Your browser doesn't support javascript.
loading
3D Super-Resolution Nuclear Q-FISH Imaging Reveals Cell-Cycle-Related Telomere Changes.
Pochechueva, Tatiana V; Schwenzer, Niko; Kohl, Tobias; Brandenburg, Sören; Kaltenecker, Gesa; Wollnik, Bernd; Lehnart, Stephan E.
Afiliación
  • Pochechueva TV; Department of Cardiology and Pneumology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075 Göttingen, Germany.
  • Schwenzer N; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Robert-Koch-Str. 42a, 37075 Göttingen, Germany.
  • Kohl T; Department of Cardiology and Pneumology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075 Göttingen, Germany.
  • Brandenburg S; Cluster of Excellence Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany.
  • Kaltenecker G; Department of Cardiology and Pneumology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075 Göttingen, Germany.
  • Wollnik B; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Robert-Koch-Str. 42a, 37075 Göttingen, Germany.
  • Lehnart SE; Department of Cardiology and Pneumology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075 Göttingen, Germany.
Int J Mol Sci ; 25(6)2024 Mar 10.
Article en En | MEDLINE | ID: mdl-38542157
ABSTRACT
We present novel workflows for Q-FISH nanoscopy with the potential for prognostic applications and resolving novel chromatin compaction changes. DNA-fluorescence in situ hybridization (DNA-FISH) is a routine application to visualize telomeres, repetitive terminal DNA sequences, in cells and tissues. Telomere attrition is associated with inherited and acquired diseases, including cancer and cardiomyopathies, and is frequently analyzed by quantitative (Q)-FISH microscopy. Recently, nanoscopic imaging techniques have resolved individual telomere dimensions and their compaction as a prognostic marker, in part leading to conflicting conclusions still unresolved to date. Here, we developed a comprehensive Q-FISH nanoscopy workflow to assess telomeres with PNA telomere probes and 3D-Stimulated Emission Depletion (STED) microscopy combined with Dynamic Intensity Minimum (DyMIN) scanning. We achieved single-telomere resolution at high, unprecedented telomere coverage. Importantly, our approach revealed a decrease in telomere signal density during mitotic cell division compared to interphase. Innovatively expanding FISH-STED applications, we conducted double FISH targeting of both telomere- and chromosome-specific sub-telomeric regions and accomplished FISH-STED in human cardiac biopsies. In summary, this work further advanced Q-FISH nanoscopy, detected a new aspect of telomere compaction related to the cell cycle, and laid the groundwork for future applications in complex cell types such as post-mitotic neurons and muscle cells.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: ADN / Telómero Límite: Humans Idioma: En Revista: Int J Mol Sci Año: 2024 Tipo del documento: Article País de afiliación: Alemania

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: ADN / Telómero Límite: Humans Idioma: En Revista: Int J Mol Sci Año: 2024 Tipo del documento: Article País de afiliación: Alemania