101
|
Boukhaled GM, Corrado M, Guak H, Krawczyk CM. Chromatin Architecture as an Essential Determinant of Dendritic Cell Function. Front Immunol 2019; 10:1119. [PMID: 31214161 PMCID: PMC6557980 DOI: 10.3389/fimmu.2019.01119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
Epigenetics has widespread implications in a variety of cellular processes ranging from cell identity and specification, to cellular adaptation to environmental stimuli. While typically associated with heritable changes in gene expression, epigenetic mechanisms are now appreciated to regulate dynamic changes in gene expression—even in post-mitotic cells. Cells of the innate immune system, including dendritic cells (DC), rapidly integrate signals from their microenvironment and respond accordingly, undergoing massive changes in transcriptional programming. This dynamic transcriptional reprogramming relies on epigenetic changes mediated by numerous enzymes and their substrates. This review highlights our current understanding of epigenetic regulation of DC function. Epigenetic mechanisms contribute to the maintenance of the steady state and are important for precise responses to proinflammatory stimuli. Interdependence between epigenetic modifications and the delicate balance of metabolites present another layer of complexity. In addition, dynamic regulation of the expression of proteins that modify chromatin architecture in DCs significantly impacts DC function. Environmental factors, including inflammation, aging, chemicals, nutrients, and lipid mediators, are increasingly appreciated to affect the epigenome in DCs, and, in doing so, regulate host immunity. Our understanding of how epigenetic mechanisms regulate DC function is in its infancy, and it must be expanded in order to discern the mechanisms underlying the balance between health and disease states.
Collapse
Affiliation(s)
- Giselle M Boukhaled
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada
| | - Mario Corrado
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada
| | - Hannah Guak
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada
| | - Connie M Krawczyk
- Department of Physiology, Goodman Cancer Research Center, McGill University, Montreal, QC, Canada.,Center for Cancer and Cell Biology, Program in Metabolic and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, United States
| |
Collapse
|
102
|
Liu C, Wang M, Wei X, Wu L, Xu J, Dai X, Xia J, Cheng M, Yuan Y, Zhang P, Li J, Feng T, Chen A, Zhang W, Chen F, Shang Z, Zhang X, Peters BA, Liu L. An ATAC-seq atlas of chromatin accessibility in mouse tissues. Sci Data 2019; 6:65. [PMID: 31110271 PMCID: PMC6527694 DOI: 10.1038/s41597-019-0071-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/10/2019] [Indexed: 01/06/2023] Open
Abstract
The Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) is a fundamental epigenomics approach and has been widely used in profiling the chromatin accessibility dynamics in multiple species. A comprehensive reference of ATAC-seq datasets for mammalian tissues is important for the understanding of regulatory specificity and developmental abnormality caused by genetic or environmental alterations. Here, we report an adult mouse ATAC-seq atlas by producing a total of 66 ATAC-seq profiles from 20 primary tissues of both male and female mice. The ATAC-seq read enrichment, fragment size distribution, and reproducibility between replicates demonstrated the high quality of the full dataset. We identified a total of 296,574 accessible elements, of which 26,916 showed tissue-specific accessibility. Further, we identified key transcription factors specific to distinct tissues and found that the enrichment of each motif reflects the developmental similarities across tissues. In summary, our study provides an important resource on the mouse epigenome and will be of great importance to various scientific disciplines such as development, cell reprogramming, and genetic disease.
Collapse
Affiliation(s)
- Chuanyu Liu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Mingyue Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Xiaoyu Wei
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Liang Wu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jiangshan Xu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Xi Dai
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jun Xia
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- MGI, BGI-Shenzhen, Shenzhen, 518083, China
| | - Mengnan Cheng
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Yue Yuan
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Pengfan Zhang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jiguang Li
- BGI-Shenzhen, Shenzhen, 518083, China
- MGI, BGI-Shenzhen, Shenzhen, 518083, China
| | - Taiqing Feng
- BGI-Shenzhen, Shenzhen, 518083, China
- MGI, BGI-Shenzhen, Shenzhen, 518083, China
| | - Ao Chen
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Wenwei Zhang
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Fang Chen
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
- MGI, BGI-Shenzhen, Shenzhen, 518083, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Zhouchun Shang
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Xiuqing Zhang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Brock A Peters
- BGI-Shenzhen, Shenzhen, 518083, China.
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.
- MGI, BGI-Shenzhen, Shenzhen, 518083, China.
- Advanced Genomics Technology Lab, Complete Genomics Inc., 2904 Orchard Parkway, San Jose, California, 95134, USA.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, 518083, China.
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.
| |
Collapse
|
103
|
Spyrou J, Gardner DK, Harvey AJ. Metabolism Is a Key Regulator of Induced Pluripotent Stem Cell Reprogramming. Stem Cells Int 2019; 2019:7360121. [PMID: 31191682 PMCID: PMC6525803 DOI: 10.1155/2019/7360121] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022] Open
Abstract
Reprogramming to pluripotency involves drastic restructuring of both metabolism and the epigenome. However, induced pluripotent stem cells (iPSC) retain transcriptional memory, epigenetic memory, and metabolic memory from their somatic cells of origin and acquire aberrant characteristics distinct from either other pluripotent cells or parental cells, reflecting incomplete reprogramming. As a critical link between the microenvironment and regulation of the epigenome, nutrient availability likely plays a significant role in the retention of somatic cell memory by iPSC. Significantly, relative nutrient availability impacts iPSC reprogramming efficiency, epigenetic regulation and cell fate, and differentially alters their ability to respond to physiological stimuli. The significance of metabolites during the reprogramming process is central to further elucidating how iPSC retain somatic cell characteristics and optimising culture conditions to generate iPSC with physiological phenotypes to ensure their reliable use in basic research and clinical applications. This review serves to integrate studies on iPSC reprogramming, memory retention and metabolism, and identifies areas in which current knowledge is limited.
Collapse
Affiliation(s)
- James Spyrou
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David K. Gardner
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexandra J. Harvey
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
104
|
Primed histone demethylation regulates shoot regenerative competency. Nat Commun 2019; 10:1786. [PMID: 30992430 PMCID: PMC6467990 DOI: 10.1038/s41467-019-09386-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 03/07/2019] [Indexed: 01/09/2023] Open
Abstract
Acquisition of pluripotency by somatic cells is a striking process that enables multicellular organisms to regenerate organs. This process includes silencing of genes to erase original tissue memory and priming of additional cell type specification genes, which are then poised for activation by external signal inputs. Here, through analysis of genome-wide histone modifications and gene expression profiles, we show that a gene priming mechanism involving LYSINE-SPECIFIC DEMETHYLASE 1-LIKE 3 (LDL3) specifically eliminates H3K4me2 during formation of the intermediate pluripotent cell mass known as callus derived from Arabidopsis root cells. While LDL3-mediated H3K4me2 removal does not immediately affect gene expression, it does facilitate the later activation of genes that act to form shoot progenitors when external cues lead to shoot induction. These results give insights into the role of H3K4 methylation in plants, and into the primed state that provides plant cells with high regenerative competency. Plant regeneration can occur via formation of a mass of pluripotent cells known as callus. Here, Ishihara et al. show that acquisition of regenerative capacity of callus-forming cells requires a lysine-specific demethylase that removes H3K4me2 to prime gene expression in response to regenerative cues.
Collapse
|
105
|
Cell Reprogramming in Tumorigenesis and Its Therapeutic Implications for Breast Cancer. Int J Mol Sci 2019; 20:ijms20081827. [PMID: 31013830 PMCID: PMC6515165 DOI: 10.3390/ijms20081827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide and can be categorized into several subtypes according to histopathological parameters or genomic signatures. Such heterogeneity of breast cancer can arise from the reactivation of mammary stem cells in situ during tumorigenesis. Moreover, different breast cancer subtypes exhibit varieties of cancer incidence, therapeutic response, and patient prognosis, suggesting that a specific therapeutic protocol is required for each breast cancer subtype. Recent studies using molecular and cellular assays identified a link between specific genetic/epigenetic alterations and distinct cells of origin of breast cancer subtypes. These alterations include oncogenes, tumor suppressor genes, and cell-lineage determinants, which can induce cell reprogramming (dedifferentiation and transdifferentiation) among two lineage-committed mammary epithelial cells, namely basal and luminal cells. The interconversion of cell states through cell reprogramming into the intermediates of mammary stem cells can give rise to heterogeneous breast cancers that complicate effective therapies of breast cancer. A better understanding of mechanisms underlying cell reprogramming in breast cancer can help in not only elucidating tumorigenesis but also developing therapeutics for breast cancer. This review introduces recent findings on cancer gene-mediated cell reprogramming in breast cancer and discusses the therapeutic potential of targeting cell reprogramming.
Collapse
|
106
|
Genomic landscape analyses of reprogrammed cells using integrative and non-integrative methods reveal variable cancer-associated alterations. Oncotarget 2019; 10:2693-2708. [PMID: 31105870 PMCID: PMC6505633 DOI: 10.18632/oncotarget.26857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 03/23/2019] [Indexed: 12/13/2022] Open
Abstract
Recent development of cell reprogramming technologies brought a major hope for future cell therapy applications by the use of these cells or their derivatives. For this purpose, one of the major requirements is the absence of genomic alterations generating a risk of cell transformation. Here we analyzed by microarray-based comparative genomic hybridization human iPSC generated by two non-integrative and one integrative method at pluripotent stage as well as in corresponding teratomas. We show that all iPSC lines exhibit copy number variations (CNV) of several genes deregulated in oncogenesis. These cancer-associated genomic alterations were more pronounced in virally programmed hiPSCs and their derivative teratoma as compared to those found in iPSC generated by mRNA-mediated reprogramming. Bioinformatics analysis showed the involvement of these genes in human leukemia and carcinoma. We conclude that genetic screening should become a standard procedure to ensure that hiPSCs are free from cancer-associated genomic alterations before clinical use.
Collapse
|
107
|
MKL1-actin pathway restricts chromatin accessibility and prevents mature pluripotency activation. Nat Commun 2019; 10:1695. [PMID: 30979898 PMCID: PMC6461646 DOI: 10.1038/s41467-019-09636-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
Actin cytoskeleton is well-known for providing structural/mechanical support, but whether and how it regulates chromatin and cell fate reprogramming is far less clear. Here, we report that MKL1, the key transcriptional co-activator of many actin cytoskeletal genes, regulates genomic accessibility and cell fate reprogramming. The MKL1-actin pathway weakens during somatic cell reprogramming by pluripotency transcription factors. Cells that reprogram efficiently display low endogenous MKL1 and inhibition of actin polymerization promotes mature pluripotency activation. Sustained MKL1 expression at a level seen in typical fibroblasts yields excessive actin cytoskeleton, decreases nuclear volume and reduces global chromatin accessibility, stalling cells on their trajectory toward mature pluripotency. In addition, the MKL1-actin imposed block of pluripotency can be bypassed, at least partially, when the Sun2-containing linker of the nucleoskeleton and cytoskeleton (LINC) complex is inhibited. Thus, we unveil a previously unappreciated aspect of control on chromatin and cell fate reprogramming exerted by the MKL1-actin pathway. MKL1 is a key transcriptional co-activator of actin cytoskeleton genes. Here the authors show that MKL1 activation in somatic cells reduces chromatin accessibility and hinders full reprogramming to pluripotency. Reduction of MKL1, disruption of actin cytoskeleton and its links to the nucleus relieve this repression.
Collapse
|
108
|
Lu QR, Qian L, Zhou X. Developmental origins and oncogenic pathways in malignant brain tumors. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e342. [PMID: 30945456 DOI: 10.1002/wdev.342] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/20/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022]
Abstract
Brain tumors such as adult glioblastomas and pediatric high-grade gliomas or medulloblastomas are among the leading causes of cancer-related deaths, exhibiting poor prognoses with little improvement in outcomes in the past several decades. These tumors are heterogeneous and can be initiated from various neural cell types, contributing to therapy resistance. How such heterogeneity arises is linked to the tumor cell of origin and their genetic alterations. Brain tumorigenesis and progression recapitulate key features associated with normal neurogenesis; however, the underlying mechanisms are quite dysregulated as tumor cells grow and divide in an uncontrolled manner. Recent comprehensive genomic, transcriptomic, and epigenomic studies at single-cell resolution have shed new light onto diverse tumor-driving events, cellular heterogeneity, and cells of origin in different brain tumors. Primary and secondary glioblastomas develop through different genetic alterations and pathways, such as EGFR amplification and IDH1/2 or TP53 mutation, respectively. Mutations such as histone H3K27M impacting epigenetic modifications define a distinct group of pediatric high-grade gliomas such as diffuse intrinsic pontine glioma. The identification of distinct genetic, epigenomic profiles and cellular heterogeneity has led to new classifications of adult and pediatric brain tumor subtypes, affording insights into molecular and lineage-specific vulnerabilities for treatment stratification. This review discusses our current understanding of tumor cells of origin, heterogeneity, recurring genetic and epigenetic alterations, oncogenic drivers and signaling pathways for adult glioblastomas, pediatric high-grade gliomas, and medulloblastomas, the genetically heterogeneous groups of malignant brain tumors. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Q Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lily Qian
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xianyao Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China.,Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
109
|
Liu S, Lu M, Li H, Zuo Y. Prediction of Gene Expression Patterns With Generalized Linear Regression Model. Front Genet 2019; 10:120. [PMID: 30886626 PMCID: PMC6409355 DOI: 10.3389/fgene.2019.00120] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/04/2019] [Indexed: 01/10/2023] Open
Abstract
Cell reprogramming has played important roles in medical science, such as tissue repair, organ reconstruction, disease treatment, new drug development, and new species breeding. Oct4, a core pluripotency factor, has especially played a key role in somatic cell reprogramming through transcriptional control and affects the expression level of genes by its combination intensity. However, the quantitative relationship between Oct4 combination intensity and target gene expression is still not clear. Therefore, firstly, a generalized linear regression method was constructed to predict gene expression values in promoter regions affected by Oct4 combination intensity. Training data, including Oct4 combination intensity and target gene expression, were from promoter regions of genes with different cell development stages. Additionally, the quantitative relationship between gene expression and Oct4 combination intensity was analyzed with the proposed model. Then, the quantitative relationship between gene expression and Oct4 combination intensity at each stage of cell development was classified into high and low levels. Experimental analysis showed that the combination height of Oct4-inhibited gene expression decremented by a temporal exponential value, whereas the combination width of Oct4-promoted gene expression incremented by a temporal logarithmic value. Experimental results showed that the proposed method can achieve goodness of fit with high confidence.
Collapse
Affiliation(s)
- Shuai Liu
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
- College of Computer Science, Inner Mongolia University, Hohhot, China
| | - Mengye Lu
- College of Computer Science, Inner Mongolia University, Hohhot, China
| | - Hanshuang Li
- College of Life Sciences, Inner Mongolia University, Hohhot, China
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
| | - Yongchun Zuo
- College of Life Sciences, Inner Mongolia University, Hohhot, China
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
| |
Collapse
|
110
|
Thompson R, Casali C, Chan C. Forskolin and IBMX Induce Neural Transdifferentiation of MSCs Through Downregulation of the NRSF. Sci Rep 2019; 9:2969. [PMID: 30814572 PMCID: PMC6393535 DOI: 10.1038/s41598-019-39544-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/10/2019] [Indexed: 01/20/2023] Open
Abstract
Neural differentiation of mesenchymal stem cells is a controversial phenomenon, as it would require transdifferentiation across the mesoderm-ectoderm barrier. However, several laboratories have observed that MSCs are able to be induced to express neural characteristics. Previously, we demonstrated that the cAMP-elevating agents, forskolin and IBMX, induced neural-like differentiation of MSCs, including expression of neural markers and increased sensitivity to neurotransmitters. However, due to the broad range of effects that forskolin and IBMX can elicit through the intracellular second messenger, cAMP, a better mechanistic understanding is required. Here, we show that neural induction by forskolin and IBMX is dependent on downregulation of expression of the master transcriptional regulator, neuron restrictive silencer factor (NRSF), and its downstream target genes. Since silencing of NRSF is known to initiate neural differentiation, it suggests that forskolin and IBMX result in transdifferentiation of MSCs into a neural lineage.
Collapse
Affiliation(s)
- Ryan Thompson
- Cell and Molecular Biology Program, Michigan State University, 567 Wilson Road, Rm 2240E, East Lansing, Michigan, 48824, USA
| | - Christina Casali
- Department of Chemical Engineering and Materials Science, Michigan State University, 428S. Shaw Lane, Rm 2527, East Lansing, Michigan, 48824, USA
| | - Christina Chan
- Cell and Molecular Biology Program, Michigan State University, 567 Wilson Road, Rm 2240E, East Lansing, Michigan, 48824, USA. .,Department of Chemical Engineering and Materials Science, Michigan State University, 428S. Shaw Lane, Rm 2527, East Lansing, Michigan, 48824, USA. .,Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI, 48824, USA.
| |
Collapse
|
111
|
Böhnke L, Traxler L, Herdy JR, Mertens J. Human neurons to model aging: A dish best served old. ACTA ACUST UNITED AC 2019; 27:43-49. [PMID: 31745399 DOI: 10.1016/j.ddmod.2019.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
With the advancing age of humans and with it, growing numbers of age-related diseases, aging has become a major focus in recent research. The lack of fitting aging models, especially in neurological diseases where access to human brain samples is limited, has highlighted direct conversion into induced neurons (iN) as an important method to overcome this challenge. Contrary to iPSC reprogramming and its corresponding cell rejuvenation, the generation of iNs enables us to retain aging signatures throughout the conversion process and beyond. In this review, we explore different cell reprogramming methods in light of age-associated neurodegenerative diseases and discuss different approaches, advances, and limitations.
Collapse
Affiliation(s)
- Lena Böhnke
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell Biology & Regenerative Medicine, Leopold-Franzens-University Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria
| | - Larissa Traxler
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell Biology & Regenerative Medicine, Leopold-Franzens-University Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria
| | - Joseph R Herdy
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jerome Mertens
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell Biology & Regenerative Medicine, Leopold-Franzens-University Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria.,Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
112
|
Nuclear-cytoplasmic shuttling of class IIa histone deacetylases regulates somatic cell reprogramming. CELL REGENERATION 2019; 8:21-29. [PMID: 31205685 PMCID: PMC6557759 DOI: 10.1016/j.cr.2018.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 11/17/2018] [Accepted: 11/17/2018] [Indexed: 12/19/2022]
Abstract
Class IIa histone deacetylases (HDACs) are a subfamily of HDACs with important functions in development and adult tissue homeostasis. As opposed to other HDACs, they lack catalytic function and bind transcription factors to recruit transcriptional co-regulators, mostly co-repressors such as nuclear receptor co-repressor (NCoR)/silencing mediator of retinoid and thyroid hormone receptor (SMRT). Class IIa HDACs enhance mouse somatic cell reprogramming to induced pluripotent stem cells (iPSCs) by repressing the function of the pro-mesenchymal transcription factor myocyte enhancer factor 2 (MEF2), which is upregulated during this process. Here, we describe, using HDAC4 and 7 as examples, that class IIa HDACs exhibit nuclear-cytoplasmic trafficking in reprogramming, being mostly cytoplasmic in donor fibroblasts and intermediate cells but translocating to the nucleus in iPSCs. Importantly, over-expressing a mutant form of HDAC4 or 7 that becomes trapped in the nucleus enhances the early phase of reprogramming but is deleterious afterwards. The latter effect is mediated through binding to the exogenous reprogramming factors at pluripotency loci, and the subsequent recruitment of NCoR/SMRT co-repressors. Thus, our findings uncover a context-dependent function of class IIa HDACs in reprogramming and further reinforce the idea that recruitment of co-repressors by the exogenous factors is a major obstacle for reactivating the pluripotency network in this process.
Collapse
|
113
|
Sugimoto K, Temman H, Kadokura S, Matsunaga S. To regenerate or not to regenerate: factors that drive plant regeneration. CURRENT OPINION IN PLANT BIOLOGY 2019; 47:138-150. [PMID: 30703741 DOI: 10.1016/j.pbi.2018.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 05/23/2023]
Abstract
Plants have a remarkable regenerative capacity, but it varies widely among species and tissue types. Whether plant cells/tissues initiate regeneration largely depends on the extent to which they are constrained to their original tissue fate. Once cells start the regeneration program, they acquire a new fate, form meristems, and develop into organs. During these processes, the cells must continuously overcome various barriers to the progression of the regeneration program until the organ (or whole plant) is complete. Recent studies have revealed key factors and signals affecting cell fate during plant regeneration. Here, we review recent research on: (i) environmental signal inputs and physical stimuli that act as initial triggers of regeneration; (ii) epigenetic and transcriptional cellular responses to those triggers leading to cellular reprograming; and (iii) molecules that direct the formation and development of the new stem cell niche. We also discuss differences and similarities between regeneration and normal development.
Collapse
Affiliation(s)
- Kaoru Sugimoto
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Haruka Temman
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Satoshi Kadokura
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Sachihiro Matsunaga
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
114
|
Dabiri Y, Gama-Brambila RA, Taškova K, Herold K, Reuter S, Adjaye J, Utikal J, Mrowka R, Wang J, Andrade-Navarro MA, Cheng X. Imidazopyridines as Potent KDM5 Demethylase Inhibitors Promoting Reprogramming Efficiency of Human iPSCs. iScience 2019; 12:168-181. [PMID: 30685712 PMCID: PMC6354736 DOI: 10.1016/j.isci.2019.01.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/11/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
Pioneering human induced pluripotent stem cell (iPSC)-based pre-clinical studies have raised safety concerns and pinpointed the need for safer and more efficient approaches to generate and maintain patient-specific iPSCs. One approach is searching for compounds that influence pluripotent stem cell reprogramming using functional screens of known drugs. Our high-throughput screening of drug-like hits showed that imidazopyridines—analogs of zolpidem, a sedative-hypnotic drug—are able to improve reprogramming efficiency and facilitate reprogramming of resistant human primary fibroblasts. The lead compound (O4I3) showed a remarkable OCT4 induction, which at least in part is due to the inhibition of H3K4 demethylase (KDM5, also known as JARID1). Experiments demonstrated that KDM5A, but not its homolog KDM5B, serves as a reprogramming barrier by interfering with the enrichment of H3K4Me3 at the OCT4 promoter. Thus our results introduce a new class of KDM5 chemical inhibitors and provide further insight into the pluripotency-related properties of KDM5 family members. O4I3 supports the maintenance and generation of human iPSCs O4I3 is a potent H3K4 demethylase KDM5 inhibitor in vitro and in cells KDM5A, but not KDM5B, serves as an epigenetic barrier of reprogramming Chemical or genetic inhibition of KDM5A tends to promote the reprogramming efficiency
Collapse
Affiliation(s)
- Yasamin Dabiri
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, ImNeuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Rodrigo A Gama-Brambila
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, ImNeuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Katerina Taškova
- Faculty of Biology, University Mainz, Gresemundweg 2, 55128 Mainz, Germany; Institute of Molecular Biology GmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Kristina Herold
- Experimentelle Nephrologie, KIM III, Universitätsklinikum, 07743 Jena, Germany
| | - Stefanie Reuter
- Experimentelle Nephrologie, KIM III, Universitätsklinikum, 07743 Jena, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 68167 Mannheim, Germany
| | - Ralf Mrowka
- Experimentelle Nephrologie, KIM III, Universitätsklinikum, 07743 Jena, Germany
| | - Jichang Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Miguel A Andrade-Navarro
- Faculty of Biology, University Mainz, Gresemundweg 2, 55128 Mainz, Germany; Institute of Molecular Biology GmbH, Ackermannweg 4, 55128 Mainz, Germany
| | - Xinlai Cheng
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, ImNeuenheimer Feld 364, 69120 Heidelberg, Germany.
| |
Collapse
|
115
|
Xu P, Wang TT, Liu XZ, Wang NY, Sun LH, Zhang ZQ, Chen HZ, Lv X, Huang Y, Liu DP. Sirt6 regulates efficiency of mouse somatic reprogramming and maintenance of pluripotency. Stem Cell Res Ther 2019; 10:9. [PMID: 30630525 PMCID: PMC6329104 DOI: 10.1186/s13287-018-1109-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/04/2018] [Accepted: 12/13/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Mouse somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by defined factors known to regulate pluripotency, including Oct4, Sox2, Klf4, and c-Myc. It has been reported that Sirtuin 6 (Sirt6), a member of the sirtuin family of NAD+-dependent protein deacetylases, is involved in embryonic stem cell differentiation. However, whether and how Sirt6 influences epigenetic reprogramming remains unknown. METHODS Mouse embryonic fibroblasts isolated from transgenic Oct4-GFP reporter mice with or without Sirt6 were used for reprogramming by Yamanaka factors. Alkaline phosphatase-positive and OCT4-GFP-positive colony were counted to calculate reprogramming efficiency. OP9 feeder cell co-culture system was used to measure the hematopoietic differentiation from mouse ES and iPS cells. RNA sequencing was measured to identify the differential expressed genes due to loss of Sirt6 in somatic and pluripotent cells. RESULTS In this study, we provide evidence that Sirt6 is involved in mouse somatic reprogramming. We found that loss of function of Sirt6 could significantly decrease reprogramming efficiency. Furthermore, we showed that Sirt6-null iPS-like cell line has intrinsically a differentiation defect even though the establishment of normal self-renewal. Particularly, by performing transcriptome analysis, we observed that several pluripotent transcriptional factors increase in knockout cell line, which explains the underlying loss of pluripotency in Sirt6-null iPS-like cell line. CONCLUSIONS Taken together, we have identified a new regulatory role of Sirt6 in reprogramming and maintenance of pluripotency.
Collapse
Affiliation(s)
- Peng Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Ting-ting Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Xiu-zhen Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Nan-Yu Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Li-hong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005 China
| | - Zhu-qin Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Hou-zao Chen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Xiang Lv
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Yue Huang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005 China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| |
Collapse
|
116
|
Pennings S, Revuelta A, McLaughlin KA, Abd Hadi NA, Petchreing P, Ottaviano R, Meehan RR. Dynamics and Mechanisms of DNA Methylation Reprogramming. EPIGENETICS AND REGENERATION 2019:19-45. [DOI: 10.1016/b978-0-12-814879-2.00002-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
117
|
Abstract
Pritumumab, a natural human IgG1kappa mAb, was isolated from the regional lymph node of a patient with cervical cancer. This antibody has been reported to bind the cytoskeletal protein vimentin, and to cell surface expressed vimentin referred to as ecto-domain vimentin (EDV). Here, we report details of the development of a potency of binding assay for pritumumab as a prerequisite before pursuing clinical trials. The enzyme linked immunosorbent assay (ELISA) to detect antibody-binding antigen can serve as a potency assay for release of manufactured samples to be used in clinical studies. Several layers of controls for this assay along with suitability testing for reagents and components of the assay must be developed before the assay can be incorporated for stability testing and release of manufatured samples.
Collapse
|
118
|
Cheng F, Zhang Y, Wang Y, Jiang Q, Zhao CJ, Deng J, Chen X, Yao Y, Xia Z, Cheng L, Dai L, Shi G, Yang Y, Zhang S, Yu D, Wei Y, Deng H. Conversion of human adipose-derived stem cells into functional and expandable endothelial-like cells for cell-based therapies. Stem Cell Res Ther 2018; 9:350. [PMID: 30558659 PMCID: PMC6296081 DOI: 10.1186/s13287-018-1088-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 02/05/2023] Open
Abstract
Background Ischemic vascular diseases are the major cause of death worldwide. In recent years, endothelial cell (EC)-based approaches to vascular regeneration are increasingly viable strategies for treating ischemic diseases, but their applications are challenged by the difficulties in their efficient generation and stable maintenance. Here, we show an alternative protocol that facilitates the generation of functional and expandable ETS variant 2 (ETV2)-induced endothelial-like cells (EiECs) from human adipose-derived stem cells (hADSCs), providing a potential source of cells for autologous ECs to treat ischemic vascular diseases. Methods hADSCs were obtained from fresh human adipose tissue. Passage 3 hADSCs were transduced with doxycycline (DOX)-inducible ETV2 transcription factor; purified ETV2-hADSCs were induced into endothelial-like cells using a two-stage induction culture system composed of small molecule compounds and cell factors. EiECs were evaluated for their surface markers, proliferation, gene expression, secretory capacity, and effects on vascular regeneration in vivo. Results We found that short-term ETV2 expression combined with TGF-β inhibition is sufficient for the generation of kinase insert domain receptor (KDR)+ cells from hADSCs within 10 days. KDR+ cells showed immature endothelial characteristics, and they can gradually mature in a chemically defined induction medium at the second stage of induction. Futher studies showed that KDR+ cells deriving EC-like cells could stably self-renew and expand about 106-fold in 1 month, and they exhibited expected genome-wide molecular features of mature ECs. Functionally, these EC-like cells significantly promoted revascularization in a hind limb ischemic model. Conclusions We isolated highly purified hADSCs and effectively converted them into functional and expandable endothelial-like cells. Thus, the study may provide an alternative strategy to obtain functional EC-like cells with potential for biomedical and pharmaceutical applications. Electronic supplementary material The online version of this article (10.1186/s13287-018-1088-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fuyi Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yujing Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qingyuan Jiang
- Department of Obstetrics, Sichuan Provincial Hospital for Women and Children, Chengdu, People's Republic of China
| | - Cheng Jian Zhao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jie Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaolei Chen
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yunqi Yao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhemin Xia
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Shuang Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
119
|
Bindhya S, Sidhanth C, Shabna A, Krishnapriya S, Garg M, Ganesan TS. Induced pluripotent stem cells: A new strategy to model human cancer. Int J Biochem Cell Biol 2018; 107:62-68. [PMID: 30557622 DOI: 10.1016/j.biocel.2018.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/16/2022]
Abstract
Induced pluripotent stem cells are derived from adult somatic cells by ectopic expression of stem cell factors OCT4, SOX2, MYC and KLF4. These cells have characteristic features similar to embryonic stem cells. Although there exists in vitro and in vivo models of cancer, recapitulating the earliest events in the pathogenesis remain challenging. More recently, induced pluripotent stem cells have been generated to model human disease and cancer. There are advantages in the cancer models derived from these cells as compared to existing conventional approaches. Induced pluripotent stem cells have been generated from cancer cell lines, primary tumours and from those with an inherited predisposition to develop cancer. In addition, these cells provide a valuable tool in understanding the pathogenesis of familial cancer in its earliest stages, and to identify additional genetic alterations that are required to develop cancer. Furthermore, these cells can serve as a resource in drug screening and developing new therapies.
Collapse
Affiliation(s)
- S Bindhya
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - C Sidhanth
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - A Shabna
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - S Krishnapriya
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - M Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Uttar Pradesh, India
| | - T S Ganesan
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India.
| |
Collapse
|
120
|
He B, Deng T, Zhu I, Furusawa T, Zhang S, Tang W, Postnikov Y, Ambs S, Li CC, Livak F, Landsman D, Bustin M. Binding of HMGN proteins to cell specific enhancers stabilizes cell identity. Nat Commun 2018; 9:5240. [PMID: 30532006 PMCID: PMC6286339 DOI: 10.1038/s41467-018-07687-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/15/2018] [Indexed: 01/10/2023] Open
Abstract
The dynamic nature of the chromatin epigenetic landscape plays a key role in the establishment and maintenance of cell identity, yet the factors that affect the dynamics of the epigenome are not fully known. Here we find that the ubiquitous nucleosome binding proteins HMGN1 and HMGN2 preferentially colocalize with epigenetic marks of active chromatin, and with cell-type specific enhancers. Loss of HMGNs enhances the rate of OSKM induced reprogramming of mouse embryonic fibroblasts (MEFs) into induced pluripotent stem cells (iPSCs), and the ASCL1 induced conversion of fibroblast into neurons. During transcription factor induced reprogramming to pluripotency, loss of HMGNs accelerates the erasure of the MEF-specific epigenetic landscape and the establishment of an iPSCs-specific chromatin landscape, without affecting the pluripotency potential and the differentiation potential of the reprogrammed cells. Thus, HMGN proteins modulate the plasticity of the chromatin epigenetic landscape thereby stabilizing, rather than determining cell identity. HMGN1 and HMGN2 are ubiquitous nucleosome binding proteins. Here the authors provide evidence that HMGN proteins preferentially localize to chromatin regulatory sites to modulate the plasticity of the epigenetic landscape, proposing that HGMNs stabilize, rather than determine, cell identity.
Collapse
Affiliation(s)
- Bing He
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tao Deng
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Iris Zhu
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Takashi Furusawa
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shaofei Zhang
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Tang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuri Postnikov
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Caiyi Cherry Li
- Laboratory of Genomic Integrity, Center for Cancer Research National Cancer Institute National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ferenc Livak
- Laboratory of Genomic Integrity, Center for Cancer Research National Cancer Institute National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Landsman
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael Bustin
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
121
|
MYC Induces a Hybrid Energetics Program Early in Cell Reprogramming. Stem Cell Reports 2018; 11:1479-1492. [PMID: 30472011 PMCID: PMC6294174 DOI: 10.1016/j.stemcr.2018.10.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 10/25/2018] [Accepted: 10/25/2018] [Indexed: 01/07/2023] Open
Abstract
Cell reprogramming is thought to be associated with a full metabolic switch from an oxidative- to a glycolytic-based metabolism. However, neither the dynamics nor the factors controlling this metabolic switch are fully understood. By using cellular, biochemical, protein array, metabolomic, and respirometry analyses, we found that c-MYC establishes a robust bivalent energetics program early in cell reprogramming. Cells prone to undergo reprogramming exhibit high mitochondrial membrane potential and display a hybrid metabolism. We conclude that MYC proteins orchestrate a rewiring of somatic cell metabolism early in cell reprogramming, whereby somatic cells acquire the phenotypic plasticity necessary for their transition to pluripotency in response to either intrinsic or external cues. Endogenous MYC biological activity is necessary for cell reprogramming MYC drives mitochondrial fission early in cell reprogramming MYC establishes a bivalent energetics state necessary for cell reprogramming MYC polarizes mitochondria, poising cells for reprogramming
Collapse
|
122
|
Sardina JL, Collombet S, Tian TV, Gómez A, Di Stefano B, Berenguer C, Brumbaugh J, Stadhouders R, Segura-Morales C, Gut M, Gut IG, Heath S, Aranda S, Di Croce L, Hochedlinger K, Thieffry D, Graf T. Transcription Factors Drive Tet2-Mediated Enhancer Demethylation to Reprogram Cell Fate. Cell Stem Cell 2018; 23:727-741.e9. [DOI: 10.1016/j.stem.2018.08.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 07/07/2018] [Accepted: 08/23/2018] [Indexed: 10/28/2022]
|
123
|
Cossec JC, Theurillat I, Chica C, Búa Aguín S, Gaume X, Andrieux A, Iturbide A, Jouvion G, Li H, Bossis G, Seeler JS, Torres-Padilla ME, Dejean A. SUMO Safeguards Somatic and Pluripotent Cell Identities by Enforcing Distinct Chromatin States. Cell Stem Cell 2018; 23:742-757.e8. [PMID: 30401455 DOI: 10.1016/j.stem.2018.10.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022]
Abstract
Understanding general principles that safeguard cellular identity should reveal critical insights into common mechanisms underlying specification of varied cell types. Here, we show that SUMO modification acts to stabilize cell fate in a variety of contexts. Hyposumoylation enhances pluripotency reprogramming in vitro and in vivo, increases lineage transdifferentiation, and facilitates leukemic cell differentiation. Suppressing sumoylation in embryonic stem cells (ESCs) promotes their conversion into 2-cell-embryo-like (2C-like) cells. During reprogramming to pluripotency, SUMO functions on fibroblastic enhancers to retain somatic transcription factors together with Oct4, Sox2, and Klf4, thus impeding somatic enhancer inactivation. In contrast, in ESCs, SUMO functions on heterochromatin to silence the 2C program, maintaining both proper H3K9me3 levels genome-wide and repression of the Dux locus by triggering recruitment of the sumoylated PRC1.6 and Kap/Setdb1 repressive complexes. Together, these studies show that SUMO acts on chromatin as a glue to stabilize key determinants of somatic and pluripotent states.
Collapse
Affiliation(s)
- Jack-Christophe Cossec
- Nuclear Organization and Oncogenesis Unit, Equipe Labellisée Ligue Nationale Contre le Cancer, Institut Pasteur, 75015 Paris, France; INSERM, U993, 75015 Paris, France
| | - Ilan Theurillat
- Nuclear Organization and Oncogenesis Unit, Equipe Labellisée Ligue Nationale Contre le Cancer, Institut Pasteur, 75015 Paris, France; INSERM, U993, 75015 Paris, France; Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Claudia Chica
- Bioinformatics and Biostatistics Hub - C3BI, USR 3756 Institut Pasteur & CNRS, 75015 Paris, France
| | - Sabela Búa Aguín
- Cellular Plasticity and Disease Modelling Unit, Institut Pasteur, 75015 Paris, France; CNRS UMR3738, 75015 Paris, France
| | - Xavier Gaume
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, München, Germany
| | - Alexandra Andrieux
- Nuclear Organization and Oncogenesis Unit, Equipe Labellisée Ligue Nationale Contre le Cancer, Institut Pasteur, 75015 Paris, France; INSERM, U993, 75015 Paris, France
| | - Ane Iturbide
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, München, Germany
| | - Gregory Jouvion
- Experimental Neuropathology Unit, Institut Pasteur, 75015 Paris, France
| | - Han Li
- Cellular Plasticity and Disease Modelling Unit, Institut Pasteur, 75015 Paris, France; CNRS UMR3738, 75015 Paris, France
| | - Guillaume Bossis
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Jacob-Sebastian Seeler
- Nuclear Organization and Oncogenesis Unit, Equipe Labellisée Ligue Nationale Contre le Cancer, Institut Pasteur, 75015 Paris, France; INSERM, U993, 75015 Paris, France
| | | | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, Equipe Labellisée Ligue Nationale Contre le Cancer, Institut Pasteur, 75015 Paris, France; INSERM, U993, 75015 Paris, France.
| |
Collapse
|
124
|
Mackey LC, Annab LA, Yang J, Rao B, Kissling GE, Schurman SH, Dixon D, Archer TK. Epigenetic Enzymes, Age, and Ancestry Regulate the Efficiency of Human iPSC Reprogramming. Stem Cells 2018; 36:1697-1708. [PMID: 30152570 DOI: 10.1002/stem.2899] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 12/17/2022]
Abstract
Epigenetic enzymes regulate higher-order chromatin architecture and cell-type specific gene expression. The ATPase BRG1 and the SWI/SNF chromatin remodeling complex are epigenetic enzymes that regulate chromatin accessibility during steady and transitional cell states. Experiments in mice show that the loss of BRG1 inhibits cellular reprogramming, while studies using human cells demonstrate that the overexpression of BRG1 enhances reprogramming. We hypothesized that the variation of SWI/SNF subunit expression in the human population would contribute to variability in the efficiency of induced pluripotent stem cells (iPSC) reprogramming. To examine the impact of an individual's sex, ancestry, and age on iPSC reprogramming, we created a novel sex and ancestry balanced cohort of 240 iPSC lines derived from human dermal fibroblasts (DF) from 80 heathy donors. We methodically assessed the reprogramming efficiency of each DF line and then quantified the individual and demographic-specific variations in SWI/SNF chromatin remodeling proteins and mRNA expression. We identified BRG1, BAF155, and BAF60a expression as strongly correlating with iPSC reprogramming efficiency. Additionally, we discovered that high efficiency iPSC reprograming is negatively correlated with donor age, positively correlated with African American descent, and uncorrelated with donor sex. These results show the variations in chromatin remodeling protein expression have a strong impact on iPSC reprogramming. Additionally, our cohort is unique in its large size, diversity, and focus on healthy donors. Consequently, this cohort can be a vital tool for researchers seeking to validate observational results from human population studies and perform detailed mechanistic studies in a controlled cell culture environment. Stem Cells 2018;36:1697-1708.
Collapse
Affiliation(s)
- Lantz C Mackey
- Epigenetics & Stem Cell Biology Laboratory, Chromatin & Gene Expression Group, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Lois A Annab
- Epigenetics & Stem Cell Biology Laboratory, Chromatin & Gene Expression Group, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Jun Yang
- Epigenetics & Stem Cell Biology Laboratory, Chromatin & Gene Expression Group, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Bhargavi Rao
- Epigenetics & Stem Cell Biology Laboratory, Chromatin & Gene Expression Group, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Grace E Kissling
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Shepard H Schurman
- Clinical Research Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Darlene Dixon
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Trevor K Archer
- Epigenetics & Stem Cell Biology Laboratory, Chromatin & Gene Expression Group, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| |
Collapse
|
125
|
Osuna-Luque J, Rodríguez-Ramos Á, Gámez-Del-Estal MDM, Ruiz-Rubio M. Behavioral Mechanisms That Depend on Dopamine and Serotonin in Caenorhabditis elegans Interact With the Antipsychotics Risperidone and Aripiprazole. J Exp Neurosci 2018; 12:1179069518798628. [PMID: 30245571 PMCID: PMC6144587 DOI: 10.1177/1179069518798628] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/14/2018] [Indexed: 11/20/2022] Open
Abstract
The neurotransmitters dopamine and serotonin participate in specific behavioral neuromuscular mechanisms in the nematode Caenorhabditis elegans. Dopamine is involved in the gentle touch response and serotonin in the pharyngeal pumping rate. In its genome, the worm presents genes encoding dopamine and serotonin receptors orthologous to those of human genes. Risperidone and aripiprazole are a class of drugs known as atypical antipsychotics commonly used to treat schizophrenia, bipolar disorder, and irritability associated with autism. Risperidone is an antagonist of the dopamine D2 and serotonin 5-HT2A receptors. Aripiprazole functions as a partial agonist of the dopamine D2 receptor and as a partial agonist and antagonist of 5-HT1A and 5-HT2A serotonin receptors, respectively. Our results show that risperidone and aripiprazole alter the touch response and pharyngeal pumping in wild-type worm animals. Furthermore, in the presence of the drugs, both behaviors change to varying degrees in dopamine (dop-1, dop-2, and dop-3), serotonin (ser-1), and tyramine (ser-2) receptor-deficient mutants. This variation in response reveals specific targets for these antipsychotics in the nematode. Interestingly, their effect on behavior persisted to some extent in successive generations, indicating that they might induce epigenetic changes throughout development. Sodium butyrate, a histone deacetylase inhibitor, eliminated the consecutive generation effect of both drugs. In addition, these transgenerational effects were also abolished after the dauer stage. These observations suggest that risperidone and aripiprazole, in addition to interacting with specific receptors impairing the function of the nervous system of the nematode, may lead to the deposition of long-lasting epigenetic marks.
Collapse
Affiliation(s)
- Jaime Osuna-Luque
- Department of Genetics, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain.,University Hospital Reina Sofía, Córdoba, Spain
| | - Ángel Rodríguez-Ramos
- Department of Genetics, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain.,University Hospital Reina Sofía, Córdoba, Spain
| | - María Del Mar Gámez-Del-Estal
- Department of Genetics, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain.,University Hospital Reina Sofía, Córdoba, Spain
| | - Manuel Ruiz-Rubio
- Department of Genetics, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain.,University Hospital Reina Sofía, Córdoba, Spain
| |
Collapse
|
126
|
Wu S, FitzGerald KT, Giordano J. On the Viability and Potential Value of Stem Cells for Repair and Treatment of Central Neurotrauma: Overview and Speculations. Front Neurol 2018; 9:602. [PMID: 30150968 PMCID: PMC6099099 DOI: 10.3389/fneur.2018.00602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/06/2018] [Indexed: 12/12/2022] Open
Abstract
Central neurotrauma, such as spinal cord injury or traumatic brain injury, can damage critical axonal pathways and neurons and lead to partial to complete loss of neural function that is difficult to address in the mature central nervous system. Improvement and innovation in the development, manufacture, and delivery of stem-cell based therapies, as well as the continued exploration of newer forms of stem cells, have allowed the professional and public spheres to resolve technical and ethical questions that previously hindered stem cell research for central nervous system injury. Recent in vitro and in vivo models have demonstrated the potential that reprogrammed autologous stem cells, in particular, have to restore functionality and induce regeneration-while potentially mitigating technical issues of immunogenicity, rejection, and ethical issues of embryonic derivation. These newer stem-cell based approaches are not, however, without concerns and problems of safety, efficacy, use and distribution. This review is an assessment of the current state of the science, the potential solutions that have been and are currently being explored, and the problems and questions that arise from what appears to be a promising way forward (i.e., autologous stem cell-based therapies)-for the purpose of advancing the research for much-needed therapeutic interventions for central neurotrauma.
Collapse
Affiliation(s)
- Samantha Wu
- Pellegrino Center for Clinical Bioethics, Georgetown University Medical Center, Washington, DC, United States
| | - Kevin T. FitzGerald
- Pellegrino Center for Clinical Bioethics, Georgetown University Medical Center, Washington, DC, United States
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - James Giordano
- Pellegrino Center for Clinical Bioethics, Georgetown University Medical Center, Washington, DC, United States
- Departments of Neurology and Biochemistry, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
127
|
Zhao W, Zhu Q, Tan P, Ajibade A, Long T, Long W, Li Q, Liu P, Ning B, Wang HY, Wang RF. Tgfbr2 inactivation facilitates cellular plasticity and development of Pten-null prostate cancer. J Mol Cell Biol 2018; 10:316-330. [PMID: 29228234 PMCID: PMC6161409 DOI: 10.1093/jmcb/mjx052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 10/31/2017] [Accepted: 12/06/2017] [Indexed: 12/26/2022] Open
Abstract
Mutations in tumors can create a state of increased cellular plasticity that promotes resistance to treatment. Thus, there is an urgent need to develop novel strategies for identifying key factors that regulate cellular plasticity in order to combat resistance to chemotherapy and radiation treatment. Here we report that prostate epithelial cell reprogramming could be exploited to identify key factors required for promoting prostate cancer tumorigenesis and cellular plasticity. Deletion of phosphatase and tensin homolog (Pten) and transforming growth factor-beta receptor type 2 (Tgfbr2) may increase prostate epithelial cell reprogramming efficiency in vitro and cause rapid tumor development and early mortality in vivo. Tgfbr2 ablation abolished TGF-β signaling but increased the bone morphogenetic protein (BMP) signaling pathway through the negative regulator Tmeff1. Furthermore, increased BMP signaling promotes expression of the tumor marker genes ID1, Oct4, Nanog, and Sox2; ID1/STAT3/NANOG expression was inversely correlated with patient survival. Thus, our findings provide information about the molecular mechanisms by which BMP signaling pathways render stemness capacity to prostate tumor cells.
Collapse
Affiliation(s)
- Wei Zhao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qingyuan Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Peng Tan
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, USA
| | - Adebusola Ajibade
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Teng Long
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenyong Long
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qingtian Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Pinghua Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Bo Ning
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, USA
| |
Collapse
|
128
|
Kim JJ, Moghe PV. Parsing Stem Cell Lineage Development Using High Content Image Analysis of Epigenetic Spatial Markers. ACTA ACUST UNITED AC 2018; 46:e54. [PMID: 29927102 DOI: 10.1002/cpsc.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This unit describes a protocol for acquiring and analyzing high-content super-resolution images of human stem cell nuclei for the characterization and classification of the cell differentiation paths based on distinct patterns of epigenetic mark organization. Here, we describe the cell culture, immunocytochemical labeling, super-resolution imaging parameters, and MATLAB-based quantitative image analysis approaches for monitoring human mesenchymal stem cells (hMSCs) and human induced pluripotent stem cells (hiPSCs) as the cells differentiate towards various lineages. Although this protocol uses specific cell types as examples, this approach could be easily extended to a variety of cell types and nuclear epigenetic and mechanosensitive biomarkers that are relevant to specific cell developmental scenarios. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Joseph J Kim
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
129
|
Apostolou E, Stadtfeld M. Cellular trajectories and molecular mechanisms of iPSC reprogramming. Curr Opin Genet Dev 2018; 52:77-85. [PMID: 29925040 DOI: 10.1016/j.gde.2018.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/24/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has solidified the concept of transcription factors as major players in controlling cell identity and provided a tractable tool to study how somatic cell identity can be dismantled and pluripotency established. A number of landmark studies have established hallmarks and roadmaps of iPSC formation by describing relative kinetics of transcriptional, protein and epigenetic changes, including alterations in DNA methylation and histone modifications. Recently, technological advancements such as single-cell analyses, high-resolution genome-wide chromatin assays and more efficient reprogramming systems have been used to challenge and refine our understanding of the reprogramming process. Here, we will outline novel insights into the molecular mechanisms underlying iPSC formation, focusing on how the core reprogramming factors OCT4, KLF4, SOX2 and MYC (OKSM) drive changes in gene expression, chromatin state and 3D genome topology. In addition, we will discuss unexpected consequences of reprogramming factor expression in in vitro and in vivo systems that may point towards new applications of iPSC technology.
Collapse
Affiliation(s)
- Effie Apostolou
- Edward and Sandra Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Matthias Stadtfeld
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology and Helen L. and Martin S. Kimmel Center for Biology and Medicine, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
130
|
Tumorigenic Cell Reprogramming and Cancer Plasticity: Interplay between Signaling, Microenvironment, and Epigenetics. Stem Cells Int 2018; 2018:4598195. [PMID: 29853913 PMCID: PMC5954911 DOI: 10.1155/2018/4598195] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/01/2018] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidences indicate that many tumors rely on subpopulations of cancer stem cells (CSCs) with the ability to propagate malignant clones indefinitely and to produce an overt cancer. Of importance, CSCs seem to be more resistant to the conventional cytotoxic treatments, driving tumor growth and contributing to relapse. CSCs can originate from normal committed cells which undergo tumor-reprogramming processes and reacquire a stem cell-like phenotype. Increasing evidences also show how tumor homeostasis and progression strongly rely on the capacity of nontumorigenic cancer cells to dedifferentiate to CSCs. Both tumor microenvironment and epigenetic reprogramming drive such dynamic mechanisms, favoring cancer cell plasticity and tumor heterogeneity. Here, we report new developments which led to an advancement in the CSC field, elucidating the concepts of cancer cell of origin and CSC plasticity in solid tumor initiation and maintenance. We further discuss the main signaling pathways which, under the influence of extrinsic environmental factors, play a critical role in the formation and maintenance of CSCs. Moreover, we propose a review of the main epigenetic mechanisms whose deregulation can favor the onset of CSC features both in tumor initiation and tumor maintenance. Finally, we provide an update of the main strategies that could be applied to target CSCs and cancer cell plasticity.
Collapse
|
131
|
Pantelic MN, Larkin LM. Stem Cells for Skeletal Muscle Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:373-391. [PMID: 29652595 DOI: 10.1089/ten.teb.2017.0451] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Volumetric muscle loss (VML) is a debilitating condition wherein muscle loss overwhelms the body's normal physiological repair mechanism. VML is particularly common among military service members who have sustained war injuries. Because of the high social and medical cost associated with VML and suboptimal current surgical treatments, there is great interest in developing better VML therapies. Skeletal muscle tissue engineering (SMTE) is a promising alternative to traditional VML surgical treatments that use autogenic tissue grafts, and rather uses isolated stem cells with myogenic potential to generate de novo skeletal muscle tissues to treat VML. Satellite cells are the native precursors to skeletal muscle tissue, and are thus the most commonly studied starting source for SMTE. However, satellite cells are difficult to isolate and purify, and it is presently unknown whether they would be a practical source in clinical SMTE applications. Alternative myogenic stem cells, including adipose-derived stem cells, bone marrow-derived mesenchymal stem cells, perivascular stem cells, umbilical cord mesenchymal stem cells, induced pluripotent stem cells, and embryonic stem cells, each have myogenic potential and have been identified as possible starting sources for SMTE, although they have yet to be studied in detail for this purpose. These alternative stem cell varieties offer unique advantages and disadvantages that are worth exploring further to advance the SMTE field toward highly functional, safe, and practical VML treatments. The following review summarizes the current state of satellite cell-based SMTE, details the properties and practical advantages of alternative myogenic stem cells, and offers guidance to tissue engineers on how alternative myogenic stem cells can be incorporated into SMTE research.
Collapse
Affiliation(s)
- Molly N Pantelic
- 1 Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - Lisa M Larkin
- 1 Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan.,2 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
132
|
Pijuan-Sala B, Guibentif C, Göttgens B. Single-cell transcriptional profiling: a window into embryonic cell-type specification. Nat Rev Mol Cell Biol 2018; 19:399-412. [DOI: 10.1038/s41580-018-0002-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
133
|
Ye B, Liu B, Hao L, Zhu X, Yang L, Wang S, Xia P, Du Y, Meng S, Huang G, Qin X, Wang Y, Yan X, Li C, Hao J, Zhu P, He L, Tian Y, Fan Z. Klf4 glutamylation is required for cell reprogramming and early embryonic development in mice. Nat Commun 2018; 9:1261. [PMID: 29593216 PMCID: PMC5871780 DOI: 10.1038/s41467-018-03008-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/10/2018] [Indexed: 12/20/2022] Open
Abstract
Temporal and spatial-specific regulation of pluripotency networks is largely dependent on the precise modifications of core transcription factors. Misregulation of glutamylation is implicated in severe physiological abnormalities. However, how glutamylation regulates cell reprogramming and pluripotency networks remains elusive. Here we show that cytosolic carboxypeptidases 1 (CCP1) or CCP6 deficiency substantially promotes induced pluripotent cell (iPSC) induction and pluripotency of embryonic stem cells (ESCs). Klf4 polyglutamylation at Glu381 by tubulin tyrosine ligase-like 4 (TTLL4) and TTLL1 during cell reprogramming impedes its lysine 48-linked ubiquitination and sustains Klf4 stability. Klf4-E381A knockin mice display impaired blastocyst development and embryonic lethality. Deletion of TTLL4 or TTLL1 abrogates cell reprogramming and early embryogenesis. Thus, Klf4 polyglutamylation plays a critical role in the regulation of cell reprogramming and pluripotency maintenance. Embryonic stem cell pluripotency depends upon precise regulation by a core transcription network. Here the authors show that polyglutamylation mediated stabilization of the transcription factor Klf4 by TTLL1 and TTLL4 promotes reprogramming, pluripotency and preimplantation embryonic development.
Collapse
Affiliation(s)
- Buqing Ye
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Benyu Liu
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lu Hao
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxiao Zhu
- Laboratory Animal Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liuliu Yang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuo Wang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pengyan Xia
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying Du
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shu Meng
- Laboratory Animal Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guanling Huang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiwen Qin
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanying Wang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinlong Yan
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chong Li
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Junfeng Hao
- Laboratory Animal Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pingping Zhu
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Luyun He
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Tian
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zusen Fan
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
134
|
Corso S, Cargnelutti M, Durando S, Menegon S, Apicella M, Migliore C, Capeloa T, Ughetto S, Isella C, Medico E, Bertotti A, Sassi F, Sarotto I, Casorzo L, Pisacane A, Mangioni M, Sottile A, Degiuli M, Fumagalli U, Sgroi G, Molfino S, De Manzoni G, Rosati R, De Simone M, Marrelli D, Saragoni L, Rausei S, Pallabazzer G, Roviello F, Cassoni P, Sapino A, Bass A, Giordano S. Rituximab Treatment Prevents Lymphoma Onset in Gastric Cancer Patient-Derived Xenografts. Neoplasia 2018; 20:443-455. [PMID: 29574251 PMCID: PMC5915970 DOI: 10.1016/j.neo.2018.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/06/2018] [Accepted: 02/14/2018] [Indexed: 12/13/2022] Open
Abstract
Patient-Derived Xenografts (PDXs), entailing implantation of cancer specimens in immunocompromised mice, are emerging as a valuable translational model that could help validate biologically relevant targets and assist the clinical development of novel therapeutic strategies for gastric cancer. More than 30% of PDXs generated from gastric carcinoma samples developed human B-cell lymphomas instead of gastric cancer. These lymphomas were monoclonal, Epstein Barr Virus (EBV) positive, originated tumorigenic cell cultures and displayed a mutational burden and an expression profile distinct from gastric adenocarcinomas. The ability of grafted samples to develop lymphomas did not correlate with patient outcome, nor with the histotype, the lymphocyte infiltration level, or the EBV status of the original gastric tumor, impeding from foreseeing lymphoma onset. Interestingly, lymphoma development was significantly more frequent when primary rather than metastatic samples were grafted. Notably, the development of such lympho-proliferative disease could be prevented by a short rituximab treatment upon mice implant, without negatively affecting gastric carcinoma engraftment. Due to the high frequency of human lymphoma onset, our data show that a careful histologic analysis is mandatory when generating gastric cancer PDXs. Such care would avoid misleading results that could occur if testing of putative gastric cancer therapies is performed in lymphoma PDXs. We propose rituximab treatment of mice to prevent lymphoma development in PDX models, averting the loss of human-derived samples.
Collapse
Affiliation(s)
- Simona Corso
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| | | | | | | | | | - Cristina Migliore
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Tania Capeloa
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Stefano Ughetto
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Andrea Bertotti
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Ivana Sarotto
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Laura Casorzo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | | | | | | | | | - Giovanni Sgroi
- Surgical Oncology Unit, Surgical Science Department, ASST Bergamo Ovest, Treviglio (BG), Italy
| | - Sarah Molfino
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, Brescia, Italy
| | - Giovanni De Manzoni
- First Department of General Surgery, Borgo Trento Hospital, University of Verona, Italy
| | - Riccardo Rosati
- Gastroenterological Surgery Unit, IRCCS San Raffaele Hospital, Vita-Salute University, Milan, Italy
| | | | - Daniele Marrelli
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Italy
| | - Luca Saragoni
- Pathology Unit, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Stefano Rausei
- Department of Surgery, University of Insubria, Varese, Italy
| | | | - Franco Roviello
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Torino, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Torino, Italy
| | - Adam Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
135
|
NCoR/SMRT co-repressors cooperate with c-MYC to create an epigenetic barrier to somatic cell reprogramming. Nat Cell Biol 2018. [PMID: 29531310 DOI: 10.1038/s41556-018-0047-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Somatic cell reprogramming by exogenous factors requires cooperation with transcriptional co-activators and co-repressors to effectively remodel the epigenetic environment. How this interplay is regulated remains poorly understood. Here, we demonstrate that NCoR/SMRT co-repressors bind to pluripotency loci to create a barrier to reprogramming with the four Yamanaka factors (OCT4, SOX2, KLF4 and c-MYC), and consequently, suppressing NCoR/SMRT significantly enhances reprogramming efficiency and kinetics. The core epigenetic subunit of the NCoR/SMRT complex, histone deacetylase 3 (HDAC3), contributes to the effects of NCoR/SMRT by inducing histone deacetylation at pluripotency loci. Among the Yamanaka factors, recruitment of NCoR/SMRT-HDAC3 to genomic loci is mostly facilitated by c-MYC. Hence, we describe how c-MYC is beneficial for the early phase of reprogramming but deleterious later. Overall, we uncover a role for NCoR/SMRT co-repressors in reprogramming and propose a dual function for c-MYC in this process.
Collapse
|
136
|
MYC-driven epigenetic reprogramming favors the onset of tumorigenesis by inducing a stem cell-like state. Nat Commun 2018. [PMID: 29523784 PMCID: PMC5844884 DOI: 10.1038/s41467-018-03264-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Breast cancer consists of highly heterogeneous tumors, whose cell of origin and driver oncogenes are difficult to be uniquely defined. Here we report that MYC acts as tumor reprogramming factor in mammary epithelial cells by inducing an alternative epigenetic program, which triggers loss of cell identity and activation of oncogenic pathways. Overexpression of MYC induces transcriptional repression of lineage-specifying transcription factors, causing decommissioning of luminal-specific enhancers. MYC-driven dedifferentiation supports the onset of a stem cell-like state by inducing the activation of de novo enhancers, which drive the transcriptional activation of oncogenic pathways. Furthermore, we demonstrate that the MYC-driven epigenetic reprogramming favors the formation and maintenance of tumor-initiating cells endowed with metastatic capacity. This study supports the notion that MYC-driven tumor initiation relies on cell reprogramming, which is mediated by the activation of MYC-dependent oncogenic enhancers, thus establishing a therapeutic rational for treating basal-like breast cancers.
Collapse
|
137
|
Liu Y, Pelham-Webb B, Di Giammartino DC, Li J, Kim D, Kita K, Saiz N, Garg V, Doane A, Giannakakou P, Hadjantonakis AK, Elemento O, Apostolou E. Widespread Mitotic Bookmarking by Histone Marks and Transcription Factors in Pluripotent Stem Cells. Cell Rep 2018; 19:1283-1293. [PMID: 28514649 DOI: 10.1016/j.celrep.2017.04.067] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/17/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022] Open
Abstract
During mitosis, transcription is halted and many chromatin features are lost, posing a challenge for the continuity of cell identity, particularly in fast cycling stem cells, which constantly balance self-renewal with differentiation. Here we show that, in pluripotent stem cells, certain histone marks and stem cell regulators remain associated with specific genomic regions of mitotic chromatin, a phenomenon known as mitotic bookmarking. Enhancers of stem cell-related genes are bookmarked by both H3K27ac and the master regulators OCT4, SOX2, and KLF4, while promoters of housekeeping genes retain high levels of mitotic H3K27ac in a cell-type invariant manner. Temporal degradation of OCT4 during mitotic exit compromises its ability both to maintain and induce pluripotency, suggesting that its regulatory function partly depends on its bookmarking activity. Together, our data document a widespread yet specific bookmarking by histone modifications and transcription factors promoting faithful and efficient propagation of stemness after cell division.
Collapse
Affiliation(s)
- Yiyuan Liu
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Bobbie Pelham-Webb
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Dafne Campigli Di Giammartino
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jiexi Li
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Daleum Kim
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Katsuhiro Kita
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nestor Saiz
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vidur Garg
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ashley Doane
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Paraskevi Giannakakou
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Effie Apostolou
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
138
|
Suppression of the ERK-SRF axis facilitates somatic cell reprogramming. Exp Mol Med 2018; 50:e448. [PMID: 29472703 PMCID: PMC5903827 DOI: 10.1038/emm.2017.279] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/18/2017] [Indexed: 01/10/2023] Open
Abstract
The molecular mechanism underlying the initiation of somatic cell reprogramming into induced pluripotent stem cells (iPSCs) has not been well described. Thus, we generated single-cell-derived clones by using a combination of drug-inducible vectors encoding transcription factors (Oct4, Sox2, Klf4 and Myc) and a single-cell expansion strategy. This system achieved a high reprogramming efficiency after metabolic and epigenetic remodeling. Functional analyses of the cloned cells revealed that extracellular signal-regulated kinase (ERK) signaling was downregulated at an early stage of reprogramming and that its inhibition was a driving force for iPSC formation. Among the reprogramming factors, Myc predominantly induced ERK suppression. ERK inhibition upregulated the conversion of somatic cells into iPSCs through concomitant suppression of serum response factor (SRF). Conversely, SRF activation suppressed the reprogramming induced by ERK inhibition and negatively regulated embryonic pluripotency by inducing differentiation via upregulation of immediate early genes, such as c-Jun, c-Fos and EGR1. These data reveal that suppression of the ERK-SRF axis is an initial molecular event that facilitates iPSC formation and may be a useful surrogate marker for cellular reprogramming.
Collapse
|
139
|
The epigenetic basis of cellular plasticity. Curr Opin Cell Biol 2018; 49:116-122. [PMID: 29413970 DOI: 10.1016/j.ceb.2018.01.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 12/23/2022]
Abstract
Cellular plasticity is now recognized as a fundamental feature of tissue biology. The steady-state differentiation of stem and progenitor cells into mature cells is, in itself, the index form of cellular plasticity in adult organisms. Following injury, when it is critical to quickly regenerate and restore tissue integrity and function, other types of cellular plasticity may be crucial for organismal survival. In these contexts, alterations in the epigenetic landscape of tissues are likely to occur in order to allow normally restricted cell fate transitions. Epigenetic mechanisms, particularly DNA methylation and histone modifications, have been shown to play an important role in regulating such plasticity. Relevant mechanisms have been well studied in the context of the direct reprograming of somatic cells into induced pluripotent stem cells. Indeed, epigenetic regulation of cell fate is part and parcel of normal embryonic development and is a central regulator of cellular diversity. This is normally thought to involve the establishment of divergent chromatin patterns that culminate in cells with distinct and what were previously thought to be irreversible fates. This brief review aims to put some of these new observations in the larger context of regeneration after injury.
Collapse
|
140
|
Apostolou E. Shaping the Pluripotent Genome: Switches, Borders, and Loops. Cell Stem Cell 2018; 22:148-150. [PMID: 29395051 DOI: 10.1016/j.stem.2018.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Three-dimensional genome organization is largely cell type specific and requires reorganization during cell fate transitions. A recent study in Nature Genetics (Stadhouders et al., 2018) offers important insights into the principles and drivers of such reorganization during reprogramming of somatic cells into pluripotent stem cells.
Collapse
Affiliation(s)
- Effie Apostolou
- Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
141
|
Stadhouders R, Vidal E, Serra F, Di Stefano B, Le Dily F, Quilez J, Gomez A, Collombet S, Berenguer C, Cuartero Y, Hecht J, Filion GJ, Beato M, Marti-Renom MA, Graf T. Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming. Nat Genet 2018; 50:238-249. [PMID: 29335546 PMCID: PMC5810905 DOI: 10.1038/s41588-017-0030-7] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 12/01/2017] [Indexed: 01/28/2023]
Abstract
Chromosomal architecture is known to influence gene expression, yet its role in controlling cell fate remains poorly understood. Reprogramming of somatic cells into pluripotent stem cells (PSCs) by the transcription factors (TFs) OCT4, SOX2, KLF4 and MYC offers an opportunity to address this question but is severely limited by the low proportion of responding cells. We have recently developed a highly efficient reprogramming protocol that synchronously converts somatic into pluripotent stem cells. Here, we used this system to integrate time-resolved changes in genome topology with gene expression, TF binding and chromatin-state dynamics. The results showed that TFs drive topological genome reorganization at multiple architectural levels, often before changes in gene expression. Removal of locus-specific topological barriers can explain why pluripotency genes are activated sequentially, instead of simultaneously, during reprogramming. Together, our results implicate genome topology as an instructive force for implementing transcriptional programs and cell fate in mammals.
Collapse
Affiliation(s)
- Ralph Stadhouders
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.
| | - Enrique Vidal
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - François Serra
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Structural Genomics Group, CNAG-CRG, BIST, Barcelona, Spain
| | - Bruno Di Stefano
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University and Harvard Medical School, Cambridge, MA, USA
| | - François Le Dily
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Structural Genomics Group, CNAG-CRG, BIST, Barcelona, Spain
| | - Javier Quilez
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Antonio Gomez
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Samuel Collombet
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, Paris, France
| | - Clara Berenguer
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Yasmina Cuartero
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Structural Genomics Group, CNAG-CRG, BIST, Barcelona, Spain
| | - Jochen Hecht
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Genomics Unit, CRG, BIST, Barcelona, Spain
| | - Guillaume J Filion
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Miguel Beato
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marc A Marti-Renom
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Structural Genomics Group, CNAG-CRG, BIST, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - Thomas Graf
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
142
|
Amlani B, Liu Y, Chen T, Ee LS, Lopez P, Heguy A, Apostolou E, Kim SY, Stadtfeld M. Nascent Induced Pluripotent Stem Cells Efficiently Generate Entirely iPSC-Derived Mice while Expressing Differentiation-Associated Genes. Cell Rep 2018; 22:876-884. [PMID: 29420174 DOI: 10.1016/j.celrep.2017.12.098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/22/2017] [Accepted: 12/26/2017] [Indexed: 01/02/2023] Open
Abstract
The ability of induced pluripotent stem cells (iPSCs) to differentiate into all adult cell types makes them attractive for research and regenerative medicine; however, it remains unknown when and how this capacity is established. We characterized the acquisition of developmental pluripotency in a suitable reprogramming system to show that iPSCs prior to passaging become capable of generating all tissues upon injection into preimplantation embryos. The developmental potential of nascent iPSCs is comparable to or even surpasses that of established pluripotent cells. Further functional assays and genome-wide molecular analyses suggest that cells acquiring developmental pluripotency exhibit a unique combination of properties that distinguish them from canonical naive and primed pluripotency states. These include reduced clonal self-renewal potential and the elevated expression of differentiation-associated transcriptional regulators. Our observations close a gap in the understanding of induced pluripotency and provide an improved roadmap of cellular reprogramming with ramifications for the use of iPSCs.
Collapse
Affiliation(s)
- Bhishma Amlani
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology and Helen L. and Martin S. Kimmel Center for Biology and Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Yiyuan Liu
- Edward and Sandra Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Taotao Chen
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology and Helen L. and Martin S. Kimmel Center for Biology and Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Ly-Sha Ee
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology and Helen L. and Martin S. Kimmel Center for Biology and Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Peter Lopez
- Cytometry and Cell Sorting Laboratory, NYU School of Medicine, New York, NY 10016, USA
| | - Adriana Heguy
- Department of Pathology and Office for Collaborative Science, NYU School of Medicine, New York, NY 10016, USA
| | - Effie Apostolou
- Edward and Sandra Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sang Yong Kim
- Department of Pathology and Office for Collaborative Science, NYU School of Medicine, New York, NY 10016, USA.
| | - Matthias Stadtfeld
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology and Helen L. and Martin S. Kimmel Center for Biology and Medicine, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
143
|
Brumbaugh J, Di Stefano B, Wang X, Borkent M, Forouzmand E, Clowers KJ, Ji F, Schwarz BA, Kalocsay M, Elledge SJ, Chen Y, Sadreyev RI, Gygi SP, Hu G, Shi Y, Hochedlinger K. Nudt21 Controls Cell Fate by Connecting Alternative Polyadenylation to Chromatin Signaling. Cell 2018; 172:106-120.e21. [PMID: 29249356 PMCID: PMC5766360 DOI: 10.1016/j.cell.2017.11.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/08/2017] [Accepted: 11/10/2017] [Indexed: 10/18/2022]
Abstract
Cell fate transitions involve rapid gene expression changes and global chromatin remodeling, yet the underlying regulatory pathways remain incompletely understood. Here, we identified the RNA-processing factor Nudt21 as a novel regulator of cell fate change using transcription-factor-induced reprogramming as a screening assay. Suppression of Nudt21 enhanced the generation of induced pluripotent stem cells, facilitated transdifferentiation into trophoblast stem cells, and impaired differentiation of myeloid precursors and embryonic stem cells, suggesting a broader role for Nudt21 in cell fate change. We show that Nudt21 directs differential polyadenylation of over 1,500 transcripts in cells acquiring pluripotency, although only a fraction changed protein levels. Remarkably, these proteins were strongly enriched for chromatin regulators, and their suppression neutralized the effect of Nudt21 during reprogramming. Collectively, our data uncover Nudt21 as a novel post-transcriptional regulator of cell fate and establish a direct, previously unappreciated link between alternative polyadenylation and chromatin signaling.
Collapse
Affiliation(s)
- Justin Brumbaugh
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Bruno Di Stefano
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Xiuye Wang
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Marti Borkent
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Elmira Forouzmand
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Katie J Clowers
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Benjamin A Schwarz
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Marian Kalocsay
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen J Elledge
- Howard Hughes Medical Institute, Brigham and Women's Hospital and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yue Chen
- Department of Biochemistry, Molecular Biology, and Biophysics, College of Biological Sciences, University of Minnesota, Saint Paul, MN 55018, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Yongsheng Shi
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA.
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
144
|
Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the hallmarks of cancer. Science 2018; 357:357/6348/eaal2380. [PMID: 28729483 DOI: 10.1126/science.aal2380] [Citation(s) in RCA: 818] [Impact Index Per Article: 136.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chromatin and associated epigenetic mechanisms stabilize gene expression and cellular states while also facilitating appropriate responses to developmental or environmental cues. Genetic, environmental, or metabolic insults can induce overly restrictive or overly permissive epigenetic landscapes that contribute to pathogenesis of cancer and other diseases. Restrictive chromatin states may prevent appropriate induction of tumor suppressor programs or block differentiation. By contrast, permissive or "plastic" states may allow stochastic oncogene activation or nonphysiologic cell fate transitions. Whereas many stochastic events will be inconsequential "passengers," some will confer a fitness advantage to a cell and be selected as "drivers." We review the broad roles played by epigenetic aberrations in tumor initiation and evolution and their potential to give rise to all classic hallmarks of cancer.
Collapse
Affiliation(s)
- William A Flavahan
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA, and Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Elizabeth Gaskell
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA, and Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bradley E Bernstein
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA, and Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
145
|
Induced Pluripotent Stem Cells and Induced Pluripotent Cancer Cells in Cancer Disease Modeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:169-183. [PMID: 30069853 DOI: 10.1007/5584_2018_257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In 2006, Noble Prize laureate Shinya Yamanaka discovered that a set of transcription factors can reprogram terminally differentiated somatic cells to a pluripotent stem cell state. Since then, induced pluripotent stem cells (iPSCs) have come into the public spotlight. Amidst a growing field of promising clinical uses of iPSCs in recent years, cancer disease modeling has emerged as a particularly promising and rapidly translatable application of iPSCs. Technological advances in genome editing over the past few years have facilitated increasingly rapid progress in generation of iPSCs with clearly defined genetic backgrounds to complement existing patient-derived models. Improved protocols for differentiation of iPSCs, engineered iPSCs and embryonic stem cells (ESCs) now permit the study of disease biology in the majority of somatic cell types. Here, we highlight current efforts to create patient-derived iPSC disease models to study various cancer types. We review the advantages and current challenges of using iPSCs in cancer disease modeling.
Collapse
|
146
|
Rodriguez-Madoz JR, San Jose-Eneriz E, Rabal O, Zapata-Linares N, Miranda E, Rodriguez S, Porciuncula A, Vilas-Zornoza A, Garate L, Segura V, Guruceaga E, Agirre X, Oyarzabal J, Prosper F. Reversible dual inhibitor against G9a and DNMT1 improves human iPSC derivation enhancing MET and facilitating transcription factor engagement to the genome. PLoS One 2017; 12:e0190275. [PMID: 29281720 PMCID: PMC5744984 DOI: 10.1371/journal.pone.0190275] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/11/2017] [Indexed: 12/29/2022] Open
Abstract
The combination of defined factors with small molecules targeting epigenetic factors is a strategy that has been shown to enhance optimal derivation of iPSCs and could be used for disease modelling, high throughput screenings and/or regenerative medicine applications. In this study, we showed that a new first-in-class reversible dual G9a/DNMT1 inhibitor compound (CM272) improves the efficiency of human cell reprogramming and iPSC generation from primary cells of healthy donors and patient samples, using both integrative and non-integrative methods. Moreover, CM272 facilitates the generation of human iPSC with only two factors allowing the removal of the most potent oncogenic factor cMYC. Furthermore, we demonstrated that mechanistically, treatment with CM272 induces heterochromatin relaxation, facilitates the engagement of OCT4 and SOX2 transcription factors to OSKM refractory binding regions that are required for iPSC establishment, and enhances mesenchymal to epithelial transition during the early phase of cell reprogramming. Thus, the use of this new G9a/DNMT reversible dual inhibitor compound may represent an interesting alternative for improving cell reprogramming and human iPSC derivation for many different applications while providing interesting insights into reprogramming mechanisms.
Collapse
Affiliation(s)
- Juan Roberto Rodriguez-Madoz
- Cell Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- * E-mail: (FP); (JRRM)
| | - Edurne San Jose-Eneriz
- Oncohematology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Obdulia Rabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Natalia Zapata-Linares
- Cell Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Estibaliz Miranda
- Oncohematology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Saray Rodriguez
- Cell Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Angelo Porciuncula
- Cell Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Amaia Vilas-Zornoza
- Oncohematology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Leire Garate
- Oncohematology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Victor Segura
- Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Elizabeth Guruceaga
- Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Xabier Agirre
- Oncohematology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Felipe Prosper
- Cell Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Oncohematology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Hematology and Area of Cell Therapy, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
- * E-mail: (FP); (JRRM)
| |
Collapse
|
147
|
TRIM28 and Interacting KRAB-ZNFs Control Self-Renewal of Human Pluripotent Stem Cells through Epigenetic Repression of Pro-differentiation Genes. Stem Cell Reports 2017; 9:2065-2080. [PMID: 29198826 PMCID: PMC5785758 DOI: 10.1016/j.stemcr.2017.10.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 10/31/2017] [Accepted: 10/31/2017] [Indexed: 11/20/2022] Open
Abstract
Reprogramming to induced pluripotent stem cells (iPSCs) and differentiation of pluripotent stem cells (PSCs) are regulated by epigenetic machinery. Tripartite motif protein 28 (TRIM28), a universal mediator of Krüppel-associated box domain zinc fingers (KRAB-ZNFs), is known to regulate both processes; however, the exact mechanism and identity of participating KRAB-ZNF genes remain unknown. Here, using a reporter system, we show that TRIM28/KRAB-ZNFs alter DNA methylation patterns in addition to H3K9me3 to cause stable gene repression during reprogramming. Using several expression datasets, we identified KRAB-ZNFs (ZNF114, ZNF483, ZNF589) in the human genome that maintain pluripotency. Moreover, we identified target genes repressed by these KRAB-ZNFs. Mechanistically, we demonstrated that these KRAB-ZNFs directly alter gene expression of important developmental genes by modulating H3K9me3 and DNA methylation of their promoters. In summary, TRIM28 employs KRAB-ZNFs to evoke epigenetic silencing of its target differentiation genes via H3K9me3 and DNA methylation. Upon reprogramming KRAB-repressor evokes stable silencing of its target genes KRAB-ZNFs repress target genes required for differentiation of pluripotent cells KRAB-ZNFs are crucial for the maintenance of pluripotency of human stem cells TRIM28/KRAB-ZNFs repress developmental genes through H3K9 and de novo DNA methylation
Collapse
|
148
|
Parrotta E, De Angelis MT, Scalise S, Candeloro P, Santamaria G, Paonessa M, Coluccio ML, Perozziello G, De Vitis S, Sgura A, Coluzzi E, Mollace V, Di Fabrizio EM, Cuda G. Two sides of the same coin? Unraveling subtle differences between human embryonic and induced pluripotent stem cells by Raman spectroscopy. Stem Cell Res Ther 2017; 8:271. [PMID: 29183402 PMCID: PMC5706396 DOI: 10.1186/s13287-017-0720-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/24/2017] [Accepted: 11/06/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, hold enormous promise for many biomedical applications, such as regenerative medicine, drug testing, and disease modeling. Although induced pluripotent stem cells resemble embryonic stem cells both morphologically and functionally, the extent to which these cell lines are truly equivalent, from a molecular point of view, remains controversial. METHODS Principal component analysis and K-means cluster analysis of collected Raman spectroscopy data were used for a comparative study of the biochemical fingerprint of human induced pluripotent stem cells and human embryonic stem cells. The Raman spectra analysis results were further validated by conventional biological assays. RESULTS Raman spectra analysis revealed that the major difference between human embryonic stem cells and induced pluripotent stem cells is due to the nucleic acid content, as shown by the strong positive peaks at 785, 1098, 1334, 1371, 1484, and 1575 cm-1, which is enriched in human induced pluripotent stem cells. CONCLUSIONS Here, we report a nonbiological approach to discriminate human induced pluripotent stem cells from their native embryonic stem cell counterparts.
Collapse
Affiliation(s)
- Elvira Parrotta
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Maria Teresa De Angelis
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Stefania Scalise
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Patrizio Candeloro
- BioNEM Lab., Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy.
| | - Gianluca Santamaria
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Mariagrazia Paonessa
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Maria Laura Coluccio
- BioNEM Lab., Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Gerardo Perozziello
- BioNEM Lab., Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Stefania De Vitis
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Antonella Sgura
- Department of Science, University of Rome "Roma tre", viale G. Marconi 446, 00146, Rome, Italy
| | - Elisa Coluzzi
- Department of Science, University of Rome "Roma tre", viale G. Marconi 446, 00146, Rome, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy
| | - Enzo Mario Di Fabrizio
- SMILEs Lab., Physical Science and Engineering Division (PSE), KAUST, 23955-6900, Thuwal, Kingdom of Saudi Arabia
| | - Giovanni Cuda
- Research Center for Advanced Biochemistry and Molecular Biology, Stem Cell Laboratory, Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy.
| |
Collapse
|
149
|
Zhao MT, Shao NY, Hu S, Ma N, Srinivasan R, Jahanbani F, Lee J, Zhang SL, Snyder MP, Wu JC. Cell Type-Specific Chromatin Signatures Underline Regulatory DNA Elements in Human Induced Pluripotent Stem Cells and Somatic Cells. Circ Res 2017; 121:1237-1250. [PMID: 29030344 DOI: 10.1161/circresaha.117.311367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/27/2017] [Accepted: 10/12/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE Regulatory DNA elements in the human genome play important roles in determining the transcriptional abundance and spatiotemporal gene expression during embryonic heart development and somatic cell reprogramming. It is not well known how chromatin marks in regulatory DNA elements are modulated to establish cell type-specific gene expression in the human heart. OBJECTIVE We aimed to decipher the cell type-specific epigenetic signatures in regulatory DNA elements and how they modulate heart-specific gene expression. METHODS AND RESULTS We profiled genome-wide transcriptional activity and a variety of epigenetic marks in the regulatory DNA elements using massive RNA-seq (n=12) and ChIP-seq (chromatin immunoprecipitation combined with high-throughput sequencing; n=84) in human endothelial cells (CD31+CD144+), cardiac progenitor cells (Sca-1+), fibroblasts (DDR2+), and their respective induced pluripotent stem cells. We uncovered 2 classes of regulatory DNA elements: class I was identified with ubiquitous enhancer (H3K4me1) and promoter (H3K4me3) marks in all cell types, whereas class II was enriched with H3K4me1 and H3K4me3 in a cell type-specific manner. Both class I and class II regulatory elements exhibited stimulatory roles in nearby gene expression in a given cell type. However, class I promoters displayed more dominant regulatory effects on transcriptional abundance regardless of distal enhancers. Transcription factor network analysis indicated that human induced pluripotent stem cells and somatic cells from the heart selected their preferential regulatory elements to maintain cell type-specific gene expression. In addition, we validated the function of these enhancer elements in transgenic mouse embryos and human cells and identified a few enhancers that could possibly regulate the cardiac-specific gene expression. CONCLUSIONS Given that a large number of genetic variants associated with human diseases are located in regulatory DNA elements, our study provides valuable resources for deciphering the epigenetic modulation of regulatory DNA elements that fine-tune spatiotemporal gene expression in human cardiac development and diseases.
Collapse
Affiliation(s)
- Ming-Tao Zhao
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Ning-Yi Shao
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Shijun Hu
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.).
| | - Ning Ma
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Rajini Srinivasan
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Fereshteh Jahanbani
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Jaecheol Lee
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Sophia L Zhang
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.)
| | - Michael P Snyder
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.).
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.-T.Z., N.-Y.S., N.M., J.L., S.L.Z., J.C.W.); Department of Cardiovascular Surgery of the Frist Affiliated Hospital, Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China (S.H.); Department of Chemical and Systems Biology, Stanford University School of Medicine, CA (R.S.); and Department of Genetics, Stanford University School of Medicine, CA (F.J., M.P.S.).
| |
Collapse
|
150
|
Chaterji S, Ahn EH, Kim DH. CRISPR Genome Engineering for Human Pluripotent Stem Cell Research. Theranostics 2017; 7:4445-4469. [PMID: 29158838 PMCID: PMC5695142 DOI: 10.7150/thno.18456] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 08/24/2017] [Indexed: 12/13/2022] Open
Abstract
The emergence of targeted and efficient genome editing technologies, such as repurposed bacterial programmable nucleases (e.g., CRISPR-Cas systems), has abetted the development of cell engineering approaches. Lessons learned from the development of RNA-interference (RNA-i) therapies can spur the translation of genome editing, such as those enabling the translation of human pluripotent stem cell engineering. In this review, we discuss the opportunities and the challenges of repurposing bacterial nucleases for genome editing, while appreciating their roles, primarily at the epigenomic granularity. First, we discuss the evolution of high-precision, genome editing technologies, highlighting CRISPR-Cas9. They exist in the form of programmable nucleases, engineered with sequence-specific localizing domains, and with the ability to revolutionize human stem cell technologies through precision targeting with greater on-target activities. Next, we highlight the major challenges that need to be met prior to bench-to-bedside translation, often learning from the path-to-clinic of complementary technologies, such as RNA-i. Finally, we suggest potential bioinformatics developments and CRISPR delivery vehicles that can be deployed to circumvent some of the challenges confronting genome editing technologies en route to the clinic.
Collapse
|