1
|
He Y, Zhu D, Greenman K, Ruiz C, Shang J, Lu Q, Kojetin DJ, Drakas R, Cameron MD, Lizarzaburu M, Solt LA, Kamenecka TM. Structure-Activity Relationship and Biological Investigation of a REV-ERBα-Selective Agonist SR-29065 ( 34) for Autoimmune Disorders. J Med Chem 2023; 66:14815-14823. [PMID: 37888788 DOI: 10.1021/acs.jmedchem.3c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Autoimmune diseases affect 50 million Americans, predominantly women, and are thought to be one of the top 10 leading causes of death among women in age groups up to 65 years. A central role for TH17 cells has been highlighted by genome-wide association studies (GWAS) linking genes preferentially expressed in TH17 cells to several human autoimmune diseases. We and others have reported that the nuclear receptors REV-ERBα and β are cell-intrinsic repressors of TH17 cell development and pathogenicity and might therefore be therapeutic targets for intervention. Herein, we describe detailed SAR studies of a novel REV-ERBα-selective scaffold. Metabolic stability of the ligands was optimized allowing for in vivo interrogation of the receptor in a mouse model of multiple sclerosis (EAE) with a ligand (34). Reduction in frequency and number of T-cells in the CNS as well as key REV-ERB target genes is a measure of target engagement in vivo.
Collapse
Affiliation(s)
- Yuanjun He
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Di Zhu
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Kevin Greenman
- ChemPartner Corporation, 280 Utah Avenue, Suite 100, South San Francisco, California 94080, United States
| | - Claudia Ruiz
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Jinsai Shang
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Qun Lu
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Douglas J Kojetin
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Robert Drakas
- ShangPharma Innovation, 280 Utah Avenue, Suite 100, South San Francisco, California 94080, United States
| | - Michael D Cameron
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Mike Lizarzaburu
- ChemPartner Corporation, 280 Utah Avenue, Suite 100, South San Francisco, California 94080, United States
| | - Laura A Solt
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Theodore M Kamenecka
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| |
Collapse
|
2
|
Oehler M, Geisser L, Diernfellner ACR, Brunner M. Transcription activator WCC recruits deacetylase HDA3 to control transcription dynamics and bursting in Neurospora. SCIENCE ADVANCES 2023; 9:eadh0721. [PMID: 37390199 PMCID: PMC10313174 DOI: 10.1126/sciadv.adh0721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/25/2023] [Indexed: 07/02/2023]
Abstract
RNA polymerase II initiates transcription either randomly or in bursts. We examined the light-dependent transcriptional activator White Collar Complex (WCC) of Neurospora to characterize the transcriptional dynamics of the strong vivid (vvd) promoter and the weaker frequency (frq) promoter. We show that WCC is not only an activator but also represses transcription by recruiting histone deacetylase 3 (HDA3). Our data suggest that bursts of frq transcription are governed by a long-lived refractory state established and maintained by WCC and HDA3 at the core promoter, whereas transcription of vvd is determined by WCC binding dynamics at an upstream activating sequence. Thus, in addition to stochastic binding of transcription factors, transcription factor-mediated repression may also influence transcriptional bursting.
Collapse
Affiliation(s)
- Michael Oehler
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, D-60120 Heidelberg, Germany
| | - Leonie Geisser
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, D-60120 Heidelberg, Germany
| | - Axel C. R. Diernfellner
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, D-60120 Heidelberg, Germany
| | | |
Collapse
|
3
|
Wang X, Jia R, Chen K, Wang J, Jiang K, Wang Z. RORα and REV-ERBα are Associated With Clinicopathological Parameters and are Independent Biomarkers of Prognosis in Gastric Cancer. Technol Cancer Res Treat 2021; 20:15330338211039670. [PMID: 34931925 PMCID: PMC8721360 DOI: 10.1177/15330338211039670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Retinoid-related orphan receptor alpha (RORα) and nuclear receptor subfamily 1 group D member 1 (REV-ERBα) play critical roles in many human cancers. Whether RORα and REV-ERBα expression levels are associated with clinical characteristics are poorly understood, and they may be independent predictors of overall survival (OS) and progression-free survival (PFS) in gastric cancer (GC). This study aimed to investigate the correlation of RORα and REV-ERBα expression levels with clinicopathological parameters, OS, and PFS in GC. Immunohistochemistry and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) were employed to assess the expression levels of RORα and REV-ERBα, which were downregulated in GC tissues compared with normal gastric tissues (P < .001; P < .001) and were associated with several clinicopathological parameters, including histological grade (P = .032; P < .001), preoperative carcinoembryonic antigen (CEA) levels (P = .004; P < .001), and tumor-node-metastasis (TNM) stage (P = .015; P < .001). Additionally, low RORα and REV-ERBα expression levels were associated with poor OS and PFS in GC patients, respectively (P < .001; P = .001). Furthermore, univariate Cox regression model analysis showed that histological grade (P < .001; P < .001), preoperative CEA levels (P < .001; P = .001), TNM stage (P < .001; P < .001), lymph node metastasis (P = .002; P = .002), RORα expression levels (P = .001; P < .001), and REV-ERBα expression levels (P < .001; P = .001) were associated with OS and PFS in GC. Multivariate Cox regression model analysis indicated that RORα expression levels and REV-ERBα expression levels are independent factors of OS and PFS in GC. Besides, RORα and REV-ERBα expression may be positively correlated (χ2 = 6.835; P = .009), and GC patients with both high RORα and REV-ERBα expression levels had the best prognosis. In conclusion, RORα and REV-ERBα may coparticipate in tumor activities and show potential to estimate the prognosis of GC.
Collapse
Affiliation(s)
- Xiaoshan Wang
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ru Jia
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ke Chen
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jingjing Wang
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Kai Jiang
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Zhengguang Wang
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
4
|
|
5
|
Amador A, Campbell S, Kazantzis M, Lan G, Burris TP, Solt LA. Distinct roles for REV-ERBα and REV-ERBβ in oxidative capacity and mitochondrial biogenesis in skeletal muscle. PLoS One 2018; 13:e0196787. [PMID: 29723273 PMCID: PMC5933789 DOI: 10.1371/journal.pone.0196787] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/19/2018] [Indexed: 12/19/2022] Open
Abstract
The nuclear receptors REV-ERBα and REV-ERBβ have been demonstrated to be core members of the circadian clock and participate in the regulation of a diverse set of metabolic functions. Due to their overlapping tissue expression patterns and gene expression profiles, REV-ERBβ is thought to be redundant to REV-ERBα. Recent work has highlighted REV-ERBα's role in the regulation of skeletal muscle oxidative capacity and mitochondrial biogenesis. Considering the similarity between the REV-ERBs and the hypothesized overlap in function, we sought to determine whether REV-ERBβ-deficiency presented with a similar skeletal muscle phenotype as REV-ERBα-deficiency. Ectopic overexpression in C2C12 cells demonstrated that REV-ERBβ drives mitochondrial biogenesis and the expression of genes involved in fatty acid oxidation. Intriguingly, knock down of REV-ERBβ in C2C12 cultures also resulted in mitochondrial biogenesis and increased expression of genes involved in fatty acid β-oxidation. To determine whether these effects occurred in vivo, we examined REV-ERBβ-deficient mice and observed a similar increase in expression of genes involved in mitochondrial biogenesis and fatty acid β-oxidation. Consistent with these results, REV-ERBβ-deficient mice exhibited an altered metabolic phenotype compared to wild-type littermate controls when measured by indirect calorimetry. This likely compensated for the increased food consumption that occurred, possibly aiding in the maintenance of their weight over time. Since feeding behaviors are a direct circadian output, this study suggests that REV-ERBβ may have more subtle effects on circadian behaviors than originally identified. Furthermore, these data implicate REV-ERBβ in the control of skeletal muscle metabolism and energy expenditure and suggest that development of REV-ERBα versus REV-ERBβ selective ligands may have therapeutic utility in the treatment of metabolic syndrome.
Collapse
MESH Headings
- Animals
- Body Weight
- Calorimetry, Indirect
- Cell Line
- Circadian Rhythm/genetics
- Circadian Rhythm/physiology
- Energy Metabolism/genetics
- Energy Metabolism/physiology
- Fatty Acids/metabolism
- Feeding Behavior/physiology
- Female
- Gene Expression Regulation
- Male
- Mice
- Mice, Knockout
- Mitochondria, Muscle/physiology
- Muscle, Skeletal/metabolism
- Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/physiology
- Organelle Biogenesis
- Oxidation-Reduction
- Oxidative Phosphorylation
- RNA Interference
- RNA, Small Interfering/genetics
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/deficiency
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/physiology
- Repressor Proteins/antagonists & inhibitors
- Repressor Proteins/deficiency
- Repressor Proteins/genetics
- Repressor Proteins/physiology
Collapse
Affiliation(s)
- Ariadna Amador
- Kellogg School of Science and Technology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Sean Campbell
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Melissa Kazantzis
- Metabolic Core, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Gary Lan
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Thomas P. Burris
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Laura A. Solt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, United States of America
| |
Collapse
|
6
|
Transgenic Adipose-specific Expression of the Nuclear Receptor RORα Drives a Striking Shift in Fat Distribution and Impairs Glycemic Control. EBioMedicine 2016; 11:101-117. [PMID: 27568222 PMCID: PMC5049998 DOI: 10.1016/j.ebiom.2016.08.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 12/17/2022] Open
Abstract
RORα is a member of the nuclear receptor (NR) superfamily and analysis of the (global) RORα-deficient mouse model revealed this NR has a role in glycemic control and fat deposition. Therefore, we generated an adipose-specific RORα ‘gain of function’ mouse model under the control of the fatty acid binding protein 4 (FABP4) promoter to elucidate the function of RORα in adipose tissue. The Tg-FABP4-RORα4 mice demonstrated a shift in fat distribution to non-adipose tissues when challenged with a high fat diet (HFD). Specifically, we observed a subcutaneous lipodystrophy, accompanied by hepatomegaly (fatty liver/mild portal fibrosis) and splenomegaly; in a background of decreased weight gain and total body fat after HFD. Moreover, we observed significantly higher fasting blood glucose and impaired clearance of glucose in Tg-FABP4-RORα4 mice. Genome wide expression and qPCR profiling analysis identified: (i) subcutaneous adipose specific decreases in the expression of genes involved in fatty acid biosynthesis, lipid droplet expansion and glycemic control, and (ii) the fibrosis pathway as the most significant pathway [including dysregulation of the collagen/extracellular matrix (ECM) pathways] in subcutaneous adipose and liver. The pathology presented in the Tg-FABP4-RORα4 mice is reminiscent of human metabolic disease (associated with aberrant ECM expression) highlighting the therapeutic potential of this NR. Adipose-specific expression of RORα is associated with subcutaneous lipodystrophy and hepatomegaly with fibrosis. The phenotype is associated with impaired glycemic control and decreased weight gain on a high fat diet. Gene expression profiling reveals significant dysregulation of extra cellular matrix signaling.
We have generated a ‘gain of function’ animal model with the nuclear hormone receptor RORα4 to understand the function of this protein in fat. Over expression of the RORα4 gene, was associated with fat deposition in non-adipose tissues on a high fat diet. Moreover, we observed a decrease in fat tissue (located under the skin) accompanied by enlargement of the liver and spleen. In addition, over expression of this receptor was associated with impaired glycemic control. The pathology in this animal model is reminiscent of metabolic disease in humans, highlighting the therapeutic potential of pharmacologically manipulating this nuclear receptor.
Collapse
|
7
|
Das UN. Molecular, Biochemical, and Physiological Basis of Beneficial Actions of Exercise. DIET AND EXERCISE IN COGNITIVE FUNCTION AND NEUROLOGICAL DISEASES 2015:183-204. [DOI: 10.1002/9781118840634.ch17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Abstract
The nuclear receptors REV-ERB (consisting of REV-ERBα and REV-ERBβ) and retinoic acid receptor-related orphan receptors (RORs; consisting of RORα, RORβ and RORγ) are involved in many physiological processes, including regulation of metabolism, development and immunity as well as the circadian rhythm. The recent characterization of endogenous ligands for these former orphan nuclear receptors has stimulated the development of synthetic ligands and opened up the possibility of targeting these receptors to treat several diseases, including diabetes, atherosclerosis, autoimmunity and cancer. This Review focuses on the latest developments in ROR and REV-ERB pharmacology indicating that these nuclear receptors are druggable targets and that ligands targeting these receptors may be useful in the treatment of several disorders.
Collapse
|
9
|
Schiano C, Casamassimi A, Vietri MT, Rienzo M, Napoli C. The roles of mediator complex in cardiovascular diseases. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1839:444-51. [PMID: 24751643 DOI: 10.1016/j.bbagrm.2014.04.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/19/2014] [Accepted: 04/11/2014] [Indexed: 02/08/2023]
Abstract
Despite recent treatment advances, an increase in cardiovascular diseases (CVD) mortality is expected for the next years. Mediator (MED) complex plays key roles in eukaryotic gene transcription. Currently, while numerous studies have correlated MED alterations with several diseases, like cancer or neurological disorders, fewer studies have investigated MED role in CVD initiation and progression. The first finding of MED involvement in these pathologies was the correlation of missense mutations in MED13L gene with transposition of the great arteries. Nowadays, also MED13 and MED15 have been associated with human congenital heart diseases and others could be added, like MED12 that is involved in early mouse development and heart formation. Interestingly, a missense mutation in MED30 gene causes a progressive cardiomyopathy in homozygous mice suggesting a potential role for this subunit also in human CVDs. Moreover, several subunits like MED1, MED13, MED14, MED15, MED23, MED25 and CDK8 exert important roles in glucose and lipid metabolism. Although these evidences derive from in vitro and animal model studies, they indicate that their deregulation may have a significant role in human CVD-related metabolic disorders. Finally, alternative transcripts of MED12, MED19 and MED30 are differently expressed in circulating endothelial progenitor cells thus suggesting they can play a role in the field of regenerative medicine. Overall, further functional studies exploring MED role in human CVD are warranted. The results could allow identifying novel biomarkers to use in combination with imaging techniques for early diagnosis; otherwise, they could be useful to develop targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Concetta Schiano
- Institute of Diagnostic and Nuclear Development (SDN), IRCCS, Via E. Gianturco 113, 80143 Naples, Italy
| | - Amelia Casamassimi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Maria Teresa Vietri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138 Naples, Italy
| | - Monica Rienzo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138 Naples, Italy
| | - Claudio Napoli
- Institute of Diagnostic and Nuclear Development (SDN), IRCCS, Via E. Gianturco 113, 80143 Naples, Italy; Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138 Naples, Italy; U.O.C. Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU), 1st School of Medicine, Second University of Naples, Piazza Miraglia 2, 80138 Naples, Italy
| |
Collapse
|
10
|
Shashni B, Sharma K, Singh R, Sakharkar KR, Dhillon SK, Nagasaki Y, Sakharkar MK. Coffee component hydroxyl hydroquinone (HHQ) as a putative ligand for PPAR gamma and implications in breast cancer. BMC Genomics 2013; 14 Suppl 5:S6. [PMID: 24564733 PMCID: PMC3852186 DOI: 10.1186/1471-2164-14-s5-s6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Coffee contains several compounds that have the potential to influence breast cancer risk and survival. However, epidemiologic data on the relation between coffee compounds and breast cancer survival are sparse and inconsistent. Results We show that coffee component HHQ has significant apoptotic effect on MDA-MB-231 and MCF-7 cells in vitro, and that ROS generation, change in mitochondrial membrane permeability, upregulation of Bax and Caspase-8 as well as down regulation of PGK1 and PKM2 expression may be important apoptosis-inducing mechanisms. The results suggest that PPARγ ligands may serve as potential therapeutic agents for breast cancer therapy. HHQ was also validated as a ligand for PPARγ by docking procedure. Conclusion This is the first report on the anti-breast cancer (in vitro) activity of HHQ.
Collapse
|
11
|
Wang SCM, Muscat GEO. Nuclear receptors and epigenetic signaling: novel regulators of glycogen metabolism in skeletal muscle. IUBMB Life 2013; 65:657-64. [PMID: 23846999 DOI: 10.1002/iub.1181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/18/2013] [Indexed: 02/04/2023]
Abstract
Glycogen is an energy storage depot for the mammalian species. This review focuses on recent developments that have identified the role of nuclear hormone receptor (NR) signaling and epigenomic control in the regulation of important genes that modulate glycogen metabolism. Specifically, new studies have revealed that the NR4A subgroup (of the NR superfamily) are strikingly sensitive to beta-adrenergic stimulation in skeletal muscle, and transgenic studies in mice have revealed the expression of these NRs affects endurance and glycogen levels in muscle. Furthermore, other studies have demonstrated that one of the NR coregulator class of enzymes that mediate chromatin remodeling, the histone methyltransferases (for example, protein arginine methyltransferase 4) regulates the expression of several genes involved in glycogen metabolism and glycogen storage diseases in skeletal muscle. Importantly, NRs and histone methyltransferases, have the potential to be pharmacologically exploited and may provide novel targets in the quest to treat disorders of glycogen storage.
Collapse
Affiliation(s)
- Shu-Ching Mary Wang
- The University of Queensland, Institute for Molecular Bioscience, Obesity Research Centre, Australia
| | | |
Collapse
|
12
|
Sakharkar MK, Shashni B, Sharma K, Dhillon SK, Ranjekar PR, Sakharkar KR. Therapeutic implications of targeting energy metabolism in breast cancer. PPAR Res 2013; 2013:109285. [PMID: 23431283 PMCID: PMC3575613 DOI: 10.1155/2013/109285] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/23/2012] [Indexed: 12/12/2022] Open
Abstract
PPARs are ligand activated transcription factors. PPARγ agonists have been reported as a new and potentially efficacious treatment of inflammation, diabetes, obesity, cancer, AD, and schizophrenia. Since cancer cells show dysregulation of glycolysis they are potentially manageable through changes in metabolic environment. Interestingly, several of the genes involved in maintaining the metabolic environment and the central energy generation pathway are regulated or predicted to be regulated by PPARγ. The use of synthetic PPARγ ligands as drugs and their recent withdrawal/restricted usage highlight the lack of understanding of the molecular basis of these drugs, their off-target effects, and their network. These data further underscores the complexity of nuclear receptor signalling mechanisms. This paper will discuss the function and role of PPARγ in energy metabolism and cancer biology in general and its emergence as a promising therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Meena K. Sakharkar
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8572, Japan
| | - Babita Shashni
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8572, Japan
| | - Karun Sharma
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba 305-8572, Japan
| | - Sarinder K. Dhillon
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Kishore R. Sakharkar
- Omicsvista, Singapore 120417
- Rajiv Gandhi Institute of Information Technology and Biotechnology, Bharati Vidyapeeth University, Pune 411046, India
| |
Collapse
|
13
|
Shashni B, Sakharkar KR, Nagasaki Y, Sakharkar MK. Glycolytic enzymes PGK1 and PKM2 as novel transcriptional targets of PPARγ in breast cancer pathophysiology. J Drug Target 2013; 21:161-74. [PMID: 23130662 DOI: 10.3109/1061186x.2012.736998] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor and plays important roles in breast cancer cell proliferation. The complexity of the underlying biochemical and molecular mechanisms of breast cancer and the involvement of PPARγ in breast cancer pathophysiology are unclear. In this study, we carried out prediction of the peroxisome proliferator response element (PPRE) motifs in 2332 genes reported to be involved in breast cancer in literature. A total of 178 genes were found to have PPRE (DR1/DR2) and/or PPAR-associated conserved motif (PACM) motifs. We further constructed protein-protein interaction network, disease gene network and gene ontology (GO) analyses to identify novel key genes for experimental validation. We identified two genes in the glycolytic pathway (phosphoglycerate kinase 1 (PGK1) and pyruvate kinase M2 (PKM2)) at the ATP production steps and experimentally validated their repression by PPARγ in two breast cancer cell lines MDA-MB-231 and MCF-7. Further analysis suggested that this repression leads to decrease in ATP levels and apoptosis. These investigations will help us in understanding the molecular mechanisms by which PPARγ regulates the cellular energy pathway and the use of its ligands in human breast cancer therapeutics.
Collapse
Affiliation(s)
- Babita Shashni
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | |
Collapse
|
14
|
Cruz-Garcia L, Sánchez-Gurmaches J, Gutiérrez J, Navarro I. Role of LXR in trout adipocytes: target genes, hormonal regulation, adipocyte differentiation and relation to lipolysis. Comp Biochem Physiol A Mol Integr Physiol 2012; 163:120-6. [PMID: 22626869 DOI: 10.1016/j.cbpa.2012.05.193] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 05/16/2012] [Accepted: 05/16/2012] [Indexed: 11/18/2022]
Abstract
In the present study, we describe an initial approach to investigate the role of LXR in fish adipose tissue. Rainbow trout (Oncorhynchus mykiss) isolated adipocytes were incubated with LXR agonists, unsaturated fatty acids, tumour necrosis factor-α (TNFα), insulin or growth hormone (GH) for 6h and LXR expression was analyzed. Lipolysis was measured after incubation with one of the LXR agonists and LXR expression was compared with levels of lipolysis. LXR expression was also analyzed during the differentiation of adipocytes in culture. The incubations with agonists in isolated adipocytes indicated that ATP-binding cassette transporter A1 (ABCA1) is an LXR target gene, but lipoprotein lipase (LPL), fatty acid synthase (FAS), hormone-sensitive lipase (HSL) and peroxisome proliferator-activated receptor (PPARs) are not. LXR agonists also induced LXR expression and raised lipolysis levels. Besides, LXR expression was upregulated in parallel with basal lipolysis. LXR mRNA expression was regulated by unsaturated fatty acids, insulin, TNFα and GH in isolated adipocytes. Besides, LXR showed an upregulation during adipocyte differentiation. All these data indicate that LXR is involved in orchestrating the transcriptional regulatory network in trout adipocyte lipid metabolism, specifically, in cholesterol transport, adipocyte differentiation and lipolysis.
Collapse
Affiliation(s)
- Lourdes Cruz-Garcia
- Departament de Fisiologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain.
| | | | | | | |
Collapse
|
15
|
Lau P, Fitzsimmons RL, Pearen MA, Watt MJ, Muscat GEO. Homozygous staggerer (sg/sg) mice display improved insulin sensitivity and enhanced glucose uptake in skeletal muscle. Diabetologia 2011; 54:1169-80. [PMID: 21279323 PMCID: PMC3071927 DOI: 10.1007/s00125-011-2046-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 12/03/2010] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Homozygous staggerer (sg/sg) mice, which have decreased and dysfunctional Rorα (also known as Rora) expression in all tissues, display a lean and dyslipidaemic phenotype. They are also resistant to (high fat) diet-induced obesity. We explored whether retinoic acid receptor-related orphan receptor (ROR) α action in skeletal muscle was involved in the regulation of glucose metabolism. METHODS We used a three-armed genomic approach, including expression profiling, ingenuity analysis and quantitative PCR validation to identify the signalling pathway(s) in skeletal muscle that are perturbed in sg/sg mice. Moreover, western analysis, functional insulin and glucose tolerance tests, and ex vivo glucose uptake assays were used to phenotypically characterise the impact of aberrant v-AKT murine thymoma viral oncogene homologue (AKT) signalling. RESULTS Homozygous and heterozygous (sg/sg and sg/+) animals exhibited decreased fasting blood glucose levels, mildly improved glucose tolerance and increased insulin sensitivity. Illumina expression profiling and bioinformatic analysis indicated the involvement of RORα in metabolic disease and phosphatidylinositol 3-kinase-AKT signalling. Quantitative PCR and western analysis validated increased AKT2 (mRNA and protein) and phosphorylation in sg/sg mice in the basal state. This was associated with increased expression of Tbc1d1 and Glut4 (also known as Slc2a4) mRNA and protein. Finally, in agreement with the phenotype, we observed increased (absolute) levels of AKT and phosphorylated AKT (in the basal and insulin stimulated states), and of (ex vivo) glucose uptake in skeletal muscle from sg/sg mice relative to wild-type littermates. CONCLUSIONS/INTERPRETATION We propose that Rorα plays an important role in regulation of the AKT2 signalling cascade, which controls glucose uptake in skeletal muscle.
Collapse
Affiliation(s)
- P. Lau
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Services Rd, St Lucia, Queensland, 4072 Australia
| | - R. L. Fitzsimmons
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Services Rd, St Lucia, Queensland, 4072 Australia
| | - M. A. Pearen
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Services Rd, St Lucia, Queensland, 4072 Australia
| | - M. J. Watt
- Department of Physiology, Monash University, Clayton, VIC Australia
| | - G. E. O. Muscat
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Services Rd, St Lucia, Queensland, 4072 Australia
| |
Collapse
|
16
|
Baranowski M, Zabielski P, Błachnio-Zabielska AU, Harasiuk D, Górski J. LXR activation prevents exhaustive exercise-induced hypoglycaemia and spares muscle glycogen but does not enhance running endurance in untrained rats. Acta Physiol (Oxf) 2011; 201:373-9. [PMID: 20887359 DOI: 10.1111/j.1748-1716.2010.02199.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM Liver X receptors (LXRs) are ligand-activated transcription factors that play an important role in regulation of hepatic lipid and carbohydrate metabolism. However, to date there is very few information on the role of LXRs in skeletal muscle. Moreover, it remains obscure whether LXR activation affects physical endurance. Therefore, we aimed to examine effects of selective LXR activator--T0901317--on running endurance and skeletal muscle exercise metabolism in rats. METHODS The animals were assigned to two groups (n=20) receiving either vehicle or T0901317 (10 mg kg(-1) day(-1) ) for 1 week. One day after the final administration, half of the rats in each group were exercised until exhaustion on the electrically driven treadmill. All animals were then anaesthetized and samples of the soleus, red and white sections of the gastrocnemius muscle, epididymal fat pad and liver were excised. RESULTS We found that LXR activation prevented exhaustive exercise-induced hypoglycaemia. T0901317 also shifted substrate utilization in working muscles in favour of fatty acids as indicated by its glycogen sparing effect, enhanced consumption of intramuscular triacylglycerol and upregulation of genes promoting fatty acid oxidation and suppressing carbohydrate oxidation. However, running time to exhaustion was not improved. CONCLUSION We conclude that LXR activation increases fatty acid utilization during exercise which, however, does not translate into measurable enhancement of exercise endurance.
Collapse
Affiliation(s)
- M Baranowski
- Department of Physiology, Medical University of Białystok, Mickiewicza 2c, Białystok, Poland.
| | | | | | | | | |
Collapse
|
17
|
Crowther LM, Wang SCM, Eriksson NA, Myers SA, Murray LA, Muscat GEO. Chicken ovalbumin upstream promoter-transcription factor II regulates nuclear receptor, myogenic, and metabolic gene expression in skeletal muscle cells. Physiol Genomics 2010; 43:213-27. [PMID: 21119012 DOI: 10.1152/physiolgenomics.00195.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We demonstrate that chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) mRNA is more abundantly expressed (than COUP-TFI mRNA) in skeletal muscle C2C12 cells and in (type I and II) skeletal muscle tissue from C57BL/10 mice. Consequently, we have utilized the ABI TaqMan Low Density Array (TLDA) platform to analyze gene expression changes specifically attributable to ectopic COUP-TFII (relative to vector only) expression in muscle cells. Utilizing a TLDA-based platform and 5 internal controls, we analyze the entire NR superfamily, 96 critical metabolic genes, and 48 important myogenic regulatory genes on the TLDA platform utilizing 5 internal controls. The low density arrays were analyzed by rigorous statistical analysis (with Genorm normalization, Bioconductor R, and the Empirical Bayes statistic) using the (integromics) statminer software. In addition, we validated the differentially expressed patho-physiologically relevant gene (identified on the TLDA platform) glucose transporter type 4 (Glut4). We demonstrated that COUP-TFII expression increased the steady state levels of Glut4 mRNA and protein, while ectopic expression of truncated COUP-TFII lacking helix 12 (COUP-TFΔH12) reduced Glut4 mRNA expression in C2C12 cells. Moreover, COUP-TFII expression trans-activated the Glut4 promoter (-997/+3), and ChIP analysis identified selective recruitment of COUP-TFII to a region encompassing a highly conserved SP1 binding site (in mouse, rat, and human) at nt positions -131/-118. Mutation of the SpI site ablated COUP-TFII mediated trans-activation of the Glut4 promoter. In conclusion, this study demonstrates that in skeletal muscle cells, COUP-TFII regulates several nuclear hormone receptors, and critical metabolic and muscle specific genes.
Collapse
Affiliation(s)
- Lisa M Crowther
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
18
|
Lewis GD, Farrell L, Wood MJ, Martinovic M, Arany Z, Rowe GC, Souza A, Cheng S, McCabe EL, Yang E, Shi X, Deo R, Roth FP, Asnani A, Rhee EP, Systrom DM, Semigran MJ, Vasan RS, Carr SA, Wang TJ, Sabatine MS, Clish CB, Gerszten RE. Metabolic signatures of exercise in human plasma. Sci Transl Med 2010; 2:33ra37. [PMID: 20505214 DOI: 10.1126/scitranslmed.3001006] [Citation(s) in RCA: 309] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Exercise provides numerous salutary effects, but our understanding of how these occur is limited. To gain a clearer picture of exercise-induced metabolic responses, we have developed comprehensive plasma metabolite signatures by using mass spectrometry to measure >200 metabolites before and after exercise. We identified plasma indicators of glycogenolysis (glucose-6-phosphate), tricarboxylic acid cycle span 2 expansion (succinate, malate, and fumarate), and lipolysis (glycerol), as well as modulators of insulin sensitivity (niacinamide) and fatty acid oxidation (pantothenic acid). Metabolites that were highly correlated with fitness parameters were found in subjects undergoing acute exercise testing and marathon running and in 302 subjects from a longitudinal cohort study. Exercise-induced increases in glycerol were strongly related to fitness levels in normal individuals and were attenuated in subjects with myocardial ischemia. A combination of metabolites that increased in plasma in response to exercise (glycerol, niacinamide, glucose-6-phosphate, pantothenate, and succinate) up-regulated the expression of nur77, a transcriptional regulator of glucose utilization and lipid metabolism genes in skeletal muscle in vitro. Plasma metabolic profiles obtained during exercise provide signatures of exercise performance and cardiovascular disease susceptibility, in addition to highlighting molecular pathways that may modulate the salutary effects of exercise.
Collapse
Affiliation(s)
- Gregory D Lewis
- Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Peltzer J, Carpentier G, Martelly I, Courty J, Keller A. Transitions towards either slow-oxidative or fast-glycolytic phenotype can be induced in the murine WTt myogenic cell line. J Cell Biochem 2010; 111:82-93. [DOI: 10.1002/jcb.22665] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Lessard SJ, Rivas DA, Chen ZP, van Denderen BJ, Watt MJ, Koch LG, Britton SL, Kemp BE, Hawley JA. Impaired skeletal muscle beta-adrenergic activation and lipolysis are associated with whole-body insulin resistance in rats bred for low intrinsic exercise capacity. Endocrinology 2009; 150:4883-91. [PMID: 19819977 PMCID: PMC2775978 DOI: 10.1210/en.2009-0158] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Rats selectively bred for high endurance running capacity (HCR) have higher insulin sensitivity and improved metabolic health compared with those bred for low endurance capacity (LCR). We investigated several skeletal muscle characteristics, in vitro and in vivo, that could contribute to the metabolic phenotypes observed in sedentary LCR and HCR rats. After 16 generations of selective breeding, HCR had approximately 400% higher running capacity (P < 0.001), improved insulin sensitivity (P < 0.001), and lower fasting plasma glucose and triglycerides (P < 0.05) compared with LCR. Skeletal muscle ceramide and diacylglycerol content, basal AMP-activated protein kinase (AMPK) activity, and basal lipolysis were similar between LCR and HCR. However, the stimulation of lipolysis in response to 10 mum isoproterenol was 70% higher in HCR (P = 0.004). Impaired isoproterenol sensitivity in LCR was associated with lower basal triacylglycerol lipase activity, Ser660 phosphorylation of HSL, and beta2-adrenergic receptor protein content in skeletal muscle. Expression of the orphan nuclear receptor Nur77, which is induced by beta-adrenergic signaling and is associated with insulin sensitivity, was lower in LCR (P < 0.05). Muscle protein content of Nur77 target genes, including uncoupling protein 3, fatty acid translocase/CD36, and the AMPK gamma3 subunit were also lower in LCR (P < 0.05). Our investigation associates whole-body insulin resistance with impaired beta-adrenergic response and reduced expression of genes that are critical regulators of glucose and lipid metabolism in skeletal muscle. We identify impaired beta-adrenergic signal transduction as a potential mechanism for impaired metabolic health after artificial selection for low intrinsic exercise capacity.
Collapse
Affiliation(s)
- Sarah J Lessard
- Joslin Diabetes Center, 1 Joslin Place, Boston, Massachusetts 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Copulation in C. elegans males requires a nuclear hormone receptor. Dev Biol 2008; 322:11-20. [DOI: 10.1016/j.ydbio.2008.06.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Revised: 06/20/2008] [Accepted: 06/27/2008] [Indexed: 11/20/2022]
|
22
|
Magner DB, Antebi A. Caenorhabditis elegans nuclear receptors: insights into life traits. Trends Endocrinol Metab 2008; 19:153-60. [PMID: 18406164 PMCID: PMC2744080 DOI: 10.1016/j.tem.2008.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 02/13/2008] [Accepted: 02/20/2008] [Indexed: 10/22/2022]
Abstract
Nuclear receptors are a class of hormone-gated transcription factors found in metazoans that regulate global changes in gene expression when bound to their cognate ligands. Despite species diversification, nuclear receptors function similarly across taxa, having fundamental roles in detecting intrinsic and environmental signals, and subsequently in coordinating transcriptional cascades that direct reproduction, development, metabolism and homeostasis. These endocrine receptors function in vivo in part as molecular switches and timers that regulate transcriptional cascades. Several Caenorhabditis elegans nuclear receptors integrate intrinsic and extrinsic signals to regulate the dauer diapause and longevity, molting, and heterochronic circuits of development, and are comparable to similar in vivo endocrine regulated processes in other animals.
Collapse
Affiliation(s)
| | - Adam Antebi
- Corresponding author: Antebi, A. (), Tel: 713-798-6661; Fax: 713-798-4161
| |
Collapse
|
23
|
Smith AG, Luk N, Newton RA, Roberts DW, Sturm RA, Muscat GEO. Melanocortin-1 receptor signaling markedly induces the expression of the NR4A nuclear receptor subgroup in melanocytic cells. J Biol Chem 2008; 283:12564-70. [PMID: 18292087 DOI: 10.1074/jbc.m800480200] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The melanocortin-1 receptor (MCIR) is a G-protein-coupled receptor expressed primarily in melanocytes and is known to play a pivotal role in the regulation of pigmentation in mammals. In humans MC1R has been found to be highly polymorphic with several functional variants associated with the phenotype of red hair color and fair skin, cutaneous UV sensitivity, and increased risk of developing melanoma and non-melanoma skin cancer. Recent evidence suggests that MC1R plays a photo-protective role in melanocytes in response to UV irradiation. Relatively few genetic targets of MC1R signaling have been identified independent of the pigmentation pathway. Here we show that MC1R signaling in B16 mouse melanoma cells and primary human melanocytes rapidly, and transiently, induces the transcription of the NR4A subfamily of orphan nuclear receptors. Furthermore, primary human melanocytes harboring homozygous RHC variant MC1R alleles exhibited an impaired induction of NR4A genes in response to the potent MC1R agonist (Nle4,D-Phe7)-alpha-melanocyte-stimulating hormone. Using small interference RNA-mediated attenuation of NR4A1 and NR4A2 expression in melanocytes, the ability to remove cyclobutane pyrimidine dimers following UV irradiation appeared to be impaired in the context of MC1R signaling. These data identify the NR4A receptor family as potential mediators of an MC1R-coordinated DNA damage response to UV exposure in melanocytic cells.
Collapse
Affiliation(s)
- Aaron G Smith
- Division of Molecular Genetics and Development, Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland 4072, Australia.
| | | | | | | | | | | |
Collapse
|
24
|
Burris TP. Nuclear hormone receptors for heme: REV-ERBalpha and REV-ERBbeta are ligand-regulated components of the mammalian clock. Mol Endocrinol 2008; 22:1509-20. [PMID: 18218725 DOI: 10.1210/me.2007-0519] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The nuclear hormone receptors (NHRs), REV-ERBalpha and REV-ERBbeta, regulate a number of physiological functions including the circadian rhythm, lipid metabolism, and cellular differentiation. These two receptors lack the activation function-2 region that is associated with the ability of NHRs to recruit coactivators and activate target gene transcription. These NHRs have been characterized as constitutive repressors of transcription due to their lack of an identified ligand and their strong ability to recruit the corepressor, nuclear receptor corepressor. Recently, the porphyrin heme was demonstrated to function as a ligand for both REV-ERBs. Heme binds directly to the ligand-binding domain and regulates the ability of these NHRs to recruit nuclear receptor corepressor to target gene promoters. This review focuses on the physiological roles that these two receptors play and the implications of heme functioning as their ligand. The prospect that these NHRs, now known to be regulated by small molecule ligands, may be targets for development of drugs for treatment of diseases associated with aberrant circadian rhythms including metabolic and psychiatric disorders as well as cancer is also addressed.
Collapse
Affiliation(s)
- Thomas P Burris
- Nuclear Receptor Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| |
Collapse
|
25
|
The MTHFR gene polymorphism is associated with lean body mass but not fat body mass. Hum Genet 2008; 123:189-96. [DOI: 10.1007/s00439-007-0463-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Accepted: 12/23/2007] [Indexed: 01/25/2023]
|
26
|
Valer Carstea B, Catunda Lemos AP, Ilie ED, Varga L, Bodó S, Kovács A, Bösze Z, Gócza E. Production of identical mouse twins and a triplet with predicted gender. CLONING AND STEM CELLS 2007; 9:247-56. [PMID: 17579557 DOI: 10.1089/clo.2006.0055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The aim of this study was to develop a method to generate identical twins and triplets with predicted gender. As a first step toward that aim, single blastomeres obtained from EGFP expressing eight-cell stage embryos and either diploid or tetraploid host embryos were used to compose chimera. We could follow the fate of EGFP expressing diploid blastomere derived cells in 3.5- and 4.5-day-old chimera embryos in vitro. We found that the diploid blastomere-derived cells had significantly higher chance to contribute to the inner cell mass if tetraploid host embryos were applied. After that, we developed a quick and reliable multiplex PCR strategy for sex diagnosis from single blastomeres by simultaneous amplification of the homologous ZFX and ZFY genes. By composed chimeras using single blastomeres, derived from sexed eight-cell stage embryos and a tetraploid host embryo, we could get preplanned sex newborns, wholly derived from these blastomeres. Among these mice, identical twins and a triplet were identified by microsatellite analysis. Unlike clones produced by nuclear transfer, these mice are identical at both the nuclear as well as mitochondrial DNA level. Therefore, the tetraploid embryo complementation method to produce monozygotic twins and triplets could be a valuable tool both in biomedical and agricultural applications.
Collapse
Affiliation(s)
- Bogdan Valer Carstea
- In vivo Gene Expression and Regulation Group, Agricultural Biotechnology Center, Gödöllö, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Shavlakadze T, Grounds M. Of bears, frogs, meat, mice and men: complexity of factors affecting skeletal muscle mass and fat. Bioessays 2006; 28:994-1009. [PMID: 16998828 DOI: 10.1002/bies.20479] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extreme loss of skeletal muscle mass (atrophy) occurs in human muscles that are not used. In striking contrast, skeletal muscles do not rapidly waste away in hibernating mammals such as bears, or aestivating frogs, subjected to many months of inactivity and starvation. What factors regulate skeletal muscle mass and what mechanisms protect against muscle atrophy in some species? Severe atrophy also occurs with ageing and there is much clinical interest in reducing such loss of muscle mass and strength (sarcopenia). In the meat industry, a key aim is optimizing the control of skeletal muscle growth and meat quality. The impaired response of muscle to insulin resulting in diabetes, that is a consequence of the metabolic impact of increasing obesity and fat deposition in humans, is also of increasing clinical concern. Intensive research in these fields, combined with mouse models, is reviewed with respect to the molecular control of muscle growth (myogenesis) and atrophy/hypertrophy and fat deposition (adipogenesis) in skeletal muscle, with a focus on IGF-1/insulin signaling.
Collapse
Affiliation(s)
- Thea Shavlakadze
- School of Anatomy and Human Biology, The University of Western Australia, 6009, Western Australia.
| | | |
Collapse
|