Your browser doesn't support javascript.
loading
A novel high throughput screen to identify candidate molecular networks that regulate spermatogenic stem cell functions†.
Lord, Tessa; Law, Nathan C; Oatley, Melissa J; Miao, Deqiang; Du, Guihua; Oatley, Jon M.
Afiliação
  • Lord T; School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
  • Law NC; Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia.
  • Oatley MJ; Hunter Medical Research Institute, Infertility and Reproduction Program, New Lambton Heights, NSW, Australia.
  • Miao D; School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
  • Du G; School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
  • Oatley JM; School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
Biol Reprod ; 106(6): 1175-1190, 2022 06 13.
Article em En | MEDLINE | ID: mdl-35244684
ABSTRACT
Spermatogenic regeneration is key for male fertility and relies on activities of an undifferentiated spermatogonial population. Here, a high-throughput approach with primary cultures of mouse spermatogonia was devised to rapidly predict alterations in functional capacity. Combining the platform with a large-scale RNAi screen of transcription factors, we generated a repository of new information from which pathway analysis was able to predict candidate molecular networks regulating regenerative functions. Extending from this database, the SRCAP-CREBBP/EP300 (Snf2-related CREBBP activator protein-CREB binding protein/E1A binding protein P300) complex was found to mediate differential levels of histone acetylation between stem cell and progenitor spermatogonia to influence expression of key self-renewal genes including the previously undescribed testis-specific transcription factor ZSCAN2 (zinc finger and SCAN domain containing 2). Single cell RNA sequencing analysis revealed that ZSCAN2 deficiency alters key cellular processes in undifferentiated spermatogonia such as translation, chromatin modification, and ubiquitination. In Zscan2 knockout mice, while spermatogenesis was moderately impacted during steady state, regeneration after cytotoxic insult was significantly impaired. Altogether, these findings have validated the utility of our high-throughput screening approach and have generated a transcription factor database that can be utilized for uncovering novel mechanisms governing spermatogonial functions.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Espermatogênese / Espermatogônias Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Biol Reprod Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Espermatogênese / Espermatogônias Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Biol Reprod Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Estados Unidos