Your browser doesn't support javascript.
loading
LaminA/C regulates epigenetic and chromatin architecture changes upon aging of hematopoietic stem cells.
Grigoryan, Ani; Guidi, Novella; Senger, Katharina; Liehr, Thomas; Soller, Karin; Marka, Gina; Vollmer, Angelika; Markaki, Yolanda; Leonhardt, Heinrich; Buske, Christian; Lipka, Daniel B; Plass, Christoph; Zheng, Yi; Mulaw, Medhanie A; Geiger, Hartmut; Florian, Maria Carolina.
Afiliação
  • Grigoryan A; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
  • Guidi N; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
  • Senger K; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
  • Liehr T; Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Kollegiengasse 10, 07743, Jena, Germany.
  • Soller K; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
  • Marka G; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
  • Vollmer A; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
  • Markaki Y; Department of Biology II and Center for Integrated Protein Science Munich (CIPSM), Ludwig Maximilians University Munich, Großhaderner Strasse 2, 82152, Planegg-Martinsried, Germany.
  • Leonhardt H; Department of Biology II and Center for Integrated Protein Science Munich (CIPSM), Ludwig Maximilians University Munich, Großhaderner Strasse 2, 82152, Planegg-Martinsried, Germany.
  • Buske C; Institute of Experimental Cancer Research, Comprehensive Cancer Center Ulm, University Hospital Ulm, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
  • Lipka DB; Regulation of Cellular Differentiation Group, INF280, 69120, Heidelberg, Germany.
  • Plass C; Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF280, 69120, Heidelberg, Germany.
  • Zheng Y; Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), INF280, 69120, Heidelberg, Germany.
  • Mulaw MA; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA.
  • Geiger H; Institute of Experimental Cancer Research, Comprehensive Cancer Center Ulm, University Hospital Ulm, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
  • Florian MC; Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Albert-Einstein-Allee 11c, 89081, Ulm, Germany.
Genome Biol ; 19(1): 189, 2018 11 07.
Article em En | MEDLINE | ID: mdl-30404662
ABSTRACT

BACKGROUND:

The decline of hematopoietic stem cell (HSC) function upon aging contributes to aging-associated immune remodeling and leukemia pathogenesis. Aged HSCs show changes to their epigenome, such as alterations in DNA methylation and histone methylation and acetylation landscapes. We previously showed a correlation between high Cdc42 activity in aged HSCs and the loss of intranuclear epigenetic polarity, or epipolarity, as indicated by the specific distribution of H4K16ac.

RESULTS:

Here, we show that not all histone modifications display a polar localization and that a reduction in H4K16ac amount and loss of epipolarity are specific to aged HSCs. Increasing the levels of H4K16ac is not sufficient to restore polarity in aged HSCs and the restoration of HSC function. The changes in H4K16ac upon aging and rejuvenation of HSCs are correlated with a change in chromosome 11 architecture and alterations in nuclear volume and shape. Surprisingly, by taking advantage of knockout mouse models, we demonstrate that increased Cdc42 activity levels correlate with the repression of the nuclear envelope protein LaminA/C, which controls chromosome 11 distribution, H4K16ac polarity, and nuclear volume and shape in aged HSCs.

CONCLUSIONS:

Collectively, our data show that chromatin architecture changes in aged stem cells are reversible by decreasing the levels of Cdc42 activity, revealing an unanticipated way to pharmacologically target LaminA/C expression and revert alterations of the epigenetic architecture in aged HSCs.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Hematopoéticas / Cromatina / Senescência Celular / Proteína cdc42 de Ligação ao GTP / Lamina Tipo A / Epigênese Genética Limite: Animals Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Hematopoéticas / Cromatina / Senescência Celular / Proteína cdc42 de Ligação ao GTP / Lamina Tipo A / Epigênese Genética Limite: Animals Idioma: En Ano de publicação: 2018 Tipo de documento: Article