1. Academic Validation
  2. Testicular Lmcd1 regulates phagocytosis by Sertoli cells through modulation of NFAT1/Txlna signaling pathway

Testicular Lmcd1 regulates phagocytosis by Sertoli cells through modulation of NFAT1/Txlna signaling pathway

  • Aging Cell. 2020 Oct;19(10):e13217. doi: 10.1111/acel.13217.
Xiaohang Jin 1 Sheng Zhang 1 Tianbing Ding 2 Pengtao Zhao 1 Chunli Zhang 1 Yuxing Zhang 2 Wei Li 3
Affiliations

Affiliations

  • 1 Department of Basic Medical Morphology, Medical College, Xijing University, Xi'an, China.
  • 2 Department of Basic Functioning Medicine, Medical College, Xijing University, Xi'an, China.
  • 3 Department of Human Anatomy, Histology and Embryology, Fourth Military Medical University, Xi'an, China.
Abstract

Increased oxidative stress is well known to cause testicular dysfunction in aging males, but the detailed relationships between aging, oxidative stress, and testicular function remain to be elucidated. LIM and cysteine-rich domains 1 (LMCD1) regulates fundamentally cellular process by interacting with transcription factors. A recent study has identified Lmcd1 as one of the most upregulated nuclear proteins associated with Sertoli cell (SC) differentiation, raising the possibility that testicular actions of LMCD1 are likely to take place. Herein, we reported that LMCD1 was exclusively expressed in the nuclei of SCs. This expression was regulated by TNF-α signaling produced by apoptotic germ cells (GCs) and was suppressed by oxidative stress in a STAT3-dependent manner. Ablation of endogenous LMCD1 expression caused lipid accumulation and senescence in GC co-incubated SCs. Using a previously validated in vivo siRNA approach, we showed that LMCD1 depletion significantly impaired male fertility by inducing oligozoospermia and asthenospermia. Mechanistically, LMCD1 upregulation was associated with the nuclear enrichment of the nuclear factor of activated T cells 1 (NFAT1), a core component of Ca2+ /calmodulin-dependent pathway. LMCD1 facilitated the dephosphorylation and nuclear translocation of NFAT1, which consequently expedited the transactivation of Txlna, a binding partner of the syntaxin family essential for testicular phagocytosis, and thus promoted the removal of apoptotic GCs by phagocytic SCs. Collectively, LMCD1 may operate as a novel pretranscriptional integrator linking SC phagocytosis, lipid homeostasis, and cell senescence.

Keywords

LMCD1; NFAT1; Sertoli cells (SCs); dephosphorylation; phagocytosis.

Figures
Products