4.7 Article

LncRNA NEAT1 controls the lineage fates of BMSCs during skeletal aging by impairing mitochondrial function and pluripotency maintenance

Journal

CELL DEATH AND DIFFERENTIATION
Volume 29, Issue 2, Pages 351-365

Publisher

SPRINGERNATURE
DOI: 10.1038/s41418-021-00858-0

Keywords

-

Funding

  1. National Natural Science Foundation of China [81771092, 81970910]
  2. Natural Science Foundation of Jiangsu Province [BK20201350]
  3. Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD) [2018-87]
  4. Postgraduate Research & Practice Innovation Program of Jiangsu Province [KYCX19_1149]
  5. Natural Science Foundation of the Jiangsu Higher Education Institutions of China [19KJA360003]

Ask authors/readers for more resources

NEAT1 serves as a key bone-fat switch in aged BMSCs by modulating mitochondrial function and pluripotency maintenance, influencing the differentiation fate of bone-fat cells.
Aged bone marrow mesenchymal stem cells (BMSCs) exhibit aberrant self-renewal and lineage specification, which contribute to imbalanced bone-fat and progressive bone loss. In addition to known master regulators of lineage commitment, it is crucial to identify pivotal switches governing the specific differentiation fate of aged BMSCs. Here, we profiled differences in epigenetic regulation between adipogenesis and osteogenesis and identified super-enhancer associated lncRNA nuclear-enriched abundant transcript 1 (NEAT1) as a key bone-fat switch in aged BMSCs. We validated that NEAT1 with high enhancer activity was transcriptionally activated by ATF2 and directed aged BMSCs to a greater propensity to differentiate toward adipocytes than osteoblasts by mediating mitochondrial function. Furthermore, we confirmed NEAT1 as a protein-binding scaffold in which phosphorylation modification of SOX2 Ser249/250 by CDK2 impaired SOX2/OCT4 complex stability and dysregulated downstream transcription networks of pluripotency maintenance. In addition, by sponging miR-27b-3p, NEAT1 upregulated BNIP3L, BMP2K, and PPARG expression to shape mitochondrial function and osteogenic/adipogenic differentiation commitment, respectively. In extracellular communication, NEAT1 promoted CSF1 secretion from aged BMSCs and then strengthened osteoclastic differentiation by extracellular vesicle delivery. Notably, Neat1 small interfering RNA delivery induced increased bone mass in aged mice and decreased fat accumulation in the bone marrow. These findings suggest that NEAT1 regulates the lineage fates of BMSCs by orchestrating mitochondrial function and pluripotency maintenance, and might be a potential therapeutic target for skeletal aging.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available