Altered Expression of MLL Methyltransferase Family Genes in Breast Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Altered Expression of MLL Methyltransferase Family Genes in Breast Cancer INTERNATIONAL JOURNAL OF ONCOLOGY 43: 653-660, 2013 Altered expression of MLL methyltransferase family genes in breast cancer DORALINA DO AMARAL RABELLO1, CAROLINA AMARO DE MOURA1, ROSANGELA VIEIRA DE ANDRADE2, ANDREA BARRETTO MOTOYAMA1 and FABIO PITTELLA SILVA1 1Laboratory of Molecular Pathology of Cancer, Faculty of Health Sciences, University of Brasília, Brasília, DF 70.910-900; 2Laboratory of Genomic Sciences and Molecular Biotechnology, Catholic University of Brasília, Brasília, DF 70790-160, Brazil Received March 28, 2013; Accepted May 8, 2013 DOI: 10.3892/ijo.2013.1981 Abstract. The histone lysine methyltransferases contain a Introduction SET domain, which catalyzes the addition of methyl groups to specific lysine residues. The MLL family of genes encodes Breast cancer is the most frequent cancer and the most common histone-modifying enzymes with histone 3-lysine 4 methyl- cause of cancer-related death in women (1). In Brazil alone, transferase activity that can regulate gene transcription. The ~50,000 new cases of breast cancer are expected per year, with MLL family exists in multi-protein complexes and has been an estimated risk of 52 cases for every 100,000 women (2). implicated in a variety of processes including normal devel- Several studies have suggested that different genetic alterations opment and cell growth. Although some of the MLL family are linked to different histological types of breast cancer, such members have already been described to be involved in cancer, as estrogen receptor expression/lobular type, c-ErbB2 expres- a clear relationship of these genes with breast cancer is not sion/invasive type, among others (3,4). Recently, molecular determined to date. In the present study, we used quantitative studies have shown that breast cancer development involves PCR to investigate the expression profile of all fiveMLL genes not only alterations in oncogenes and tumor suppressor genes, [MLL (ALL-1), MLL2, MLL3, MLL4 and MLL5] in 7 breast but also epigenetic alterations in genes related to cellular cancer cell lines, 8 breast tumors and adjacent non-tumor growth, survival, motility and differentiation (5). Disturbance tissues and in 12 normal tissues. We observed a diminished of epigenetic regulation of gene expression through methyla- expression of all five genes in the breast cancer cell lines tion might contribute to tumorigenesis. In this context, altered when compared to normal breast tissue. We found a signifi- histone modifying enzymes such as methyltransferases may cantly decreased expression of MLL2 in the tumor samples play important roles in the process of carcinogenesis. compared to the non-tumor controls. In tumor samples, MLL5 The mixed lineage leukemia (MLL) family is character- also showed a clear suppression tendency. Among the normal ized by a conserved catalytic SET domain, responsible for tissues analyzed, all genes showed a markedly higher expres- the methyltransferase activity and comprises five genes sion in skeletal muscle and brain. Although further studies [MLL (ALL-1), MLL2, MLL3, MLL4 and MLL5]. are required to determine the exact role of these methyltrans- The mixed lineage leukemia gene (MLL or ALL-1, HRX ferases in cancer development, our results indicate that the and Htrx1) (Gene ID: 4297) is located in human chromo- suppression of MLL genes, especially MLL2 and 5, take part some 11q23 and is frequently involved in chromosomal in modulating breast carcinogenesis. Our assessment of the translocations in leukemia (6,7). MLL seems to function as a MLL family gene expression patterns in a diverse set of breast mammalian counterpart of Drosophila trx, which maintains cancer cell lines and in a multitude of tissue types and breast the expression of multiple Hox genes during development. tumors should lead to increasingly detailed information on the Mll-deficiency in mice results in failure to express Hox genes involvement of these genes in cancer progression. and in embryonic lethality. Heterozygosity for disrupted MLL, by homologous recombination in mouse embryonic stem cells, leads to retarded growth, hematopoietic abnormalities and skeleton malformations (8). Correspondence to: Professor Fábio Pittella Silva, Laboratory The MLL2 gene (Gene ID: 8085) is located on human of Molecular Pathology of Cancer, Faculty of Health Sciences, chromosome 12q13.12 and encodes a protein with domains University of Brasília, Campus Universitário Darcy Ribeiro, Asa similar to MLL. MLL2 was shown to be associated with Norte, Brasília, DF 70.910-900, Brazil Pax7 (paired-box transcription factor), forming the Wdr5- E-mail: [email protected] Ash2L-MLL2 histone methyltransferase (HMT) complex that directs tri-methylation of histone H3 lysine 4, when bound Key words: gene expression. MLL (ALL-1), MLL2, MLL3, MLL4 to Myf5. These chromatin modifications stimulate transcrip- and MLL5, breast cancer tional activation of target genes to regulate entry into the myogenic developmental program (9). Another study, based on 654 RABELLO et al: MLL METHYLTRANSFERASE EXPRESSION IN BREAST CANCER muscle-specific myogenin gene, suggested that transcriptional better understanding of the role of the MLL genes in breast activators Mef2d and Six4 mediate recruitment of trithorax carcinogenesis. group proteins Ash2L/MLL2 and UTX to MyoD-bound promoters to overcome the polycomb-mediated repression of Materials and methods muscle genes, allowing transcriptional activation of muscle- specific genes (10).MLL and MLL2 genes show highly similar Cell lines and culture. Seven breast cancer lines were used exon/intron structures. The proteins encoded by these genes on this study: HCC-1954, MCF-7, CAMA-1, SKBR-3, are ubiquitously expressed among adult human tissues and MDA-MB-231, MDA-MB-436 and MDA-MB-468. The cell although they share similar domains, they seem to act in a lines HCC-1954, MCF-7 and MDA-MB-231, obtained from different manner. MLL2 could not compensate for the hetero- the ATCC, were cultured in our laboratory in recommended zygosity of the disrupted MLL, by homologous recombination media and checked periodically for mycoplasma contamina- in mice (8), which indicates that their functions seem to be at tion. The main characteristics of the cell lines are summarized least partially non-overlapping. in Table I (20). Cells were grown in Dulbecco's modified MLL3 (Gene ID: 58508) maps to chromosome 7q36.1, a Eagle's medium (DMEM, Sigma-Aldrich, St. Louis, MO, USA) region usually deleted in hematological malignancies (11). supplemented with 10% fetal bovine serum (Sigma-Aldrich) MLL4 (Gene ID: 9757) is located on chromosome 19q13.1. and penicillin/streptomycin (100 U/ml; Life Technologies, MLL3 and MLL4 are found as part of a complex named Carlsbad, CA, USA) at 37˚C and 5% CO2. Total RNA from ASCOM (for ASC-2 COMplex), that acts as a p53 co-activator. these cells was extracted and used for expression analysis as This complex is required for the expression of endogenous described below. Additionally, total RNA from the remainder p53-target genes in response to the DNA damage (12). cells were kindly provided by the Ludwig Institute for Cancer Deletions or truncations in Mll, Mll2 and Mll3 in mice show Research, São Paulo, and were included in the gene expression different phenotypes, suggesting that MLL protein functions analysis. are not redundant (8,13). MLL5 (Gene ID: 55904) is located in human chromosome Normal tissues. A commercial panel (OriGene Technologies, 7q22.1. The nuclear protein encoded shows single PHD and Rockville, MD, USA) of total RNA from twelve human normal SET domains and lacks other conserved sequence elements, tissues from different organs (lung, small intestine (SI), brain, like its Drosophila homolog CG9007. MLL5 shows ubiqui- colon, kidney, muscle, liver, spleen, heart, testis, stomach and tous tissue expression and is highly conserved. It is the latest placenta) was used in this study. Total RNA of each normal addition to the mammalian Trithorax/MLL gene family and tissue was used to verify the expression levels of the genes recent studies show that the MLL5 may function in a different studied. manner, without methyltranferase activity, which is consistent with its different sequence of the SET domain compared to Tumor specimens. The sample collection protocol was the rest of the MLL family of H3K4 methyltransferases (14). It approved by the Research Ethics Committee of the Faculty was suggested that MLL5 would have an indirect mechanism of Health Sciences, University of Brasília, Brazil, based on regulating expression of histone-modifying enzymes, such as resolution 196/96 of the National Health Council/Brazilian LSD1 and SET7/9 (15). MLL5 was shown to promote myogenic Ministry of Health, project no. 025/09. Written informed differentiation through positive regulation of the pro-myogenic consent was obtained from all participants. The samples were genes Pax7, Myf5 and myogenin, which are impaired in MLL5 collected from surgically removed breast tissue from female knockdown, leading to serious differentiation defects (15). patients diagnosed with breast cancer at the Hospital of the Mll5 deficiency in mice leads to growth retardation, male University of Brasilia. A total of 14 samples (6 normal and infertility and defects in multiple hematopoietic lineages (16). 8 tumor samples) was used. The identification of tumor tissue Therefore, the defects found could be associated with deregu- from the removed samples was based on the histopathological lation of cell cycle control. examination and all types of breast cancer were included in our Some studies have shown that MLLs are critical analyses. Samples were selected based on tumor content (mini- co-activators in estrogen (E2)-dependent regulation of mally 80% tumor) as determined by microscopic pathological E2-responsive genes, through LXXLL motifs (NR boxes) analysis. A sample of normal tissue was collected from each (17,18). The suppression of MLL2 decreased expression of patient whenever possible. The clinical and histopathological estrogen receptor (ER) target genes (18). MLL3 and MLL4 characteristics of the patients are summarized in Table IV. coordinate with ERs and act in transcriptional regulation of HOXC10 in the presence of estrogen (19).
Recommended publications
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • Utx Is Required for Proper Induction of Ectoderm and Mesoderm During Differentiation of Embryonic Stem Cells
    Utx Is Required for Proper Induction of Ectoderm and Mesoderm during Differentiation of Embryonic Stem Cells Cristina Morales Torres1,2, Anne Laugesen1,2, Kristian Helin1,2,3* 1 Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark, 2 Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark, 3 The Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark Abstract Embryonic development requires chromatin remodeling for dynamic regulation of gene expression patterns to ensure silencing of pluripotent transcription factors and activation of developmental regulators. Demethylation of H3K27me3 by the histone demethylases Utx and Jmjd3 is important for the activation of lineage choice genes in response to developmental signals. To further understand the function of Utx in pluripotency and differentiation we generated Utx knockout embryonic stem cells (ESCs). Here we show that Utx is not required for the proliferation of ESCs, however, Utx contributes to the establishment of ectoderm and mesoderm in vitro. Interestingly, this contribution is independent of the catalytic activity of Utx. Furthermore, we provide data showing that the Utx homologue, Uty, which is devoid of detectable demethylase activity, and Jmjd3 partly compensate for the loss of Utx. Taken together our results show that Utx is required for proper formation of ectoderm and mesoderm in vitro, and that Utx, similar to its C.elegans homologue, has demethylase dependent and independent functions. Citation: Morales Torres C, Laugesen A, Helin K (2013) Utx Is Required for Proper Induction of Ectoderm and Mesoderm during Differentiation of Embryonic Stem Cells. PLoS ONE 8(4): e60020. doi:10.1371/journal.pone.0060020 Editor: Qiang Wu, National University of Singapore, Singapore Received December 13, 2012; Accepted February 21, 2013; Published April 3, 2013 Copyright: ß 2013 Morales Torres et al.
    [Show full text]
  • Potential Genotoxicity from Integration Sites in CLAD Dogs Treated Successfully with Gammaretroviral Vector-Mediated Gene Therapy
    Gene Therapy (2008) 15, 1067–1071 & 2008 Nature Publishing Group All rights reserved 0969-7128/08 $30.00 www.nature.com/gt SHORT COMMUNICATION Potential genotoxicity from integration sites in CLAD dogs treated successfully with gammaretroviral vector-mediated gene therapy M Hai1,3, RL Adler1,3, TR Bauer Jr1,3, LM Tuschong1, Y-C Gu1,XWu2 and DD Hickstein1 1Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA and 2Laboratory of Molecular Technology, Scientific Applications International Corporation-Frederick, National Cancer Institute-Frederick, Frederick, Maryland, USA Integration site analysis was performed on six dogs with in hematopoietic stem cells. Integrations clustered around canine leukocyte adhesion deficiency (CLAD) that survived common insertion sites more frequently than random. greater than 1 year after infusion of autologous CD34+ bone Despite potential genotoxicity from RIS, to date there has marrow cells transduced with a gammaretroviral vector been no progression to oligoclonal hematopoiesis and no expressing canine CD18. A total of 387 retroviral insertion evidence that vector integration sites influenced cell survival sites (RIS) were identified in the peripheral blood leukocytes or proliferation. Continued follow-up in disease-specific from the six dogs at 1 year postinfusion. A total of 129 RIS animal models such as CLAD will be required to provide an were identified in CD3+ T-lymphocytes and 102 RIS in accurate estimate
    [Show full text]
  • MLL3/MLL4 Methyltransferase Activities Regulate Embryonic Stem Cell
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.14.296558; this version posted September 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 MLL3/MLL4 methyltransferase activities regulate embryonic stem cell 2 differentiation independent of enhancer H3K4me1 3 4 Guojia Xie1, Ji-Eun Lee1, Kaitlin McKernan1, Young-Kwon Park1, Younghoon Jang1, Chengyu Liu2, Weiqun 5 Peng3 and Kai Ge1* 6 7 1Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney 8 Diseases, National Institutes of Health, Bethesda, MD 20892, USA 9 2Transgenic Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 10 20892, USA 11 3Departments of Physics and Anatomy and Cell Biology, The George Washington University, Washington, 12 DC 20052, USA 13 14 *To whom correspondence should be addressed. (Email: [email protected]) 15 16 17 18 19 Highlights 20 ● Simultaneous elimination of MLL3 and MLL4 enzymatic activities leads to early embryonic lethality in 21 mice 22 ● MLL3/4 enzymatic activities are dispensable for ESC differentiation towards the three germ layers 23 ● ESCs lacking MLL3/4 enzymatic activities show cavitation defects during EB differentiation, likely due 24 to impaired VE induction 25 ● MLL3/4-catalyzed H3K4me1 is dispensable for enhancer activation in ESC differentiation 26 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.09.14.296558; this version posted September 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.
    [Show full text]
  • Functional Annotation of Human Long Noncoding Rnas Using Chromatin
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.13.426305; this version posted January 14, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Funconal annotaon of human long noncoding RNAs using chroman conformaon data Saumya Agrawal1, Tanvir Alam2, Masaru Koido1,3, Ivan V. Kulakovskiy4,5, Jessica Severin1, Imad ABugessaisa1, Andrey Buyan5,6, Josee Dos&e7, Masayoshi Itoh1,8, Naoto Kondo9, Yunjing Li10, Mickaël Mendez11, Jordan A. Ramilowski1,12, Ken Yagi1, Kayoko Yasuzawa1, CHi Wai Yip1, Yasushi Okazaki1, MicHael M. Ho9man11,13,14,15, Lisa Strug10, CHung CHau Hon1, CHikashi Terao1, Takeya Kasukawa1, Vsevolod J. Makeev4,16, Jay W. SHin1, Piero Carninci1, MicHiel JL de Hoon1 1RIKEN Center for Integra&ve Medical Sciences, YokoHama, Japan. 2College of Science and Engineering, Hamad Bin KHalifa University, DoHa, Qatar. 3Ins&tute of Medical Science, THe University of Tokyo, Tokyo, Japan. 4Vavilov Ins&tute of General Gene&cs, Russian Academy of Sciences, Moscow, Russia. 5Ins&tute of Protein ResearcH, Russian Academy of Sciences, PusHcHino, Russia. 6Faculty of Bioengineering and Bioinforma&cs, Lomonosov Moscow State University, Moscow, Russia. 7Department of BiocHemistry, Rosalind and Morris Goodman Cancer ResearcH Center, McGill University, Montréal, QuéBec, Canada. 8RIKEN Preven&ve Medicine and Diagnosis Innova&on Program, Wako, Japan. 9RIKEN Center for Life Science TecHnologies, YokoHama, Japan. 10Division of Biosta&s&cs, Dalla Lana ScHool of PuBlic HealtH, University of Toronto, Toronto, Ontario, Canada. 11Department of Computer Science, University of Toronto, Toronto, Ontario, Canada. 12Advanced Medical ResearcH Center, YokoHama City University, YokoHama, Japan.
    [Show full text]
  • A Small UTX Stabilization Domain of Trr Is Conserved Within Mammalian MLL3-4/COMPASS and Is Sufficient to Rescue Loss of Viability in Null Animals
    Downloaded from genesdev.cshlp.org on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press A small UTX stabilization domain of Trr is conserved within mammalian MLL3-4/ COMPASS and is sufficient to rescue loss of viability in null animals Ryan Rickels,1 Lu Wang,1 Marta Iwanaszko,1 Patrick A. Ozark,1 Marc A. Morgan,1 Andrea Piunti,1 Natalia Khalatyan,2 Shimaa H.A. Soliman,1 Emily J. Rendleman,1 Jeffrey N. Savas,2 Edwin R. Smith,1 and Ali Shilatifard1 1Simpson Querrey Institute for Epigenetics, Department of Biochemistry and Molecular Genetics, 2Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA Catalytic-inactivating mutations within the Drosophila enhancer H3K4 mono-methyltransferase Trr and its mammalian homologs, MLL3/4, cause only minor changes in gene expression compared with whole-gene deletions for these COMPASS members. To identify essential histone methyltransferase-independent functions of Trr, we screened to identify a minimal Trr domain sufficient to rescue Trr-null lethality and demonstrate that this domain binds and stabilizes Utx in vivo. Using the homologous MLL3/MLL4 human sequences, we mapped a short ∼80- amino-acid UTX stabilization domain (USD) that promotes UTX stability in the absence of the rest of MLL3/4. Nuclear UTX stability is enhanced when the USD is fused with the MLL4 HMG-box. Thus, COMPASS-dependent UTX stabilization is an essential noncatalytic function of Trr/MLL3/MLL4, suggesting that stabilizing UTX could be a therapeutic strategy for cancers with MLL3/4 loss-of-function mutations. [Keywords: COMPASS; chromatin; epigenetics; transcription] Supplemental material is available for this article.
    [Show full text]
  • Enhancer Priming by H3K4 Methyltransferase MLL4 Controls Cell Fate Transition
    Enhancer priming by H3K4 methyltransferase MLL4 controls cell fate transition Chaochen Wanga, Ji-Eun Leea, Binbin Laia, Todd S. Macfarlanb, Shiliyang Xua, Lenan Zhuanga, Chengyu Liuc, Weiqun Pengd,e, and Kai Gea,1 aLaboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892; bDivision of Developmental Biology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892; cTransgenic Core, Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; dDepartment of Physics, The George Washington University, Washington, DC 20052; and eDepartment of Anatomy and Regenerative Biology, The George Washington University, Washington, DC 20052 Edited by Eric N. Olson, University of Texas Southwestern Medical Center, Dallas, TX, and approved August 26, 2016 (received for review April 29, 2016) Transcriptional enhancers control cell-type–specific gene expres- excellent models for studying cell fate transition and the underlying sion. Primed enhancers are marked by histone H3 lysine 4 (H3K4) molecular mechanism. mono/di-methylation (H3K4me1/2). Active enhancers are further MLL4 is a major enhancer H3K4 mono- and di-methyltransfer- marked by H3K27 acetylation (H3K27ac). Mixed-lineage leukemia ase with partial functional redundancy with MLL3 in mammalian 4 (MLL4/KMT2D) is a major enhancer H3K4me1/2 methyltransferase cells (7, 8). MLL4 colocalizes with lineage-determining TFs on AEs with functional redundancy with MLL3 (KMT2C). However, its role in during adipogenesis and myogenesis. Furthermore, MLL4 is re- cell fate maintenance and transition is poorly understood. Here, we quired for enhancer activation, cell-type–specific gene expression, show in mouse embryonic stem cells (ESCs) that MLL4 associates and cell differentiation during adipogenesis and myogenesis (8).
    [Show full text]
  • Enhancer Priming by H3K4 Methyltransferase MLL4 Controls Cell Fate Transition
    Enhancer priming by H3K4 methyltransferase MLL4 controls cell fate transition Chaochen Wanga, Ji-Eun Leea, Binbin Laia, Todd S. Macfarlanb, Shiliyang Xua, Lenan Zhuanga, Chengyu Liuc, Weiqun Pengd,e, and Kai Gea,1 aLaboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892; bDivision of Developmental Biology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892; cTransgenic Core, Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; dDepartment of Physics, The George Washington University, Washington, DC 20052; and eDepartment of Anatomy and Regenerative Biology, The George Washington University, Washington, DC 20052 Edited by Eric N. Olson, University of Texas Southwestern Medical Center, Dallas, TX, and approved August 26, 2016 (received for review April 29, 2016) Transcriptional enhancers control cell-type–specific gene expres- excellent models for studying cell fate transition and the underlying sion. Primed enhancers are marked by histone H3 lysine 4 (H3K4) molecular mechanism. mono/di-methylation (H3K4me1/2). Active enhancers are further MLL4 is a major enhancer H3K4 mono- and di-methyltransfer- marked by H3K27 acetylation (H3K27ac). Mixed-lineage leukemia ase with partial functional redundancy with MLL3 in mammalian 4 (MLL4/KMT2D) is a major enhancer H3K4me1/2 methyltransferase cells (7, 8). MLL4 colocalizes with lineage-determining TFs on AEs with functional redundancy with MLL3 (KMT2C). However, its role in during adipogenesis and myogenesis. Furthermore, MLL4 is re- cell fate maintenance and transition is poorly understood. Here, we quired for enhancer activation, cell-type–specific gene expression, show in mouse embryonic stem cells (ESCs) that MLL4 associates and cell differentiation during adipogenesis and myogenesis (8).
    [Show full text]
  • Transcriptional and Epigenetic Control of Brown and Beige Adipose Cell Fate and Function
    REVIEWS Transcriptional and epigenetic control of brown and beige adipose cell fate and function Takeshi Inagaki1,2, Juro Sakai1,2 and Shingo Kajimura3 Abstract | White adipocytes store excess energy in the form of triglycerides, whereas brown and beige adipocytes dissipate energy in the form of heat. This thermogenic function relies on the activation of brown and beige adipocyte-specific gene programmes that are coordinately regulated by adipose-selective chromatin architectures and by a set of unique transcriptional and epigenetic regulators. A number of transcriptional and epigenetic regulators are also required for promoting beige adipocyte biogenesis in response to various environmental stimuli. A better understanding of the molecular mechanisms governing the generation and function of brown and beige adipocytes is necessary to allow us to control adipose cell fate and stimulate thermogenesis. This may provide a therapeutic approach for the treatment of obesity and obesity-associated diseases, such as type 2 diabetes. Interscapular BAT Adipose tissue has a central role in whole-body energy subjects who had previously lacked detectable BAT Brown adipose tissue (BAT) is a homeostasis. White adipose tissue (WAT) is the major depots before cold exposure, presumably owing to the specialized organ that adipose organ in mammals. It represents 10% or more emergence of new thermogenic adipocytes. This, then, produces heat. BAT is localized of the body weight of healthy adult humans and is leads to an increase in non-shivering thermogenesis in the interscapular and 6–9 perirenal regions of rodents specialized for the storage of excess energy. Humans and/or an improvement in insulin sensitivity . These and infants.
    [Show full text]
  • Histone Methylation Regulation in Neurodegenerative Disorders
    International Journal of Molecular Sciences Review Histone Methylation Regulation in Neurodegenerative Disorders Balapal S. Basavarajappa 1,2,3,4,* and Shivakumar Subbanna 1 1 Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; [email protected] 2 New York State Psychiatric Institute, New York, NY 10032, USA 3 Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA 4 New York University Langone Medical Center, Department of Psychiatry, New York, NY 10016, USA * Correspondence: [email protected]; Tel.: +1-845-398-3234; Fax: +1-845-398-5451 Abstract: Advances achieved with molecular biology and genomics technologies have permitted investigators to discover epigenetic mechanisms, such as DNA methylation and histone posttransla- tional modifications, which are critical for gene expression in almost all tissues and in brain health and disease. These advances have influenced much interest in understanding the dysregulation of epigenetic mechanisms in neurodegenerative disorders. Although these disorders diverge in their fundamental causes and pathophysiology, several involve the dysregulation of histone methylation- mediated gene expression. Interestingly, epigenetic remodeling via histone methylation in specific brain regions has been suggested to play a critical function in the neurobiology of psychiatric disor- ders, including that related to neurodegenerative diseases. Prominently, epigenetic dysregulation currently brings considerable interest as an essential player in neurodegenerative disorders, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), Amyotrophic lateral sclerosis (ALS) and drugs of abuse, including alcohol abuse disorder, where it may facilitate connections between genetic and environmental risk factors or directly influence disease-specific Citation: Basavarajappa, B.S.; Subbanna, S.
    [Show full text]
  • Loss of the Transcription Factor MAFB Limits β-Cell
    UCSF UC San Francisco Previously Published Works Title Loss of the transcription factor MAFB limits β-cell derivation from human PSCs. Permalink https://escholarship.org/uc/item/0zb338n9 Journal Nature communications, 11(1) ISSN 2041-1723 Authors Russell, Ronan Carnese, Phichitpol P Hennings, Thomas G et al. Publication Date 2020-06-02 DOI 10.1038/s41467-020-16550-9 Peer reviewed eScholarship.org Powered by the California Digital Library University of California ARTICLE https://doi.org/10.1038/s41467-020-16550-9 OPEN Loss of the transcription factor MAFB limits β-cell derivation from human PSCs Ronan Russell1, Phichitpol P. Carnese1, Thomas G. Hennings1, Emily M. Walker 2, Holger A. Russ 1,3, ✉ Jennifer S. Liu1, Simone Giacometti1, Roland Stein2 & Matthias Hebrok 1 Next generation sequencing studies have highlighted discrepancies in β-cells which exist between mice and men. Numerous reports have identified MAF BZIP Transcription Factor B 1234567890():,; (MAFB) to be present in human β-cells postnatally, while its expression is restricted to embryonic and neo-natal β-cells in mice. Using CRISPR/Cas9-mediated gene editing, coupled with endocrine cell differentiation strategies, we dissect the contribution of MAFB to β-cell development and function specifically in humans. Here we report that MAFB knockout hPSCs have normal pancreatic differentiation capacity up to the progenitor stage, but favor soma- tostatin- and pancreatic polypeptide–positive cells at the expense of insulin- and glucagon- producing cells during endocrine cell development. Our results describe a requirement for MAFB late in the human pancreatic developmental program and identify it as a distinguishing transcription factor within islet cell subtype specification.
    [Show full text]
  • The Role of the MLL-HOXA9 Axis in Normal and Malignant
    The Role of the MLL-HOXA9 Axis in Normal and Malignant Hematopoiesis by Jingya Wang A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Molecular and Cellular Pathology) in the University of Michigan 2013 Doctoral Committee: Professor Jay L. Hess, Chair Assistant Professor Tomasz Cierpicki Professor Eric R. Fearon Assistant Professor Maria E. Figueroa Professor Tom K. Kerppola Associate Professor Xiaochun Yu © Jingya Wang 2013 To My parents and My husband ii Acknowledgement This dissertation would not have been possible without the tremendous support and guidance from my advisor, Dr. Jay L. Hess. Jay’s enthusiasm and dedication towards scientific research have always been a great source of inspiration. His scientific vision and openness to novel ideas greatly influenced my scientific career. I would also like to thank Jay for encouraging me to pursue my interest in statistics and the opportunity to apply my knowledge to biomedical research. I deeply appreciate Jay’s invaluable advice and generous support for the next stage of my career. My sincere appreciations also go to Drs. Andrew Muntean and Maria (“Ken”) Figueroa. Throughout my graduate study, Andy was actively involved in many aspects of my projects and my scientific career, including providing technical assistance and scientific thinking, and most importantly, sharing his insights of science and being a better scientist. I could not have been where I am without his generous help. Ken led me into the field of high throughput sequencing data analysis, and patiently discussed experimental design to algorithm in details. Her presence also contributes greatly to the completion of this thesis.
    [Show full text]