1
|
Fleishman JS, Kumar S. Bile acid metabolism and signaling in health and disease: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:97. [PMID: 38664391 PMCID: PMC11045871 DOI: 10.1038/s41392-024-01811-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.
Collapse
Affiliation(s)
- Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
2
|
Tate T, Plumber SA, Al-Ahmadie H, Chen X, Choi W, Lu C, Viny A, Batourina E, Gartensson K, Alija B, Molotkov A, Wiessner G, McKiernan J, McConkey D, Dinney C, Czerniak B, Mendelsohn CL. Combined Mek inhibition and Pparg activation Eradicates Muscle Invasive Bladder cancer in a Mouse Model of BBN-induced Carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.19.553961. [PMID: 37662238 PMCID: PMC10473651 DOI: 10.1101/2023.08.19.553961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Bladder cancers (BCs) can be divided into 2 major subgroups displaying distinct clinical behaviors and mutational profiles: basal/squamous (BASQ) tumors that tend to be muscle invasive, and luminal/papillary (LP) tumors that are exophytic and tend to be non-invasive. Pparg is a likely driver of LP BC and has been suggested to act as a tumor suppressor in BASQ tumors, where it is likely suppressed by MEK-dependent phosphorylation. Here we tested the effects of rosiglitazone, a Pparg agonist, in a mouse model of BBN-induced muscle invasive BC. Rosiglitazone activated Pparg signaling in suprabasal epithelial layers of tumors but not in basal-most layers containing highly proliferative invasive cells, reducing proliferation but not affecting tumor survival. Addition of trametinib, a MEK inhibitor, induced Pparg signaling throughout all tumor layers, and eradicated 91% of tumors within 7-days of treatment. The 2-drug combination also activated a luminal differentiation program, reversing squamous metaplasia in the urothelium of tumor-bearing mice. Paired ATAC-RNA-seq analysis revealed that tumor apoptosis was most likely linked to down-regulation of Bcl-2 and other pro-survival genes, while the shift from BASQ to luminal differentiation was associated with activation of the retinoic acid pathway and upregulation of Kdm6a, a lysine demethylase that facilitates retinoid-signaling. Our data suggest that rosiglitazone, trametinib, and retinoids, which are all FDA approved, may be clinically active in BASQ tumors in patients. That muscle invasive tumors are populated by basal and suprabasal cell types with different responsiveness to PPARG agonists will be an important consideration when designing new treatments.
Collapse
|
3
|
Shah SS, Fulton A, Jabroun M, Brightman D, Simpson BN, Bodamer OA. Insights into the genotype-phenotype relationship of ocular manifestations in Kabuki syndrome. Am J Med Genet A 2023; 191:1325-1338. [PMID: 36891680 DOI: 10.1002/ajmg.a.63155] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 03/10/2023]
Abstract
We aim to assess if genotype-phenotype correlations are present within ocular manifestations of Kabuki syndrome (KS) among a large multicenter cohort. We conducted a retrospective, medical record review including clinical history and comprehensive ophthalmological examinations of a total of 47 individuals with molecularly confirmed KS and ocular manifestations at Boston Children's Hospital and Cincinnati Children's Hospital Medical Center. We assessed information regarding ocular structural, functional, and adnexal elements as well as pertinent associated phenotypic features associated with KS. For both type 1 KS (KS1) and type 2 KS (KS2), we observed more severe eye pathology in nonsense variants towards the C-terminus of each gene, KMT2D and KDM6A, respectively. Furthermore, frameshift variants appeared to be not associated with structural ocular elements. Between both types of KS, ocular structural elements were more frequently identified in KS1 compared with KS2, which only involved the optic disc in our cohort. These results reinforce the need for a comprehensive ophthalmologic exam upon diagnosis of KS and regular follow-up exams. The specific genotype may allow risk stratification of the severity of the ophthalmologic manifestation. However, additional studies involving larger cohorts are needed to replicate our observations and conduct powered analyses to more formally risk-stratify based on genotype, highlighting the importance of multicenter collaborations in rare disease research.
Collapse
Affiliation(s)
- Suraj S Shah
- Tufts University School of Medicine, Boston, Massachusetts, USA.,Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Anne Fulton
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Mireille Jabroun
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona, USA
| | - Diana Brightman
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio, USA
| | - Brittany N Simpson
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio, USA
| | - Olaf A Bodamer
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA.,The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
Baxter M, Poolman T, Cunningham P, Hunter L, Voronkov M, Kitchen GB, Goosey L, Begley N, Kay D, Hespe A, Maidstone R, Loudon ASI, Ray DW. Circadian clock function does not require the histone methyltransferase MLL3. FASEB J 2022; 36:e22356. [PMID: 35704036 PMCID: PMC9328146 DOI: 10.1096/fj.202200368r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/26/2022] [Accepted: 05/06/2022] [Indexed: 11/11/2022]
Abstract
The circadian clock controls the physiological function of tissues through the regulation of thousands of genes in a cell-type-specific manner. The core cellular circadian clock is a transcription-translation negative feedback loop, which can recruit epigenetic regulators to facilitate temporal control of gene expression. Histone methyltransferase, mixed lineage leukemia gene 3 (MLL3) was reported to be required for the maintenance of circadian oscillations in cultured cells. Here, we test the role of MLL3 in circadian organization in whole animals. Using mice expressing catalytically inactive MLL3, we show that MLL3 methyltransferase activity is in fact not required for circadian oscillations in vitro in a range of tissues, nor for the maintenance of circadian behavioral rhythms in vivo. In contrast to a previous report, loss of MLL3-dependent methylation did not affect the global levels of H3K4 methylation in liver, indicating substantial compensation from other methyltransferases. Furthermore, we found little evidence of genomic repositioning of H3K4me3 marks. We did, however, observe repositioning of H3K4me1 from intronic regions to intergenic regions and gene promoters; however, there were no changes in H3K4me1 mark abundance around core circadian clock genes. Output functions of the circadian clock, such as control of inflammation, were largely intact in MLL3-methyltransferase-deficient mice, although some gene-specific changes were observed, with sexually dimorphic loss of circadian regulation of specific cytokines. Taken together, these observations indicate that MLL3-directed histone methylation is not essential for core circadian clock function; however, it may influence the inflammatory response.
Collapse
Affiliation(s)
- Matthew Baxter
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Toryn Poolman
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Peter Cunningham
- Centre for Biological TimingFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Louise Hunter
- Centre for Biological TimingFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Maria Voronkov
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Gareth B. Kitchen
- Centre for Biological TimingFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Laurence Goosey
- Centre for Biological TimingFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Nicola Begley
- Centre for Biological TimingFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Danielle Kay
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Abby Hespe
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Robert Maidstone
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Andrew S. I. Loudon
- Centre for Biological TimingFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - David W. Ray
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| |
Collapse
|
5
|
CaSWC4 regulates the immunity-thermotolerance tradeoff by recruiting CabZIP63/CaWRKY40 to target genes and activating chromatin in pepper. PLoS Genet 2022; 18:e1010023. [PMID: 35226664 PMCID: PMC8884482 DOI: 10.1371/journal.pgen.1010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/10/2022] [Indexed: 11/19/2022] Open
Abstract
Pepper (Capsicum annuum) responds differently to high temperature stress (HTS) and Ralstonia solanacearum infection (RSI) but employs some shared transcription factors (TFs), such as CabZIP63 and CaWRKY40, in both cases. How the plant activates and balances these distinct responses, however, was unclear. Here, we show that the protein CaSWC4 interacts with CaRUVBL2 and CaTAF14b and they all act positively in pepper response to RSI and thermotolerance. CaSWC4 activates chromatin of immunity or thermotolerance related target genes of CaWRKY40 or CabZIP63 by promoting deposition of H2A.Z, H3K9ac and H4K5ac, simultaneously recruits CabZIP63 and CaWRKY40 through physical interaction and brings them to their targets (immunity- or thermotolerance-related genes) via binding AT-rich DNA element. The above process relies on the recruitment of CaRUVBL2 and TAF14 by CaSWC4 via physical interaction, which occurs at loci of immunity related target genes only when the plants are challenged with RSI, and at loci of thermotolerance related target genes only upon HTS. Collectively, our data suggest that CaSWC4 regulates rapid, accurate responses to both RSI and HTS by modulating chromatin of specific target genes opening and recruiting the TFs, CaRUVBL2 and CaTAF14b to the specific target genes, thereby helping achieve the balance between immunity and thermotolerance.
Collapse
|
6
|
Yang Y, Luan Y, Feng Q, Chen X, Qin B, Ren KD, Luan Y. Epigenetics and Beyond: Targeting Histone Methylation to Treat Type 2 Diabetes Mellitus. Front Pharmacol 2022; 12:807413. [PMID: 35087408 PMCID: PMC8788853 DOI: 10.3389/fphar.2021.807413] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022] Open
Abstract
Diabetes mellitus is a global public health challenge with high morbidity. Type 2 diabetes mellitus (T2DM) accounts for 90% of the global prevalence of diabetes. T2DM is featured by a combination of defective insulin secretion by pancreatic β-cells and the inability of insulin-sensitive tissues to respond appropriately to insulin. However, the pathogenesis of this disease is complicated by genetic and environmental factors, which needs further study. Numerous studies have demonstrated an epigenetic influence on the course of this disease via altering the expression of downstream diabetes-related proteins. Further studies in the field of epigenetics can help to elucidate the mechanisms and identify appropriate treatments. Histone methylation is defined as a common histone mark by adding a methyl group (-CH3) onto a lysine or arginine residue, which can alter the expression of downstream proteins and affect cellular processes. Thus, in tthis study will discuss types and functions of histone methylation and its role in T2DM wilsed. We will review the involvement of histone methyltransferases and histone demethylases in the progression of T2DM and analyze epigenetic-based therapies. We will also discuss the potential application of histone methylation modification as targets for the treatment of T2DM.
Collapse
Affiliation(s)
- Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Luan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qi Feng
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
| | - Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Qin
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai-Di Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Yi Luan
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Kaur G, Iyer LM, Burroughs AM, Aravind L. Bacterial death and TRADD-N domains help define novel apoptosis and immunity mechanisms shared by prokaryotes and metazoans. eLife 2021; 10:70394. [PMID: 34061031 PMCID: PMC8195603 DOI: 10.7554/elife.70394] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022] Open
Abstract
Several homologous domains are shared by eukaryotic immunity and programmed cell-death systems and poorly understood bacterial proteins. Recent studies show these to be components of a network of highly regulated systems connecting apoptotic processes to counter-invader immunity, in prokaryotes with a multicellular habit. However, the provenance of key adaptor domains, namely those of the Death-like and TRADD-N superfamilies, a quintessential feature of metazoan apoptotic systems, remained murky. Here, we use sensitive sequence analysis and comparative genomics methods to identify unambiguous bacterial homologs of the Death-like and TRADD-N superfamilies. We show the former to have arisen as part of a radiation of effector-associated α-helical adaptor domains that likely mediate homotypic interactions bringing together diverse effector and signaling domains in predicted bacterial apoptosis- and counter-invader systems. Similarly, we show that the TRADD-N domain defines a key, widespread signaling bridge that links effector deployment to invader-sensing in multicellular bacterial and metazoan counter-invader systems. TRADD-N domains are expanded in aggregating marine invertebrates and point to distinctive diversifying immune strategies probably directed both at RNA and retroviruses and cellular pathogens that might infect such communities. These TRADD-N and Death-like domains helped identify several new bacterial and metazoan counter-invader systems featuring underappreciated, common functional principles: the use of intracellular invader-sensing lectin-like (NPCBM and FGS), transcription elongation GreA/B-C, glycosyltransferase-4 family, inactive NTPase (serving as nucleic acid receptors), and invader-sensing GTPase switch domains. Finally, these findings point to the possibility of multicellular bacteria-stem metazoan symbiosis in the emergence of the immune/apoptotic systems of the latter.
Collapse
Affiliation(s)
- Gurmeet Kaur
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Lakshminarayan M Iyer
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - A Maxwell Burroughs
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - L Aravind
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| |
Collapse
|
8
|
Huisman C, Kim YA, Jeon S, Shin B, Choi J, Lim SJ, Youn SM, Park Y, K C M, Kim S, Lee SK, Lee S, Lee JW. The histone H3-lysine 4-methyltransferase Mll4 regulates the development of growth hormone-releasing hormone-producing neurons in the mouse hypothalamus. Nat Commun 2021; 12:256. [PMID: 33431871 PMCID: PMC7801453 DOI: 10.1038/s41467-020-20511-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023] Open
Abstract
In humans, inactivating mutations in MLL4, which encodes a histone H3-lysine 4-methyltransferase, lead to Kabuki syndrome (KS). While dwarfism is a cardinal feature of KS, the underlying etiology remains unclear. Here we report that Mll4 regulates the development of growth hormone-releasing hormone (GHRH)-producing neurons in the mouse hypothalamus. Our two Mll4 mutant mouse models exhibit dwarfism phenotype and impairment of the developmental programs for GHRH-neurons. Our ChIP-seq analysis reveals that, in the developing mouse hypothalamus, Mll4 interacts with the transcription factor Nrf1 to trigger the expression of GHRH-neuronal genes. Interestingly, the deficiency of Mll4 results in a marked reduction of histone marks of active transcription, while treatment with the histone deacetylase inhibitor AR-42 rescues the histone mark signature and restores GHRH-neuronal production in Mll4 mutant mice. Our results suggest that the developmental dysregulation of Mll4-directed epigenetic control of transcription plays a role in the development of GHRH-neurons and dwarfism phenotype in mice.
Collapse
Affiliation(s)
- Christian Huisman
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Young A Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Shin Jeon
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Bongjin Shin
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Jeonghoon Choi
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Su Jeong Lim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Sung Min Youn
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Younjung Park
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Medha K C
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Sangsoo Kim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Soo-Kyung Lee
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea.
| | - Jae W Lee
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA.
| |
Collapse
|
9
|
Lavery WJ, Barski A, Wiley S, Schorry EK, Lindsley AW. KMT2C/D COMPASS complex-associated diseases [K CDCOM-ADs]: an emerging class of congenital regulopathies. Clin Epigenetics 2020; 12:10. [PMID: 31924266 PMCID: PMC6954584 DOI: 10.1186/s13148-019-0802-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022] Open
Abstract
The type 2 lysine methyltransferases KMT2C and KMT2D are large, enzymatically active scaffold proteins that form the core of nuclear regulatory structures known as KMT2C/D COMPASS complexes (complex of proteins associating with Set1). These evolutionarily conserved proteins regulate DNA promoter and enhancer elements, modulating the activity of diverse cell types critical for embryonic morphogenesis, central nervous system development, and post-natal survival. KMT2C/D COMPASS complexes and their binding partners enhance active gene expression of specific loci via the targeted modification of histone-3 tail residues, in general promoting active euchromatic conformations. Over the last 20 years, mutations in five key COMPASS complex genes have been linked to three human congenital syndromes: Kabuki syndrome (type 1 [KMT2D] and 2 [KDM6A]), Rubinstein-Taybi syndrome (type 1 [CBP] and 2 [EP300]), and Kleefstra syndrome type 2 (KMT2C). Here, we review the composition and biochemical function of the KMT2 complexes. The specific cellular and embryonic roles of the KMT2C/D COMPASS complex are highlight with a focus on clinically relevant mechanisms sensitive to haploinsufficiency. The phenotypic similarities and differences between the members of this new family of disorders are outlined and emerging therapeutic strategies are detailed.
Collapse
Affiliation(s)
- William J Lavery
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH, 45229-3026, USA
| | - Artem Barski
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH, 45229-3026, USA
- Division of Human Genetics, CCHMC, Cincinnati, OH, USA
| | - Susan Wiley
- Division of Developmental and Behavioral Pediatrics, CCHMC, Cincinnati, OH, USA
| | | | - Andrew W Lindsley
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH, 45229-3026, USA.
| |
Collapse
|
10
|
Li L, Zhao Z, Jiang W, Guo J, Zhang S. Identification and functional characterization of Lys-trimethylation of lactate dehydrogenase A. Onco Targets Ther 2019; 12:5395-5404. [PMID: 31371982 PMCID: PMC6626897 DOI: 10.2147/ott.s208637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/13/2019] [Indexed: 01/10/2023] Open
Abstract
Background: Trimethylation of histones has been extensively studied, where histone methyltransferases catalyze the transfer of methyl groups from S-adenosyl methionine. Thus far, there have been no researches on the trimethylation of non-histone proteins. The precise mechanisms by which trimethylation affects cell progress and the related protein functions remain unclear. Purpose: The objective of this study was to identify the Lys-trimethylated proteins in kidney-derived cells and tissues, as well as to better understand the mechanisms underlying Lys-trimethylation-mediated cell metabolism. Methods: The levels of Lys-trimethylation in kidney-derived cells and tissues were assayed by Western blotting. Additionally, high-resolution mass spectrometry was used to analyze kidney-derived cells and tissues, and the eukaryotic expression vectors that led to the mutations of lysine were constructed and transfected into HEK293T cells. The LDHA activity of HEK293T cells was detected under conditions of Lys-trimethylation inhibition, and the proliferation of HEK293T cells was measured using EdU and Western blotting analyses. Results: The different proteins in kidney-derived cells and tissues showed different levels of Lys-trimethylation. In particular, lactate dehydrogenase A (LDHA) was Lys-trimethylated on lysine (K5). Inhibition of the Lys-trimethylation in LDHA increased the LDH activity of HEK293T cells and upregulated their proliferation. Conclusion: We suggested that LDHA affects the metabolism and proliferation of cells via a Lys-trimethylation-mediated mechanism; Lys-trimethylation might be a potential target for therapeutic research or used as a prognostic and treatment biomarker of several diseases.
Collapse
Affiliation(s)
- Lin Li
- Cancer Research Center, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, People's Republic of China.,Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, People's Republic of China
| | - Zuohui Zhao
- Departments of Urology and Pediatric Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250021, People's Republic of China
| | - Wenguo Jiang
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, People's Republic of China
| | - Jisheng Guo
- Cancer Research Center, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Shuping Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, People's Republic of China
| |
Collapse
|
11
|
Kim J, Lee B, Kim DH, Yeon JG, Lee J, Park Y, Lee Y, Lee SK, Lee S, Lee JW. UBE3A Suppresses Overnutrition-Induced Expression of the Steatosis Target Genes of MLL4 by Degrading MLL4. Hepatology 2019; 69:1122-1134. [PMID: 30230575 PMCID: PMC6393921 DOI: 10.1002/hep.30284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/05/2018] [Indexed: 12/28/2022]
Abstract
Regulation of the protein stability of epigenetic regulators remains ill-defined despite its potential applicability in epigenetic therapies. The histone H3-lysine 4-methyltransferase MLL4 is an epigenetic transcriptional coactivator that directs overnutrition-induced obesity and fatty liver formation, and Mll4+/- mice are resistant to both. Here we show that the E3 ubiquitin ligase UBE3A targets MLL4 for degradation, thereby suppressing high-fat diet (HFD)-induced expression of the hepatic steatosis target genes of MLL4. In contrast to Mll4+/- mice, Ube3a+/- mice are hypersensitive to HFD-induced obesity and fatty liver development. Ube3a+/-;Mll4+/- mice lose this hypersensitivity, supporting roles of increased MLL4 levels in both phenotypes of Ube3a+/- mice. Correspondingly, our comparative studies with wild-type, Ube3a+/- and Ube3a-/- and UBE3A-overexpressing transgenic mouse livers demonstrate an inverse correlation of UBE3A protein levels with MLL4 protein levels, expression of the steatosis target genes of MLL4, and their decoration by H3-lysine 4-monomethylation, a surrogate marker for the epigenetic action of MLL4. Conclusion: UBE3A indirectly exerts an epigenetic regulation of obesity and steatosis by degrading MLL4. This UBE3A-MLL4 regulatory axis provides a potential therapeutic venue for treating various MLL4-directed pathogeneses, including obesity and hepatic steatosis.
Collapse
Affiliation(s)
- Janghyun Kim
- Neuroscience Section, Papé Family Pediatric Research
Institute, Department of Pediatrics, Oregon Health & Science University,
Portland, OR 97239, USA
| | - Bora Lee
- Center for Neuroscience, Korea Institute of Science and
Technology, Seoul 02792, Korea
| | - Dae-Hwan Kim
- Neuroscience Section, Papé Family Pediatric Research
Institute, Department of Pediatrics, Oregon Health & Science University,
Portland, OR 97239, USA
| | - Je Gwang Yeon
- College of Pharmacy and Research Institute of
Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Jeongkyung Lee
- Division of Endocrinology & Metabolism, Department of
Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Younjung Park
- Neuroscience Section, Papé Family Pediatric Research
Institute, Department of Pediatrics, Oregon Health & Science University,
Portland, OR 97239, USA
| | - Yuna Lee
- Neuroscience Section, Papé Family Pediatric Research
Institute, Department of Pediatrics, Oregon Health & Science University,
Portland, OR 97239, USA
| | - Soo-Kyung Lee
- Neuroscience Section, Papé Family Pediatric Research
Institute, Department of Pediatrics, Oregon Health & Science University,
Portland, OR 97239, USA,Vollum Institute, Oregon Health & Science University,
Portland, OR 97239, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of
Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea,Correspondences: Seunghee Lee
() or Jae W. Lee
()
| | - Jae W. Lee
- Neuroscience Section, Papé Family Pediatric Research
Institute, Department of Pediatrics, Oregon Health & Science University,
Portland, OR 97239, USA,Correspondences: Seunghee Lee
() or Jae W. Lee
()
| |
Collapse
|
12
|
Kim DH, Kim J, Kwon JS, Sandhu J, Tontonoz P, Lee SK, Lee S, Lee JW. Critical Roles of the Histone Methyltransferase MLL4/KMT2D in Murine Hepatic Steatosis Directed by ABL1 and PPARγ2. Cell Rep 2017; 17:1671-1682. [PMID: 27806304 DOI: 10.1016/j.celrep.2016.10.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/12/2016] [Accepted: 10/07/2016] [Indexed: 10/20/2022] Open
Abstract
The pathophysiologic continuum of non-alcoholic fatty liver disease begins with steatosis. Despite recent advances in our understanding of the gene regulatory program directing steatosis, how it is orchestrated at the chromatin level is unclear. PPARγ2 is a hepatic steatotic transcription factor induced by overnutrition. Here, we report that the histone H3 lysine 4 methyltransferase MLL4/KMT2D directs overnutrition-induced murine steatosis via its coactivator function for PPARγ2. We demonstrate that overnutrition facilitates the recruitment of MLL4 to steatotic target genes of PPARγ2 and their transactivation via H3 lysine 4 methylation because PPARγ2 phosphorylated by overnutrition-activated ABL1 kinase shows enhanced interaction with MLL4. We further show that Pparg2 (encoding PPARγ2) is also a hepatic target gene of ABL1-PPARγ2-MLL4. Consistently, inhibition of ABL1 improves the fatty liver condition of mice with overnutrition by suppressing the pro-steatotic action of MLL4. Our results uncover a murine hepatic steatosis regulatory axis consisting of ABL1-PPARγ2-MLL4, which may serve as a target of anti-steatosis drug development.
Collapse
Affiliation(s)
- Dae-Hwan Kim
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Janghyun Kim
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ji-Sun Kwon
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jaspreet Sandhu
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Soo-Kyung Lee
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA; Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 08826 Seoul, Korea.
| | - Jae W Lee
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
13
|
Yan L, Wang Y, Liu J, Nie Y, Zhong XB, Kan Q, Zhang L. Alterations of Histone Modifications Contribute to Pregnane X Receptor-Mediated Induction of CYP3A4 by Rifampicin. Mol Pharmacol 2017; 92:113-123. [PMID: 28546420 DOI: 10.1124/mol.117.108225] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/22/2017] [Indexed: 01/28/2023] Open
Abstract
CYP3A4 is one of the major drug-metabolizing enzymes in human and is responsible for the metabolism of 60% of clinically used drugs. Many drugs are able to induce the expression of CYP3A4, which usually causes drug-drug interactions and adverse drug reactions. This study aims to explore the role of histone modifications in rifampicin-induced expression of CYP3A4 in LS174T cells. We found that the induction of CYP3A4 mRNA (4- to 15-fold) by rifampicin in LS174T cells was associated with increased levels of histone H3 lysine 4 trimethylation (H3K4me3, above 1.8-fold) and H3 acetylation (above 2-fold) and a decreased level of histone H3 lysine 27 trimethylation (H3K27me3, about 50%) in the CYP3A4 promoter. Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Silencing NCOA6 or p300 by short-hairpin RNAs resulted in inhibition of the CYP3A4 induction as well as altered levels of H3K4me3, H3K27me3, or H3 acetylation in the CYP3A4 promoter. Knockdown of pregnane X receptor (PXR) expression not only suppressed the recruitment of NCOA6 and p300 but also abolished the changes caused by rifampicin in H3K4me3, H3K27me3, and H3 acetylation levels in the CYP3A4 promoter. Moreover, rifampicin treatment enhanced the nuclear accumulation and interactions between PXR and NCOA6/p300. In conclusion, we show that the alterations of histone modifications contribute to the PXR-mediated induction of CYP3A4 by rifampicin.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Yiting Wang
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Jingyang Liu
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Yali Nie
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Xiao-Bo Zhong
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Quancheng Kan
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Lirong Zhang
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| |
Collapse
|
14
|
Yang W, Ernst P. SET/MLL family proteins in hematopoiesis and leukemia. Int J Hematol 2016; 105:7-16. [DOI: 10.1007/s12185-016-2118-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 10/20/2016] [Indexed: 01/18/2023]
|
15
|
Canovas S, Ross PJ. Epigenetics in preimplantation mammalian development. Theriogenology 2016; 86:69-79. [PMID: 27165992 DOI: 10.1016/j.theriogenology.2016.04.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 02/27/2016] [Accepted: 03/14/2016] [Indexed: 12/11/2022]
Abstract
Fertilization is a very dynamic period of comprehensive chromatin remodeling, from which two specialized cells result in a totipotent zygote. The formation of a totipotent cell requires extensive epigenetic remodeling that, although independent of modifications in the DNA sequence, still entails a profound cell-fate change, supported by transcriptional profile modifications. As a result of finely tuned interactions between numerous mechanisms, the goal of fertilization is to form a full healthy new individual. To avoid the persistence of alterations in epigenetic marks, the epigenetic information contained in each gamete is reset during early embryogenesis. Covalent modification of DNA by methylation, as well as posttranslational modifications of histone proteins and noncoding RNAs, appears to be the main epigenetic mechanisms that control gene expression. These allow different cells in an organism to express different transcription profiles, despite each cell containing the same DNA sequence. In the context of replacement of spermatic protamine with histones from the oocyte, active cell division, and specification of different lineages, active and passive mechanisms of epigenetic remodeling have been revealed as critical for editing the epigenetic profile of the early embryo. Importantly, redundant factors and mechanisms are likely in place, and only a few have been reported as critical for fertilization or embryo survival by the use of knockout models. The aim of this review is to highlight the main mechanisms of epigenetic remodeling that ensue after fertilization in mammals.
Collapse
Affiliation(s)
- Sebastian Canovas
- LARCEL (Laboratorio Andaluz de Reprogramacion Celular), BIONAND, Centro Andaluz de Nanomedicina y Biotecnologia Campanillas, Malaga, Spain.
| | - Pablo Juan Ross
- Department of Animal Science, University of California, Davis, California, USA.
| |
Collapse
|
16
|
Chandel N, Ayasolla KS, Lan X, Sultana-Syed M, Chawla A, Lederman R, Vethantham V, Saleem MA, Chander PN, Malhotra A, Singhal PC. Epigenetic Modulation of Human Podocyte Vitamin D Receptor in HIV Milieu. J Mol Biol 2015; 427:3201-3215. [PMID: 26210663 PMCID: PMC4586951 DOI: 10.1016/j.jmb.2015.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/09/2015] [Accepted: 07/14/2015] [Indexed: 01/08/2023]
Abstract
HIV (human immunodeficiency virus) has been reported to induce podocyte injury through down regulation of vitamin D receptor (VDR) and activation of renin angiotensin system; however, the involved mechanism is not clear. Since HIV has been reported to modulate gene expression via epigenetic phenomena, we asked whether epigenetic factors contribute to down regulation of VDR. Kidney cells in HIV transgenic mice and HIV-infected podocytes (HIV/HPs) displayed enhanced expression of SNAIL, a repressor of VDR. To elucidate the mechanism, we studied the effect of HIV on expression of molecules involved in SNAIL repressor complex formation and demonstrated that HIV enhances expression of the histone deacetylase HDAC1 and DNA methyl transferases DNMT3b and DNMT1. 293T cells, when stably transfected with SNAIL (SNAIL/293T), displayed suppressed transcription and translation of VDR. In SNAIL/293T cells, co-immunoprecipitation studies revealed the association of HDAC1, DNMT3b, DNMT1, and mSin3A with SNAIL. Chromatin immunoprecipitation experiments confirmed the presence of the SNAIL repressor complex at the VDR promoter. Consistent with the enhanced DNA methyl transferase expression in HIV/HPs, there was an increased CpG methylation at the VDR promoter. Chromatin immunoprecipitation assay confirmed occurrence of H3K4 trimethylation on SNAIL promoter. Neither a VDR agonist (VDA) nor an HDAC inhibitor (HDACI) nor a demethylating agent (DAC) individually could optimally up regulate VDR in HIV milieu. However, VDA and HDACI when combined were successful in de-repressing VDR expression. Our findings demonstrate that SNAIL recruits multiple chromatin enzymes to form a repressor complex in HIV milieu that down regulates VDR expression.
Collapse
Affiliation(s)
- Nirupama Chandel
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Kameshwar S Ayasolla
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Xiqian Lan
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Maria Sultana-Syed
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Amrita Chawla
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Rivka Lederman
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Vasupradha Vethantham
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Moin A Saleem
- Renal Academic Unit, University of Bristol, City of Bristol BS8 1TH, United Kingdom
| | - Praveen N Chander
- Department of Pathology, New York Medical College, Valhalla, NY 10595, USA
| | - Ashwani Malhotra
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA
| | - Pravin C Singhal
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA; Hofstra North Shore LIJ Medical School, Hempstead, NY 11549-1000, USA.
| |
Collapse
|
17
|
Critical Roles of the LIM Domains of Lhx3 in Recruiting Coactivators to the Motor Neuron-Specifying Isl1-Lhx3 Complex. Mol Cell Biol 2015; 35:3579-89. [PMID: 26260513 DOI: 10.1128/mcb.00335-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 08/03/2015] [Indexed: 11/20/2022] Open
Abstract
During spinal cord development, the LIM domains of the LIM homeodomain factor Lhx3 bind to either the LIM cofactor nuclear LIM interactor (NLI) or another LIM homeodomain factor, Isl1, assembling the tetrameric V2 interneuron-specifying Lhx3 complex (2NLI:2Lhx3) or the hexameric motor neuron-specifying Isl1-Lhx3 complex (2NLI:2Isl1:2Lhx3). However, the detailed molecular basis by which the Lhx3-LIM domains contribute to motor neuron specification still remains poorly understood. Here, we show that the Lhx3-LIM domains are essential for recruiting transcriptional coactivators to the Isl1-Lhx3 complex. Using a yeast genetic screening system, we identify Lhx3 point mutants that bind to NLI but not Isl1. Accordingly, these mutants fail to assemble the Isl1-Lhx3 complex. However, their interaction with coactivators is relatively intact, and they are fully functional in the Lhx3 complex and V2 interneuron specification. Interestingly, when these Lhx3 mutants are directly fused to Isl1, their transcriptional activity in the Isl1-Lhx3 complex is restored. We further show that this restoration reflects an unexpected role of the Lhx3-LIM domains, likely together with Isl1, to form an interaction interface for coactivators. Our results suggest that the Lhx3-LIM domains play critical roles in transactivation of the Isl1-Lhx3 complex by not only directing the assembly of the Isl1-Lhx3 complex but also recruiting coactivators to the complex.
Collapse
|
18
|
Romagnolo DF, Zempleni J, Selmin OI. Nuclear receptors and epigenetic regulation: opportunities for nutritional targeting and disease prevention. Adv Nutr 2014; 5:373-85. [PMID: 25022987 PMCID: PMC4085186 DOI: 10.3945/an.114.005868] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Posttranslational modifications of histones, alterations in the recruitment and functions of non-histone proteins, DNA methylation, and changes in expression of noncoding RNAs contribute to current models of epigenetic regulation. Nuclear receptors (NRs) are a group of transcription factors that, through ligand-binding, act as sensors to changes in nutritional, environmental, developmental, pathophysiologic, and endocrine conditions and drive adaptive responses via gene regulation. One mechanism through which NRs direct gene expression is the assembly of transcription complexes with cofactors and coregulators that possess chromatin-modifying properties. Chromatin modifications can be transient or become part of the cellular "memory" and contribute to genomic imprinting. Because many food components bind to NRs, they can ultimately influence transcription of genes associated with biologic processes, such as inflammation, proliferation, apoptosis, and hormonal response, and alter the susceptibility to chronic diseases (e.g., cancer, diabetes, obesity). The objective of this review is to highlight how NRs influence epigenetic regulation and the relevance of dietary compound-NR interactions in human nutrition and for disease prevention and treatment. Identifying gene targets of unliganded and bound NRs may assist in the development of epigenetic maps for food components and dietary patterns. Progress in these areas may lead to the formulation of disease-prevention models based on epigenetic control by individual or associations of food ligands of NRs.
Collapse
Affiliation(s)
- Donato F Romagnolo
- Department of Nutritional Sciences and University of Arizona Cancer Center, University of Arizona, Tucson, AZ; and
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE
| | - Ornella I Selmin
- Department of Nutritional Sciences and University of Arizona Cancer Center, University of Arizona, Tucson, AZ; and
| |
Collapse
|
19
|
Roidl D, Hacker C. Histone methylation during neural development. Cell Tissue Res 2014; 356:539-52. [PMID: 24817100 DOI: 10.1007/s00441-014-1842-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/03/2014] [Indexed: 12/15/2022]
Abstract
Post-translational modification of histone proteins, such as the methylation of lysine and arginine residues, influences the higher order of chromatin and leads to gene activation or silencing. Histone methyltransferases or demethylases actively add or remove various methylation marks in a cell-type-specific and context-dependent way. They are therefore important players in regulating the transcriptional program of a cell. Some control of the various cellular programs is necessary during the differentiation of stem cells along a specific lineage, when differentiation to alternative lineages needs to be suppressed. One example is the development of neurons from neural stem cells during neurogenesis. Neurogenesis is a highly organized process that requires the proper coordination of survival, proliferation, differentiation and migration signals. This holds true for both embryonic and neural stem cells that give rise to the various cell types of the central nervous system. The control of embryonic and neural stem cell self-renewal and differentiation is achieved by both extrinsic and intrinsic signals that regulate gene expression precisely. Recent advances in neuroscience support the importance of epigenetic modifications, such as the methylation and acetylation of histones, as an important intrinsic mechanism for the regulation of central nervous system development. This review summarizes our current knowledge of histone methylation processes during neural development and provides insights into the function of histone methylation enzymes and their role during central nervous system development.
Collapse
Affiliation(s)
- Deborah Roidl
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | | |
Collapse
|
20
|
Kim JY, Banerjee T, Vinckevicius A, Luo Q, Parker JB, Baker MR, Radhakrishnan I, Wei JJ, Barish GD, Chakravarti D. A role for WDR5 in integrating threonine 11 phosphorylation to lysine 4 methylation on histone H3 during androgen signaling and in prostate cancer. Mol Cell 2014; 54:613-25. [PMID: 24793694 DOI: 10.1016/j.molcel.2014.03.043] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 12/04/2013] [Accepted: 03/04/2014] [Indexed: 11/16/2022]
Abstract
Upon androgen stimulation, PKN1-mediated histone H3 threonine 11 phosphorylation (H3T11P) promotes AR target gene activation. However, the underlying mechanism is not completely understood. Here, we show that WDR5, a subunit of the SET1/MLL complex, interacts with H3T11P, and this interaction facilitates the recruitment of the MLL1 complex and subsequent H3K4 tri-methylation (H3K4me3). Using ChIP-seq, we find that androgen stimulation results in a 6-fold increase in the number of H3T11P-marked regions and induces WDR5 colocalization to one third of H3T11P-enriched promoters, thus establishing a genome-wide relationship between H3T11P and recruitment of WDR5. Accordingly, PKN1 knockdown or chemical inhibition severely blocks WDR5 chromatin association and H3K4me3 on AR target genes. Finally, WDR5 is critical in prostate cancer cell proliferation and is hyperexpressed in human prostate cancers. Together, these results identify WDR5 as a critical epigenomic integrator of histone phosphorylation and methylation and as a major driver of androgen-dependent prostate cancer cell proliferation.
Collapse
Affiliation(s)
- Ji-Young Kim
- Division of Reproductive Science in Medicine, Department of OB/GYN, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Taraswi Banerjee
- Division of Reproductive Science in Medicine, Department of OB/GYN, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Aurimas Vinckevicius
- Division of Reproductive Science in Medicine, Department of OB/GYN, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Driskill Graduate Program, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Qianyi Luo
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - J Brandon Parker
- Division of Reproductive Science in Medicine, Department of OB/GYN, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Mairead R Baker
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ishwar Radhakrishnan
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Jian-Jun Wei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Grant D Barish
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Debabrata Chakravarti
- Division of Reproductive Science in Medicine, Department of OB/GYN, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
21
|
Praktiknjo SD, Llamas B, Scott-Boyer MP, Picard S, Robert F, Langlais D, Haibe-Kains B, Faubert D, Silversides DW, Deschepper CF. Novel effects of chromosome Y on cardiac regulation, chromatin remodeling, and neonatal programming in male mice. Endocrinology 2013; 154:4746-56. [PMID: 24105479 DOI: 10.1210/en.2013-1699] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Little is known about the functions of chromosome Y (chrY) genes beyond their effects on sex and reproduction. In hearts, postpubertal testosterone affects the size of cells and the expression of genes differently in male C57BL/6J than in their C57.Y(A) counterparts, where the original chrY has been substituted with that from A/J mice. We further compared the 2 strains to better understand how chrY polymorphisms may affect cardiac properties, the latter being sexually dimorphic but unrelated to sex and reproduction. Genomic regions showing occupancy with androgen receptors (ARs) were identified in adult male hearts from both strains by chromatin immunoprecipitation. AR chromatin immunoprecipitation peaks (showing significant enrichment for consensus AR binding sites) were mostly strain specific. Measurements of anogenital distances in male pups showed that the biologic effects of perinatal androgens were greater in C57BL/6J than in C57.Y(A). Although perinatal endocrine manipulations showed that these differences contributed to the strain-specific differences in the response of adult cardiac cells to testosterone, the amounts of androgens produced by fetal testes were not different in each strain. Nonetheless, chrY polymorphisms associated in newborn pups' hearts with strain-specific differences in genomic regions showing either AR occupancy, accessible chromatin sites, or trimethylation of histone H3 Lysine 4 marks, as well as with differential expression of 2 chrY-encoded histone demethylases. In conclusion, the effects of chrY on adult cardiac phenotypes appeared to result from an interaction of this chromosome with the organizational programming effects exerted by the neonatal testosterone surge and show several characteristics of being mediated by an epigenetic remodeling of chromatin.
Collapse
Affiliation(s)
- Samantha D Praktiknjo
- Institut de Recherches Cliniques de Montréal, 110 Pine Avenue West, Montréal, Québec, Canada H2W 1R7.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Histone H3K27 trimethylation inhibits H3 binding and function of SET1-like H3K4 methyltransferase complexes. Mol Cell Biol 2013; 33:4936-46. [PMID: 24126056 DOI: 10.1128/mcb.00601-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Trimethylated histone H3 lysine 4 (H3K4) and H3K27 generally mark transcriptionally active and repressive chromatins, respectively. In most cell types, these two modifications are mutually exclusive, and this segregation is crucial for the regulation of gene expression. However, how this anticorrelation is achieved has not been fully understood. Here, we show that removal of the H3K27 trimethyl mark facilitates recruitment of SET1-like H3K4 methyltransferase complexes to their target genes by eliciting a novel interaction between histone H3 and two common subunits, WDR5 and RBBP5, of SET1-like complexes. Consistent with this result, H3K27 trimethylation destabilizes interactions of H3 with SET1-like complexes and antagonizes their ability to carry out H3K4 trimethylation of peptide (H3 residues 1 to 36), histone octamer, and mononucleosome substrates. Altogether, our studies reveal that H3K27 trimethylation of histone H3 represses a previously unrecognized interaction between H3 and SET1-like complexes. This provides an important mechanism that directs the anticorrelation between H3K4 and H3K27 trimethylation.
Collapse
|
23
|
Smith Z, Ryerson D, Kemper JK. Epigenomic regulation of bile acid metabolism: emerging role of transcriptional cofactors. Mol Cell Endocrinol 2013; 368:59-70. [PMID: 22579755 PMCID: PMC3473118 DOI: 10.1016/j.mce.2012.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 04/20/2012] [Accepted: 04/24/2012] [Indexed: 01/07/2023]
Abstract
The traditional role of bile acids is to simply facilitate absorption and digestion of lipid nutrients, but bile acids also act as endocrine signaling molecules that activate nuclear and membrane receptors to control integrative metabolism and energy balance. The mechanisms by which bile acid signals are integrated to regulate target genes are, however, largely unknown. Recently emerging evidence has shown that transcriptional cofactors sense metabolic changes and modulate gene transcription by mediating reversible epigenomic post-translational modifications (PTMs) of histones and chromatin remodeling. Importantly, targeting these epigenomic changes has been a successful approach for treating human diseases, especially cancer. Here, we review emerging roles of transcriptional cofactors in the epigenomic regulation of liver metabolism, especially focusing on bile acid metabolism. Targeting PTMs of histones and chromatin remodelers, together with the bile acid-activated receptors, may provide new therapeutic options for bile acid-related disease, such as cholestasis, obesity, diabetes, and entero-hepatic cancers.
Collapse
Affiliation(s)
- Zachary Smith
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, IL 61801, USA
| | | | | |
Collapse
|
24
|
Bögershausen N, Bruford E, Wollnik B. Skirting the pitfalls: a clear-cut nomenclature for H3K4 methyltransferases. Clin Genet 2012; 83:212-4. [PMID: 23130995 PMCID: PMC3597981 DOI: 10.1111/cge.12050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 10/30/2012] [Accepted: 10/30/2012] [Indexed: 01/16/2023]
Abstract
To unravel the system of epigenetic control of transcriptional regulation is a fascinating and important scientific pursuit. Surprisingly, recent successes in gene identification using high-throughput sequencing strategies showed that, despite their ubiquitous role in transcriptional control, dysfunction of chromatin-modifying enzymes can cause very specific human developmental phenotypes. An intriguing example is the identification of de novo dominant mutations in MLL2 as a cause of Kabuki syndrome, a well-known congenital syndrome that is associated with a very recognizable facial gestalt. However, the existing confusion in the nomenclature of the human and mouse MLL gene family impedes correct interpretation of scientific findings for these genes and their encoded proteins. This Review aims to point out this nomenclature pitfall, to explain its historical background, and to promote an unequivocal nomenclature system for chromatin-modifying enzymes as proposed by Allis et al. (2007).
Collapse
Affiliation(s)
- N Bögershausen
- Institute of Human Genetics, University of Cologne, Cologne, Germany
| | | | | |
Collapse
|
25
|
Wang J, Muntean AG, Wu L, Hess JL. A subset of mixed lineage leukemia proteins has plant homeodomain (PHD)-mediated E3 ligase activity. J Biol Chem 2012; 287:43410-6. [PMID: 23129768 DOI: 10.1074/jbc.m112.423855] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The mixed lineage leukemia protein MLL1 contains four highly conserved plant homeodomain (PHD) fingers, which are invariably deleted in oncogenic MLL1 fusion proteins in human leukemia. Here we show that the second PHD finger (PHD2) of MLL1 is an E3 ubiquitin ligase in the presence of the E2-conjugating enzyme CDC34. This activity is conserved in the second PHD finger of MLL4, the closest homolog to MLL1 but not in MLL2 or MLL3. Mutation of PHD2 leads to MLL1 stabilization, as well as increased transactivation ability and MLL1 recruitment to the target gene loci, suggesting that PHD2 negatively regulates MLL1 activity.
Collapse
Affiliation(s)
- Jingya Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | |
Collapse
|
26
|
Abstract
Cellular reprogramming involves the artificial dedifferentiation of somatic cells to a pluripotent state. When affected by overexpressing specific transcription factors, the process is highly inefficient, as only 0.1-1% of cells typically undergo the transformation. This low efficiency has been attributed to high kinetic barriers that affect all cells equally and can only be overcome by rare stochastic events. The barriers to reprogramming are likely to involve transformations of chromatin state because (i) inhibitors of chromatin-modifying enzymes can enhance the efficiency of reprogramming and (ii) knockdown or knock-out of chromatin-modifying enzymes can lower the efficiency of reprogramming. Here, we review the relationship between chromatin state transformations (chromatin reprogramming) and cellular reprogramming, with an emphasis on transcription factors, chromatin remodeling factors, histone modifications and DNA methylation.
Collapse
|
27
|
X-linked H3K27me3 demethylase Utx is required for embryonic development in a sex-specific manner. Proc Natl Acad Sci U S A 2012; 109:13004-9. [PMID: 22826230 DOI: 10.1073/pnas.1210787109] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Embryogenesis requires the timely and coordinated activation of developmental regulators. It has been suggested that the recently discovered class of histone demethylases (UTX and JMJD3) that specifically target the repressive H3K27me3 modification play an important role in the activation of "bivalent" genes in response to specific developmental cues. To determine the requirements for UTX in pluripotency and development, we have generated Utx-null ES cells and mutant mice. The loss of UTX had a profound effect during embryogenesis. Utx-null embryos had reduced somite counts, neural tube closure defects and heart malformation that presented between E9.5 and E13.5. Unexpectedly, homozygous mutant female embryos were more severely affected than hemizygous mutant male embryos. In fact, we observed the survival of a subset of UTX-deficient males that were smaller in size and had reduced lifespan. Interestingly, these animals were fertile with normal spermatogenesis. Consistent with a midgestation lethality, UTX-null male and female ES cells gave rise to all three germ layers in teratoma assays, though sex-specific differences could be observed in the activation of developmental regulators in embryoid body assays. Lastly, ChIP-seq analysis revealed an increase in H3K27me3 in Utx-null male ES cells. In summary, our data demonstrate sex-specific requirements for this X-linked gene while suggesting a role for UTY during development.
Collapse
|
28
|
Chen W, Roeder RG. Mediator-dependent nuclear receptor function. Semin Cell Dev Biol 2011; 22:749-58. [PMID: 21854863 DOI: 10.1016/j.semcdb.2011.07.026] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 12/24/2022]
Abstract
As gene-specific transcription factors, nuclear receptors are broadly involved in many important biological processes. Their function on target genes requires the stepwise assembly of different coactivator complexes that facilitate chromatin remodeling and subsequent preinitiation complex (PIC) formation and function. Mediator has proved to be a crucial, and general, nuclear receptor-interacting coactivator, with demonstrated functions in transcription steps ranging from chromatin remodeling to subsequent PIC formation and function. Here we discuss our current understanding of (i) pathways involved in Mediator recruitment and function through nuclear receptor target gene enhancers and promoters, (ii) conditional requirements for the strong nuclear receptor-Mediator interactions mediated by NR AF2 domains and the MED1 LXXLL motifs, (iii) Mediator functions, through different nuclear receptor-interacting subunits, in different metabolic pathways, (iv) emerging functions of Mediator as a corepressor in addition to its major role as a coactivator and (v) mechanisms by which Mediator acts to transmit signals from enhancer-bound nuclear receptors to the general transcription machinery at core promoters to effect PIC formation and function. As a nuclear receptor coregulator with increasingly diverse functions, Mediator may thus modulate nuclear receptor signaling through several different mechanisms.
Collapse
Affiliation(s)
- Wei Chen
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, 1230 York Ave., New York, NY 10065, USA.
| | | |
Collapse
|
29
|
Lee S, Lee SK. Crucial roles of histone-modifying enzymes in mediating neural cell-type specification. Curr Opin Neurobiol 2010; 20:29-36. [PMID: 20137907 DOI: 10.1016/j.conb.2010.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 01/12/2010] [Accepted: 01/13/2010] [Indexed: 11/29/2022]
Abstract
The development of the central nervous system (CNS) is governed by networks of extrinsic and intrinsic molecular programs that together orchestrate precise gene regulation. For the past few years, significant progress has been made in the characterization of histone-modifying enzymes and the roles they play in transcriptional control by affecting chromatin structure. Importantly, recent studies have revealed dynamic changes in histone modifications over the course of neural cell-fate specification. Further understanding of physiological functions of histone-modifying enzymes and their molecular mechanisms of action in CNS development will provide crucial insights into the process of generating neural cell types with tremendous diversity. Here we discuss the recent advancement in understanding the roles of enzymes involved in histone acetylation and methylation during neural cell-type specification.
Collapse
Affiliation(s)
- Seunghee Lee
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|