• 제목/요약/키워드: epigenetic reprogramming

검색결과 44건 처리시간 0.022초

Inhibition of Class I Histone Deacetylase Enhances Self-Reprogramming of Spermatogonial Stem Cells into Pluripotent Stem Cells

  • Yukyeong Lee;Seung-Won Lee;Dahee Jeong;Hye Jeong Lee;Na Young Choi;Jin Seok Bang;Seokbeom Ham;Kinarm, Ko
    • International Journal of Stem Cells
    • /
    • 제16권1호
    • /
    • pp.27-35
    • /
    • 2023
  • Background and Objectives: Spermatogonial stem cells (SSCs) are the most primitive cells in spermatogenesis and are the only adult stem cells capable of passing on the genome of a given species to the next generation. SSCs are the only adult stem cells known to exhibit high Oct4 expression and can be induced to self-reprogram into pluripotent cells depending on culture conditions. Epigenetic modulation is well known to be involved in the induction of pluripotency of somatic cells. However, epigenetic modulation in self-reprogramming of SSCs into pluripotent cells has not been studied. Methods and Results: In this study, we examined the involvement of epigenetic modulation by assessing whether selfreprogramming of SSCs is enhanced by treatment with epigenetic modulators. We found that second-generation selective class I HDAC inhibitors increased SSC reprogramming efficiency, whereas non-selective HDAC inhibitors had no effect. Conclusions: We showed that pluripotent stem cells derived from adult SSCs by treatment with small molecules with epigenetic modulator functions exhibit pluripotency in vitro and in vivo. Our results suggest that the mechanism of SSC reprogramming by epigenetic modulator can be used for important applications in epigenetic reprogramming research.

Epigenetic Reprogramming in Cloned Embryos

  • Kang, Yong-Kook;Han, Yong-Mahn;Lee, Kyung-Kwang
    • 한국동물번식학회:학술대회논문집
    • /
    • 한국동물번식학회 2001년도 발생공학 국제심포지움 및 학술대회 발표자료집
    • /
    • pp.25-31
    • /
    • 2001
  • During early development, a dramatic reduction in methylation levels occurs in mouse (Monk et al., 1987). The process of epigenetic reprogramming in early embryos erases gamete-specific methylation patterns inherited from the parents (Howlett & Reik 1991, Monk et al., 1987, Oswald et al., 2000, Sanford et al., 1984). This genome-wide demethylation process may be a prerequisite for the formation of pluripotent stem cells that are important for the later development (Reik & Surani 1997). During post-implantation development, a wave of de novo methylation takes place; most of the genomic DNA is methylated at defined developmental timepoints, whereas tissue-specific genes undergo demethylation in their tissues of expression (Kafri et al., 1992, Razin & Kafri 1994). Another demethylation-remethylation cycle of epigenetic reprogramming takes place during gametogenesis and is necessary for resetting of genomic imprinting (Solter 1988). The dynamic epigenetic reprogramming events appear to be basic and are probably conserved in eutherian mammals (see below). (omitted)

  • PDF

Strategic Application of Epigenetic Regulators for Efficient Neuronal Reprogramming of Human Fibroblasts

  • Gary Stanley Fernandes;Rishabh Deo Singh;Debojyoti De;Kyeong Kyu Kim
    • International Journal of Stem Cells
    • /
    • 제16권2호
    • /
    • pp.156-167
    • /
    • 2023
  • Background and Objectives: Cellular reprogramming in regenerative medicine holds great promise for treating patients with neurological disorders. In this regard, small molecule-mediated cellular conversion has attracted special attention because of its ease of reproducibility, applicability, and fewer safety concerns. However, currently available protocols for the direct conversion of somatic cells to neurons are limited in clinical application due of their complex nature, lengthy process, and low conversion efficiency. Methods and Results: Here, we report a new protocol involving chemical-based direct conversion of human fibroblasts (HF) to matured neuron-like cells with a short duration and high conversion efficiency using temporal and strategic dual epigenetic regulation. In this protocol, epigenetic modulation by inhibition of histone deacetylase and bromodomain enabled to overcome "recalcitrant" nature of adult fibroblasts and shorten the duration of neuronal reprogramming. We further observed that an extended epigenetic regulation is necessary to maintain the induced neuronal program to generate a homogenous population of neuron-like cells. Conclusions: Therefore, our study provides a new protocol to produce neurons-like cells and highlights the need of proper epigenetic resetting to establish and maintain neuronal program in HF.

후성 유전학적 리프로그래밍과 클로닝 (Epigenetic Reprogramming and Cloning)

  • 한용만;강용국;구덕본;이경광
    • 한국발생생물학회지:발생과생식
    • /
    • 제7권2호
    • /
    • pp.61-68
    • /
    • 2003
  • 포유동물의 초기 발생과정 중 접합체가 전능성이나 다능성을 가지기 위해서는 전반적인 DNA 메틸화를 포함하는 후성 유전학적 리프로그래밍의 복잡한 과정을 거쳐야만 한다. 본 연구팀에서는 공여핵의 후성 유전학적 리프로그래밍 과정을 조사하기 위하여 소 복제수정란에서 메틸화 양상을 분석하였다. 복제수정란의 비정상적인 메틸화 양상이 다양한 반복염기서열에서 관찰되었지만 single-copy유전자들의 염기서열은 정상적인 메틸화 양상을 보여주었다. 전반적으로 복제수정란의 전반적인 메틸화 상태는 정상수정란과 완전히 다른 양상을 보여주었다. 또한 복제 배반포의 영양외배엽세포에서 특이적으로 높은 메틸화 수준은 현 복제동물에서 빈번히 나타나는 불완전한 태반형성에 작용할 수 있을 것이다. 결론적으로 복제수정란의 비정상적 발생은 공여핵의 불완전한 후성 유전학적 리프로그래밍에 기인할 수 있다는 사실을 제시하게 되었다. 이러한 공여핵의 후성 유전학적 과정의 이해는 복제수정란의 비정상적 발생을 보다 분명히 밝힐 수 있을 것이다.

  • PDF

Metabolic Signaling to Epigenetic Alterations in Cancer

  • Kim, Jung-Ae;Yeom, Young Il
    • Biomolecules & Therapeutics
    • /
    • 제26권1호
    • /
    • pp.69-80
    • /
    • 2018
  • Cancer cells reprogram cellular metabolism to support the malignant features of tumors, such as rapid growth and proliferation. The cancer promoting effects of metabolic reprogramming are found in many aspects: generating additional energy, providing more anabolic molecules for biosynthesis, and rebalancing cellular redox states in cancer cells. Metabolic pathways are considered the pipelines to supply metabolic cofactors of epigenetic modifiers. In this regard, cancer metabolism, whereby cellular metabolite levels are greatly altered compared to normal levels, is closely associated with cancer epigenetics, which is implicated in many stages of tumorigenesis. In this review, we provide an overview of cancer metabolism and its involvement in epigenetic modifications and suggest that the metabolic adaptation leading to epigenetic changes in cancer cells is an important non-genetic factor for tumor progression, which cooperates with genetic causes. Understanding the interaction of metabolic reprogramming with epigenetics in cancers may help to develop novel or highly improved therapeutic strategies that target cancer metabolism.

Enhanced Green Fluorescent Protein Gene under the Regulation of Human Oct4 Promoter as a Marker to Identify Reprogramming of Human Fibroblasts

  • Heo, Soon-Young;Ahn, Kwang-Sung;Kang, Jee-Hyun;Shim, Ho-Sup
    • Reproductive and Developmental Biology
    • /
    • 제32권2호
    • /
    • pp.135-140
    • /
    • 2008
  • Recent studies on nuclear transfer and induced pluripotent stem cells have demonstrated that differentiated somatic cells can be returned to the undifferentiated state by reversing their developmental process. These epigenetically reprogrammed somatic cells may again be differentiated into various cell types, and used for cell replacement therapies through autologous transplantation to treat many degenerative diseases. To date, however, reprogramming of somatic cells into undifferentiated cells has been extremely inefficient. Hence, reliable markers to identify the event of reprogramming would assist effective selection of reprogrammed cells. In this study, a transgene construct encoding enhanced green fluorescent protein (EGFP) under the regulation of human Oct4 promoter was developed as a reporter for the reprogramming of somatic cells. Microinjection of the transgene construct into pronuclei of fertilized mouse eggs resulted in the emission of green fluorescence, suggesting that the undifferentiated cytoplasmic environment provided by fertilized eggs induces the expression of EGFP. Next, the transgene construct was introduced into human embryonic fibroblasts, and the nuclei from these cells were transferred into enucleated porcine oocytes. Along with their in vitro development, nuclear transfer embryos emitted green fluorescence, suggesting the reprogramming of donor nuclei in nuclear transfer embryos. The results of the present study demonstrate that expression of the transgene under the regulation of human Oct4 promoter coincides with epigenetic reprogramming, and may be used as a convenient marker that non-invasively reflects reprogramming of somatic cells.

Factors Involving Reprogramming in Cloned Embryos

  • Kim, N. H;X. S. Cui;Kim, I. H.;Y. M. Han
    • 한국가축번식학회지
    • /
    • 제27권4호
    • /
    • pp.349-357
    • /
    • 2003
  • Although nuclear transfer (NT) techniques are used to clone animals, its efficiency is very low. Moreover, nuclear transfer has resulted in offspring with severe developmental problems, probably due to incomplete nuclear reprogramming. Nuclear reprogramming is characterized by functional modification of the transferred nucleus to allow it to direct normal embryo development with the potential to grow to term. Although the nature of the reprogramming factor(s) in mammals is not clear, various nuclear as well as cytoplasmic components are involved in the processes. In this article we review recent data on factors involved in the nuclear reprogramming of cloned embryos.

Epigenetic Reprogramming and Cloning

  • Han, Yong-Mahn
    • 한국발생생물학회:학술대회논문집
    • /
    • 한국발생생물학회 2003년도 전기 한국발생생물학회 제16차 학술대회논문집
    • /
    • pp.6-6
    • /
    • 2003
  • No Abstract, See Full Text

  • PDF

Cooperative Instruction of Signaling and Metabolic Pathways on the Epigenetic Landscape

  • Kim, Jung-Ae
    • Molecules and Cells
    • /
    • 제41권4호
    • /
    • pp.264-270
    • /
    • 2018
  • Cells cope with diverse intrinsic and extrinsic stimuli in order to make adaptations for survival. The epigenetic landscape plays a crucial role in cellular adaptation, as it integrates the information generated from stimuli. Signaling pathways induced by stimuli communicate with chromatin to change the epigenetic landscape through regulation of epigenetic modifiers. Metabolic dynamics altered by these stimuli also affect the activity of epigenetic modifiers. Here, I review the current understanding of epigenetic regulation via signaling and metabolic pathways. In addition, I will discuss possible ways to achieve specificity of epigenetic modifications through the cooperation of stimuli-induced signal transduction and metabolic reprogramming.