RRC ID 45190
著者 Kobayashi J, Yoshida M, Tarui S, Hirata M, Nagai Y, Kasahara S, Naruse K, Ito H, Sano S, Oh H.
タイトル Directed differentiation of patient-specific induced pluripotent stem cells identifies the transcriptional repression and epigenetic modification of NKX2-5, HAND1, and NOTCH1 in hypoplastic left heart syndrome.
ジャーナル PLoS One
Abstract The genetic basis of hypoplastic left heart syndrome (HLHS) remains unknown, and the lack of animal models to reconstitute the cardiac maldevelopment has hampered the study of this disease. This study investigated the altered control of transcriptional and epigenetic programs that may affect the development of HLHS by using disease-specific induced pluripotent stem (iPS) cells. Cardiac progenitor cells (CPCs) were isolated from patients with congenital heart diseases to generate patient-specific iPS cells. Comparative gene expression analysis of HLHS- and biventricle (BV) heart-derived iPS cells was performed to dissect the complex genetic circuits that may promote the disease phenotype. Both HLHS- and BV heart-derived CPCs were reprogrammed to generate disease-specific iPS cells, which showed characteristic human embryonic stem cell signatures, expressed pluripotency markers, and could give rise to cardiomyocytes. However, HLHS-iPS cells exhibited lower cardiomyogenic differentiation potential than BV-iPS cells. Quantitative gene expression analysis demonstrated that HLHS-derived iPS cells showed transcriptional repression of NKX2-5, reduced levels of TBX2 and NOTCH/HEY signaling, and inhibited HAND1/2 transcripts compared with control cells. Although both HLHS-derived CPCs and iPS cells showed reduced SRE and TNNT2 transcriptional activation compared with BV-derived cells, co-transfection of NKX2-5, HAND1, and NOTCH1 into HLHS-derived cells resulted in synergistic restoration of these promoters activation. Notably, gain- and loss-of-function studies revealed that NKX2-5 had a predominant impact on NPPA transcriptional activation. Moreover, differentiated HLHS-derived iPS cells showed reduced H3K4 dimethylation as well as histone H3 acetylation but increased H3K27 trimethylation to inhibit transcriptional activation on the NKX2-5 promoter. These findings suggest that patient-specific iPS cells may provide molecular insights into complex transcriptional and epigenetic mechanisms, at least in part, through combinatorial expression of NKX2-5, HAND1, and NOTCH1 that coordinately contribute to cardiac malformations in HLHS.
巻・号 9(7)
ページ e102796
公開日 2014-1-1
DOI 10.1371/journal.pone.0102796
PII PONE-D-13-20040
PMID 25050861
PMC PMC4106834
MeSH Animals Basic Helix-Loop-Helix Transcription Factors / genetics* Basic Helix-Loop-Helix Transcription Factors / metabolism Cell Differentiation Cells, Cultured Epigenesis, Genetic* Histones / metabolism Homeobox Protein Nkx-2.5 Homeodomain Proteins / genetics* Homeodomain Proteins / metabolism Humans Hypoplastic Left Heart Syndrome / genetics* Hypoplastic Left Heart Syndrome / metabolism Hypoplastic Left Heart Syndrome / pathology Induced Pluripotent Stem Cells / physiology* Mice, Inbred NOD Mice, SCID Myocytes, Cardiac / metabolism Promoter Regions, Genetic Protein Processing, Post-Translational Receptor, Notch1 / genetics* Receptor, Notch1 / metabolism Transcription Factors / genetics* Transcription Factors / metabolism Transcription, Genetic
IF 2.74
引用数 23
WOS 分野 CARDIAC & CARDIOVASCULAR SYSTEMS
リソース情報
ヒト・動物細胞 201B7(HPS0063)