1
|
Dowker-Key PD, Jadi PK, Gill NB, Hubbard KN, Elshaarrawi A, Alfatlawy ND, Bettaieb A. A Closer Look into White Adipose Tissue Biology and the Molecular Regulation of Stem Cell Commitment and Differentiation. Genes (Basel) 2024; 15:1017. [PMID: 39202377 PMCID: PMC11353785 DOI: 10.3390/genes15081017] [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: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Praveen Kumar Jadi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Katelin N. Hubbard
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Elshaarrawi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Naba D. Alfatlawy
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996-0840, USA
| |
Collapse
|
2
|
Nevzorova YA, Cubero FJ. Obesity under the moonlight of c-MYC. Front Cell Dev Biol 2023; 11:1293218. [PMID: 38116204 PMCID: PMC10728299 DOI: 10.3389/fcell.2023.1293218] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023] Open
Abstract
The moonlighting protein c-Myc is a master regulator of multiple biological processes including cell proliferation, differentiation, angiogenesis, apoptosis and metabolism. It is constitutively and aberrantly expressed in more than 70% of human cancers. Overwhelming evidence suggests that c-Myc dysregulation is involved in several inflammatory, autoimmune, metabolic and other non-cancerous diseases. In this review, we addressed the role of c-Myc in obesity. Obesity is a systemic disease, accompanied by multi-organ dysfunction apart from white adipose tissue (WAT), such as the liver, the pancreas, and the intestine. c-Myc plays a big diversity of functions regulating cellular proliferation, the maturation of progenitor cells, fatty acids (FAs) metabolism, and extracellular matrix (ECM) remodeling. Moreover, c-Myc drives the expression of a wide range of metabolic genes, modulates the inflammatory response, induces insulin resistance (IR), and contributes to the regulation of intestinal dysbiosis. Altogether, c-Myc is an interesting diagnostic tool and/or therapeutic target in order to mitigate obesity and its consequences.
Collapse
Affiliation(s)
- Yulia A. Nevzorova
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
3
|
Björk C, Subramanian N, Liu J, Acosta JR, Tavira B, Eriksson AB, Arner P, Laurencikiene J. An RNAi Screening of Clinically Relevant Transcription Factors Regulating Human Adipogenesis and Adipocyte Metabolism. Endocrinology 2021; 162:6272286. [PMID: 33963396 PMCID: PMC8197287 DOI: 10.1210/endocr/bqab096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Indexed: 12/13/2022]
Abstract
CONTEXT Healthy hyperplasic (many but smaller fat cells) white adipose tissue (WAT) expansion is mediated by recruitment, proliferation and/or differentiation of new fat cells. This process (adipogenesis) is controlled by transcriptional programs that have been mostly identified in rodents. OBJECTIVE A systemic investigation of adipogenic human transcription factors (TFs) that are relevant for metabolic conditions has not been revealed previously. METHODS TFs regulated in WAT by obesity, adipose morphology, cancer cachexia, and insulin resistance were selected from microarrays. Their role in differentiation of human adipose tissue-derived stem cells (hASC) was investigated by RNA interference (RNAi) screen. Lipid accumulation, cell number, and lipolysis were measured for all screened factors (148 TFs). RNA (RNAseq), protein (Western blot) expression, insulin, and catecholamine responsiveness were examined in hASC following siRNA treatment of selected target TFs. RESULTS Analysis of TFs regulated by metabolic conditions in human WAT revealed that many of them belong to adipogenesis-regulating pathways. The RNAi screen identified 39 genes that affected fat cell differentiation in vitro, where 11 genes were novel. Of the latter JARID2 stood out as being necessary for formation of healthy fat cell metabolic phenotype by regulating expression of multiple fat cell phenotype-specific genes. CONCLUSION This comprehensive RNAi screening in hASC suggests that a large proportion of WAT TFs that are impacted by metabolic conditions might be important for hyperplastic adipose tissue expansion. The screen also identified JARID2 as a novel TF essential for the development of functional adipocytes.
Collapse
Affiliation(s)
- Christel Björk
- Lipid laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Narmadha Subramanian
- Lipid laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Jianping Liu
- Karolinska High Throughput Center, Department of Medical Biochemistry and Biophysics (MBB), Division of Functional Genomics, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Juan Ramon Acosta
- Lipid laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Beatriz Tavira
- Lipid laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Anders B Eriksson
- Karolinska High Throughput Center, Department of Medical Biochemistry and Biophysics (MBB), Division of Functional Genomics, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Peter Arner
- Lipid laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Jurga Laurencikiene
- Lipid laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, SE-14186, Sweden
- Correspondence: Jurga Laurencikiene, PhD, Karolinska Institutet, Lipid laboratory, Dept. of Medicine Huddinge (MedH), NEO, Hälsovägen 9/Blickagången 16, 14183 Huddinge, Sweden.
| |
Collapse
|
4
|
Zhang K, Yang X, Zhao Q, Li Z, Fu F, Zhang H, Zheng M, Zhang S. Molecular Mechanism of Stem Cell Differentiation into Adipocytes and Adipocyte Differentiation of Malignant Tumor. Stem Cells Int 2020; 2020:8892300. [PMID: 32849880 PMCID: PMC7441422 DOI: 10.1155/2020/8892300] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/07/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Adipogenesis is the process through which preadipocytes differentiate into adipocytes. During this process, the preadipocytes cease to proliferate, begin to accumulate lipid droplets, and develop morphologic and biochemical characteristics of mature adipocytes. Mesenchymal stem cells (MSCs) are a type of adult stem cells known for their high plasticity and capacity to generate mesodermal and nonmesodermal tissues. Many mature cell types can be generated from MSCs, including adipocyte, osteocyte, and chondrocyte. The differentiation of stem cells into multiple mature phenotypes is at the basis for tissue regeneration and repair. Cancer stem cells (CSCs) play a very important role in tumor development and have the potential to differentiate into multiple cell lineages. Accumulating evidence has shown that cancer cells can be induced to differentiate into various benign cells, such as adipocytes, fibrocytes, osteoblast, by a variety of small molecular compounds, which may provide new strategies for cancer treatment. Recent studies have reported that tumor cells undergoing epithelial-to-mesenchymal transition can be induced to differentiate into adipocytes. In this review, molecular mechanisms, signal pathways, and the roles of various biological processes in adipose differentiation are summarized. Understanding the molecular mechanism of adipogenesis and adipose differentiation of cancer cells may contribute to cancer treatments that involve inducing differentiation into benign cells.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Xudong Yang
- Tianjin Rehabilitation Center, Tianjin, China
| | - Qi Zhao
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Zugui Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fangmei Fu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hao Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
5
|
Al Hasan M, Roy P, Dolan S, Martin PE, Patterson S, Bartholomew C. Adhesion G-protein coupled receptor 56 is required for 3T3-L1 adipogenesis. J Cell Physiol 2019; 235:1601-1614. [PMID: 31304602 DOI: 10.1002/jcp.29079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022]
Abstract
Obesity-associated conditions represent major global health and financial burdens and understanding processes regulating adipogenesis could lead to novel intervention strategies. This study shows that adhesion G-protein coupled receptor 56 (GPR56) gene transcripts are reduced in abdominal visceral white adipose tissue derived from obese Zucker rats versus lean controls. Immunostaining in 3T3-L1 preadipocytes reveals both mitotic cell restricted surface and low level general expression patterns of Gpr56. Gpr56 transcripts are differentially expressed in 3T3-L1 cells during adipogenesis. Transient knockdown (KD) of Gpr56 in 3T3-L1 cells dramatically inhibits differentiation through reducing the accumulation of both neutral cellular lipids (56%) and production of established adipogenesis Pparγ 2 (60-80%), C/ebpα (40-78%) mediator, and Ap2 (56-80%) marker proteins. Furthermore, genome editing of Gpr56 in 3T3-L1 cells created CW2.2.4 and RM4.2.5.5 clones (Gpr56 -/- cells) with compound heterozygous deletion frameshift mutations which abolish adipogenesis. Genome edited cells have sustained levels of the adipogenesis inhibitor β-catenin, reduced proliferation, reduced adhesion, altered profiles, and or abundance of extracellular matrix component gene transcripts for fibronectin, types I, III, and IV collagens and loss of actin stress fibers. β-catenin KD alone is insufficient to restore adipogenesis in Gpr56 -/- cells. Together these data show that Gpr56 is required for adipogenesis in 3T3-L1 cells. This report is the first demonstration that Gpr56 participates in regulation of the adipogenesis developmental program. Modulation of the levels of this protein and/or its biological activity may represent a novel target for development of therapeutic agents for the treatment of obesity.
Collapse
Affiliation(s)
- Mohammad Al Hasan
- Department of Biological & Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Poornima Roy
- Department of Biological & Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Sharron Dolan
- Department of Biological & Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Patricia E Martin
- Department of Biological & Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Steven Patterson
- Department of Biological & Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Chris Bartholomew
- Department of Biological & Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| |
Collapse
|
6
|
Xiong T, Li J, Chen F, Zhang F. PCAT-1: A Novel Oncogenic Long Non-Coding RNA in Human Cancers. Int J Biol Sci 2019; 15:847-856. [PMID: 30906215 PMCID: PMC6429018 DOI: 10.7150/ijbs.30970] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides (nts) without obvious protein coding potential. lncRNAs act as multiple roles in biological processes of diseases, especially carcinomas. Prostate cancer associated transcript-1 (PCAT-1) is an oncogenic lncRNA that identified by RNA-Sequence in prostate cancer. High expression of PCAT-1 is observed in different types of cancers, including prostate cancer, colorectal cancer, hepatocellular cancer and gastric cancer. High expressed PCAT-1 is correlated with poor overall survival. Furthermore, PCAT-1 regulates cancer cell proliferation, apoptosis, migration and invasion. Additionally, PCAT-1 is involved in EMT and Wnt/β-catenin-signaling pathway. In this review, we focus on the implication of PCAT-1 in human cancers.
Collapse
Affiliation(s)
| | | | - Fangfang Chen
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen 518036
| | - Fangting Zhang
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen 518036
| |
Collapse
|
7
|
Abstract
Propose Obesity is a fast growing epidemic worldwide. During obesity, the increase in adipose tissue mass arise from two different mechanisms, namely, hyperplasia and hypertrophy. Hyperplasia which is the increase in adipocyte number is characteristic of severe obese patients. Recently, there has been much interest in targeting adipogenesis as therapeutic strategy against obesity. Flavonoids have been shown to regulate several pathways and affect a number of molecular targets during specific stages of adipocyte development. Methods Presently, we provide a review of key studies evaluating the effects of dietary flavonoids in different stages of adipocyte development with a particular emphasis on the investigations that explore the underlying mechanisms of action of these compounds in human or animal cell lines as well as animal models. Results Flavonoids have been shown to regulate several pathways and affect a number of molecular targets during specific stages of adipocyte development. Although most of the studies reveal anti-adipogenic effect of flavonoids, some flavonoids demonstrated proadipogenic effect in mesenchymal stem cells or preadipocytes. Conclusion The anti-adipogenic effect of flavonoids is mainly via their effect on regulation of several pathways such as induction of apoptosis, suppression of key adipogenic transcription factors, activation of AMPK and Wnt pathways, inhibition of clonal expansion, and cell-cycle arrest.
Collapse
|
8
|
The H3K27 demethylase, Utx, regulates adipogenesis in a differentiation stage-dependent manner. PLoS One 2017; 12:e0173713. [PMID: 28319137 PMCID: PMC5358847 DOI: 10.1371/journal.pone.0173713] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/24/2017] [Indexed: 12/24/2022] Open
Abstract
Understanding the molecular mechanisms that drive adipogenesis is important in developing new treatments for obesity and diabetes. Epigenetic regulations determine the capacity of adipogenesis. In this study, we examined the role of a histone H3 lysine 27 demethylase, the ubiquitously transcribed tetratricopeptide repeat protein on the X chromosome (Utx), in the differentiation of mouse embryonic stem cells (mESCs) to adipocytes. Using gene trapping, we examined Utx-deficient male mESCs to determine whether loss of Utx would enhance or inhibit the differentiation of mESCs to adipocytes. Utx-deficient mESCs showed diminished potential to differentiate to adipocytes compared to that of controls. In contrast, Utx-deficient preadipocytes showed enhanced differentiation to adipocytes. Microarray analyses indicated that the β-catenin/c-Myc signaling pathway was differentially regulated in Utx-deficient cells during adipocyte differentiation. Therefore, our data suggest that Utx governs adipogenesis by regulating c-Myc in a differentiation stage-specific manner and that targeting the Utx signaling pathway could be beneficial for the treatment of obesity, diabetes, and congenital utx-deficiency disorders.
Collapse
|
9
|
Jeon M, Rahman N, Kim YS. Wnt/β-catenin signaling plays a distinct role in methyl gallate-mediated inhibition of adipogenesis. Biochem Biophys Res Commun 2016; 479:22-7. [PMID: 27592552 DOI: 10.1016/j.bbrc.2016.08.178] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/31/2016] [Indexed: 01/14/2023]
Abstract
The canonical Wnt/β-catenin signaling not only features in many developmental processes but also recently emerged as an attractive negative regulator of differentiation of preadipocytes into adipocytes. Here, we show that β-catenin signaling plays a distinct role in methyl gallate (MG)-mediated inhibition of 3T3-L1 adipocytes differentiation. We found that the expression of β-catenin decreased after adipogenic hormonal induction, whereas incubation of the differentiating cells with a physiological concentration of MG during adipogenic hormonal induction significantly prevented β-catenin degradation by activating Wnt signaling components such as Wnt1, Wnt10b, Fzd1, Fzd2, Lrp5, Lrp6, Dvl1, and Dvl2. Mechanistic experiments revealed that MG treatment during early adipocytic differentiation specifically inhibited degradation of β-catenin caused by phosphorylation at serine-33. In addition, MG treatment led to phosphorylation of GSK3β, which is one of β-catenin-degrading enzymes. Consequently, MG treatment facilitated translocation of the stabilized β-catenin from the cytoplasm to nucleus, and activates its target genes cyclin D1 and c-Myc. Furthermore, MG-induced stabilization of β-catenin suppresses PPARγ expression. Moreover, pharmacological activation or inhibition of β-catenin signaling during adipocytes differentiation decreased and increased, respectively, the level of the key adipogenic marker, PPARγ, and of its downstream targets, aP2 and adiponectin while MG treatment effectively reversed their expression level. Collectively, our data suggest that MG is a novel pharmacological stimulator of canonical Wnt/β-catenin signaling, and therefore represents a promising therapeutic agent in obesity.
Collapse
Affiliation(s)
- Miso Jeon
- Department of Microbiology, College of Medicine, Soonchunhyang University, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, South Korea
| | - Naimur Rahman
- Department of Microbiology, College of Medicine, Soonchunhyang University, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, South Korea
| | - Yong-Sik Kim
- Department of Microbiology, College of Medicine, Soonchunhyang University, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, South Korea.
| |
Collapse
|
10
|
Basse AL, Dixen K, Yadav R, Tygesen MP, Qvortrup K, Kristiansen K, Quistorff B, Gupta R, Wang J, Hansen JB. Global gene expression profiling of brown to white adipose tissue transformation in sheep reveals novel transcriptional components linked to adipose remodeling. BMC Genomics 2015; 16:215. [PMID: 25887780 PMCID: PMC4407871 DOI: 10.1186/s12864-015-1405-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/26/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Large mammals are capable of thermoregulation shortly after birth due to the presence of brown adipose tissue (BAT). The majority of BAT disappears after birth and is replaced by white adipose tissue (WAT). RESULTS We analyzed the postnatal transformation of adipose in sheep with a time course study of the perirenal adipose depot. We observed changes in tissue morphology, gene expression and metabolism within the first two weeks of postnatal life consistent with the expected transition from BAT to WAT. The transformation was characterized by massively decreased mitochondrial abundance and down-regulation of gene expression related to mitochondrial function and oxidative phosphorylation. Global gene expression profiling demonstrated that the time points grouped into three phases: a brown adipose phase, a transition phase and a white adipose phase. Between the brown adipose and the transition phase 170 genes were differentially expressed, and 717 genes were differentially expressed between the transition and the white adipose phase. Thirty-eight genes were shared among the two sets of differentially expressed genes. We identified a number of regulated transcription factors, including NR1H3, MYC, KLF4, ESR1, RELA and BCL6, which were linked to the overall changes in gene expression during the adipose tissue remodeling. Finally, the perirenal adipose tissue expressed both brown and brite/beige adipocyte marker genes at birth, the expression of which changed substantially over time. CONCLUSIONS Using global gene expression profiling of the postnatal BAT to WAT transformation in sheep, we provide novel insight into adipose tissue plasticity in a large mammal, including identification of novel transcriptional components linked to adipose tissue remodeling. Moreover, our data set provides a useful resource for further studies in adipose tissue plasticity.
Collapse
Affiliation(s)
- Astrid L Basse
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Karen Dixen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Rachita Yadav
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Malin P Tygesen
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg, Denmark.
| | - Klaus Qvortrup
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| | - Karsten Kristiansen
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| | - Bjørn Quistorff
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| | - Ramneek Gupta
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
| | - Jun Wang
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
- BGI-Shenzhen, Shenzhen, 518083, China.
- Princess Al Jawhara Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
- Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China.
| | - Jacob B Hansen
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark.
| |
Collapse
|
11
|
MYC is an early response regulator of human adipogenesis in adipose stem cells. PLoS One 2014; 9:e114133. [PMID: 25437437 PMCID: PMC4250176 DOI: 10.1371/journal.pone.0114133] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/02/2014] [Indexed: 12/13/2022] Open
Abstract
Adipose stem cell (ASC) differentiation is necessary for the proper maintenance and function of adipose tissue. The procurement and characterization of multipotent ASCs has enabled investigation into the molecular determinants driving human adipogenesis. Here, the transcription factor MYC was identified as a significant regulator of ASC differentiation. Expression of MYC transcript and protein was found to accumulate during the initial course of differentiation. Loss-of-function analysis using siRNA mediated knockdown of MYC demonstrated inhibition of hormonally stimulated adipogenesis. MYC exhibited an early and sustained expression pattern that preceded down regulation of key suppressor genes, as well as induction of transcriptional and functional effectors. Glucocorticoid stimulation was identified as a necessary component for MYC induction and was found to impact adipogenesis in a concentration-dependent manner. Global gene expression analysis of MYC knockdown in ASC enriched for functional pathways related to cell adhesion, cytoskeletal remodeling, and transcriptional components of adipogenesis. These results identify a functional role for MYC in promotion of multipotent ASC to the adipogenic lineage.
Collapse
|
12
|
Rodriguez R, Tornin J, Suarez C, Astudillo A, Rubio R, Yauk C, Williams A, Rosu-Myles M, Funes JM, Boshoff C, Menendez P. Expression of FUS-CHOP fusion protein in immortalized/transformed human mesenchymal stem cells drives mixoid liposarcoma formation. Stem Cells 2014; 31:2061-72. [PMID: 23836491 DOI: 10.1002/stem.1472] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Accepted: 06/08/2013] [Indexed: 12/11/2022]
Abstract
Increasing evidence supports that mesenchymal stromal/stem cells (MSCs) may represent the target cell for sarcoma development. Although different sarcomas have been modeled in mice upon expression of fusion oncogenes in MSCs, sarcomagenesis has not been successfully modeled in human MSCs (hMSCs). We report that FUS-CHOP, a hallmark fusion gene in mixoid liposarcoma (MLS), has an instructive role in lineage commitment, and its expression in hMSC sequentially immortalized/transformed with up to five oncogenic hits (p53 and Rb deficiency, hTERT over-expression, c-myc stabilization, and H-RAS(v12) mutation) drives the formation of serially transplantable MLS. This is the first model of sarcoma based on the expression of a sarcoma-associated fusion protein in hMSC, and allowed us to unravel the differentiation processes and signaling pathways altered in the MLS-initiating cells. This study will contribute to test novel therapeutic approaches and constitutes a proof-of-concept to use hMSCs as target cell for modeling other fusion gene-associated human sarcomas.
Collapse
Affiliation(s)
- Rene Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Shimada Y, Kuroyanagi J, Zhang B, Ariyoshi M, Umemoto N, Nishimura Y, Tanaka T. Downregulation of Max dimerization protein 3 is involved in decreased visceral adipose tissue by inhibiting adipocyte differentiation in zebrafish and mice. Int J Obes (Lond) 2013; 38:1053-60. [PMID: 24254064 DOI: 10.1038/ijo.2013.217] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 10/16/2013] [Accepted: 11/05/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND The diet-induced obesity model of zebrafish (DIO-zebrafish) share a common pathophysiological pathway with mammalian obesity. OBJECTIVES We aimed to investigate the role of Max dimerization protein 3 (MXD3) in visceral fat accumulation and adipocyte differentiation, by conducting knockdown experiments using zebrafish and mouse preadipocytes. METHODS To identify genes related to visceral adiposity, we conducted transcriptome analyses of human and zebrafish obese populations using the Gene Expression Omnibus and DNA microarray. We then intraperitoneally injected morpholino antisense oligonucleotides (MO-mxd3) to knockdown mxd3 gene expression in DIO-zebrafish and measured several parameters, which reflected human obesity and associated metabolic diseases. Finally, lentiviral Mxd3 shRNA knockdown in mouse 3T3-L1 preadipocytes was conducted. Quantitative PCR analyses of several differentiation markers were conducted during these gene knockdown experiments. RESULTS We found that MXD3 expression was increased in the obese population in humans and zebrafish. Intraperitoneal MO-mxd3 administration to DIO-zebrafish suppressed the increase in body weight, visceral fat accumulation and the size of mature adipocytes. Subsequently, dyslipidemia and liver steatosis were also ameliorated by MO-mxd3. In mouse adipocytes, Mxd3 expression was drastically increased in the early differentiation stage. Mxd3 shRNA inhibited preadipocyte proliferation and adipocyte maturation. Quantitative PCR analyses showed that the early differentiation marker, CCAAT/enhancer-binding protein delta (Cebpd) and late differentiation markers (CCAAT/enhancer-binding protein, alpha and peroxisome proliferator-activated receptor gamma) were downregulated by Mxd3 knockdown in 3T3-L1 cells and DIO-zebrafish. Subsequently, mature adipocyte markers (adiponectin and caveolin 1 for zebrafish, and fatty acid binding protein 4 and stearoyl-coenzyme A desaturase 1 for mouse adipocytes) were also decreased. CONCLUSION Mxd3 regulates preadipocyte proliferation and early adipocyte differentiation via Cebpd downregulation in vitro and in vivo. Integrated analysis of human and zebrafish transcriptomes allows identification of a novel therapeutic target against human obesity and further associated metabolic disease.
Collapse
Affiliation(s)
- Y Shimada
- 1] Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan [2] Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan [3] Mie University Medical Zebrafish Research Center, Mie, Japan [4] Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan [5] Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie, Japan
| | - J Kuroyanagi
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan
| | - B Zhang
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan
| | - M Ariyoshi
- 1] Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan [2] Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan
| | - N Umemoto
- 1] Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan [2] Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan
| | - Y Nishimura
- 1] Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan [2] Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan [3] Mie University Medical Zebrafish Research Center, Mie, Japan [4] Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan [5] Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie, Japan
| | - T Tanaka
- 1] Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, Mie, Japan [2] Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan [3] Mie University Medical Zebrafish Research Center, Mie, Japan [4] Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan [5] Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie, Japan
| |
Collapse
|
14
|
Fromm-Dornieden C, von der Heyde S, Lytovchenko O, Salinas-Riester G, Brenig B, Beissbarth T, Baumgartner BG. Novel polysome messages and changes in translational activity appear after induction of adipogenesis in 3T3-L1 cells. BMC Mol Biol 2012; 13:9. [PMID: 22436005 PMCID: PMC3347988 DOI: 10.1186/1471-2199-13-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 03/21/2012] [Indexed: 01/06/2023] Open
Abstract
Background Control of translation allows for rapid adaptation of the cell to stimuli, rather than the slower transcriptional control. We presume that translational control is an essential process in the control of adipogenesis, especially in the first hours after hormonal stimulation. 3T3-L1 preadipocytes were cultured to confluency and adipogenesis was induced by standard protocols using a hormonal cocktail. Cells were harvested before and 6 hours after hormonal induction. mRNAs attached to ribosomes (polysomal mRNAs) were separated from unbound mRNAs by velocity sedimentation. Pools of polysomal and unbound mRNA fractions were analyzed by microarray analysis. Changes in relative abundance in unbound and polysomal mRNA pools were calculated to detect putative changes in translational activity. Changes of expression levels of selected genes were verified by qPCR and Western blotting. Results We identified 43 genes that shifted towards the polysomal fraction (up-regulated) and 2 genes that shifted towards free mRNA fraction (down-regulated). Interestingly, we found Ghrelin to be down-regulated. Up-regulated genes comprise factors that are nucleic acid binding (eIF4B, HSF1, IRF6, MYC, POLR2a, RPL18, RPL27a, RPL6, RPL7a, RPS18, RPSa, TSC22d3), form part of ribosomes (RPL18, RPL27a, RPL6, RPL7a, RPS18, RPSa), act on the regulation of translation (eIF4B) or transcription (HSF1, IRF6, MYC, TSC22d3). Others act as chaperones (BAG3, HSPA8, HSP90ab1) or in other metabolic or signals transducing processes. Conclusions We conclude that a moderate reorganisation of the functionality of the ribosomal machinery and translational activity are very important steps for growth and gene expression control in the initial phase of adipogenesis.
Collapse
|
15
|
Xiao H, Leblanc SE, Wu Q, Konda S, Salma N, Marfella CGA, Ohkawa Y, Imbalzano AN. Chromatin accessibility and transcription factor binding at the PPARγ2 promoter during adipogenesis is protein kinase A-dependent. J Cell Physiol 2010; 226:86-93. [PMID: 20625991 DOI: 10.1002/jcp.22308] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated transcription factor that specifies formation of the adipocyte lineage. PPARγ also serves as a primary target for the treatment of type 2 diabetes, illustrating both its medical relevance as well as the need to understand fundamental aspects of PPARγ expression and function. Here, we characterize molecular changes that occur at the PPARγ2 promoter within the first several hours of adipocyte differentiation in culture. Our results demonstrate that changes in chromatin accessibility at the PPARγ2 promoter and occupancy of the promoter by the c-Fos transcription factor occur within an hour of the onset of differentiation, followed closely by the binding of the CCAAT/enhancer binding protein beta (C/EBPβ) transcription factor. All three events show a remarkable dependency on protein kinase A (PKA) activity. These results reflect novel requirements for the PKA signaling pathway and reinforce the importance of PKA function during the onset of adipocyte differentiation.
Collapse
Affiliation(s)
- Hengyi Xiao
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
v-Myc inhibits C/EBPβ activity by preventing C/EBPβ-induced phosphorylation of the co-activator p300. Oncogene 2009; 28:2446-55. [DOI: 10.1038/onc.2009.90] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Bonal C, Thorel F, Ait-Lounis A, Reith W, Trumpp A, Herrera PL. Pancreatic inactivation of c-Myc decreases acinar mass and transdifferentiates acinar cells into adipocytes in mice. Gastroenterology 2009; 136:309-319.e9. [PMID: 19022256 DOI: 10.1053/j.gastro.2008.10.015] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 09/29/2008] [Accepted: 10/02/2008] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS The pancreatic mass is determined by the coordinated expansion and differentiation of progenitor cells and is maintained via tight control of cell replacement rates. The basic helix-loop-helix transcription factor c-Myc is one of the main regulators of these processes in many organs. We studied the requirement of c-Myc in controlling the generation and maintenance of pancreatic mass. METHODS We conditionally inactivated c-Myc in Pdx1+ pancreatic progenitor cells. Pancreata of mice lacking c-Myc (c-Myc(P-/-) mice) were analyzed during development and ageing. RESULTS Pancreatic growth in c-Myc(P-/-) mice was impaired starting on E12.5, in early primordia, because of decreased proliferation and altered differentiation of exocrine progenitors; islet progenitors were spared. Acinar cell maturation was defective in the adult hypotrophic pancreas, which hampered exocrine mass maintenance in aged animals. From 2 to 10 months of age, the c-Myc(P-/-) pancreas was progressively remodeled without inflammatory injury. Loss of acinar cells increased with time, concomitantly with adipose tissue accumulation. Using a genetic cell lineage tracing analysis, we demonstrated that pancreatic adipose cells were derived directly from transdifferentiating acinar cells. This epithelial-to-mesenchyme transition was also observed in normal aged specimens and in pancreatitis. CONCLUSIONS These results provide evidence indicating that c-Myc activity is required for growth and maturation of the exocrine pancreas, and sheds new light on the ontogeny of pancreatic adipose cells in processes of organ degenerescence and tissue involution.
Collapse
Affiliation(s)
- Claire Bonal
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
18
|
Nakai T, Mochida J, Sakai D. Synergistic role of c-Myc and ERK1/2 in the mitogenic response to TGF beta-1 in cultured rat nucleus pulposus cells. Arthritis Res Ther 2008; 10:R140. [PMID: 19061498 PMCID: PMC2656245 DOI: 10.1186/ar2567] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 11/29/2008] [Accepted: 12/05/2008] [Indexed: 12/24/2022] Open
Abstract
Introduction Although transforming growth factor β1 (TGFβ1) is known to be a potent inhibitor of proliferation in most cell types, it accelerates proliferation in certain mesenchymal cells, such as articular chondrocytes and nucleus pulposus cells. The low ability for self-renewal of nucleus pulposus cells is one obstacle in developing new therapeutic options for intervertebral disc diseases, and utilizing cytokines is one of the strategies to regulate nucleus pulposus cell proliferation. However, the precise cell cycle progression and molecular mechanisms by which TGFβ1 stimulates cell growth remain unclear. The aim of this study was to elucidate a mechanism that enables cell proliferation with TGFβ1 stimulation. Methods We tested cultured rat nucleus pulposus cells for proliferation and cell cycle distribution under exogenous TGFβ1 stimulation with and without putative pharmaceutical inhibitors. To understand the molecular mechanism, we evaluated the expression levels of key regulatory G1 phase proteins, c-Myc and the cyclin-dependent kinase inhibitors. Results We found that TGFβ1 promoted proliferation and cell cycle progression while reducing expression of the cyclin-dependent kinase inhibitors p21 and p27, which are downregulators of the cell cycle. Robust c-Myc expression for 2 h and immediate phosphorylation of extra cellular signal regulated kinase (ERK1/2) were detected in cultures when TGFβ1 was added. However, pretreatment with 10058-F4 (an inhibitor of c-Myc transcriptional activity) or PD98059 (an inhibitor of ERK1/2) suppressed c-Myc expression and ERK1/2 phosphorylation, and inhibited cell cycle promotion by TGFβ1. Conclusions Our experimental results indicate that TGFβ1 promotes cell proliferation and cell cycle progression in rat nucleus pulposus cells and that c-Myc and phosphorylated ERK1/2 play important roles in this mechanism. While the difference between rat and human disc tissues requires future studies using different species, investigation of distinct response in the rat model provides fundamental information to elucidate a specific regulatory pathway of TGFβ1.
Collapse
Affiliation(s)
- Tomoko Nakai
- Division of Organogenesis, Research Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan.
| | | | | |
Collapse
|
19
|
Kim SJ, Lee KH, Lee YS, Mun EG, Kwon DY, Cha YS. Transcriptome analysis and promoter sequence studies on early adipogenesis in 3T3-L1 cells. Nutr Res Pract 2007; 1:19-28. [PMID: 20535381 PMCID: PMC2882572 DOI: 10.4162/nrp.2007.1.1.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 03/02/2007] [Accepted: 03/05/2007] [Indexed: 12/13/2022] Open
Abstract
To identify regulatory molecules which play key roles in the development of obesity, we investigated the transcriptional profiles in 3T3-L1 cells at early stage of differentiation and analyzed the promoter sequences of differentially regulated genes. One hundred and sixty-one (161) genes were found to have significant changes in expression at the 2nd day following treatment with differentiation cocktail. Among them, 86 transcripts were up-regulated and 75 transcripts were down-regulated. The 161 transcripts were classified into 10 categories according to their functional roles; cytoskeleton, cell adhesion, immune, defense response, metabolism, protein modification, protein metabolism, regulation of transcription, signal transduction and transporter. To identify transcription factors likely involved in regulating these differentially expressed genes, we analyzed the promoter sequences of up- or -down regulated genes for the presence of transcription factor binding sites (TFBSs). Based on coincidence of regulatory sites, we have identified candidate transcription factors (TFs), which include those previously known to be involved in adipogenesis (CREB, OCT-1 and c-Myc). Among them, c-Myc was also identified by our microarray data. Our approach to take advantage of the resource of the human genome sequences and the results from our microarray experiments should be validated by further studies of promoter occupancy and TF perturbation.
Collapse
Affiliation(s)
- Su-Jong Kim
- Department of Biochemistry, College of Medicine, Hanyang University, Seoul 133-791, Korea
| | | | | | | | | | | |
Collapse
|
20
|
Gupta P, Park SW, Farooqui M, Wei LN. Orphan nuclear receptor TR2, a mediator of preadipocyte proliferation, is differentially regulated by RA through exchange of coactivator PCAF with corepressor RIP140 on a platform molecule GRIP1. Nucleic Acids Res 2007; 35:2269-82. [PMID: 17389641 PMCID: PMC1874640 DOI: 10.1093/nar/gkl1147] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 11/12/2006] [Accepted: 12/15/2006] [Indexed: 01/15/2023] Open
Abstract
Orphan nuclear receptor TR2 is a preadipocyte proliferator. Knockdown of TR2 in 3T3-L1 preadipocytes reduced their proliferation efficiency, whereas specific elevation of TR2 in these cells facilitated their proliferation. All-trans retinoic acid (RA) stimulates cellular proliferation in 3T3-L1 preadipocytes by activating TR2 through an IR0-type RA response element, which further activates c-Myc expression. In post-differentiated adipocytes, RA becomes a repressive signal for TR2 and rapidly down-regulates its expression. The biphasic effect of RA on TR2 expression in 3T3-L1 is mediated by differential RA-dependent coregulator recruitment to the receptor/Glucocorticoid Receptor-Interacting Protein 1 (GRIP1) complex that binds IR0 on the TR2 promoter. RA induces the recruitment of histone acetyl transferase-containing/GRIP1/p300/CBP-associated factor (PCAF) complex to the TR2 promoter in undifferentiated cells, whereas it triggers recruitment of histone deacetylase-containing/GRIP1/receptor-interacting protein 140 (RIP140) complex in differentiated cells. GRIP1 directly interacts with RIP140 through its carboxyl terminal AD2 domain. GRIP1 interacts with PCAF and RIP140 directly and differentially, functioning as a platform molecule to mediate differential RA-induced coregulator recruitment to TR2 promoter target. This results in a biphasic effect of RA on the expression of TR2 in undifferentiated and differentiated cells, which is required for RA-stimulated preadipocyte proliferation.
Collapse
Affiliation(s)
| | | | | | - L.-N. Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
Blakely CM, Sintasath L, D'Cruz CM, Hahn KT, Dugan KD, Belka GK, Chodosh LA. Developmental stage determines the effects of MYC in the mammary epithelium. Development 2005; 132:1147-60. [PMID: 15689376 DOI: 10.1242/dev.01655] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Epidemiological findings suggest that the consequences of a given oncogenic stimulus vary depending upon the developmental state of the target tissue at the time of exposure. This is particularly evident in the mammary gland, where both age at exposure to a carcinogenic stimulus and the timing of a first full-term pregnancy can markedly alter the risk of developing breast cancer. Analogous to this, the biological consequences of activating oncogenes, such as MYC, can be influenced by cellular context both in terms of cell lineage and cellular environment. In light of this, we hypothesized that the consequences of aberrant MYC activation in the mammary gland might be determined by the developmental state of the gland at the time of MYC exposure. To test this hypothesis directly, we have used a doxycycline-inducible transgenic mouse model to overexpress MYC during different stages of mammary gland development. Using this model, we find that the ability of MYC to inhibit postpartum lactation is due entirely to its activation within a specific 72-hour window during mid-pregnancy; by contrast, MYC activation either prior to or following this 72-hour window has little or no effect on postpartum lactation. Surprisingly, we find that MYC does not block postpartum lactation by inhibiting mammary epithelial differentiation, but rather by promoting differentiation and precocious lactation during pregnancy, which in turn leads to premature involution of the gland. We further show that this developmental stage-specific ability of MYC to promote mammary epithelial differentiation is tightly linked to its ability to downregulate caveolin 1 and activate Stat5 in a developmental stage-specific manner. Our findings provide unique in vivo molecular evidence for developmental stage-specific effects of oncogene activation, as well as the first evidence linking MYC with activation of the Jak2-Stat5 signaling pathway.
Collapse
Affiliation(s)
- Collin M Blakely
- Departments of Cancer Biology, Cell and Developmental Biology, Medicine, and The Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6160, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Ouyang GL, Li QF, Peng XX, Liu QR, Hong SG. Effects of tachyplesin on proliferation and differentiation of human hepatocellular carcinoma SMMC-7721 cells. World J Gastroenterol 2002; 8:1053-8. [PMID: 12439924 PMCID: PMC4656379 DOI: 10.3748/wjg.v8.i6.1053] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the antitumor activities of tachyplesin on human hepatocellular carcinoma (HCC) cells.
METHODS: Tachyplesin, isolated from acid extracts of Chinese horseshoe crab (Tachypleus tridentatus) hemocytes, was used to treat the human HCC cell line SMMC-7721. Effects of tachyplesin on the proliferation of SMMC-7721 cells were measured with trypan blue dye exclusion test and HE staining. The morphology and ultrastructure of the cells were examined by light microscopy and transmission electron microscopy, respectively. The activities of γ-glutamyltransferase (γ-GT) and tyrosine aminotransferase (TAT) were assayed with biochemical methods. The levels of alpha fetoprotein (α-FP), proliferating cell nuclear antigen (PCNA), p21WAF1/CIP1 and c-myc were examined by immunocytochemistry.
RESULTS: After treatment with tachyplesin 3.0 mg/L, the proliferation of SMMC-7721 cells was inhibited significantly, with the cell growth inhibitory rate amounted to 55.57% and the maximum cell mitotic index declined by 43.68%. The morphology and ultrastructure underwent restorational alteration. The activity of γ-GT declined while TAT activity increased obviously, and the levels of α-FP and PCNA decreased. Moreover, the expression of p21WAF1/CIP1 protein was up-regulated and that of c-myc protein was down-regulated.
CONCLUSION: Tachyplesin could effectively inhibit the proliferation of hepatocarcinoma cells, reverse the malignant morphological and ultrastructural characteristics, alter the levels of enzymes and antigens, regulate the expression of differentiation-associated oncogene and tumor suppressor gene, and induce hepatocarcinama cell differentiation.
Collapse
Affiliation(s)
- Gao-Liang Ouyang
- Laboratory of Cell Biology, School of Life Sciences, Xiamen University, Fuijan Province, China
| | | | | | | | | |
Collapse
|
23
|
Affiliation(s)
- H Gadgil
- Department of Biochemistry, University of Tennessee, 858 Madison Avenue, Memphis, Tennessee 38163, USA
| | | | | |
Collapse
|
24
|
Heath VJ, Gillespie DA, Crouch DH. Inhibition of adipocyte differentiation by cMyc is not accompanied by alterations in cell cycle control. Biochem Biophys Res Commun 2000; 269:438-43. [PMID: 10708572 DOI: 10.1006/bbrc.2000.2316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Using a preadipocyte cell line constitutively expressing cMyc, we set out to determine if the ability of cMyc to inhibit adipogenic differentiation was functionally distinct from its role in cell cycle progression and transformation. We now report that in this system differentiation control and cellular transformation are separable functions of cMyc. Furthermore, the Myc-induced inhibition of adipocyte differentiation appears to be mediated via suppression of C/EBP-alpha and p21 gene expression late in the adipogenic differentiation programme, without deregulation of either cell cycle control or gene expression during the early stages of the process.
Collapse
Affiliation(s)
- V J Heath
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, United Kingdom
| | | | | |
Collapse
|