
id doaj.art-6c1d9d271d7e4d99a5ad40f404f1daf7
institution Directory Open Access Journal
issn 2296-634X
language English
last_indexed 2024-04-12T02:00:19Z
publishDate 2022-12-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Cell and Developmental Biology
spelling doaj.art-6c1d9d271d7e4d99a5ad40f404f1daf72022-12-22T03:52:41ZengFrontiers Media S.A.Frontiers in Cell and Developmental Biology2296-634X2022-12-011010.3389/fcell.2022.10523631052363Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiationYi Zhang0Yi Zhang1Ilimbek Beketaev2Yanlin Ma3Yanlin Ma4Jun Wang5Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, ChinaStem Cell Engineering, Texas Heart Institute, Houston, TX, United StatesStem Cell Engineering, Texas Heart Institute, Houston, TX, United StatesHainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Reproductive Medical Center, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, ChinaKey Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, ChinaStem Cell Engineering, Texas Heart Institute, Houston, TX, United StatesPhosphoglycerate mutase 2 (PGAM2) is a critical glycolytic enzyme that is highly expressed in skeletal muscle. In humans, naturally occurring mutations in Phosphoglycerate mutase 2 have been etiologically linked to glycogen storage disease X (GSDX). Phosphoglycerate mutase 2 activity is regulated by several posttranslational modifications such as ubiquitination and acetylation. Here, we report that Phosphoglycerate mutase 2 activity is regulated by sumoylation—a covalent conjugation involved in a wide spectrum of cellular events. We found that Phosphoglycerate mutase 2 contains two primary SUMO acceptor sites, lysine (K)49 and K176, and that the mutation of either K to arginine (R) abolished Phosphoglycerate mutase 2 sumoylation. Given that K176 is more highly evolutionarily conserved across paralogs and orthologs than K49 is, we used the CRISPR-mediated homologous recombination technique in myogenic C2C12 cells to generate homozygous K176R knock-in cells (PGAM2K176R/K176R). Compared with wild-type (WT) C2C12 cells, PGAM2K176R/K176R C2C12 cells exhibited impaired myogenic differentiation, as indicated by decreased differentiation and fusion indexes. Furthermore, the results of glycolytic and mitochondrial stress assays with the XF96 Extracellular Flux analyzer revealed a reduced proton efflux rate (PER), glycolytic PER (glycoPER), extracellular acidification rate (ECAR), and oxygen consumption rate (OCR) in PGAM2K176R/K176R C2C12 cells, both at baseline and in response to stress. Impaired mitochondrial function was also observed in PGAM2K176R/K176R P19 cells, a carcinoma cell line. These findings indicate that the PGAM2-K176R mutation impaired glycolysis and mitochondrial function. Gene ontology term analysis of RNA sequencing data further revealed that several downregulated genes in PGAM2K176R/K176R C2C12 cells were associated with muscle differentiation/development/contraction programs. Finally, PGAM2 with either of two naturally occurring missense mutations linked to GSDX, E89A (conversion of glutamic acid 89 to alanine) or R90W (conversion of arginine 90 to tryptophan), exhibited reduced Phosphoglycerate mutase 2 sumoylation. Thus, sumoylation is an important mechanism that mediates Phosphoglycerate mutase 2 activity and is potentially implicated in Phosphoglycerate mutase 2 mutation-linked disease in humans.https://www.frontiersin.org/articles/10.3389/fcell.2022.1052363/fullphosphoglycerate mutase 2sumoylationposttranslational modificationmyogenic differentiationmitochondrial function
spellingShingle Yi Zhang
Yi Zhang
Ilimbek Beketaev
Yanlin Ma
Yanlin Ma
Jun Wang
Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation
Frontiers in Cell and Developmental Biology
phosphoglycerate mutase 2
sumoylation
posttranslational modification
myogenic differentiation
mitochondrial function
title Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation
title_full Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation
title_fullStr Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation
title_full_unstemmed Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation
title_short Sumoylation-deficient phosphoglycerate mutase 2 impairs myogenic differentiation
title_sort sumoylation deficient phosphoglycerate mutase 2 impairs myogenic differentiation
topic phosphoglycerate mutase 2
sumoylation
posttranslational modification
myogenic differentiation
mitochondrial function
url https://www.frontiersin.org/articles/10.3389/fcell.2022.1052363/full
work_keys_str_mv AT yizhang sumoylationdeficientphosphoglyceratemutase2impairsmyogenicdifferentiation
AT yizhang sumoylationdeficientphosphoglyceratemutase2impairsmyogenicdifferentiation
AT ilimbekbeketaev sumoylationdeficientphosphoglyceratemutase2impairsmyogenicdifferentiation
AT yanlinma sumoylationdeficientphosphoglyceratemutase2impairsmyogenicdifferentiation
AT yanlinma sumoylationdeficientphosphoglyceratemutase2impairsmyogenicdifferentiation
AT junwang sumoylationdeficientphosphoglyceratemutase2impairsmyogenicdifferentiation