1
|
Xie Y, Zhao R, Zheng Y, Li Y, Wu F, Lei Y, Li L, Zeng H, Chen Z, Hou Y. Targeting KPNB1 suppresses AML cells by inhibiting HMGB2 nuclear import. Oncogene 2025; 44:1646-1661. [PMID: 40082556 DOI: 10.1038/s41388-025-03340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Acute myeloid leukemia (AML) represents the most prevalent malignancy within the hematologic system, characterized by refractory relapses and a scarcity of effective treatment options. Karyopherin subunit beta-1 (KPNB1) is a member of karyopherin β family, mediating the nuclear import of its cargoes. In this study, we found that elevated expression levels of KPNB1 are associated with unfavorable outcomes in patients with AML. The knockdown of KPNB1 resulted in growth inhibition and apoptosis in AML cells. Additionally, pharmacological inhibition of KPNB1 using the specific inhibitor importazole (IPZ) significantly reduced tumor burden and prolonged survival in MLL-AF9-induced AML mice. Notably, the inhibition of KPNB1 by IPZ significantly enhanced the sensitivity of both AML cell lines and patient-derived cells to venetoclax in vitro and in xenograft mice models. At the molecular level, we identified an unrecognized cargo of KPNB1, high mobility group 2 (HMGB2), which plays a crucial role in DNA damage repair. Inhibition of KPNB1 resulted in impaired nuclear import of HMGB2, eventually leading to compromised DNA damage repair in AML cells. Overall, our findings elucidate the essential roles of KPNB1 in AML cells through the HMGB2-DNA damage repair axis and highlight a promising therapeutic target for AML intervention.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Animals
- Mice
- beta Karyopherins/antagonists & inhibitors
- beta Karyopherins/genetics
- beta Karyopherins/metabolism
- HMGB2 Protein/metabolism
- HMGB2 Protein/genetics
- Active Transport, Cell Nucleus/drug effects
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Apoptosis/drug effects
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Nucleus/metabolism
- Cell Proliferation/drug effects
- Quinazolines
Collapse
Affiliation(s)
- Yuxin Xie
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Runlong Zhao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yingjiao Zheng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yan Li
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Feng Wu
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yufei Lei
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Lei Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Hanqing Zeng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
| | - Zhe Chen
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
| | - Yu Hou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China.
| |
Collapse
|
2
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
3
|
Arrieta A, Chapski DJ, Reese A, Kimball TH, Song K, Rosa-Garrido M, Vondriska TM. Circadian control of histone turnover during cardiac development and growth. J Biol Chem 2024; 300:107434. [PMID: 38830405 PMCID: PMC11261805 DOI: 10.1016/j.jbc.2024.107434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
During postnatal cardiac hypertrophy, cardiomyocytes undergo mitotic exit, relying on DNA replication-independent mechanisms of histone turnover to maintain chromatin organization and gene transcription. In other tissues, circadian oscillations in nucleosome occupancy influence clock-controlled gene expression, suggesting a role for the circadian clock in temporal control of histone turnover and coordinated cardiomyocyte gene expression. We sought to elucidate roles for the master circadian transcription factor, Bmal1, in histone turnover, chromatin organization, and myocyte-specific gene expression and cell growth in the neonatal period. Bmal1 knockdown in neonatal rat ventricular myocytes decreased myocyte size, total cellular protein synthesis, and transcription of the fetal hypertrophic gene Nppb after treatment with serum or the α-adrenergic agonist phenylephrine. Depletion of Bmal1 decreased the expression of clock-controlled genes Per2 and Tcap, as well as Sik1, a Bmal1 target upregulated in adult versus embryonic hearts. Bmal1 knockdown impaired Per2 and Sik1 promoter accessibility as measured by micrococcal nuclease-quantitative PCR and impaired histone turnover as measured by metabolic labeling of acid-soluble chromatin fractions. Sik1 knockdown in turn decreased myocyte size, while simultaneously inhibiting natriuretic peptide B transcription and activating Per2 transcription. Linking these changes to chromatin remodeling, depletion of the replication-independent histone variant H3.3a inhibited myocyte hypertrophy and prevented phenylephrine-induced changes in clock-controlled gene transcription. Bmal1 is required for neonatal myocyte growth, replication-independent histone turnover, and chromatin organization at the Sik1 promoter. Sik1 represents a novel clock-controlled gene that coordinates myocyte growth with hypertrophic and clock-controlled gene transcription. Replication-independent histone turnover is required for transcriptional remodeling of clock-controlled genes in cardiac myocytes in response to growth stimuli.
Collapse
Affiliation(s)
- Adrian Arrieta
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Douglas J Chapski
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Anna Reese
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Todd H Kimball
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Kunhua Song
- Internal Medicine, Heart Institute, Center for Regenerative Medicine, University of South Florida, Tampa, Florida, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Division of Cardiology, Department of Medicine, UCLA, Los Angeles, California, USA; Department of Physiology, UCLA, Los Angeles, California, USA; Molecular Biology Institute, UCLA, Los Angeles, California, USA.
| |
Collapse
|
4
|
Arrieta A, Chapski DJ, Reese A, Kimball T, Song K, Rosa-Garrido M, Vondriska TM. Circadian Control of Histone Turnover During Cardiac Development and Growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567086. [PMID: 38014083 PMCID: PMC10680691 DOI: 10.1101/2023.11.14.567086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Rationale: During postnatal cardiac hypertrophy, cardiomyocytes undergo mitotic exit, relying on DNA replication-independent mechanisms of histone turnover to maintain chromatin organization and gene transcription. In other tissues, circadian oscillations in nucleosome occupancy influence clock-controlled gene expression, suggesting an unrecognized role for the circadian clock in temporal control of histone turnover and coordinate cardiomyocyte gene expression. Objective: To elucidate roles for the master circadian transcription factor, Bmal1, in histone turnover, chromatin organization, and myocyte-specific gene expression and cell growth in the neonatal period. Methods and Results: Bmal1 knockdown in neonatal rat ventricular myocytes (NRVM) decreased myocyte size, total cellular protein, and transcription of the fetal hypertrophic gene Nppb following treatment with increasing serum concentrations or the α-adrenergic agonist phenylephrine (PE). Bmal1 knockdown decreased expression of clock-controlled genes Per2 and Tcap, and salt-inducible kinase 1 (Sik1) which was identified via gene ontology analysis of Bmal1 targets upregulated in adult versus embryonic hearts. Epigenomic analyses revealed co-localized chromatin accessibility and Bmal1 localization in the Sik1 promoter. Bmal1 knockdown impaired Per2 and Sik1 promoter accessibility as measured by MNase-qPCR and impaired histone turnover indicated by metabolic labeling of acid-soluble chromatin fractions and immunoblots of total and chromatin-associated core histones. Sik1 knockdown basally increased myocyte size, while simultaneously impairing and driving Nppb and Per2 transcription, respectively. Conclusions: Bmal1 is required for neonatal myocyte growth, replication-independent histone turnover, and chromatin organization at the Sik1 promoter. Sik1 represents a novel clock-controlled gene that coordinates myocyte growth with hypertrophic and clock-controlled gene transcription.
Collapse
|
5
|
Zheng X, Lu J, Liu J, Zhou L, He Y. HMGB family proteins: Potential biomarkers and mechanistic factors in cardiovascular diseases. Biomed Pharmacother 2023; 165:115118. [PMID: 37437373 DOI: 10.1016/j.biopha.2023.115118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/14/2023] Open
Abstract
Cardiovascular disease (CVD) is the most fatal disease that causes sudden death, and inflammation contributes substantially to its occurrence and progression. The prevalence of CVD increases as the population ages, and the pathophysiology is complex. Anti-inflammatory and immunological modulation are the potential methods for CVD prevention and treatment. High-Mobility Group (HMG) chromosomal proteins are one of the most abundant nuclear nonhistone proteins which act as inflammatory mediators in DNA replication, transcription, and repair by producing cytokines and serving as damage-associated molecular patterns in inflammatory responses. The most common and well-studied HMG proteins are those with an HMGB domain, which participate in a variety of biological processes. HMGB1 and HMGB2 were the first members of the HMGB family to be identified and are present in all investigated eukaryotes. Our review is primarily concerned with the involvement of HMGB1 and HMGB2 in CVD. The purpose of this review is to provide a theoretical framework for diagnosing and treating CVD by discussing the structure and function of HMGB1 and HMGB2.
Collapse
Affiliation(s)
- Xialei Zheng
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Junmi Lu
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jing Liu
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Liufang Zhou
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Cardiovascular Medicine, the Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi 533000, China
| | - Yuhu He
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
6
|
Mathai C, Jourd'heuil F, Pham LGC, Gilliard K, Howard D, Balnis J, Jaitovich A, Chittur SV, Rilley M, Peredo-Wende R, Ammoura I, Shin SJ, Barroso M, Barra J, Shishkova E, Coon JJ, Lopez-Soler RI, Jourd'heuil D. Regulation of DNA damage and transcriptional output in the vasculature through a cytoglobin-HMGB2 axis. Redox Biol 2023; 65:102838. [PMID: 37573836 PMCID: PMC10428073 DOI: 10.1016/j.redox.2023.102838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023] Open
Abstract
Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.
Collapse
Affiliation(s)
- Clinton Mathai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Frances Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Le Gia Cat Pham
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Kurrim Gilliard
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dennis Howard
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Joseph Balnis
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Ariel Jaitovich
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Sridar V Chittur
- Center for Functional Genomics, Cancer Research Center, University at Albany, New York, 12144, USA
| | - Mark Rilley
- Division of Rheumatology, Department of Medicine, Samuel Stratton VA Medical Center, Albany, NY, 12208, USA
| | - Ruben Peredo-Wende
- Division of Rheumatology, Department of Medicine, Samuel Stratton VA Medical Center, Albany, NY, 12208, USA
| | - Ibrahim Ammoura
- Department of Pathology and Medicine, Albany Medical Center, Albany, NY, 12208, USA
| | - Sandra J Shin
- Department of Pathology and Medicine, Albany Medical Center, Albany, NY, 12208, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jonathan Barra
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI, 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53506, USA
| | - Joshua J Coon
- Department of Pathology and Medicine, Albany Medical Center, Albany, NY, 12208, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53506, USA; Morgridge Institute for Research, Madison, WI, 53515, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53506, USA
| | - Reynold I Lopez-Soler
- Section of Renal Transplantation, Edward Hines VA Jr. Hospital, Hines, IL, 60141, USA; Department of Surgery, Division of Intra-Abdominal Transplantation, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
7
|
Mathai C, Jourd'heuil F, Pham LGC, Gilliard K, Howard D, Balnis J, Jaitovich A, Chittur SV, Rilley M, Peredo-Wende R, Ammoura I, Shin SJ, Barroso M, Barra J, Shishkova E, Coon JJ, Lopez-Soler RI, Jourd'heuil D. Nuclear cytoglobin associates with HMGB2 and regulates DNA damage and genome-wide transcriptional output in the vasculature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540045. [PMID: 37214992 PMCID: PMC10197644 DOI: 10.1101/2023.05.10.540045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.
Collapse
|
8
|
Starkova T, Polyanichko A, Tomilin AN, Chikhirzhina E. Structure and Functions of HMGB2 Protein. Int J Mol Sci 2023; 24:ijms24098334. [PMID: 37176041 PMCID: PMC10179549 DOI: 10.3390/ijms24098334] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
High-Mobility Group (HMG) chromosomal proteins are the most numerous nuclear non-histone proteins. HMGB domain proteins are the most abundant and well-studied HMG proteins. They are involved in variety of biological processes. HMGB1 and HMGB2 were the first members of HMGB-family to be discovered and are found in all studied eukaryotes. Despite the high degree of homology, HMGB1 and HMGB2 proteins differ from each other both in structure and functions. In contrast to HMGB2, there is a large pool of works devoted to the HMGB1 protein whose structure-function properties have been described in detail in our previous review in 2020. In this review, we attempted to bring together diverse data about the structure and functions of the HMGB2 protein. The review also describes post-translational modifications of the HMGB2 protein and its role in the development of a number of diseases. Particular attention is paid to its interaction with various targets, including DNA and protein partners. The influence of the level of HMGB2 expression on various processes associated with cell differentiation and aging and its ability to mediate the differentiation of embryonic and adult stem cells are also discussed.
Collapse
Affiliation(s)
- Tatiana Starkova
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | - Alexander Polyanichko
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | - Alexey N Tomilin
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | - Elena Chikhirzhina
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| |
Collapse
|
9
|
Gromova T, Gehred ND, Vondriska TM. Single-cell transcriptomes in the heart: when every epigenome counts. Cardiovasc Res 2023; 119:64-78. [PMID: 35325060 PMCID: PMC10233279 DOI: 10.1093/cvr/cvac040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The response of an organ to stimuli emerges from the actions of individual cells. Recent cardiac single-cell RNA-sequencing studies of development, injury, and reprogramming have uncovered heterogeneous populations even among previously well-defined cell types, raising questions about what level of experimental resolution corresponds to disease-relevant, tissue-level phenotypes. In this review, we explore the biological meaning behind this cellular heterogeneity by undertaking an exhaustive analysis of single-cell transcriptomics in the heart (including a comprehensive, annotated compendium of studies published to date) and evaluating new models for the cardiac function that have emerged from these studies (including discussion and schematics that depict new hypotheses in the field). We evaluate the evidence to support the biological actions of newly identified cell populations and debate questions related to the role of cell-to-cell variability in development and disease. Finally, we present emerging epigenomic approaches that, when combined with single-cell RNA-sequencing, can resolve basic mechanisms of gene regulation and variability in cell phenotype.
Collapse
Affiliation(s)
- Tatiana Gromova
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Natalie D Gehred
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Arrieta A, Vondriska TM. Nucleosome proteostasis and histone turnover. Front Mol Biosci 2022; 9:990006. [PMID: 36250018 PMCID: PMC9563994 DOI: 10.3389/fmolb.2022.990006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Maintenance of protein folding homeostasis, or proteostasis is critical for cell survival as well as for execution of cell type specific biological processes such as muscle cell contractility, neuronal synapse and memory formation, and cell transition from a mitotic to post-mitotic cell type. Cell type specification is driven largely by chromatin organization, which dictates which genes are turned off or on, depending on cell needs and function. Loss of chromatin organization can have catastrophic consequences either on cell survival or cell type specific function. Chromatin organization is highly dependent on organization of nucleosomes, spatiotemporal nucleosome assembly and disassembly, and histone turnover. In this review our goal is to highlight why nucleosome proteostasis is critical for chromatin organization, how this process is mediated by histone chaperones and ATP-dependent chromatin remodelers and outline potential and established mechanisms of disrupted nucleosome proteostasis during disease. Finally, we highlight how these mechanisms of histone turnover and nucleosome proteostasis may conspire with unfolded protein response programs to drive histone turnover in cell growth and development.
Collapse
Affiliation(s)
- Adrian Arrieta
- Departments of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Adrian Arrieta,
| | - Thomas M. Vondriska
- Departments of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Departments of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Departments of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Chen F, Li W, Zhang D, Fu Y, Yuan W, Luo G, Liu F, Luo J. MALAT1 regulates hypertrophy of cardiomyocytes by modulating the miR-181a/HMGB2 pathway. Eur J Histochem 2022; 66:3426. [PMID: 35726535 PMCID: PMC9251611 DOI: 10.4081/ejh.2022.3426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/25/2022] [Indexed: 12/27/2022] Open
Abstract
Noncoding RNAs are important for regulation of cardiac hypertrophy. The function of MALAT1 (a long noncoding mRNA), miR-181a, and HMGB2; their contribution to cardiac hypertrophy; and the regulatory relationship between them during this process remain unknown. In the present study, we treated primary cardiomyocytes with angiotensin II (Ang II) to mimic cardiac hypertrophy. MALAT1 expression was significantly downregulated in Ang II-treated cardiomyocytes compared with control cardiomyocytes. Ang II-induced cardiac hypertrophy was suppressed by overexpression of MALAT1 and promoted by genetic knockdown of MALAT1. A dual-luciferase reporter assay demonstrated that MALAT1 acted as a sponge for miR-181a and inhibited its expression during cardiac hypertrophy. Cardiac hypertrophy was suppressed by overexpression of a miR-181a inhibitor and enhanced by overexpression of a miR-181a mimic. HMGB2 was downregulated during cardiac hypertrophy and was identified as a target of miR-181a by bioinformatics analysis and a dual-luciferase reporter assay. miR-181a overexpression decreased the mRNA and protein levels of HMGB2. Rescue experiments indicated that MALAT1 overexpression reversed the effect of miR-181a on HMGB2 expression. In summary, the results of the present study show that MALAT1 acts as a sponge for miR-181a and thereby regulates expression of HMGB2 and development of cardiac hypertrophy. The novel MALAT1/miR-181a/HMGB2 axis might play a crucial role in cardiac hypertrophy and serve as a new therapeutic target.
Collapse
Affiliation(s)
- Feng Chen
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong; Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi .
| | - Wenfeng Li
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong; Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi .
| | - Dandan Zhang
- Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi.
| | - Youlin Fu
- Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi.
| | - Wenjin Yuan
- Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi.
| | - Gang Luo
- Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi.
| | - Fuwei Liu
- Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi.
| | - Jun Luo
- Department of Cardiology, Ganzhou People's Hospital, Nanchang University, Ganzhou, Jiangxi.
| |
Collapse
|
12
|
Hu S, Vondriska TM. How Chromatin Stiffens Fibroblasts. CURRENT OPINION IN PHYSIOLOGY 2022; 26. [DOI: 10.1016/j.cophys.2022.100537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Jo HR, Jeong JH. MicroRNA-Mediated Downregulation of HMGB2 Contributes to Cellular Senescence in Microvascular Endothelial Cells. Cells 2022; 11:cells11030584. [PMID: 35159393 PMCID: PMC8834370 DOI: 10.3390/cells11030584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/29/2022] [Accepted: 02/07/2022] [Indexed: 11/24/2022] Open
Abstract
High mobility group box 2 (HMGB2) is a non-histone chromosomal protein involved in various biological processes, including cellular senescence. However, its role in cellular senescence has not been evaluated extensively. To determine the regulatory role and mechanism of HMGB2 in cellular senescence, we performed gene expression analysis, senescence staining, and tube formation assays using young and senescent microvascular endothelial cells (MVECs) after small RNA treatment or HMGB2 overexpression. HMGB2 expression decreased with age and was regulated at the transcriptional level. siRNA-mediated downregulation inhibited cell proliferation and accelerated cellular senescence. In contrast, ectopic overexpression delayed senescence and maintained relatively higher tube-forming activity. To determine the HMGB2 downregulation mechanism, we screened miRNAs that were significantly upregulated in senescent MVECs and selected HMGB2-targeting miRNAs. Six miRNAs, miR-23a-3p, 23b-3p, -181a-5p, -181b-5p, -221-3p, and -222-3p, were overexpressed in senescent MVECs. Ectopic introduction of miR-23a-3p, -23b-3p, -181a-5p, -181b-5p, and -221-3p, with the exception of miR-222-3p, led to the downregulation of HMGB2, upregulation of senescence-associated markers, and decreased tube formation activity. Inhibition of miR-23a-3p, -181a-5p, -181b-5p, and -221-3p delayed cellular senescence. Restoration of HMGB2 expression using miRNA inhibitors represents a potential strategy to overcome the detrimental effects of cellular senescence in endothelial cells.
Collapse
Affiliation(s)
- Hye-Ram Jo
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Science, Seoul 01812, Korea;
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Korea
| | - Jae-Hoon Jeong
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Science, Seoul 01812, Korea;
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: ; Tel.: +82-2970-1386
| |
Collapse
|
14
|
Interactions of HMGB Proteins with the Genome and the Impact on Disease. Biomolecules 2021; 11:biom11101451. [PMID: 34680084 PMCID: PMC8533419 DOI: 10.3390/biom11101451] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023] Open
Abstract
High Mobility Group Box (HMGB) proteins are small architectural DNA binding proteins that regulate multiple genomic processes such as DNA damage repair, nucleosome sliding, telomere homeostasis, and transcription. In doing so they control both normal cellular functions and impact a myriad of disease states, including cancers and autoimmune diseases. HMGB proteins bind to DNA and nucleosomes to modulate the local chromatin environment, which facilitates the binding of regulatory protein factors to the genome and modulates higher order chromosomal organization. Numerous studies over the years have characterized the structure and function of interactions between HMGB proteins and DNA, both biochemically and inside cells, providing valuable mechanistic insight as well as evidence these interactions influence pathological processes. This review highlights recent studies supporting the roles of HMGB1 and HMGB2 in global organization of the genome, as well as roles in transcriptional regulation and telomere maintenance via interactions with G-quadruplex structures. Moreover, emerging models for how HMGB proteins function as RNA binding proteins are presented. Nuclear HMGB proteins have broad regulatory potential to impact numerous aspects of cellular metabolism in normal and disease states.
Collapse
|
15
|
Pagiatakis C, Di Mauro V. The Emerging Role of Epigenetics in Therapeutic Targeting of Cardiomyopathies. Int J Mol Sci 2021; 22:ijms22168721. [PMID: 34445422 PMCID: PMC8395924 DOI: 10.3390/ijms22168721] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiomyopathies (CMPs) are a heterogeneous group of myocardial diseases accountable for the majority of cases of heart failure (HF) and/or sudden cardiac death (SCD) worldwide. With the recent advances in genomics, the original classification of CMPs on the basis of morphological and functional criteria (dilated (DCM), hypertrophic (HCM), restrictive (RCM), and arrhythmogenic ventricular cardiomyopathy (AVC)) was further refined into genetic (inherited or familial) and acquired (non-inherited or secondary) forms. Despite substantial progress in the identification of novel CMP-associated genetic variations, as well as improved clinical recognition diagnoses, the functional consequences of these mutations and the exact details of the signaling pathways leading to hypertrophy, dilation, and/or contractile impairment remain elusive. To date, global research has mainly focused on the genetic factors underlying CMP pathogenesis. However, growing evidence shows that alterations in molecular mediators associated with the diagnosis of CMPs are not always correlated with genetic mutations, suggesting that additional mechanisms, such as epigenetics, may play a role in the onset or progression of CMPs. This review summarizes published findings of inherited CMPs with a specific focus on the potential role of epigenetic mechanisms in regulating these cardiac disorders.
Collapse
Affiliation(s)
- Christina Pagiatakis
- IRCCS-Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Correspondence: (C.P.); (V.D.M.)
| | - Vittoria Di Mauro
- IRCCS-Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Institute of Genetic and Biomedical Research (IRGB), Milan Unit, National Research Council, Via Fantoli 16/15, 20138 Milan, Italy
- Correspondence: (C.P.); (V.D.M.)
| |
Collapse
|
16
|
Papantonis A. HMGs as rheostats of chromosomal structure and cell proliferation. Trends Genet 2021; 37:986-994. [PMID: 34311989 DOI: 10.1016/j.tig.2021.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 11/18/2022]
Abstract
High mobility group proteins (HMGs) are the most abundant nuclear proteins next to histones and are robustly expressed across tissues and organs. HMGs can uniquely bend or bind distorted DNA, and are central to such processes as transcription, recombination, and DNA repair. However, their dynamic association with chromatin renders capturing HMGs on chromosomes challenging. Recent work has changed this and now implicates these factors in spatial genome organization. Here, I revisit older and review recent literature to describe how HMGs rewire spatial chromatin interactions to sustain homeostasis or promote cellular aging. I propose a 'rheostat' model to explain how HMG-box proteins (HMGBs), and to some extent HMG A proteins (HMGAs), may control cellular aging and, likely, cancer progression.
Collapse
Affiliation(s)
- Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
17
|
Liu Y, Jing J, Yu H, Zhang J, Cao Q, Zhang X, Liu J, Zhang S, Cheng W. Expression profiles of long non-coding RNAs in the cartilage of patients with knee osteoarthritis and normal individuals. Exp Ther Med 2021; 21:365. [PMID: 33732338 PMCID: PMC7903471 DOI: 10.3892/etm.2021.9796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022] Open
Abstract
Knee osteoarthritis is caused by a multifactorial imbalance in the synthesis and degradation of knee chondrocytes, subchondral bone and extracellular matrix. Abnormal expression of long non-coding RNAs (lncRNAs) affects the metabolism, synovitis, autophagy and apoptosis of chondrocytes, as well as the production of cartilage matrix. The aim of the present study was to identify novel targets for the treatment of osteoarthritis and to examine the pathogenesis of the disease. The lncRNA expression profiles of seven patients with knee osteoarthritis and six healthy controls were examined by RNA-sequencing. Differentially expressed lncRNAs were selected for bioinformatics analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Reverse transcription-quantitative PCR (RT-qPCR) was used to further investigate the differential expression of the lncRNAs. A total of 23,583 lncRNAs were identified in osteoarthritis cartilage, including 5,255 upregulated and 5,690 downregulated lncRNAs, compared with normal cartilage. Although there were more downregulated lncRNAs compared with upregulated lncRNAs, among the changed lncRNAs (fold-change >6), there were more upregulated lncRNAs compared with downregulated lncRNAs. Several lncRNAs exhibiting differences were identified as potential therapeutic targets in knee osteoarthritis. GO and KEGG pathway analyses were performed for the target genes of the differentially expressed lncRNAs. RT-qPCR validation was performed on three randomly selected upregulated and downregulated lncRNAs. The results of RT-qPCR were consistent with the findings obtained by RNA-sequencing analysis. The findings from the present study may contribute to the diagnosis of osteoarthritis and may predict the development of osteoarthritis. Furthermore, the differentially expressed lncRNAs may aid in the identification of novel candidate targets for the treatment of knee osteoarthritis.
Collapse
Affiliation(s)
- Yanchang Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Juehua Jing
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Haoran Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Jisen Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Qiliang Cao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Xin Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Jianjun Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Shuo Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Wendan Cheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
18
|
Bertero A, Rosa-Garrido M. Three-dimensional chromatin organization in cardiac development and disease. J Mol Cell Cardiol 2021; 151:89-105. [PMID: 33242466 PMCID: PMC11056610 DOI: 10.1016/j.yjmcc.2020.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Recent technological advancements in the field of chromatin biology have rewritten the textbook on nuclear organization. We now appreciate that the folding of chromatin in the three-dimensional space (i.e. its 3D "architecture") is non-random, hierarchical, and highly complex. While 3D chromatin structure is partially encoded in the primary sequence and thereby broadly conserved across cell types and states, a substantial portion of the genome seems to be dynamic during development or in disease. Moreover, there is growing evidence that at least some of the 3D structure of chromatin is functionally linked to gene regulation, both being modulated by and impacting on multiple nuclear processes (including DNA replication, transcription, and RNA splicing). In recent years, these new concepts have nourished several investigations about the functional role of 3D chromatin topology dynamics in the heart during development and disease. This review aims to provide a comprehensive overview of our current understanding in this field, and to discuss how this knowledge can inform further research as well as clinical practice.
Collapse
Affiliation(s)
- Alessandro Bertero
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, 850 Republican Street, Brotman Building, Seattle, WA 98109, USA.
| | - Manuel Rosa-Garrido
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, 650 Charles Young Dr, Los Angeles, CA 90095, USA.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Characterized by enlarged ventricle and loss of systolic function, dilated cardiomyopathy (DCM) has the highest morbidity among all the cardiomyopathies. Although it is well established that DCM is typically caused by mutations in a large number of genes, there is an emerging appreciation for the contribution of epigenetic alteration in the development of DCM. RECENT FINDINGS We present some of the recent progress in the field of epigenetics in DCM by focusing on the four major epigenetic modifications, that is, DNA methylation, histone modification, chromatin remodeling as well as the noncoding RNAs. The major players involved in these DCM-related epigenetic reprogramming will be highlighted. Finally, the diagnostic and the therapeutic implications for DCM based on new knowledge of epigenetic regulation will also be discussed. SUMMARY As a rapidly expanding field, epigenetic studies in DCM have the promise to yield both novel mechanistic insights as well as potential new avenues for more effective treatment of the disease.
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Comprehensive analyses of the genome, transcriptome, proteome and metabolome are instrumental in identifying biomarkers of disease, to gain insight into mechanisms underlying the development of cardiovascular disease, and show promise for better stratifying patients according to disease subtypes. This review highlights recent 'omics' studies, including integration of multiple 'omics' that have advanced mechanistic understanding and diagnosis in humans and animal models. RECENT FINDINGS Transcriptome-based discovery continues to be a primary method to obtain data for hypothesis generation and the understanding of disease pathogenesis has been enhanced by single cell-based methods capable of revealing heterogeneity in cellular responses. Advances in proteome coverage and quantitation of individual protein species, together with enhanced methods for detecting posttranslational modifications, have improved discovery of protein-based biomarkers. SUMMARY High-throughput assays capable of quantitating the vast majority of any particular type of biomolecule within a tissue sample, isolated cells or plasma are now available. In order to make best use of the large amount of data that can be generated on given molecule types, as well as their interrelationships in disease, continued development of pattern-recognition algorithms ('machine learning') will be required and the subclassification of disease that is made possible by such algorithms will be likely to inform clinical practice, and vice versa.
Collapse
|
21
|
Zeng Z, Huang N, Zhang Y, Wang Y, Su Y, Zhang H, An Y. CTCF inhibits endoplasmic reticulum stress and apoptosis in cardiomyocytes by upregulating RYR2 via inhibiting S100A1. Life Sci 2019; 242:117158. [PMID: 31837328 DOI: 10.1016/j.lfs.2019.117158] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
AIMS Pediatric heart failure is a common cardiovascular disease in clinical pediatrics. CCCTC-binding factor (CTCF), a novel transcriptional repressor, was reported to participate in the occurrence of various cardiovascular diseases. The present study focuses on exploring the effects of CTCF on tunicamycin (TM)-induced endoplasmic reticulum (ER) stress, and investigating the underlying mechanisms. MATERIALS AND METHOD Expression of CTCF in blood samples of heart failure children and TM-induced cardiomyocytes were evaluated by real-time quantitative PCR (RT-qPCR). Apoptotic rate of cardiomyocytes was detected by Annexin v assay. Western blotting and enzyme-linked immunosorbent assay (ELISA) were applied to examine the effect of CTCF on ER stress. Co-immunoprecipitation and western blotting were devoted to explore the mechanism by which CTCF contributes to ER stress. KEY FINDINGS We proved that CTCF was lowly expressed in blood samples of heart failure children and TM-induced cardiomyocytes, and overexpression of CTCF weaken the TM-induced ER stress. Using co-immunoprecipitation and protein blots, we demonstrated that CTCF upregulates RYR2 by inhibiting S100A1, thus mediating the PERK signaling pathway and regulating ER stress. SIGNIFICANCE Our data revealed that CTCF protects cardiomyocytes from ER stress through S100A1-RYR2 axis, and can be applied as a therapeutic target for the treatment of pediatric heart failure in future.
Collapse
Affiliation(s)
- Zhu Zeng
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003
| | - Nina Huang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003
| | - Yudan Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003
| | - Ying Wang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003
| | - Yufei Su
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003
| | - Huifang Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003
| | - Yuan An
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China 710003.
| |
Collapse
|
22
|
Abstract
Supplemental Digital Content is available in the text. If unifying principles could be revealed for how the same genome encodes different eukaryotic cells and for how genetic variability and environmental input are integrated to impact cardiovascular health, grand challenges in basic cell biology and translational medicine may succumb to experimental dissection. A rich body of work in model systems has implicated chromatin-modifying enzymes, DNA methylation, noncoding RNAs, and other transcriptome-shaping factors in adult health and in the development, progression, and mitigation of cardiovascular disease. Meanwhile, deployment of epigenomic tools, powered by next-generation sequencing technologies in cardiovascular models and human populations, has enabled description of epigenomic landscapes underpinning cellular function in the cardiovascular system. This essay aims to unpack the conceptual framework in which epigenomes are studied and to stimulate discussion on how principles of chromatin function may inform investigations of cardiovascular disease and the development of new therapies.
Collapse
Affiliation(s)
- Manuel Rosa-Garrido
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Douglas J Chapski
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Thomas M Vondriska
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles.
| |
Collapse
|
23
|
Rajderkar S, Mann JM, Panaretos C, Yumoto K, Li HD, Mishina Y, Ralston B, Kaartinen V. Trim33 is required for appropriate development of pre-cardiogenic mesoderm. Dev Biol 2019; 450:101-114. [PMID: 30940539 DOI: 10.1016/j.ydbio.2019.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 11/25/2022]
Abstract
Congenital cardiac malformations are among the most common birth defects in humans. Here we show that Trim33, a member of the Tif1 subfamily of tripartite domain containing transcriptional cofactors, is required for appropriate differentiation of the pre-cardiogenic mesoderm during a narrow time window in late gastrulation. While mesoderm-specific Trim33 mutants did not display noticeable phenotypes, epiblast-specific Trim33 mutant embryos developed ventricular septal defects, showed sparse trabeculation and abnormally thin compact myocardium, and died as a result of cardiac failure during late gestation. Differentiating embryoid bodies deficient in Trim33 showed an enrichment of gene sets associated with cardiac differentiation and contractility, while the total number of cardiac precursor cells was reduced. Concordantly, cardiac progenitor cell proliferation was reduced in Trim33-deficient embryos. ChIP-Seq performed using antibodies against Trim33 in differentiating embryoid bodies revealed more than 4000 peaks, which were significantly enriched close to genes implicated in stem cell maintenance and mesoderm development. Nearly half of the Trim33 peaks overlapped with binding sites of the Ctcf insulator protein. Our results suggest that Trim33 is required for appropriate differentiation of precardiogenic mesoderm during late gastrulation and that it will likely mediate some of its functions via multi-protein complexes, many of which include the chromatin architectural and insulator protein Ctcf.
Collapse
Affiliation(s)
- Sudha Rajderkar
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeffrey M Mann
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Christopher Panaretos
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kenji Yumoto
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hong-Dong Li
- Center for Bioinformatics, School of Information Science and Engineering, Central South University, Changsha, Hunan, 410083, PR China
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin Ralston
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
Inhibition of Triple-Negative Breast Cancer Cell Aggressiveness by Cathepsin D Blockage: Role of Annexin A1. Int J Mol Sci 2019; 20:ijms20061337. [PMID: 30884823 PMCID: PMC6471925 DOI: 10.3390/ijms20061337] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/13/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) are more aggressive than other breast cancer (BC) subtypes and lack effective therapeutic options. Unraveling marker events of TNBCs may provide new directions for development of strategies for targeted TNBC therapy. Herein, we reported that Annexin A1 (AnxA1) and Cathepsin D (CatD) are highly expressed in MDA-MB-231 (TNBC lineage), compared to MCF-10A and MCF-7. Since the proposed concept was that CatD has protumorigenic activity associated with its ability to cleave AnxA1 (generating a 35.5 KDa fragment), we investigated this mechanism more deeply using the inhibitor of CatD, Pepstatin A (PepA). Fourier Transform Infrared (FTIR) spectroscopy demonstrated that PepA inhibits CatD activity by occupying its active site; the OH bond from PepA interacts with a CO bond from carboxylic acids of CatD catalytic aspartate dyad, favoring the deprotonation of Asp33 and consequently inhibiting CatD. Treatment of MDA-MB-231 cells with PepA induced apoptosis and autophagy processes while reducing the proliferation, invasion, and migration. Finally, in silico molecular docking demonstrated that the catalytic inhibition comprises Asp231 protonated and Asp33 deprotonated, proving all functional results obtained. Our findings elucidated critical CatD activity in TNBC cell trough AnxA1 cleavage, indicating the inhibition of CatD as a possible strategy for TNBC treatment.
Collapse
|
25
|
Karbassi E, Rosa-Garrido M, Chapski DJ, Wu Y, Ren S, Wang Y, Stefani E, Vondriska TM. Direct visualization of cardiac transcription factories reveals regulatory principles of nuclear architecture during pathological remodeling. J Mol Cell Cardiol 2019; 128:198-211. [PMID: 30742811 PMCID: PMC6644685 DOI: 10.1016/j.yjmcc.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022]
Abstract
Heart failure is associated with hypertrophying of cardiomyocytes and changes in transcriptional activity. Studies from rapidly dividing cells in culture have suggested that transcription may be compartmentalized into factories within the nucleus, but this phenomenon has not been tested in vivo and the role of nuclear architecture in cardiac gene regulation is unknown. While alterations to transcription have been linked to disease, little is known about the regulation of the spatial organization of transcription and its properties in the pathological setting. In the present study, we investigate the structural features of endogenous transcription factories in the heart and determine the principles connecting chromatin structure to transcriptional regulation in vivo. Super-resolution imaging of endogenous RNA polymerase II clusters in neonatal and adult cardiomyocytes revealed distinct properties of transcription factories in response to pathological stress: neonatal nuclei demonstrated changes in number of clusters, with parallel increases in nuclear area, while the adult nuclei underwent changes in size and intensity of RNA polymerase II foci. Fluorescence in situ hybridization-based labeling of genes revealed locus-specific relationships between expression change and anatomical localization-with respect to nuclear periphery and heterochromatin regions, both sites associated with gene silencing-in the nuclei of cardiomyocytes in hearts (but not liver hepatocytes) of mice subjected to pathologic stimuli that induce heart failure. These findings demonstrate a role for chromatin organization and rearrangement of nuclear architecture for cell type-specific transcription in vivo during disease. RNA polymerase II ChIP and chromatin conformation capture studies in the same model system demonstrate formation and reorganization of distinct nuclear compartments regulating gene expression. These findings reveal locus-specific compartmentalization of stress-activated, housekeeping and silenced genes in the anatomical context of the endogenous nucleus, revealing basic principles of global chromatin structure and nuclear architecture in the regulation of gene expression in healthy and diseased conditions.
Collapse
Affiliation(s)
- Elaheh Karbassi
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Manuel Rosa-Garrido
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Douglas J Chapski
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Yong Wu
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Shuxun Ren
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Yibin Wang
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Enrico Stefani
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States
| | - Thomas M Vondriska
- Departments of Anesthesiology, Medicine/Cardiology, Physiology, David Geffen School of Medicine at UCLA, 650 Charles Young Dr., Los Angeles, CA 90095, United States.
| |
Collapse
|
26
|
Abstract
Spatiotemporal gene expression during cardiac development is a highly regulated process. Activation of key signaling pathways involved in electrophysiological programming, such as Notch and Wnt signaling, occurs in early cardiovascular development and these pathways are reactivated during pathologic remodeling. Direct targets of these signaling pathways have also been associated with inherited arrhythmias such as Brugada syndrome and arrhythmogenic cardiomyopathy. In addition, evidence is emerging from animal models that reactivation of Notch and Wnt signaling during cardiac pathology may predispose to acquired arrhythmias, underscoring the importance of elucidating the transcriptional and epigenetic effects on cardiac gene regulation. Here, we highlight specific examples where gene expression dictates electrophysiological properties in both normal and diseased hearts.
Collapse
|
27
|
Cui M, Wang Z, Bassel-Duby R, Olson EN. Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease. Development 2018; 145:145/24/dev171983. [PMID: 30573475 DOI: 10.1242/dev.171983] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Embryonic and postnatal life depend on the uninterrupted function of cardiac muscle cells. These cells, termed cardiomyocytes, display many fascinating behaviors, including complex morphogenic movements, interactions with other cell types of the heart, persistent contractility and quiescence after birth. Each of these behaviors depends on complex interactions between both cardiac-restricted and widely expressed transcription factors, as well as on epigenetic modifications. Here, we review recent advances in our understanding of the genetic and epigenetic control of cardiomyocyte differentiation and proliferation during heart development, regeneration and disease. We focus on those regulators that are required for both heart development and disease, and highlight the regenerative principles that might be manipulated to restore function to the injured adult heart.
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
28
|
Xu J, Ma H, Jin J, Uttam S, Fu R, Huang Y, Liu Y. Super-Resolution Imaging of Higher-Order Chromatin Structures at Different Epigenomic States in Single Mammalian Cells. Cell Rep 2018; 24:873-882. [PMID: 30044984 PMCID: PMC6154382 DOI: 10.1016/j.celrep.2018.06.085] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/18/2018] [Accepted: 06/20/2018] [Indexed: 01/10/2023] Open
Abstract
Histone modifications influence higher-order chromatin structures at individual epigenomic states and chromatin environments to regulate gene expression. However, genome-wide higher-order chromatin structures shaped by different histone modifications remain poorly characterized. With stochastic optical reconstruction microscopy (STORM), we characterized the higher-order chromatin structures at their epigenomic states, categorized into three major types in interphase: histone acetylation marks form spatially segregated nanoclusters, active histone methylation marks form spatially dispersed larger nanodomains, and repressive histone methylation marks form condensed large aggregates. These distinct structural characteristics are also observed in mitotic chromosomes. Furthermore, active histone marks coincide with less compact chromatin and exhibit a higher degree of co-localization with other active marks and RNA polymerase II (RNAP II), while repressive marks coincide with densely packed chromatin and spatially distant from repressive marks and active RNAP II. Taken together, super-resolution imaging reveals three distinct chromatin structures at various epigenomic states, which may be spatially coordinated to impact transcription.
Collapse
Affiliation(s)
- Jianquan Xu
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hongqiang Ma
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jingyi Jin
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; School of Medicine, Tsinghua University, No.1 Tsinghua Yuan, Haidian District, Beijing 100084, China
| | - Shikhar Uttam
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rao Fu
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; College of Chemical Engineering, Northeast Electric Power University, Jilin City, Jilin Province 132012, China
| | - Yi Huang
- Magee-Women's Research Institute, University of Pittsburgh Cancer Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| | - Yang Liu
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
29
|
Sizer AJ, Martin KA. Respecting boundaries: CTCF, chromatin structural organization, and heart failure. J Thorac Dis 2018; 9:4889-4892. [PMID: 29312684 DOI: 10.21037/jtd.2017.11.77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ashley J Sizer
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
30
|
Rosa-Garrido M, Chapski DJ, Schmitt AD, Kimball TH, Karbassi E, Monte E, Balderas E, Pellegrini M, Shih TT, Soehalim E, Liem D, Ping P, Galjart NJ, Ren S, Wang Y, Ren B, Vondriska TM. High-Resolution Mapping of Chromatin Conformation in Cardiac Myocytes Reveals Structural Remodeling of the Epigenome in Heart Failure. Circulation 2017; 136:1613-1625. [PMID: 28802249 PMCID: PMC5648689 DOI: 10.1161/circulationaha.117.029430] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/31/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cardiovascular disease is associated with epigenomic changes in the heart; however, the endogenous structure of cardiac myocyte chromatin has never been determined. METHODS To investigate the mechanisms of epigenomic function in the heart, genome-wide chromatin conformation capture (Hi-C) and DNA sequencing were performed in adult cardiac myocytes following development of pressure overload-induced hypertrophy. Mice with cardiac-specific deletion of CTCF (a ubiquitous chromatin structural protein) were generated to explore the role of this protein in chromatin structure and cardiac phenotype. Transcriptome analyses by RNA-seq were conducted as a functional readout of the epigenomic structural changes. RESULTS Depletion of CTCF was sufficient to induce heart failure in mice, and human patients with heart failure receiving mechanical unloading via left ventricular assist devices show increased CTCF abundance. Chromatin structural analyses revealed interactions within the cardiac myocyte genome at 5-kb resolution, enabling examination of intra- and interchromosomal events, and providing a resource for future cardiac epigenomic investigations. Pressure overload or CTCF depletion selectively altered boundary strength between topologically associating domains and A/B compartmentalization, measurements of genome accessibility. Heart failure involved decreased stability of chromatin interactions around disease-causing genes. In addition, pressure overload or CTCF depletion remodeled long-range interactions of cardiac enhancers, resulting in a significant decrease in local chromatin interactions around these functional elements. CONCLUSIONS These findings provide a high-resolution chromatin architecture resource for cardiac epigenomic investigations and demonstrate that global structural remodeling of chromatin underpins heart failure. The newly identified principles of endogenous chromatin structure have key implications for epigenetic therapy.
Collapse
Affiliation(s)
- Manuel Rosa-Garrido
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Douglas J Chapski
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Anthony D Schmitt
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Todd H Kimball
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Elaheh Karbassi
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Emma Monte
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Enrique Balderas
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Matteo Pellegrini
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Tsai-Ting Shih
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Elizabeth Soehalim
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - David Liem
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Peipei Ping
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Niels J Galjart
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Shuxun Ren
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Yibin Wang
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Bing Ren
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.)
| | - Thomas M Vondriska
- From Departments of Anesthesiology and Perioperative Medicine (M.R.-G., D.J.C., T.H.K., E.K., E.M., T.-T.S., E.S., S.R., Y.W., T.M.V.), Medicine (D.L., P.P., Y.W., T.M.V.), Physiology (P.P.,Y.W., T.M.V.), Molecular, Cellular and Developmental Biology (M.P.), UCLA, Los Angeles, CA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City (E.B.); Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, and Moores Cancer Center, UCSD, San Diego, CA (A.D.S., B.R.); and Department of Cell Biology and Genetics, Erasmus MC, Rotterdam, The Netherlands (N.J.G.).
| |
Collapse
|
31
|
Bronstein R, Kyle J, Abraham AB, Tsirka SE. Neurogenic to Gliogenic Fate Transition Perturbed by Loss of HMGB2. Front Mol Neurosci 2017; 10:153. [PMID: 28588451 PMCID: PMC5440561 DOI: 10.3389/fnmol.2017.00153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/05/2017] [Indexed: 01/21/2023] Open
Abstract
Mouse cortical development relies heavily on a delicate balance between neurogenesis and gliogenesis. The lateral ventricular zone produces different classes of excitatory pyramidal cells until just before birth, when the production of astroglia begins to prevail. Epigenetic control of this fate shift is of critical importance and chromatin regulatory elements driving neuronal or astroglial development play an vital role. Different classes of chromatin binding proteins orchestrate the transcriptional repression of neuronal-specific genes, while allowing for the activation of astrocyte-specific genes. Through proteomic analysis of embryonic neural progenitor cells (NPCs) our group had previously identified high mobility group B2 (HMGB2), a chromatin protein dynamically expressed throughout embryonic development. In the current study using cultures of perinatal NPCs from HMGB2+/+ and HMGB2-/- mice we discovered that vital elements of the polycomb group (PcG) epigenetic complexes polycomb repressive complexes 1 and 2 (PRC1/2) were downregulated during the differentiation process of HMGB2-null NPCs. These epigenetic changes led to downstream changes in specific histone modification levels, specifically the trimethylation of H3K27, and a subsequent shift in the perinatal neurogenesis to gliogenesis fate transition. Collectively these results demonstrate that chromatin binding proteins, such as HMGB2, can have significant effects on the epigenetic landscape of perinatal neural stem/progenitor cells.
Collapse
Affiliation(s)
- Robert Bronstein
- Program in Neuroscience, Stony Brook University, Stony BrookNY, United States
- Cold Spring Harbor Laboratory, Cold Spring HarborNY, United States
| | - Jackson Kyle
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony BrookNY, United States
| | - Ariel B. Abraham
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony BrookNY, United States
| | - Stella E. Tsirka
- Program in Neuroscience, Stony Brook University, Stony BrookNY, United States
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony BrookNY, United States
| |
Collapse
|