Login / Signup

Zbtb40 Deficiency Leads to Morphological and Phenotypic Abnormalities of Spermatocytes and Spermatozoa and Causes Male Infertility.

Yinghong CuiMingqing ZhouQuanyuan HeZuping He
Published in: Cells (2023)
Studies on the gene regulation of spermatogenesis are of unusual significance for maintaining male reproduction and treating male infertility. Here, we have demonstrated, for the first time, that a loss of ZBTB40 function leads to abnormalities in the morphological and phenotypic characteristics of mouse spermatocytes and spermatids as well as male infertility. We revealed that Zbtb40 was expressed in spermatocytes of mouse testes, and it was co-localized with γH2AX in mouse secondary spermatocytes. Interestingly, spermatocytes of Zbtb40 knockout mice had longer telomeres, compromised double-strand break (DSB) repair in the sex chromosome, and a higher apoptosis ratio compared to wild-type (WT) mice. The testis weight, testicular volume, and cauda epididymis body weight of the Zbtb40 +/- male mice were significantly lower than in WT mice. Mating tests indicated that Zbtb40 +/- male mice were able to mate normally, but they failed to produce any pups. Notably, sperm of Zbtb40 +/- mice showed flagellum deformities and abnormal acrosome biogenesis. Furthermore, a ZBTB40 mutation was associated with non-obstructive azoospermia. Our results implicate that ZBTB40 deficiency leads to morphological and phenotypic abnormalities of spermatocytes and spermatids and causes male infertility. This study thus offers a new genetic mechanism regulating mammalian spermatogenesis and provides a novel target for gene therapy in male infertility.
Keyphrases
  • wild type
  • body weight
  • gene therapy
  • polycystic ovary syndrome
  • high fat diet induced
  • body mass index
  • oxidative stress
  • cell death
  • endoplasmic reticulum stress
  • cell proliferation
  • cell cycle arrest