1
|
Cheng K, Zhou Y, Hao Y, Wu S, Wang N, Zhang P, Wang Y. Magnolol inhibits appetite and causes visceral fat loss through Growth/differentiation factor-15 (GDF-15) by activating transcription factor 4-CCAAT enhancer binding protein γ-mediated endoplasmic reticulum stress responses. Chin J Nat Med 2025; 23:334-345. [PMID: 40122663 DOI: 10.1016/s1875-5364(25)60835-1] [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: 12/23/2023] [Revised: 03/11/2024] [Accepted: 05/07/2024] [Indexed: 03/25/2025]
Abstract
Magnolol, a compound extracted from Magnolia officinalis, demonstrates potential efficacy in addressing metabolic dysfunction and cardiovascular diseases. Its biological activities encompass anti-inflammatory, antioxidant, anticoagulant, and anti-diabetic effects. Growth/differentiation factor-15 (GDF-15), a member of the transforming growth factor β superfamily, is considered a potential therapeutic target for metabolic disorders. This study investigated the impact of magnolol on GDF-15 production and its underlying mechanism. The research examined the pharmacological effect of magnolol on GDF-15 expression in vitro and in vivo, and determined the involvement of endoplasmic reticulum (ER) stress signaling in this process. Luciferase reporter assays, chromatin immunoprecipitation, and in vitro DNA binding assays were employed to examine the regulation of GDF-15 by activating transcription factor 4 (ATF4), CCAAT enhancer binding protein γ (CEBPG), and CCCTC-binding factor (CTCF). The study also investigated the effect of magnolol and ATF4 on the activity of a putative enhancer located in the intron of the GDF-15 gene, as well as the influence of single nucleotide polymorphisms (SNPs) on magnolol and ATF4-induced transcription activity. Results demonstrated that magnolol triggers GDF-15 production in endothelial cells (ECs), hepatoma cell line G2 (HepG2) and hepatoma cell line 3B (Hep3B) cell lines, and primary mouse hepatocytes. The cooperative binding of ATF4 and CEBPG upstream of the GDF-15 gene or the E1944285 enhancer located in the intron led to full-power transcription of the GDF-15 gene. SNP alleles were found to impact the magnolol and ATF4-induced transcription activity of GDF-15. In high-fat diet ApoE-/- mice, administration of magnolol induced GDF-15 production and partially suppressed appetite through GDF-15. These findings suggest that magnolol regulates GDF-15 expression through priming of promoter and enhancer activity, indicating its potential as a drug for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Keru Cheng
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yanyun Zhou
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yilong Hao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Shengyun Wu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Nanping Wang
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Peng Zhang
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Yinfang Wang
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
2
|
Chen Y, Liu L, Calhoun R, Cheng L, Merrick D, Steger DJ, Seale P. Transcriptional regulation of adipocyte lipolysis by IRF2BP2. SCIENCE ADVANCES 2025; 11:eads5963. [PMID: 39752494 PMCID: PMC11698119 DOI: 10.1126/sciadv.ads5963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/03/2024] [Indexed: 01/06/2025]
Abstract
Adipocyte lipolysis controls systemic energy levels and metabolic homeostasis. Lipolysis is regulated by posttranslational modifications of key lipolytic enzymes. However, less is known about the transcriptional mechanisms that regulate lipolysis. Here, we identify interferon regulatory factor-2 binding protein 2 (IRF2BP2) as a transcriptional repressor of adipocyte lipolysis. Deletion of IRF2BP2 in human adipocytes increases lipolysis without affecting glucose uptake, whereas IRF2BP2 overexpression decreases lipolysis. RNA sequencing, and chromatin immunoprecipitation sequencing analyses show that IRF2BP2 represses lipolysis-related genes, including LIPE, which encodes hormone sensitive lipase, the rate-limiting enzyme in lipolysis. Adipocyte-selective deletion of Irf2bp2 in mice increases Lipe expression and free fatty acid levels, resulting in adipose tissue inflammation and glucose intolerance. Together, these findings demonstrate that IRF2BP2 restrains adipocyte lipolysis and opens avenues to target lipolysis for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Yang Chen
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Liu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Calhoun
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lan Cheng
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Merrick
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Steger
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Chen Y, Liu L, Calhoun R, Cheng L, Merrick D, Steger DJ, Seale P. Transcriptional regulation of adipocyte lipolysis by IRF2BP2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605689. [PMID: 39211193 PMCID: PMC11360913 DOI: 10.1101/2024.07.31.605689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Adipocyte lipolysis controls systemic energy levels and metabolic homeostasis. Lipolysis is regulated by post-translational modifications of key lipolytic enzymes. However, less is known about the transcriptional mechanisms that regulate lipolysis. Here, we identify the transcriptional factor interferon regulatory factor-2 binding protein 2 (IRF2BP2) as a repressor of adipocyte lipolysis. Deletion of IRF2BP2 in primary human adipocytes increases lipolysis without affecting glucose uptake, whereas IRF2BP2 overexpression decreases lipolysis. RNA-seq and ChIP-seq analyses reveal that IRF2BP2 directly represses several lipolysis-related genes, including LIPE ( HSL , hormone sensitive lipase), which encodes the rate-limiting enzyme in lipolysis. Adipocyte-selective deletion of Irf2bp2 in mice increases Lipe expression and free fatty acid levels, resulting in elevated adipose tissue inflammation and glucose intolerance. Altogether, these findings demonstrate that IRF2BP2 restrains adipocyte lipolysis and opens new avenues to target lipolysis for the treatment of metabolic disease.
Collapse
|
4
|
Deng J, Liu J, Chen W, Liang Q, He Y, Sun G. Effects of Natural Products through Inhibiting Endoplasmic Reticulum Stress on Attenuation of Idiopathic Pulmonary Fibrosis. Drug Des Devel Ther 2024; 18:1627-1650. [PMID: 38774483 PMCID: PMC11108075 DOI: 10.2147/dddt.s388920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 04/23/2024] [Indexed: 05/24/2024] Open
Abstract
With ever-increasing intensive studies of idiopathic pulmonary fibrosis (IPF), significant progresses have been made. Endoplasmic reticulum stress (ERS)/unfolded protein reaction (UPR) is associated with the development and progression of IPF, and targeting ERS/UPR may be beneficial in the treatment of IPF. Natural product is a tremendous source of new drug discovery, and accumulating studies have reported that many natural products show potential therapeutic effects for IPF via modulating one or more branches of the ERS signaling pathway. Therefore, this review focuses on critical roles of ERS in IPF development, and summarizes herbal preparations and bioactive compounds which protect against IPF through regulating ERS.
Collapse
Affiliation(s)
- JiuLing Deng
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, People’s Republic of China
| | - Jing Liu
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, People’s Republic of China
| | - WanSheng Chen
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People’s Republic of China
| | - Qing Liang
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, People’s Republic of China
| | - YuQiong He
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People’s Republic of China
| | - GuangChun Sun
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, People’s Republic of China
| |
Collapse
|
5
|
Grmai L, Michaca M, Lackner E, Nampoothiri V P N, Vasudevan D. Integrated stress response signaling acts as a metabolic sensor in fat tissues to regulate oocyte maturation and ovulation. Cell Rep 2024; 43:113863. [PMID: 38457339 PMCID: PMC11077669 DOI: 10.1016/j.celrep.2024.113863] [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: 03/21/2023] [Revised: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/10/2024] Open
Abstract
Reproduction is an energy-intensive process requiring systemic coordination. However, the inter-organ signaling mechanisms that relay nutrient status to modulate reproductive output are poorly understood. Here, we use Drosophila melanogaster as a model to establish the integrated stress response (ISR) transcription factor, Atf4, as a fat tissue metabolic sensor that instructs oogenesis. We demonstrate that Atf4 regulates lipase activity to mediate yolk lipoprotein synthesis in the fat body. Depletion of Atf4 in the fat body also blunts oogenesis recovery after amino acid deprivation and re-feeding, suggestive of a nutrient-sensing role for Atf4. We also discovered that Atf4 promotes secretion of a fat-body-derived neuropeptide, CNMamide, which modulates neural circuits that promote egg-laying behavior (ovulation). Thus, we posit that ISR signaling in fat tissue acts as a "metabolic sensor" that instructs female reproduction-directly by impacting yolk lipoprotein production and follicle maturation and systemically by regulating ovulation.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Manuel Michaca
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Emily Lackner
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Deepika Vasudevan
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Xiao Y, Xie X, Chen Z, Yin G, Kong W, Zhou J. Advances in the roles of ATF4 in osteoporosis. Biomed Pharmacother 2023; 169:115864. [PMID: 37948991 DOI: 10.1016/j.biopha.2023.115864] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
Osteoporosis (OP) is characterized by reduced bone mass, decreased strength, and enhanced bone fragility fracture risk. Activating transcription factor 4 (ATF4) plays a role in cell differentiation, proliferation, apoptosis, redox balance, amino acid uptake, and glycolipid metabolism. ATF4 induces the differentiation of bone marrow mesenchymal stem cells (BM-MSCs) into osteoblasts, increases osteoblast activity, and inhibits osteoclast formation, promoting bone formation and remodeling. In addition, ATF4 mediates the energy metabolism in osteoblasts and promotes angiogenesis. ATF4 is also involved in the mediation of adipogenesis. ATF4 can selectively accumulate in osteoblasts. ATF4 can directly interact with RUNT-related transcription factor 2 (RUNX2) and up-regulate the expression of osteocalcin (OCN) and osterix (Osx). Several upstream factors, such as Wnt/β-catenin and BMP2/Smad signaling pathways, have been involved in ATF4-mediated osteoblast differentiation. ATF4 promotes osteoclastogenesis by mediating the receptor activator of nuclear factor κ-B (NF-κB) ligand (RANKL) signaling. Several agents, such as parathyroid (PTH), melatonin, and natural compounds, have been reported to regulate ATF4 expression and mediate bone metabolism. In this review, we comprehensively discuss the biological activities of ATF4 in maintaining bone homeostasis and inhibiting OP development. ATF4 has become a therapeutic target for OP treatment.
Collapse
Affiliation(s)
- Yaosheng Xiao
- Department of Orthopaetics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Zhixi Chen
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
7
|
Kim G, Lee J, Ha J, Kang I, Choe W. Endoplasmic Reticulum Stress and Its Impact on Adipogenesis: Molecular Mechanisms Implicated. Nutrients 2023; 15:5082. [PMID: 38140341 PMCID: PMC10745682 DOI: 10.3390/nu15245082] [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: 10/28/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Endoplasmic reticulum (ER) stress plays a pivotal role in adipogenesis, which encompasses the differentiation of adipocytes and lipid accumulation. Sustained ER stress has the potential to disrupt the signaling of the unfolded protein response (UPR), thereby influencing adipogenesis. This comprehensive review illuminates the molecular mechanisms that underpin the interplay between ER stress and adipogenesis. We delve into the dysregulation of UPR pathways, namely, IRE1-XBP1, PERK and ATF6 in relation to adipocyte differentiation, lipid metabolism, and tissue inflammation. Moreover, we scrutinize how ER stress impacts key adipogenic transcription factors such as proliferator-activated receptor γ (PPARγ) and CCAAT-enhancer-binding proteins (C/EBPs) along with their interaction with other signaling pathways. The cellular ramifications include alterations in lipid metabolism, dysregulation of adipokines, and aged adipose tissue inflammation. We also discuss the potential roles the molecular chaperones cyclophilin A and cyclophilin B play in adipogenesis. By shedding light on the intricate relationship between ER stress and adipogenesis, this review paves the way for devising innovative therapeutic interventions.
Collapse
Affiliation(s)
- Gyuhui Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jiyoon Lee
- Department of Biological Sciences, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30609, USA;
| | - Joohun Ha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
8
|
Liu F, Liu Z, Cheng W, Zhao Q, Zhang X, Zhang H, Yu M, Xu H, Gao Y, Jiang Q, Shi G, Wang L, Gu S, Wang J, Cao N, Chen Z. The PERK Branch of the Unfolded Protein Response Safeguards Protein Homeostasis and Mesendoderm Specification of Human Pluripotent Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303799. [PMID: 37890465 PMCID: PMC10724406 DOI: 10.1002/advs.202303799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/14/2023] [Indexed: 10/29/2023]
Abstract
Cardiac development involves large-scale rearrangements of the proteome. How the developing cardiac cells maintain the integrity of the proteome during the rapid lineage transition remains unclear. Here it is shown that proteotoxic stress visualized by the misfolded and/or aggregated proteins appears during early cardiac differentiation of human pluripotent stem cells and is resolved by activation of the PERK branch of unfolded protein response (UPR). PERK depletion increases misfolded and/or aggregated protein accumulation, leading to pluripotency exit defect and impaired mesendoderm specification of human pluripotent stem cells. Mechanistically, it is found that PERK safeguards mesendoderm specification through its conserved downstream effector ATF4, which subsequently activates a novel transcriptional target WARS1, to cope with the differentiation-induced proteotoxic stress. The results indicate that protein quality control represents a previously unrecognized core component of the cardiogenic regulatory network. Broadly, these findings provide a framework for understanding how UPR is integrated into the developmental program by activating the PERK-ATF4-WARS1 axis.
Collapse
Affiliation(s)
- Fang Liu
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
- Department of Clinical LaboratoryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022P. R. China
| | - Zhun Liu
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Weisheng Cheng
- Prenatal Diagnosis CenterDepartment of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230022P. R. China
- Department of Medical InformaticsZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080P. R. China
| | - Qingquan Zhao
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Xinyu Zhang
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - He Zhang
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Miao Yu
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - He Xu
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Yichen Gao
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Qianrui Jiang
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Guojun Shi
- Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity ResearchGuangdong Provincial Key Laboratory of DiabetologyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangdong510080P. R. China
| | - Likun Wang
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijing100101P. R. China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shanshan Gu
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Jia Wang
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesShandong266071China
| | - Nan Cao
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| | - Zhongyan Chen
- Advanced Medical Technology CenterZhongshan School of Medicine and the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080P. R. China
- Key Laboratory for Stem Cells and Tissue EngineeringSun Yat‐Sen UniversityMinistry of EducationGuangzhou510080P. R. China
| |
Collapse
|
9
|
Örd T, Örd D, Adler P, Örd T. Genome-wide census of ATF4 binding sites and functional profiling of trait-associated genetic variants overlapping ATF4 binding motifs. PLoS Genet 2023; 19:e1011014. [PMID: 37906604 PMCID: PMC10637723 DOI: 10.1371/journal.pgen.1011014] [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: 05/25/2023] [Revised: 11/10/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023] Open
Abstract
Activating Transcription Factor 4 (ATF4) is an important regulator of gene expression in stress responses and developmental processes in many cell types. Here, we catalogued ATF4 binding sites in the human genome and identified overlaps with trait-associated genetic variants. We probed these genetic variants for allelic regulatory activity using a massively parallel reporter assay (MPRA) in HepG2 hepatoma cells exposed to tunicamycin to induce endoplasmic reticulum stress and ATF4 upregulation. The results revealed that in the majority of cases, the MPRA allelic activity of these SNPs was in agreement with the nucleotide preference seen in the ATF4 binding motif from ChIP-Seq. Luciferase and electrophoretic mobility shift assays in additional cellular models further confirmed ATF4-dependent regulatory effects for the SNPs rs532446 (GADD45A intronic; linked to hematological parameters), rs7011846 (LPL upstream; myocardial infarction), rs2718215 (diastolic blood pressure), rs281758 (psychiatric disorders) and rs6491544 (educational attainment). CRISPR-Cas9 disruption and/or deletion of the regulatory elements harboring rs532446 and rs7011846 led to the downregulation of GADD45A and LPL, respectively. Thus, these SNPs could represent examples of GWAS genetic variants that affect gene expression by altering ATF4-mediated transcriptional activation.
Collapse
Affiliation(s)
- Tiit Örd
- Institute of Genomics, University of Tartu, Tartu, Estonia
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Daima Örd
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Priit Adler
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Tõnis Örd
- Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
10
|
Grmai L, Michaca M, Lackner E, Nampoothiri V P N, Vasudevan D. Integrated Stress Response signaling acts as a metabolic sensor in fat tissues to regulate oocyte maturation and ovulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530289. [PMID: 36909541 PMCID: PMC10002630 DOI: 10.1101/2023.02.27.530289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Reproduction is an energy-intensive process requiring systemic coordination. However, the inter-organ signaling mechanisms that relay nutrient status to modulate reproductive output are poorly understood. Here, we use Drosophila melanogaster as a model to establish the Integrated Stress response (ISR) transcription factor, Atf4, as a fat tissue metabolic sensor which instructs oogenesis. We demonstrate that Atf4 regulates the lipase Brummer to mediate yolk lipoprotein synthesis in the fat body. Depletion of Atf4 in the fat body also blunts oogenesis recovery after amino acid deprivation and re-feeding, suggestive of a nutrient sensing role for Atf4. We also discovered that Atf4 promotes secretion of a fat body-derived neuropeptide, CNMamide, which modulates neural circuits that promote egg-laying behavior (ovulation). Thus, we posit that ISR signaling in fat tissue acts as a "metabolic sensor" that instructs female reproduction: directly, by impacting yolk lipoprotein production and follicle maturation, and systemically, by regulating ovulation.
Collapse
|
11
|
Neill G, Masson GR. A stay of execution: ATF4 regulation and potential outcomes for the integrated stress response. Front Mol Neurosci 2023; 16:1112253. [PMID: 36825279 PMCID: PMC9941348 DOI: 10.3389/fnmol.2023.1112253] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/19/2023] [Indexed: 02/10/2023] Open
Abstract
ATF4 is a cellular stress induced bZIP transcription factor that is a hallmark effector of the integrated stress response. The integrated stress response is triggered by phosphorylation of the alpha subunit of the eukaryotic initiation factor 2 complex that can be carried out by the cellular stress responsive kinases; GCN2, PERK, PKR, and HRI. eIF2α phosphorylation downregulates mRNA translation initiation en masse, however ATF4 translation is upregulated. The integrated stress response can output two contradicting outcomes in cells; pro-survival or apoptosis. The mechanism for choice between these outcomes is unknown, however combinations of ATF4 heterodimerisation partners and post-translational modifications have been linked to this regulation. This semi-systematic review article covers ATF4 target genes, heterodimerisation partners and post-translational modifications. Together, this review aims to be a useful resource to elucidate the mechanisms controlling the effects of the integrated stress response. Additional putative roles of the ATF4 protein in cell division and synaptic plasticity are outlined.
Collapse
Affiliation(s)
- Graham Neill
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | | |
Collapse
|
12
|
GCN4 Enhances the Transcriptional Regulation of AreA by Interacting with SKO1 To Mediate Nitrogen Utilization in Ganoderma lucidum. Appl Environ Microbiol 2022; 88:e0132222. [PMID: 36342130 PMCID: PMC9680636 DOI: 10.1128/aem.01322-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Nitrogen is an essential nutrient for cell growth and proliferation. Limitations of nitrogen availability in organisms elicit a series of rapid transcriptional reprogramming mechanisms, which involve the participation of many transcription factors.
Collapse
|
13
|
Chen Y, He R, Han Z, Wu Y, Wang Q, Zhu X, Huang Z, Ye J, Tang Y, Huang H, Chen J, Shan H, Xiao F. Cooperation of ATF4 and CTCF promotes adipogenesis through transcriptional regulation. Cell Biol Toxicol 2022; 38:741-763. [PMID: 33950334 DOI: 10.1007/s10565-021-09608-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Adipogenesis is a multi-step process orchestrated by activation of numerous TFs, whose cooperation and regulatory network remain elusive. Activating transcription factor 4 (ATF4) is critical for adipogenesis, yet its regulatory network is unclarified. Here, we mapped genome-wide ATF4 binding landscape and its regulatory network by Chip-seq and RNA-seq and found ATF4 directly modulated transcription of genes enriching in fat cell differentiation. Motifs of TFs especially CTCF were found from ATF4 binding sites, suggesting a direct role of ATF4 in regulating adipogenesis associated with CTCF and other TFs. Deletion of CTCF attenuated adipogenesis while overexpression enhanced adipocyte differentiation, indicating CTCF is indispensable for adipogenesis. Intriguingly, combined analysis of Chip-seq data of these two TFs showed that ATF4 co-localized with CTCF in the promoters of key adipogenic genes including Cebpd and PPARg and co-regulated their transactivation. Moreover, ATF4 directly regulated CTCF expression and interacted with CTCF in differentiated 3T3-L1 cells. In vivo, downregulation of ATF4 suppressed the expression of CTCF, Cebpd, and PPARg, leading to reduced adipose tissue expansion in refeeding mice. Consistently, mRNA expression of ATF4 and CTCF was positively correlated with each other in human subcutaneous adipose tissue and inversely associated with BMI, indicating a possible involvement of these two TFs in adipose development. Taken together, our data propose for the first time that ATF4 and CTCF work cooperatively to control adipogenesis and adipose development via orchestrating transcription of adipogenic genes. Our findings reveal novel therapeutic targets in obesity treatment.
Collapse
Affiliation(s)
- Yingchun Chen
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Rongquan He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Zhiqiang Han
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Yanyan Wu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Xiujuan Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Zhiguang Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Juan Ye
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Yao Tang
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Hongbin Huang
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Jianxu Chen
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China.
| | - Fei Xiao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China.
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China.
| |
Collapse
|
14
|
Kim D, Kim J, Yu YS, Kim YR, Baek SH, Won KJ. Systemic approaches using single cell transcriptome reveal that C/EBPγ regulates autophagy under amino acid starved condition. Nucleic Acids Res 2022; 50:7298-7309. [PMID: 35801910 PMCID: PMC9303372 DOI: 10.1093/nar/gkac593] [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: 12/22/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/14/2022] Open
Abstract
Autophagy, a catabolic process to remove unnecessary or dysfunctional organelles, is triggered by various signals including nutrient starvation. Depending on the types of the nutrient deficiency, diverse sensing mechanisms and signaling pathways orchestrate for transcriptional and epigenetic regulation of autophagy. However, our knowledge about nutrient type-specific transcriptional regulation during autophagy is limited. To understand nutrient type-dependent transcriptional mechanisms during autophagy, we performed single cell RNA sequencing (scRNAseq) in the mouse embryonic fibroblasts (MEFs) with or without glucose starvation (GS) as well as amino acid starvation (AAS). Trajectory analysis using scRNAseq identified sequential induction of potential transcriptional regulators for each condition. Gene regulatory rules inferred using TENET newly identified CCAAT/enhancer binding protein γ (C/EBPγ) as a regulator of autophagy in AAS, but not GS, condition, and knockdown experiment confirmed the TENET result. Cell biological and biochemical studies validated that activating transcription factor 4 (ATF4) is responsible for conferring specificity to C/EBPγ for the activation of autophagy genes under AAS, but not under GS condition. Together, our data identified C/EBPγ as a previously unidentified key regulator under AAS-induced autophagy.
Collapse
Affiliation(s)
- Dongha Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.,Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junil Kim
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.,School of Systems Biomedical Science, Soongsil University, 369 Sangdo-Ro, Dongjak-Gu, Seoul 06978, Republic of Korea
| | - Young Suk Yu
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong Ryoul Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung-Jae Won
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
15
|
THE INTEGRATED STRESS RESPONSE AS A KEY PATHWAY DOWNSTREAM OF MITOCHONDRIAL DYSFUNCTION. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Naqvi I, Giroux N, Olson L, Morrison SA, Llanga T, Akinade TO, Zhu Y, Zhong Y, Bose S, Arvai S, Abramson K, Chen L, Que L, Kraft B, Shen X, Lee J, Leong KW, Nair SK, Sullenger B. DAMPs/PAMPs induce monocytic TLR activation and tolerance in COVID-19 patients; nucleic acid binding scavengers can counteract such TLR agonists. Biomaterials 2022; 283:121393. [PMID: 35349874 PMCID: PMC8797062 DOI: 10.1016/j.biomaterials.2022.121393] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022]
Abstract
Millions of COVID-19 patients have succumbed to respiratory and systemic inflammation. Hyperstimulation of toll-like receptor (TLR) signaling is a key driver of immunopathology following infection by viruses. We found that severely ill COVID-19 patients in the Intensive Care Unit (ICU) display hallmarks of such hyper-stimulation with abundant agonists of nucleic acid-sensing TLRs present in their blood and lungs. These nucleic acid-containing Damage and Pathogen Associated Molecular Patterns (DAMPs/PAMPs) can be depleted using nucleic acid-binding microfibers to limit the patient samples' ability to hyperactivate such innate immune receptors. Single-cell RNA-sequencing revealed that CD16+ monocytes from deceased but not recovered ICU patients exhibit a TLR-tolerant phenotype and a deficient anti-viral response after ex vivo TLR stimulation. Plasma proteomics confirmed such myeloid hyperactivation and revealed DAMP/PAMP carrier consumption in deceased patients. Treatment of these COVID-19 patient samples with MnO nanoparticles effectively neutralizes TLR activation by the abundant nucleic acid-containing DAMPs/PAMPs present in their lungs and blood. Finally, MnO nanoscavenger treatment limits the ability of DAMPs/PAMPs to induce TLR tolerance in monocytes. Thus, treatment with microfiber- or nanoparticle-based DAMP/PAMP scavengers may prove useful for limiting SARS-CoV-2 induced hyperinflammation, preventing monocytic TLR tolerance, and improving outcomes in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Ibtehaj Naqvi
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | - Nicholas Giroux
- Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA; Duke University, Graduate School, USA
| | - Lyra Olson
- Duke University, Graduate School, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA
| | - Sarah Ahn Morrison
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | | | - Tolu O Akinade
- Columbia University, Department of Biomedical Engineering, USA
| | - Yuefei Zhu
- Columbia University, Department of Biomedical Engineering, USA
| | - Yiling Zhong
- Columbia University, Department of Biomedical Engineering, USA
| | - Shree Bose
- Duke University, Graduate School, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA
| | - Stephanie Arvai
- Duke University Center for Genomic and Computational Biology, RNA Sequencing Core, USA
| | - Karen Abramson
- Duke University Center for Genomic and Computational Biology, RNA Sequencing Core, USA
| | - Lingye Chen
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Loretta Que
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Bryan Kraft
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Xiling Shen
- Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA
| | - Jaewoo Lee
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | - Kam W Leong
- Columbia University, Department of Biomedical Engineering, USA
| | - Smita K Nair
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA; Duke University School of Medicine, Department of Pathology, USA; Duke University School of Medicine, Department of Neurosurgery, USA.
| | - Bruce Sullenger
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA; Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA; Duke University School of Medicine, Department of Neurosurgery, USA.
| |
Collapse
|
17
|
Ebert SM, Rasmussen BB, Judge AR, Judge SM, Larsson L, Wek RC, Anthony TG, Marcotte GR, Miller MJ, Yorek MA, Vella A, Volpi E, Stern JI, Strub MD, Ryan Z, Talley JJ, Adams CM. Biology of Activating Transcription Factor 4 (ATF4) and Its Role in Skeletal Muscle Atrophy. J Nutr 2022; 152:926-938. [PMID: 34958390 PMCID: PMC8970988 DOI: 10.1093/jn/nxab440] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
Activating transcription factor 4 (ATF4) is a multifunctional transcription regulatory protein in the basic leucine zipper superfamily. ATF4 can be expressed in most if not all mammalian cell types, and it can participate in a variety of cellular responses to specific environmental stresses, intracellular derangements, or growth factors. Because ATF4 is involved in a wide range of biological processes, its roles in human health and disease are not yet fully understood. Much of our current knowledge about ATF4 comes from investigations in cultured cell models, where ATF4 was originally characterized and where further investigations continue to provide new insights. ATF4 is also an increasingly prominent topic of in vivo investigations in fully differentiated mammalian cell types, where our current understanding of ATF4 is less complete. Here, we review some important high-level concepts and questions concerning the basic biology of ATF4. We then discuss current knowledge and emerging questions about the in vivo role of ATF4 in one fully differentiated cell type, mammalian skeletal muscle fibers.
Collapse
Affiliation(s)
- Scott M Ebert
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Emmyon, Inc., Rochester, MN, USA
| | - Blake B Rasmussen
- Emmyon, Inc., Rochester, MN, USA
- Department of Nutrition, Metabolism and Rehabilitation Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Andrew R Judge
- Emmyon, Inc., Rochester, MN, USA
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska, Stockholm, Sweden
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - George R Marcotte
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Matthew J Miller
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Mark A Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
- Department of Internal Medicine, Iowa City VA Medical Center, Iowa City, IA, USA
| | - Adrian Vella
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Emmyon, Inc., Rochester, MN, USA
| | - Elena Volpi
- Department of Nutrition, Metabolism and Rehabilitation Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennifer I Stern
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Matthew D Strub
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Zachary Ryan
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | | | - Christopher M Adams
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Emmyon, Inc., Rochester, MN, USA
- Department of Internal Medicine, Iowa City VA Medical Center, Iowa City, IA, USA
| |
Collapse
|
18
|
Dziadowicz SA, Wang L, Akhter H, Aesoph D, Sharma T, Adjeroh DA, Hazlehurst LA, Hu G. Bone Marrow Stroma-Induced Transcriptome and Regulome Signatures of Multiple Myeloma. Cancers (Basel) 2022; 14:927. [PMID: 35205675 PMCID: PMC8870223 DOI: 10.3390/cancers14040927] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Multiple myeloma (MM) is a hematological cancer with inevitable drug resistance. MM cells interacting with bone marrow stromal cells (BMSCs) undergo substantial changes in the transcriptome and develop de novo multi-drug resistance. As a critical component in transcriptional regulation, how the chromatin landscape is transformed in MM cells exposed to BMSCs and contributes to the transcriptional response to BMSCs remains elusive. We profiled the transcriptome and regulome for MM cells using a transwell coculture system with BMSCs. The transcriptome and regulome of MM cells from the upper transwell resembled MM cells that coexisted with BMSCs from the lower chamber but were distinctive to monoculture. BMSC-induced genes were enriched in the JAK2/STAT3 signaling pathway, unfolded protein stress, signatures of early plasma cells, and response to proteasome inhibitors. Genes with increasing accessibility at multiple regulatory sites were preferentially induced by BMSCs; these genes were enriched in functions linked to responses to drugs and unfavorable clinic outcomes. We proposed JUNB and ATF4::CEBPβ as candidate transcription factors (TFs) that modulate the BMSC-induced transformation of the regulome linked to the transcriptional response. Together, we characterized the BMSC-induced transcriptome and regulome signatures of MM cells to facilitate research on epigenetic mechanisms of BMSC-induced multi-drug resistance in MM.
Collapse
Affiliation(s)
- Sebastian A. Dziadowicz
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26505, USA; (S.A.D.); (L.W.); (H.A.); (D.A.); (T.S.)
| | - Lei Wang
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26505, USA; (S.A.D.); (L.W.); (H.A.); (D.A.); (T.S.)
| | - Halima Akhter
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26505, USA; (S.A.D.); (L.W.); (H.A.); (D.A.); (T.S.)
- Lane Department of Computer Science & Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| | - Drake Aesoph
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26505, USA; (S.A.D.); (L.W.); (H.A.); (D.A.); (T.S.)
- Lane Department of Computer Science & Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| | - Tulika Sharma
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26505, USA; (S.A.D.); (L.W.); (H.A.); (D.A.); (T.S.)
| | - Donald A. Adjeroh
- Lane Department of Computer Science & Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| | - Lori A. Hazlehurst
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA;
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morganton, WV 26506, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26505, USA; (S.A.D.); (L.W.); (H.A.); (D.A.); (T.S.)
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
19
|
Orchard P, Manickam N, Ventresca C, Vadlamudi S, Varshney A, Rai V, Kaplan J, Lalancette C, Mohlke KL, Gallagher K, Burant CF, Parker SCJ. Human and rat skeletal muscle single-nuclei multi-omic integrative analyses nominate causal cell types, regulatory elements, and SNPs for complex traits. Genome Res 2021; 31:2258-2275. [PMID: 34815310 PMCID: PMC8647829 DOI: 10.1101/gr.268482.120] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/16/2021] [Indexed: 12/12/2022]
Abstract
Skeletal muscle accounts for the largest proportion of human body mass, on average, and is a key tissue in complex diseases and mobility. It is composed of several different cell and muscle fiber types. Here, we optimize single-nucleus ATAC-seq (snATAC-seq) to map skeletal muscle cell-specific chromatin accessibility landscapes in frozen human and rat samples, and single-nucleus RNA-seq (snRNA-seq) to map cell-specific transcriptomes in human. We additionally perform multi-omics profiling (gene expression and chromatin accessibility) on human and rat muscle samples. We capture type I and type II muscle fiber signatures, which are generally missed by existing single-cell RNA-seq methods. We perform cross-modality and cross-species integrative analyses on 33,862 nuclei and identify seven cell types ranging in abundance from 59.6% to 1.0% of all nuclei. We introduce a regression-based approach to infer cell types by comparing transcription start site-distal ATAC-seq peaks to reference enhancer maps and show consistency with RNA-based marker gene cell type assignments. We find heterogeneity in enrichment of genetic variants linked to complex phenotypes from the UK Biobank and diabetes genome-wide association studies in cell-specific ATAC-seq peaks, with the most striking enrichment patterns in muscle mesenchymal stem cells (∼3.5% of nuclei). Finally, we overlay these chromatin accessibility maps on GWAS data to nominate causal cell types, SNPs, transcription factor motifs, and target genes for type 2 diabetes signals. These chromatin accessibility profiles for human and rat skeletal muscle cell types are a useful resource for nominating causal GWAS SNPs and cell types.
Collapse
Affiliation(s)
- Peter Orchard
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Nandini Manickam
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Christa Ventresca
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Swarooparani Vadlamudi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Arushi Varshney
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Vivek Rai
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jeremy Kaplan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Claudia Lalancette
- Epigenomics Core, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Katherine Gallagher
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
20
|
Lin T, Zhang Y, Zhang T, Steckler RA, Yang X. Hop2 interacts with the transcription factor CEBPα and suppresses adipocyte differentiation. J Biol Chem 2021; 297:101264. [PMID: 34600885 PMCID: PMC8528721 DOI: 10.1016/j.jbc.2021.101264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/09/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
CCAAT enhancer binding protein (CEBP) transcription factors (TFs) are known to promote adipocyte differentiation; however, suppressors of CEBP TFs have not been reported thus far. Here, we find that homologous chromosome pairing protein 2 (Hop2) functions as an inhibitor for the TF CEBPα. We found that Hop2 mRNA is highly and specifically expressed in adipose tissue, and that ectopic Hop2 expression suppresses reporter activity induced by CEBP as revealed by DNA transfection. Recombinant and ectopically expressed Hop2 was shown to interact with CEBPα in pull-down and coimmunoprecipitation assays, and interaction between endogenous Hop2 and CEBPα was observed in the nuclei of 3T3 preadipocytes and adipocytes by immunofluorescence and coimmunoprecipitation of nuclear extracts. In addition, Hop2 stable overexpression in 3T3 preadipocytes inhibited adipocyte differentiation and adipocyte marker gene expression. These in vitro data suggest that Hop2 inhibits adipogenesis by suppressing CEBP-mediated transactivation. Consistent with a negative role for Hop2 in adipogenesis, ablation of Hop2 (Hop2-/-) in mice led to increased body weight, adipose volume, adipocyte size, and adipogenic marker gene expression. Adipogenic differentiation of isolated adipose-derived mesenchymal stem cells showed a greater number of lipid droplet-containing colonies formed in Hop2-/- adipose-derived mesenchymal stem cell cultures than in wt controls, which is associated with the increased expression of adipogenic marker genes. Finally, chromatin immunoprecipitation revealed a higher binding activity of endogenous CEBPα to peroxisome proliferator-activated receptor γ, a master adipogenic TF, and a known CEBPα target gene. Therefore, our study identifies for the first time that Hop2 is an intrinsic suppressor of CEBPα and thus adipogenesis in adipocytes.
Collapse
Affiliation(s)
- Tonghui Lin
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Yang Zhang
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Tingting Zhang
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Rita A Steckler
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Xiangli Yang
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA.
| |
Collapse
|
21
|
Vildanova M, Vishnyakova P, Saidova A, Konduktorova V, Onishchenko G, Smirnova E. Gibberellic Acid Initiates ER Stress and Activation of Differentiation in Cultured Human Immortalized Keratinocytes HaCaT and Epidermoid Carcinoma Cells A431. Pharmaceutics 2021; 13:pharmaceutics13111813. [PMID: 34834228 PMCID: PMC8622727 DOI: 10.3390/pharmaceutics13111813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/30/2022] Open
Abstract
Diterpenoid plant hormone gibberellic acid (GA) plays an important role in regulation of plant growth and development and is commonly used in agriculture for activation of plant growth and food production. It is known that many plant-derived compounds have miscellaneous biological effects on animals and humans, influencing specific cellular functions and metabolic pathways. However, the effect of GA on animal and human cells remains controversial. We investigated the effect of GA on cultured human cell lines of epidermoid origin-immortalized non-tumorigenic keratinocytes HaCaT and carcinoma A431 cells. We found that at a non-toxic dose, GA upregulated the expression of genes associated with the ER stress response-CHOP, sXBP1, GRP87 in both cell lines, and ATF4 predominantly in A431 cells. We also showed that GA was more effective in upregulating the production of ER stress marker GRP78, autophagy marker LC3B-II, and differentiation markers involucrin and filaggrin in A431 cells than in HaCaT. We conclude that GA induces mild ER stress in both cell lines, followed by the activation of differentiation via upregulation of autophagy. However, in comparison with immortalized keratinocytes HaCaT, GA is more effective in inducing differentiation of carcinoma A431 cells, probably due to the inherently lower differentiation status of A431 cells. The activation of differentiation in poorly differentiated and highly malignant A431 cells by GA may lower the level of malignancy of these cells and decrease their tumorigenic potential.
Collapse
Affiliation(s)
- Mariya Vildanova
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119234 Moscow, Russia; (A.S.); (V.K.); (G.O.); (E.S.)
- Correspondence: or
| | - Polina Vishnyakova
- National Medical Research Center for Obstetrics, Laboratory of Regenerative Medicine, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia;
- Histology Department, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
| | - Aleena Saidova
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119234 Moscow, Russia; (A.S.); (V.K.); (G.O.); (E.S.)
| | - Victoria Konduktorova
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119234 Moscow, Russia; (A.S.); (V.K.); (G.O.); (E.S.)
| | - Galina Onishchenko
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119234 Moscow, Russia; (A.S.); (V.K.); (G.O.); (E.S.)
| | - Elena Smirnova
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 1, Bld. 12, 119234 Moscow, Russia; (A.S.); (V.K.); (G.O.); (E.S.)
| |
Collapse
|
22
|
Giroud M, Tsokanos FF, Caratti G, Kotschi S, Khani S, Jouffe C, Vogl ES, Irmler M, Glantschnig C, Gil-Lozano M, Hass D, Khan AA, Garcia MR, Mattijssen F, Maida A, Tews D, Fischer-Posovszky P, Feuchtinger A, Virtanen KA, Beckers J, Wabitsch M, Uhlenhaut H, Blüher M, Tuckermann J, Scheideler M, Bartelt A, Herzig S. HAND2 is a novel obesity-linked adipogenic transcription factor regulated by glucocorticoid signalling. Diabetologia 2021; 64:1850-1865. [PMID: 34014371 PMCID: PMC8245394 DOI: 10.1007/s00125-021-05470-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/18/2021] [Indexed: 11/01/2022]
Abstract
AIMS/HYPOTHESIS Adipocytes are critical cornerstones of energy metabolism. While obesity-induced adipocyte dysfunction is associated with insulin resistance and systemic metabolic disturbances, adipogenesis, the formation of new adipocytes and healthy adipose tissue expansion are associated with metabolic benefits. Understanding the molecular mechanisms governing adipogenesis is of great clinical potential to efficiently restore metabolic health in obesity. Here we investigate the role of heart and neural crest derivatives-expressed 2 (HAND2) in adipogenesis. METHODS Human white adipose tissue (WAT) was collected from two cross-sectional studies of 318 and 96 individuals. In vitro, for mechanistic experiments we used primary adipocytes from humans and mice as well as human multipotent adipose-derived stem (hMADS) cells. Gene silencing was performed using siRNA or genetic inactivation in primary adipocytes from loxP and or tamoxifen-inducible Cre-ERT2 mouse models with Cre-encoding mRNA or tamoxifen, respectively. Adipogenesis and adipocyte metabolism were measured by Oil Red O staining, quantitative PCR (qPCR), microarray, glucose uptake assay, western blot and lipolysis assay. A combinatorial RNA sequencing (RNAseq) and ChIP qPCR approach was used to identify target genes regulated by HAND2. In vivo, we created a conditional adipocyte Hand2 deletion mouse model using Cre under control of the Adipoq promoter (Hand2AdipoqCre) and performed a large panel of metabolic tests. RESULTS We found that HAND2 is an obesity-linked white adipocyte transcription factor regulated by glucocorticoids that was necessary but insufficient for adipocyte differentiation in vitro. In a large cohort of humans, WAT HAND2 expression was correlated to BMI. The HAND2 gene was enriched in white adipocytes compared with brown, induced early in differentiation and responded to dexamethasone (DEX), a typical glucocorticoid receptor (GR, encoded by NR3C1) agonist. Silencing of NR3C1 in hMADS cells or deletion of GR in a transgenic conditional mouse model results in diminished HAND2 expression, establishing that adipocyte HAND2 is regulated by glucocorticoids via GR in vitro and in vivo. Furthermore, we identified gene clusters indirectly regulated by the GR-HAND2 pathway. Interestingly, silencing of HAND2 impaired adipocyte differentiation in hMADS and primary mouse adipocytes. However, a conditional adipocyte Hand2 deletion mouse model using Cre under control of the Adipoq promoter did not mirror these effects on adipose tissue differentiation, indicating that HAND2 was required at stages prior to Adipoq expression. CONCLUSIONS/INTERPRETATION In summary, our study identifies HAND2 as a novel obesity-linked adipocyte transcription factor, highlighting new mechanisms of GR-dependent adipogenesis in humans and mice. DATA AVAILABILITY Array data have been submitted to the GEO database at NCBI (GSE148699).
Collapse
Affiliation(s)
- Maude Giroud
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Foivos-Filippos Tsokanos
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Giorgio Caratti
- Institute for Comparative Molecular Endocrinology, Universität Ulm, Ulm, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Sajjad Khani
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Céline Jouffe
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Elena S Vogl
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christina Glantschnig
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Manuel Gil-Lozano
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniela Hass
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Asrar Ali Khan
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcos Rios Garcia
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Frits Mattijssen
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Adriano Maida
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Tews
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, Neuherberg, Germany
| | | | - Johannes Beckers
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Henriette Uhlenhaut
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Metabolic Programming, TUM School of Life Sciences Weihenstephan and ZIEL Institute for Food & Health, Munich, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Jan Tuckermann
- Institute for Comparative Molecular Endocrinology, Universität Ulm, Ulm, Germany
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Bartelt
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC); Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.
- Molecular Metabolic Control, Medical Faculty, Technical University Munich, Munich, Germany.
| |
Collapse
|
23
|
Torrence ME, MacArthur MR, Hosios AM, Valvezan AJ, Asara JM, Mitchell JR, Manning BD. The mTORC1-mediated activation of ATF4 promotes protein and glutathione synthesis downstream of growth signals. eLife 2021; 10:e63326. [PMID: 33646118 PMCID: PMC7997658 DOI: 10.7554/elife.63326] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) stimulates a coordinated anabolic program in response to growth-promoting signals. Paradoxically, recent studies indicate that mTORC1 can activate the transcription factor ATF4 through mechanisms distinct from its canonical induction by the integrated stress response (ISR). However, its broader roles as a downstream target of mTORC1 are unknown. Therefore, we directly compared ATF4-dependent transcriptional changes induced upon insulin-stimulated mTORC1 signaling to those activated by the ISR. In multiple mouse embryo fibroblast and human cancer cell lines, the mTORC1-ATF4 pathway stimulated expression of only a subset of the ATF4 target genes induced by the ISR, including genes involved in amino acid uptake, synthesis, and tRNA charging. We demonstrate that ATF4 is a metabolic effector of mTORC1 involved in both its established role in promoting protein synthesis and in a previously unappreciated function for mTORC1 in stimulating cellular cystine uptake and glutathione synthesis.
Collapse
Affiliation(s)
- Margaret E Torrence
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public HealthBostonUnited States
| | - Michael R MacArthur
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public HealthBostonUnited States
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Aaron M Hosios
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public HealthBostonUnited States
| | - Alexander J Valvezan
- Center for Advanced Biotechnology and Medicine, Department of Pharmacology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical SchoolBostonUnited States
| | - James R Mitchell
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public HealthBostonUnited States
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Brendan D Manning
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public HealthBostonUnited States
| |
Collapse
|
24
|
Ronzio M, Bernardini A, Pavesi G, Mantovani R, Dolfini D. On the NF-Y regulome as in ENCODE (2019). PLoS Comput Biol 2020; 16:e1008488. [PMID: 33370256 PMCID: PMC7793273 DOI: 10.1371/journal.pcbi.1008488] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/08/2021] [Accepted: 11/04/2020] [Indexed: 11/19/2022] Open
Abstract
NF-Y is a trimeric Transcription Factor -TF- which binds with high selectivity to the conserved CCAAT element. Individual ChIP-seq analysis as well as ENCODE have progressively identified locations shared by other TFs. Here, we have analyzed data introduced by ENCODE over the last five years in K562, HeLa-S3 and GM12878, including several chromatin features, as well RNA-seq profiling of HeLa cells after NF-Y inactivation. We double the number of sequence-specific TFs and co-factors reported. We catalogue them in 4 classes based on co-association criteria, infer target genes categorizations, identify positional bias of binding sites and gene expression changes. Larger and novel co-associations emerge, specifically concerning subunits of repressive complexes as well as RNA-binding proteins. On the one hand, these data better define NF-Y association with single members of major classes of TFs, on the other, they suggest that it might have a wider role in the control of mRNA production. The ongoing ENCODE consortium represents a useful compendium of locations of TFs, chromatin marks, gene expression data. In previous reports, we identified modules of CCAAT-binding NF-Y with individual TFs. Here, we analyzed all 363 factors currently present: 68 with enrichment of CCAAT in their locations, 38 with overlap of peaks. New sequence-specific TFs, co-activators and co-repressors are reported. Co-association patterns correspond to specific targeted genes categorizations and gene expression changes, as assessed by RNA-seq after NF-Y inactivation. These data widen and better define a coherent model of synergy of NF-Y with selected groups of TFs and co-factors.
Collapse
Affiliation(s)
- Mirko Ronzio
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Andrea Bernardini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Giulio Pavesi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
- * E-mail:
| |
Collapse
|
25
|
Xu Z, Zhou Y, Nong Q, You W, Wang L, Wang Y, Shan T. LKB1 Differently Regulates Adipogenesis in Intramuscular and Subcutaneous Adipocytes through Metabolic and Cytokine-Related Signaling Pathways. Cells 2020; 9:cells9122599. [PMID: 33291665 PMCID: PMC7761942 DOI: 10.3390/cells9122599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Liver kinase B1 (LKB1) plays important and various roles in the differentiation and lipid metabolism of adipocytes. However, the current knowledge of the respective roles of LKB1 in subcutaneous fat (SCF) and intramuscular fat (IMF) adipocytes remains unclear. This study aimed to discover the different regulatory mechanisms of LKB1 in SCF and IMF adipocytes. We found that LKB1 overexpression inhibited adipogenesis in both SCF and IMF adipocytes, and SCF adipocytes were more sensitive to regulation by LKB1. Transcriptomics results showed that IMF adipocytes had many more differentially expressed genes (DEGs) than SCF adipocytes. Pathway analysis of the shared and distinct DEGs revealed that the main adipogenesis mechanism was similar between SCF and IMF adipocytes upon LKB1 overexpression, while regulatory and metabolic signaling pathways, such as MAPK, PPAR signaling pathways, were differently regulated by LKB1. Several cytokine-related pathways were only enriched in LKB1-overexpressing IMF adipocytes. Our study reveals different regulators and signaling pathways between SCF and IMF adipocytes under LKB1 overexpression, which may be potential targets to differentially control SCF and IMF deposition and improve our understanding of the regulatory mechanisms of IMF deposition.
Collapse
Affiliation(s)
- Ziye Xu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Yanbing Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Qiuyun Nong
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Z.X.); (Y.Z.); (Q.N.); (W.Y.); (L.W.); (Y.W.)
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Correspondence: ; Tel.: +86-0571-8898-2102
| |
Collapse
|
26
|
MLL3/MLL4-Associated PAGR1 Regulates Adipogenesis by Controlling Induction of C/EBPβ and C/EBPδ. Mol Cell Biol 2020; 40:MCB.00209-20. [PMID: 32601106 DOI: 10.1128/mcb.00209-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/19/2020] [Indexed: 01/12/2023] Open
Abstract
Transcription factors C/EBPβ and C/EBPδ are induced within hours after initiation of adipogenesis in culture. They directly promote the expression of master adipogenic transcription factors peroxisome proliferator-activated receptor γ (PPARγ) and C/EBPα and are required for adipogenesis in vivo However, the mechanism that controls the induction of C/EBPβ and C/EBPδ remains elusive. We previously showed that histone methyltransferases MLL3/MLL4 and associated PTIP are required for the induction of PPARγ and C/EBPα during adipogenesis. Here, we show MLL3/MLL4/PTIP-associated protein PAGR1 (also known as PA1) cooperates with phosphorylated CREB and ligand-activated glucocorticoid receptor to directly control the induction of C/EBPβ and C/EBPδ in the early phase of adipogenesis. Deletion of Pagr1 in white and brown preadipocytes prevents the induction of C/EBPβ and C/EBPδ and leads to severe defects in adipogenesis. Adipogenesis defects in PAGR1-deficient cells can be rescued by the ectopic expression of C/EBPβ or PPARγ. Finally, the deletion of Pagr1 in Myf5+ precursor cells impairs brown adipose tissue and muscle development. Thus, by controlling the induction of C/EBPβ and C/EBPδ, PAGR1 plays a critical role in adipogenesis.
Collapse
|
27
|
Chen Y, Wang Y, Lin W, Sheng R, Wu Y, Xu R, Zhou C, Yuan Q. AFF1 inhibits adipogenic differentiation via targeting TGM2 transcription. Cell Prolif 2020; 53:e12831. [PMID: 32441391 PMCID: PMC7309944 DOI: 10.1111/cpr.12831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/16/2020] [Accepted: 04/28/2020] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES AF4/FMR2 family member 1 (AFF1), known as a central scaffolding protein of super elongation complex (SEC), regulates gene transcription. We previously reported that AFF1 inhibited osteogenic differentiation of human mesenchymal stromal/stem cells (hMSCs). However, its role in adipogenic differentiation has not been elucidated. MATERIALS AND METHODS hMSCs and 3T3-L1 pre-adipocytes were cultured and induced for adipogenic differentiation. Small interfering RNAs (siRNAs) were applied to deplete AFF1 while lentiviruses expressing HA-Aff1 were used for overexpression. Oil Red O staining, triglyceride (TAG) quantification, quantitative real-time PCR (qPCR), Western blot analysis, immunofluorescence staining, RNA sequencing (RNA-seq) analysis and ChIP-qPCR were performed. To evaluate the adipogenesis in vivo, BALB/c nude mice were subcutaneously injected with Aff1-overexpressed 3T3-L1 pre-adipocytes. RESULTS AFF1 depletion leads to an enhanced adipogenesis in both hMSCs and 3T3-L1 pre-adipocytes. Overexpression of Aff1 in 3T3-L1 cells results in the reduction of adipogenic differentiation and less adipose tissue formation in vivo. Mechanistically, AFF1 binds to the promoter region of Tgm2 gene and regulates its transcription. Overexpression of Tgm2 largely rescues adipogenic differentiation of Aff1-deficient cells. CONCLUSIONS Our data indicate that AFF1 inhibits adipogenic differentiation by regulating the transcription of TGM2.
Collapse
Affiliation(s)
- Yaqian Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Sheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunshu Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ruoshi Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Ebert SM, Bullard SA, Basisty N, Marcotte GR, Skopec ZP, Dierdorff JM, Al-Zougbi A, Tomcheck KC, DeLau AD, Rathmacher JA, Bodine SC, Schilling B, Adams CM. Activating transcription factor 4 (ATF4) promotes skeletal muscle atrophy by forming a heterodimer with the transcriptional regulator C/EBPβ. J Biol Chem 2020; 295:2787-2803. [PMID: 31953319 PMCID: PMC7049960 DOI: 10.1074/jbc.ra119.012095] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/10/2020] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle atrophy is a highly-prevalent and debilitating condition that remains poorly understood at the molecular level. Previous work found that aging, fasting, and immobilization promote skeletal muscle atrophy via expression of activating transcription factor 4 (ATF4) in skeletal muscle fibers. However, the direct biochemical mechanism by which ATF4 promotes muscle atrophy is unknown. ATF4 is a member of the basic leucine zipper transcription factor (bZIP) superfamily. Because bZIP transcription factors are obligate dimers, and because ATF4 is unable to form highly-stable homodimers, we hypothesized that ATF4 may promote muscle atrophy by forming a heterodimer with another bZIP family member. To test this hypothesis, we biochemically isolated skeletal muscle proteins that associate with the dimerization- and DNA-binding domain of ATF4 (the bZIP domain) in mouse skeletal muscle fibers in vivo Interestingly, we found that ATF4 forms at least five distinct heterodimeric bZIP transcription factors in skeletal muscle fibers. Furthermore, one of these heterodimers, composed of ATF4 and CCAAT enhancer-binding protein β (C/EBPβ), mediates muscle atrophy. Within skeletal muscle fibers, the ATF4-C/EBPβ heterodimer interacts with a previously unrecognized and evolutionarily conserved ATF-C/EBP composite site in exon 4 of the Gadd45a gene. This three-way interaction between ATF4, C/EBPβ, and the ATF-C/EBP composite site activates the Gadd45a gene, which encodes a critical mediator of muscle atrophy. Together, these results identify a biochemical mechanism by which ATF4 induces skeletal muscle atrophy, providing molecular-level insights into the etiology of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Scott M Ebert
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246; Emmyon, Inc., Coralville, Iowa 52241
| | - Steven A Bullard
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Nathan Basisty
- Buck Institute for Research on Aging, Novato, California 94945
| | - George R Marcotte
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Zachary P Skopec
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Jason M Dierdorff
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Asma Al-Zougbi
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Kristin C Tomcheck
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Austin D DeLau
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Jacob A Rathmacher
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Sue C Bodine
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Emmyon, Inc., Coralville, Iowa 52241
| | | | - Christopher M Adams
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246; Emmyon, Inc., Coralville, Iowa 52241.
| |
Collapse
|
29
|
Abstract
The 3T3-L1 pre-adipocyte cell line is widely used to study the fat cell differentiation in vitro. Researchers also use this cell model to study obesity and insulin resistance. We surveyed the literature, the gene expression omnibus and the sequence read archive for RNA-Seq and ChIP-Seq datasets of MDI-induced 3T3-L1 differentiating cells sampled at one or more time points. The metadata of the relevant datasets were manually curated using unified language across the original studies. The raw reads were collected and pre-processed using a reproducible state-of-the-art pipeline. The final datasets are presented as reads count in genes for the RNA-Seq and reads count in peaks for the ChIP-Seq dataset. The curated datasets are available as two Bioconductor experimental data packages curatedAdipoRNA and curatedAdipoChIP. In addition, the packages document the source code of the data collection and the pre-processing pipelines. Here, we provide a descriptive analysis of the datasets with context and technical validation.
Collapse
Affiliation(s)
- Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| |
Collapse
|
30
|
Pällmann N, Livgård M, Tesikova M, Zeynep Nenseth H, Akkus E, Sikkeland J, Jin Y, Koc D, Kuzu OF, Pradhan M, Danielsen HE, Kahraman N, Mokhlis HM, Ozpolat B, Banerjee PP, Uren A, Fazli L, Rennie PS, Jin Y, Saatcioglu F. Regulation of the unfolded protein response through ATF4 and FAM129A in prostate cancer. Oncogene 2019; 38:6301-6318. [DOI: 10.1038/s41388-019-0879-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/31/2019] [Accepted: 04/10/2019] [Indexed: 12/26/2022]
|
31
|
Cohen DM, Lim HW, Won KJ, Steger DJ. Shared nucleotide flanks confer transcriptional competency to bZip core motifs. Nucleic Acids Res 2019; 46:8371-8384. [PMID: 30085281 PMCID: PMC6144830 DOI: 10.1093/nar/gky681] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/17/2018] [Indexed: 12/31/2022] Open
Abstract
Sequence-specific DNA binding recruits transcription factors (TFs) to the genome to regulate gene expression. Here, we perform high resolution mapping of CEBP proteins to determine how sequence dictates genomic occupancy. We demonstrate a fundamental difference between the sequence repertoire utilized by CEBPs in vivo versus the palindromic sequence preference reported by classical in vitro models, by identifying a palindromic motif at <1% of the genomic binding sites. On the native genome, CEBPs bind a diversity of related 10 bp sequences resulting from the fusion of degenerate and canonical half-sites. Altered DNA specificity of CEBPs in cells occurs through heterodimerization with other bZip TFs, and approximately 40% of CEBP-binding sites in primary human cells harbor motifs characteristic of CEBP heterodimers. In addition, we uncover an important role for sequence bias at core-motif-flanking bases for CEBPs and demonstrate that flanking bases regulate motif function across mammalian bZip TFs. Favorable flanking bases confer efficient TF occupancy and transcriptional activity, and DNA shape may explain how the flanks alter TF binding. Importantly, motif optimization within the 10-mer is strongly correlated with cell-type-independent recruitment of CEBPβ, providing key insight into how sequence sub-optimization affects genomic occupancy of widely expressed CEBPs across cell types.
Collapse
Affiliation(s)
- Daniel M Cohen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hee-Woong Lim
- The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyoung-Jae Won
- The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - David J Steger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
Zhang DG, Cheng J, Tai ZP, Luo Z. Identification of five genes in endoplasmic reticulum (ER) stress-apoptosis pathways in yellow catfish Pelteobagrus fulvidraco and their transcriptional responses to dietary lipid levels. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1117-1127. [PMID: 30847627 DOI: 10.1007/s10695-019-00627-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/26/2019] [Indexed: 06/09/2023]
Abstract
The activating transcription factor 4 (ATF4), DNA damage-inducible transcript 3 (DDIT3), growth arrest, and DNA damage-inducible protein 34 (GADD34), endoplasmic reticulum oxidoreductin 1α (ERO1α), and tumor necrosis factor receptor associated factor 2 (TRAF2) cDNAs were first characterized from yellow catfish Pelteobagrus fulvidraco. Compared to corresponding genes of mammals, all of these proteins shared similar conserved domains. Their mRNAs were widely expressed in various tissues, but at variable levels. Dietary lipid levels did not significantly influence ATF4 mRNA expression. mRNA expression of DDIT3 and GADD34 was highest for fish fed the low-lipid diets and lowest for fish fed middle-lipid diets. The mRNA levels of ERO1α and TRAF2 declined with increasing dietary lipid levels. For the first time, we characterized the full-length cDNA sequences of ATF4, DDIT3, GADD34, ERO1α, and TRAF2 and determined their tissue expression profiles and transcriptional responses to dietary lipid levels, which would contribute to our exploration into their biological functions, and providing new insights on relations between ER stress and lipid metabolism in fish.
Collapse
Affiliation(s)
- Dian-Guang Zhang
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China
| | - Jie Cheng
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China
| | - Zhi-Peng Tai
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China
| | - Zhi Luo
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China.
| |
Collapse
|
33
|
Abstract
Understanding adipogenesis, the process of adipocyte development, may provide new ways to treat obesity and related metabolic diseases. Adipogenesis is controlled by coordinated actions of lineage-determining transcription factors and epigenomic regulators. Peroxisome proliferator-activated receptor gamma (PPARγ) and C/EBPα are master "adipogenic" transcription factors. In recent years, a growing number of studies have reported the identification of novel transcriptional and epigenomic regulators of adipogenesis. However, many of these novel regulators have not been validated in adipocyte development in vivo and their working mechanisms are often far from clear. In this minireview, we discuss recent advances in transcriptional and epigenomic regulation of adipogenesis, with a focus on factors and mechanisms shared by both white adipogenesis and brown adipogenesis. Studies on the transcriptional regulation of adipogenesis highlight the importance of investigating adipocyte differentiation in vivo rather than drawing conclusions based on knockdown experiments in cell culture. Advances in understanding of epigenomic regulation of adipogenesis have revealed critical roles of histone methylation/demethylation, histone acetylation/deacetylation, chromatin remodeling, DNA methylation, and microRNAs in adipocyte differentiation. We also discuss future research directions that may help identify novel factors and mechanisms regulating adipogenesis.
Collapse
|
34
|
Zhang S, An Q, Hu P, Wu X, Pan X, Peng W, Wang R, Gan J, Chen D, Li Z, Wang T, Zhou G. Core regulatory RNA molecules identified in articular cartilage stem/progenitor cells during osteoarthritis progression. Epigenomics 2019; 11:669-684. [PMID: 30775942 DOI: 10.2217/epi-2018-0212] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To assess cartilage-derived stem/progenitor cells (CSPCs) in osteoarthritis (OA) by employing mRNA-miRNA-circRNA-lncRNA network biology approach. Methods: Differentially expressed (DE) RNAs in CSPCs from 2-/4-/8-month-old STR/Ort and CBA mice were identified to construct networks via RNA sequencing. Results: Compared with age-matched CBA mice, 4-/8-month-old STR/Ort mice had cartilage lesions and their CSPCs exhibited lower proliferative and differentiation capacity (decreased CD44 and CD90), and identified 7082 DE RNAs in STR/Ort mice were associated with strain differences or OA progression. OA-related core RNAs were identified via the networks constructed with the predominant DE RNAs, which were involved in the signaling pathways (NF-κB/MAPK/Hippo/Wnt/TGF-β/cytoskeleton organization). The core RNAs (miR-322-5p/miR-493-5p/miR-378c/CPNE1/Cdh2/PRDM16/CTGF/NCAM1) were validated in CSPCs from OA patients. Conclusion: RNA-based networks identifying core RNAs and signaling pathways contribute to CSPC-dependent OA mechanisms.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Qier An
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Peilin Hu
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Xiaomin Wu
- Department of Orthopedic & Traumatology, Shenzhen BaoAn People Hospital Affiliated Southern Medical University, Shenzhen, Guangdong 518101, PR China
| | - Xiaohua Pan
- Department of Orthopedic & Traumatology, Shenzhen BaoAn People Hospital Affiliated Southern Medical University, Shenzhen, Guangdong 518101, PR China
| | - Wenjin Peng
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Rikang Wang
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Jingyi Gan
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Zhen Li
- Shenzhen Alps Cell Sci-Tech Co. Ltd, Longhua District, Shenzhen, PR China
| | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements & Ultrasound Imaging, School of Biomedical Engineering, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology & Genetics, Guangdong Key Laboratory of Genomic Stability & Disease Prevention, Shenzhen Key Laboratory of Anti-aging & Regenerative Medicine, & Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| |
Collapse
|
35
|
Kuri-Harcuch W, Velez-delValle C, Vazquez-Sandoval A, Hernández-Mosqueira C, Fernandez-Sanchez V. A cellular perspective of adipogenesis transcriptional regulation. J Cell Physiol 2018; 234:1111-1129. [PMID: 30146705 DOI: 10.1002/jcp.27060] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022]
Abstract
Adipose cells store lipids in the cytoplasm and signal systemically through secretion of adipokines and other molecules that regulate body energy metabolism. Differentiation of fat cells and its regulation has been the focus of extensive research since the early 1970s. In this review, we had attempted to examine the research bearing on the control of adipose cell differentiation, some of it dating back to the early days when Howard Green and his group described the preadipocyte cell lines 3T3-L1 and 3T3-F442A during 1974-1975. We also concentrated our attention on research published during the last few years, emphasizing data described on transcription factors that regulate adipose differentiation, outside of those that were reported earlier as part of the canonical adipogenic transcriptional cascade, which has been the subject of ample reviews by several groups of researchers. We focused on the studies carried out with the two preadipocyte cell culture models, the 3T3-L1 and 3T3-F442A cells that have provided essential data on adipose biology.
Collapse
Affiliation(s)
- Walid Kuri-Harcuch
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Cristina Velez-delValle
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Alfredo Vazquez-Sandoval
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Claudia Hernández-Mosqueira
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Veronica Fernandez-Sanchez
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
36
|
Zhou J, Wan J, Shu XE, Mao Y, Liu XM, Yuan X, Zhang X, Hess ME, Brüning JC, Qian SB. N 6-Methyladenosine Guides mRNA Alternative Translation during Integrated Stress Response. Mol Cell 2018; 69:636-647.e7. [PMID: 29429926 DOI: 10.1016/j.molcel.2018.01.019] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 10/20/2017] [Accepted: 01/12/2018] [Indexed: 11/28/2022]
Abstract
The integrated stress response (ISR) facilitates cellular adaptation to stress conditions via the common target eIF2α. During ISR, the selective translation of stress-related mRNAs often relies on alternative mechanisms, such as leaky scanning or reinitiation, but the underlying mechanism remains incompletely understood. Here we report that, in response to amino acid starvation, the reinitiation of ATF4 is not only governed by the eIF2α signaling pathway, but is also subjected to regulation by mRNA methylation in the form of N6-methyladenosine (m6A). While depleting m6A demethylases represses ATF4 reinitiation, knocking down m6A methyltransferases promotes ATF4 translation. We demonstrate that m6A in the 5' UTR controls ribosome scanning and subsequent start codon selection. Global profiling of initiating ribosomes reveals widespread alternative translation events influenced by dynamic mRNA methylation. Consistently, Fto transgenic mice manifest enhanced ATF4 expression, highlighting the critical role of m6A in translational regulation of ISR at cellular and organismal levels.
Collapse
Affiliation(s)
- Jun Zhou
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Ji Wan
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Xin Erica Shu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Yuanhui Mao
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Xiao-Min Liu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Xin Yuan
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Xingqian Zhang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Martin E Hess
- Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Shu-Bing Qian
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
37
|
Yonekura S, Tsuchiya M, Tokutake Y, Mizusawa M, Nakano M, Miyaji M, Ishizaki H, Haga S. The unfolded protein response is involved in both differentiation and apoptosis of bovine mammary epithelial cells. J Dairy Sci 2018; 101:3568-3578. [PMID: 29428758 DOI: 10.3168/jds.2017-13718] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/29/2022]
Abstract
The unfolded protein response (UPR) describes a process involved in the homeostasis of endoplasmic reticulum (ER) and the differentiation of secretory cells. At present, the roles of UPR in the mammary gland tissue of dairy cattle are unknown. In the current study, we investigated the expression of UPR-related genes in Holstein cows during the developmental and lactating stages of the mammary gland tissue. To investigate the roles of UPR during the differentiation of mammary epithelial cells (MEC), we used MAC-T cells, a line of MEC. We collected samples of mammary gland tissue in dairy cows by biopsy during the late gestation and lactation periods and examined the expression of UPR-related genes by quantitative real-time PCR. Expression levels of the spliced X-box binding protein 1 (XBP1) and activating transcription factor 4 (ATF4) were found to be significantly higher in the mammary gland tissue 10 d before delivery compared with 40 d before delivery. An investigation before and after differentiation in MAC-T cells showed that the expression of ATF4 increased after differentiation of MEC, whereas that of the spliced XBP1 did not significantly change. Western blot analysis revealed that the differentiation-inducing stimulus induced phosphorylation of eukaryotic initiation factor 2α (eIF2α) but reduced that of protein kinase RNA-like endoplasmic reticulum kinase (PERK). Additionally, in ATF4-knockdown bovine MEC, differentiation was significantly suppressed; ATF4 knockdown also significantly suppressed the expression of glucocorticoid and insulin receptors. These results revealed that ER stress-independent ATF4 is involved in the cell differentiation mechanism, either directly or indirectly, via the control of the expression of lactogenic hormone receptors in bovine MEC. Immediately after parturition, gene expression levels of the spliced XBP1, ATF4, and C/EBP homologous protein (CHOP) markedly increased in mammary gland tissue, with a strong negative correlation between expression of CHOP and initial milk yield; CHOP is an apoptosis-related protein induced by ER stress. The above findings indicate that UPR is intrinsically associated with apoptosis of MEC, thus affecting the differentiation of these cells, as well as milk yield.
Collapse
Affiliation(s)
- Shinichi Yonekura
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan; Graduate School of Agriculture, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan; Interdisciplinary Graduate School of Science and Technology, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan.
| | - Megumi Tsuchiya
- Graduate School of Agriculture, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan
| | - Yukako Tokutake
- Interdisciplinary Graduate School of Science and Technology, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan
| | - Moeko Mizusawa
- Graduate School of Agriculture, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan
| | - Miwa Nakano
- Grazing Animal Unit, Division of Grassland Farming, Institute of Livestock and Grassland Science, NARO, 768 Senbonmatsu, Nasushiobara, Tochigi 329-2793, Japan
| | - Makoto Miyaji
- Feed Production and Utilization Group, Division of Dairy Production Research, Hokkaido Agricultural Research Center, NARO, 1 Hitsujigaoka, Toyohira, Sapporo, Hokkaido 062-8555, Japan
| | - Hiroshi Ishizaki
- Grazing Animal Unit, Division of Grassland Farming, Institute of Livestock and Grassland Science, NARO, 768 Senbonmatsu, Nasushiobara, Tochigi 329-2793, Japan
| | - Satoshi Haga
- Grazing Animal Unit, Division of Grassland Farming, Institute of Livestock and Grassland Science, NARO, 768 Senbonmatsu, Nasushiobara, Tochigi 329-2793, Japan
| |
Collapse
|
38
|
Lee JE, Park YK, Park S, Jang Y, Waring N, Dey A, Ozato K, Lai B, Peng W, Ge K. Brd4 binds to active enhancers to control cell identity gene induction in adipogenesis and myogenesis. Nat Commun 2017; 8:2217. [PMID: 29263365 PMCID: PMC5738375 DOI: 10.1038/s41467-017-02403-5] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/28/2017] [Indexed: 01/13/2023] Open
Abstract
The epigenomic reader Brd4 is an important drug target for cancers. However, its role in cell differentiation and animal development remains largely unclear. Using two conditional knockout mouse strains and derived cells, we demonstrate that Brd4 controls cell identity gene induction and is essential for adipogenesis and myogenesis. Brd4 co-localizes with lineage-determining transcription factors (LDTFs) on active enhancers during differentiation. LDTFs coordinate with H3K4 mono-methyltransferases MLL3/MLL4 (KMT2C/KMT2D) and H3K27 acetyltransferases CBP/p300 to recruit Brd4 to enhancers activated during differentiation. Brd4 deletion prevents the enrichment of Mediator and RNA polymerase II transcription machinery, but not that of LDTFs, MLL3/MLL4-mediated H3K4me1, and CBP/p300-mediated H3K27ac, on enhancers. Consequently, Brd4 deletion prevents enhancer RNA production, cell identity gene induction and cell differentiation. Interestingly, Brd4 is dispensable for maintaining cell identity genes in differentiated cells. These findings identify Brd4 as an enhancer epigenomic reader that links active enhancers with cell identity gene induction in differentiation. Despite being an important cancer drug target, the role of epigenetic reader Brd4 in cell differentiation and development remains unclear. Here, the authors provide evidence that Brd4 plays an important role in adipogenesis and myogenesis by binding to active enhancers to regulate gene expression.
Collapse
Affiliation(s)
- Ji-Eun Lee
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Young-Kwon Park
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sarah Park
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Younghoon Jang
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nicholas Waring
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.,Departments of Physics and Anatomy and Regenerative Biology, The George Washington University, Washington, DC, 20052, USA
| | - Anup Dey
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Keiko Ozato
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Binbin Lai
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.,Departments of Physics and Anatomy and Regenerative Biology, The George Washington University, Washington, DC, 20052, USA
| | - Weiqun Peng
- Departments of Physics and Anatomy and Regenerative Biology, The George Washington University, Washington, DC, 20052, USA
| | - Kai Ge
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
39
|
Linares JF, Cordes T, Duran A, Reina-Campos M, Valencia T, Ahn CS, Castilla EA, Moscat J, Metallo CM, Diaz-Meco MT. ATF4-Induced Metabolic Reprograming Is a Synthetic Vulnerability of the p62-Deficient Tumor Stroma. Cell Metab 2017; 26:817-829.e6. [PMID: 28988820 PMCID: PMC5718961 DOI: 10.1016/j.cmet.2017.09.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/27/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022]
Abstract
Tumors undergo nutrient stress and need to reprogram their metabolism to survive. The stroma may play a critical role in this process by providing nutrients to support the epithelial compartment of the tumor. Here we show that p62 deficiency in stromal fibroblasts promotes resistance to glutamine deprivation by the direct control of ATF4 stability through its p62-mediated polyubiquitination. ATF4 upregulation by p62 deficiency in the stroma activates glucose carbon flux through a pyruvate carboxylase-asparagine synthase cascade that results in asparagine generation as a source of nitrogen for stroma and tumor epithelial proliferation. Thus, p62 directly targets nuclear transcription factors to control metabolic reprogramming in the microenvironment and repress tumorigenesis, and identifies ATF4 as a synthetic vulnerability in p62-deficient tumor stroma.
Collapse
Affiliation(s)
- Juan F Linares
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Thekla Cordes
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Angeles Duran
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Miguel Reina-Campos
- Sanford Burnham Prebys Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Tania Valencia
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Christopher S Ahn
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Elias A Castilla
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jorge Moscat
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Maria T Diaz-Meco
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
40
|
Yeh KY, Lai CY, Lin CY, Hsu CC, Lo CP, Her GM. ATF4 overexpression induces early onset of hyperlipidaemia and hepatic steatosis and enhances adipogenesis in zebrafish. Sci Rep 2017; 7:16362. [PMID: 29180630 PMCID: PMC5703967 DOI: 10.1038/s41598-017-16587-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/13/2017] [Indexed: 02/08/2023] Open
Abstract
Activating transcription factor 4 (ATF4) is constitutively expressed in a variety of tissues, and regulates several pathological features associated with metabolic diseases such as non-alcoholic fatty liver diseases (NAFLD) and obesity. However, the role of ATF4 in animal model systems is poorly understood. To investigate ATF4 functions in zebrafish, we conditionally expressed ATF4 proteins, using a Tet-off transgenic system. We observed early-onset hyperlipidaemia and liver steatosis in ATF4 transgenic zebrafish (ATs) without doxycycline treatment (ATs − Dox). Oil Red O (ORO)-stained signals were predominant in the intravascular blood vessels and liver buds of larval ATs − Dox, indicating that ATF4 functionally promotes lipogenesis. Further, ATF4 overexpression accompanied the stimulation of the unfolded protein response. Therefore, adult ATs − Dox showed increased lipid accumulation, which led, in turn, to liver steatosis. Liver histology and ORO staining of ATs − Dox hepatocytes also indicated oxidative stress and induced NASH-like phenotypes. Moreover, ATF4 overexpression accelerated adipocyte differentiation via CCAAT enhancer binding protein-beta and peroxisome proliferator activated receptor-gamma inducible expression. ATs-Dox zebrafish showed increased weight gain with larger fat pads due to adipocyte hyperplasia. In this study, we report that ATF4 is a potential stimulator of lipid biosynthesis and adipogenesis in zebrafish.
Collapse
Affiliation(s)
- Kun-Yun Yeh
- Division of Hemato-Oncology, Department of Internal Medicine, Chang-Chung Memorial Hospital, 222 Maijin Road, Keelung, 204, Taiwan
| | - Chi-Yu Lai
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, 202, Taiwan
| | - Chiu-Ya Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, 202, Taiwan
| | - Chia-Chun Hsu
- Department of Radiology, Buddhist Tzu Chi General Hospital, Taichung Branch, No. 66 Fēngxìng Road Section 1, Taichung, 427, Taiwan.,School of Medicine, Tzu Chi University, No.701, Sec. 3, Jhongyang Road, Hualien, 97004, Taiwan
| | - Chung-Ping Lo
- Department of Radiology, Buddhist Tzu Chi General Hospital, Taichung Branch, No. 66 Fēngxìng Road Section 1, Taichung, 427, Taiwan.,School of Medicine, Tzu Chi University, No.701, Sec. 3, Jhongyang Road, Hualien, 97004, Taiwan
| | - Guor Mour Her
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, 202, Taiwan.
| |
Collapse
|
41
|
MicroRNA-214 Suppresses Osteogenic Differentiation of Human Periodontal Ligament Stem Cells by Targeting ATF4. Stem Cells Int 2017; 2017:3028647. [PMID: 29213288 PMCID: PMC5682087 DOI: 10.1155/2017/3028647] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/19/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Periodontitis is the main cause of adult tooth loss. Stem cell-based tissue engineering has become a promising therapy for periodontitis treatment. To date, human periodontal ligament stem cells (hPDLSCs) have been shown to be a favorable source for tissue engineering, but modulatory mechanisms of hPDLSCs remain unclear. Approximately 60% of mammalian genes are the targets of over 2000 miRNAs in multiple human cell types, and miRNAs are able to influence various biological processes in the human body, including bone formation. In this study, we found that after osteogenic induction, miR-214 was significantly decreased in hPDLSCs; therefore, we examined the effects of miR-214 on osteogenic differentiation. Computational miRNA target prediction analyses and luciferase reporter assays revealed that activating transcription factor 4 (ATF4) is a direct target of miR-214. We prepared cells overexpressing miR-214 and found that miR-214 negatively regulates osteogenic differentiation of hPDLSCs. For the target of miR-214, ATF4 protein expression level was decreased after induction. In conclusion, we found that miR-214-ATF4 axis is a novel pathway for regulating hPDLSC osteogenic differentiation.
Collapse
|
42
|
Yang F, Ma Q, Liu Z, Li W, Tan Y, Jin C, Ma W, Hu Y, Shen J, Ohgi KA, Telese F, Liu W, Rosenfeld MG. Glucocorticoid Receptor:MegaTrans Switching Mediates the Repression of an ERα-Regulated Transcriptional Program. Mol Cell 2017; 66:321-331.e6. [PMID: 28475868 PMCID: PMC5510478 DOI: 10.1016/j.molcel.2017.03.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/01/2016] [Accepted: 03/31/2017] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms underlying the opposing functions of glucocorticoid receptors (GRs) and estrogen receptor α (ERα) in breast cancer development remain poorly understood. Here we report that, in breast cancer cells, liganded GR represses a large ERα-activated transcriptional program by binding, in trans, to ERα-occupied enhancers. This abolishes effective activation of these enhancers and their cognate target genes, and it leads to the inhibition of ERα-dependent binding of components of the MegaTrans complex. Consistent with the effects of SUMOylation on other classes of nuclear receptors, dexamethasone (Dex)-induced trans-repression of the estrogen E2 program appears to depend on GR SUMOylation, which leads to stable trans-recruitment of the GR-N-CoR/SMRT-HDAC3 corepressor complex on these enhancers. Together, these results uncover a mechanism by which competitive recruitment of DNA-binding nuclear receptors/transcription factors in trans to hot spot enhancers serves as an effective biological strategy for trans-repression, with clear implications for breast cancer and other diseases.
Collapse
Affiliation(s)
- Feng Yang
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qi Ma
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhijie Liu
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Yuliang Tan
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chunyu Jin
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wubin Ma
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yiren Hu
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jia Shen
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kenneth A Ohgi
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Francesca Telese
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wen Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian 361102, China
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
43
|
Cohen DM, Steger DJ. Nuclear Receptor Function through Genomics: Lessons from the Glucocorticoid Receptor. Trends Endocrinol Metab 2017; 28:531-540. [PMID: 28495406 PMCID: PMC5505657 DOI: 10.1016/j.tem.2017.04.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/14/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022]
Abstract
Unlocking the therapeutic potential of the glucocorticoid receptor (GR) has motivated a search for small molecules that selectively modulate its ability to activate or repress gene transcription. Recently, breakthrough studies in the field of genomics have reinvigorated debate over longstanding transcriptional models explaining how GR controls tissue-specific gene expression. Here, we highlight these genomic studies with the dual goals of advancing understanding of nuclear receptor-mediated transcription and stimulating thought on the development of anti-inflammatory and immunosuppressive ligands for GR that have reduced harmful effects on metabolism.
Collapse
Affiliation(s)
- Daniel M Cohen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J Steger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
44
|
Rodríguez-Martínez JA, Reinke AW, Bhimsaria D, Keating AE, Ansari AZ. Combinatorial bZIP dimers display complex DNA-binding specificity landscapes. eLife 2017; 6:e19272. [PMID: 28186491 PMCID: PMC5349851 DOI: 10.7554/elife.19272] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
How transcription factor dimerization impacts DNA-binding specificity is poorly understood. Guided by protein dimerization properties, we examined DNA binding specificities of 270 human bZIP pairs. DNA interactomes of 80 heterodimers and 22 homodimers revealed that 72% of heterodimer motifs correspond to conjoined half-sites preferred by partnering monomers. Remarkably, the remaining motifs are composed of variably-spaced half-sites (12%) or 'emergent' sites (16%) that cannot be readily inferred from half-site preferences of partnering monomers. These binding sites were biochemically validated by EMSA-FRET analysis and validated in vivo by ChIP-seq data from human cell lines. Focusing on ATF3, we observed distinct cognate site preferences conferred by different bZIP partners, and demonstrated that genome-wide binding of ATF3 is best explained by considering many dimers in which it participates. Importantly, our compendium of bZIP-DNA interactomes predicted bZIP binding to 156 disease associated SNPs, of which only 20 were previously annotated with known bZIP motifs.
Collapse
Affiliation(s)
| | - Aaron W Reinke
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Devesh Bhimsaria
- Department of Biochemistry, University of Wisconsin-Madison, Madison, United States
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, Unites States
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Aseem Z Ansari
- Department of Biochemistry, University of Wisconsin-Madison, Madison, United States
- The Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, United States
| |
Collapse
|
45
|
Chen H, Yuan R, Zhang Y, Zhang X, Chen L, Zhou X, Yuan Z, Nie Y, Li M, Mo D, Chen Y. ATF4 regulates SREBP1c expression to control fatty acids synthesis in 3T3-L1 adipocytes differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1459-1469. [PMID: 27452504 DOI: 10.1016/j.bbagrm.2016.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/07/2016] [Accepted: 07/20/2016] [Indexed: 11/26/2022]
Abstract
Activating transcription factor 4 (ATF4), which is highly expressed in 3T3-L1 adipocytes after adipogenic induction, is essential for adipocytes differentiation. ATF4 also plays a vital role in regulating fatty acids biosynthesis, whereas the detailed mechanism of this process is still unclear. Here we demonstrated that siRNA-based ATF4 depletion in 3T3-L1 adipocytes significantly reduced the accumulation of fatty acids and triglycerides. Moreover, SREBP1c protein, which is an important transcription factor of lipogenesis, appreciably decreased while Srebp1c mRNA increased. Then we identified that ATF4 could maintain SREBP1c protein stability by directly activating the expression of USP7 which deubiquitinates SREBP1c and increases its protein content in cell. Besides, USP7 could restore the synthesis of fatty acids and triglycerides in the absence of ATF4. On the other hand, we found that ATF4 might inhibit the transcription of Srebp1c through TRB3, which is repressed by IBMX and DEX during early adipogenesis. Thus, our data indicate that ATF4 regulates SREBP1c expression to control fatty acids synthesis.
Collapse
Affiliation(s)
- Hu Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Renqiang Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ying Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xumeng Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xingyu Zhou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhuning Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yaping Nie
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ming Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
46
|
Huggins CJ, Mayekar MK, Martin N, Saylor KL, Gonit M, Jailwala P, Kasoji M, Haines DC, Quiñones OA, Johnson PF. C/EBPγ Is a Critical Regulator of Cellular Stress Response Networks through Heterodimerization with ATF4. Mol Cell Biol 2015; 36:693-713. [PMID: 26667036 PMCID: PMC4760225 DOI: 10.1128/mcb.00911-15] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 10/30/2015] [Accepted: 12/07/2015] [Indexed: 12/24/2022] Open
Abstract
The integrated stress response (ISR) controls cellular adaptations to nutrient deprivation, redox imbalances, and endoplasmic reticulum (ER) stress. ISR genes are upregulated in stressed cells, primarily by the bZIP transcription factor ATF4 through its recruitment to cis-regulatory C/EBP:ATF response elements (CAREs) together with a dimeric partner of uncertain identity. Here, we show that C/EBPγ:ATF4 heterodimers, but not C/EBPβ:ATF4 dimers, are the predominant CARE-binding species in stressed cells. C/EBPγ and ATF4 associate with genomic CAREs in a mutually dependent manner and coregulate many ISR genes. In contrast, the C/EBP family members C/EBPβ and C/EBP homologous protein (CHOP) were largely dispensable for induction of stress genes. Cebpg(-/-) mouse embryonic fibroblasts (MEFs) proliferate poorly and exhibit oxidative stress due to reduced glutathione levels and impaired expression of several glutathione biosynthesis pathway genes. Cebpg(-/-) mice (C57BL/6 background) display reduced body size and microphthalmia, similar to ATF4-null animals. In addition, C/EBPγ-deficient newborns die from atelectasis and respiratory failure, which can be mitigated by in utero exposure to the antioxidant, N-acetyl-cysteine. Cebpg(-/-) mice on a mixed strain background showed improved viability but, upon aging, developed significantly fewer malignant solid tumors than WT animals. Our findings identify C/EBPγ as a novel antioxidant regulator and an obligatory ATF4 partner that controls redox homeostasis in normal and cancerous cells.
Collapse
Affiliation(s)
- Christopher J Huggins
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Manasi K Mayekar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Nancy Martin
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Karen L Saylor
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Mesfin Gonit
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Parthav Jailwala
- Advanced Biomedical Computing Center, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Manjula Kasoji
- Advanced Biomedical Computing Center, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Diana C Haines
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Octavio A Quiñones
- DMS, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Peter F Johnson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| |
Collapse
|