1
|
Gao Y, Hossain M, Zhao L, Liu X, Chen Y, Deavila J, Zhu M, Murdoch G, Du M. Balancing LncRNA H19 and miR-675 Bioconversion as a Key Regulator of Embryonic Myogenesis Under Maternal Obesity. J Cachexia Sarcopenia Muscle 2025; 16:e13791. [PMID: 40162559 PMCID: PMC11955836 DOI: 10.1002/jcsm.13791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Maternal obesity (MO) impairs fetal skeletal muscle development, but the underlying mechanisms remain poorly defined. The regulatory roles of lncRNA H19 and its first exon derived microRNA675 (miR675) in prenatal muscle development remain to be examined. H19/Igf2 are in the same imprinting cluster with H19 expressed from the maternal allele while Igf2 expresses paternally. H19 contains a G-rich loop, and KH-type splicing regulatory protein (KHSRP) mediates the biogenesis of pre-miRNAs containing G-rich loops, which depends on its phosphorylation by AKT, a key mediator of IGF2 signalling. This study aims to depict the elusive function of these regulators that are affected by MO during embryonic myogenesis. METHODS Single-cell transcriptomic sequencing and GeoMx spatial RNA sequencing were performed to identify the differentially expressed genes between embryos from MO and control (CT) mice. Both E11.5 and E13.5 embryos were collected and analysed to validate the sequencing data. The roles of H19 and miR657 in myogenesis were further analysed in P19 embryonic cells via CRISPR/dCas9-mediated H19 activation and inhibition. The epigenetic changes of H19 were analysed by methylated DNA immunoprecipitation, and allele-targeted analysis of H19 was performed by crossing C57BL/6J and CAST/EiJ mice. RESULTS Transcriptomic analysis showed that MO embryos contained less differentiated myocytes (1.34%) than CT embryos (2.86%). Myogenesis-related GO biological processes were down-regulated in the MO embryonic myotome region. MO embryos showed lower expression of myogenic transcription factors such as Myf5, Myod1, Myog, Mef2c and Myh3 (p < 0.05). MO altered epigenetic modifications of the H19 genomic cluster, showing a decreased methylation level in H19 imprinting control region (p < 0.05) and a diallelic expression pattern of H19, which elevated its expression in MO embryos. Overexpression of H19 inhibited myogenesis in P19 cells, but miR675 promoted myogenesis, suggesting the critical regulatory roles of bioconversion of H19 to miR675. A KHSRP mediates the biogenesis of miR675, a process that relies on its phosphorylation by IGF2/AKT signalling. Knocking-down of KHSRP and inhibition of AKT abolished miR675 biogenesis. MO suppressed IGF2/AKT signalling and blocked KHSRP-dependent miR675 biogenesis in embryos. CONCLUSIONS We found differential effects of H19 and miR675 on embryonic myogenesis. MO up-regulates H19 but blocks its miR675 bioconversion via suppressing IGF2/AKT/KHSRP signalling axis. Myogenesis in MO embryos was impeded due to the highly accumulated H19 and blocked miR675 biogenesis.
Collapse
Affiliation(s)
- Yao Gao
- Nutrigenomics and Growth Biology Laboratory, Department of Animal SciencesWashington State UniversityPullmanWashingtonUSA
| | - Md Nazmul Hossain
- Nutrigenomics and Growth Biology Laboratory, Department of Animal SciencesWashington State UniversityPullmanWashingtonUSA
| | - Liang Zhao
- College of Animal Science and TechnologyNanjing Agricultural UniversityNanjingJiangsuChina
| | | | - Yanting Chen
- College of Animal Science and TechnologyNanjing Agricultural UniversityNanjingJiangsuChina
| | - Jeanene Marie Deavila
- Nutrigenomics and Growth Biology Laboratory, Department of Animal SciencesWashington State UniversityPullmanWashingtonUSA
| | - Mei‐Jun Zhu
- School of Food ScienceWashington State UniversityPullmanWashingtonUSA
| | - Gordon K. Murdoch
- Nutrigenomics and Growth Biology Laboratory, Department of Animal SciencesWashington State UniversityPullmanWashingtonUSA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal SciencesWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
2
|
Ben-Zaken S, Meckel Y, Remmel L, Nemet D, Jürimäe J, Eliakim A. The prevalence of IGF-I axis genetic polymorphisms among decathlon athletes. Growth Horm IGF Res 2022; 64:101468. [PMID: 35605565 DOI: 10.1016/j.ghir.2022.101468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Decathlon is a combined track and field competition, consisting of ten, mainly anaerobic events. Insulin-like growth factor-I (IGF1) axis plays a pivotal role in athletes' structural and functional muscle adaptation to exercise training, and in their competitive performance. Based on the great demand for speed physiological characteristics among decathlon athletes, the aim of this study was to assess the prevalence of IGF genetic polymorphisms among decathletes, to present an optimal genetic profile for enhancing performance. METHODS The participants included 151 male athletes and 75 male non-athletic controls from Israel and Estonia. Athletes were divided into four groups, according to the field of expertise: (a) 40 sprinters and long jumpers; (b) 40 middle distance runners; (c) 44 Weightlifters; and (d) 27 decathletes. Genomic DNA was extracted from the participants' buccal epithelial cells using standard protocol and then analyzed for IGF1 axis related genetic polymorphism using the allelic discrimination assay. RESULTS A significantly higher prevalence of the IGF1 rs35767 TT genotype was found among decathletes compared to the other athletes, as well as a lower prevalence of the IGF1 rs7136446 GG genotype, a higher prevalence of the IGF1R rs1464430 AA genotype, and a higher prevalence of the IGF2 rs680 GG genotype. Moreover, among the decathletes, carriers of the IGF1 rs7136446 GG genotype achieved higher decathlon scores compared to A-allele carriers. CONCLUSIONS The findings of this study suggest a potential beneficial role for some IGF-axis polymorphisms (mainly the IGF1 1245 TT and the IGF2 GG) among decathletes, both of which are associated with improved speed performance.
Collapse
Affiliation(s)
- Sigal Ben-Zaken
- Genetics and Molecular Biology Laboratory, The Academic College for Physical Education and Sports Sciences at the Wingate Institute, Netanya 42902, Israel.
| | - Yoav Meckel
- Genetics and Molecular Biology Laboratory, The Academic College for Physical Education and Sports Sciences at the Wingate Institute, Netanya 42902, Israel
| | | | - Dan Nemet
- Child Health and Sports Center, Pediatric Department, Meir Medical Center, Kfar-Saba, Israel
| | | | - Alon Eliakim
- Genetics and Molecular Biology Laboratory, The Academic College for Physical Education and Sports Sciences at the Wingate Institute, Netanya 42902, Israel; Child Health and Sports Center, Pediatric Department, Meir Medical Center, Kfar-Saba, Israel
| |
Collapse
|
3
|
Katoku-Kikyo N, Paatela E, Houtz DL, Lee B, Munson D, Wang X, Hussein M, Bhatia J, Lim S, Yuan C, Asakura Y, Asakura A, Kikyo N. Per1/Per2-Igf2 axis-mediated circadian regulation of myogenic differentiation. J Cell Biol 2021; 220:212164. [PMID: 34009269 PMCID: PMC8138781 DOI: 10.1083/jcb.202101057] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 01/02/2023] Open
Abstract
Circadian rhythms regulate cell proliferation and differentiation, but circadian control of tissue regeneration remains elusive at the molecular level. Here, we show that proper myoblast differentiation and muscle regeneration are regulated by the circadian master regulators Per1 and Per2. Depletion of Per1 or Per2 suppressed myoblast differentiation in vitro and muscle regeneration in vivo, demonstrating their nonredundant functions. Both Per1 and Per2 were required for the activation of Igf2, an autocrine promoter of myoblast differentiation, accompanied by Per-dependent recruitment of RNA polymerase II, dynamic histone modifications at the Igf2 promoter and enhancer, and the promoter–enhancer interaction. This circadian epigenetic priming created a preferred time window for initiating myoblast differentiation. Consistently, muscle regeneration was faster if initiated at night, when Per1, Per2, and Igf2 were highly expressed compared with morning. This study reveals the circadian timing as a significant factor for effective muscle cell differentiation and regeneration.
Collapse
Affiliation(s)
- Nobuko Katoku-Kikyo
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN
| | - Ellen Paatela
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN
| | - Daniel L Houtz
- Stem Cell Institute, University of Minnesota, Minneapolis, MN
| | - Britney Lee
- Stem Cell Institute, University of Minnesota, Minneapolis, MN
| | - Dane Munson
- Stem Cell Institute, University of Minnesota, Minneapolis, MN
| | - Xuerui Wang
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN.,Department of Neurology, University of Minnesota, Minneapolis, MN
| | - Mohammed Hussein
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN.,Department of Neurology, University of Minnesota, Minneapolis, MN
| | - Jasmeet Bhatia
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN.,Department of Neurology, University of Minnesota, Minneapolis, MN
| | - Seunghyun Lim
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Bioinformatics and Computational Biology Graduate Program, University of Minnesota, Minneapolis, MN
| | - Ce Yuan
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Bioinformatics and Computational Biology Graduate Program, University of Minnesota, Minneapolis, MN
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN.,Department of Neurology, University of Minnesota, Minneapolis, MN
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN.,Department of Neurology, University of Minnesota, Minneapolis, MN
| | - Nobuaki Kikyo
- Stem Cell Institute, University of Minnesota, Minneapolis, MN.,Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN
| |
Collapse
|
4
|
Ojima K, Muroya S, Wada H, Ogawa K, Oe M, Takimoto K, Nishimura T. Immature adipocyte-derived exosomes inhibit expression of muscle differentiation markers. FEBS Open Bio 2021; 11:768-781. [PMID: 33527775 PMCID: PMC7931241 DOI: 10.1002/2211-5463.13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/24/2020] [Accepted: 01/27/2021] [Indexed: 11/16/2022] Open
Abstract
Exosomes are released from a variety of cells to communicate with recipient cells. Exosomes contain microRNAs (miRNAs), which are noncoding RNAs that suppress target genes. Our previous proteomic study (FEBS Open Bio 2016, 6, 816–826) demonstrated that 3T3‐L1 adipocytes secrete exosome components as well as growth factors, inspiring us to investigate what type of miRNA is involved in adipocyte‐secreted exosomes and what functions they carry out in recipient cells. Here, we profiled miRNAs in 3T3‐L1 adipocyte‐secreted exosomes and revealed suppression of muscle differentiation by adipocyte‐derived exosomes. Through our microarray analysis, we detected over 300 exosomal miRNAs during adipocyte differentiation. Exosomal miRNAs present during adipocyte differentiation included not only pro‐adipogenic miRNAs but also miRNAs associated with muscular dystrophy. Gene ontology analysis predicted that the target genes of miRNAs are associated primarily with transcriptional regulation. To further investigate whether adipocyte‐secreted exosomes regulate the expression levels of genes involved in muscle differentiation, we treated cultured myoblasts with adipocyte‐derived exosome fractions. Intriguingly, the expression levels of myogenic regulatory factors, Myog and Myf6, and other muscle differentiation markers, myosin heavy‐chain 3 and insulin‐like growth factor 2, were significantly downregulated in myoblasts treated with adipocyte‐derived exosomes. Immature adipocyte‐derived exosomes exhibited a stronger suppressive effect than mature adipocyte‐derived exosomes. Our results suggest that adipocytes suppress the expression levels of muscle differentiation‐associated genes in myoblasts via adipocyte‐secreted exosomes containing miRNAs.
Collapse
Affiliation(s)
- Koichi Ojima
- Muscle Biology Research Unit, Division of Animal Products Research, National Institute of Livestock and Grassland Science, NARO, Tsukuba, Japan
| | - Susumu Muroya
- Muscle Biology Research Unit, Division of Animal Products Research, National Institute of Livestock and Grassland Science, NARO, Tsukuba, Japan
| | - Hiromu Wada
- Ion Channel Laboratory, Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Japan
| | - Kotaro Ogawa
- Ion Channel Laboratory, Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Japan
| | - Mika Oe
- Muscle Biology Research Unit, Division of Animal Products Research, National Institute of Livestock and Grassland Science, NARO, Tsukuba, Japan
| | - Koichi Takimoto
- Ion Channel Laboratory, Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
5
|
Rubanova N, Pinna G, Kropp J, Campalans A, Radicella JP, Polesskaya A, Harel-Bellan A, Morozova N. MasterPATH: network analysis of functional genomics screening data. BMC Genomics 2020; 21:632. [PMID: 32928103 PMCID: PMC7491077 DOI: 10.1186/s12864-020-07047-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
Background Functional genomics employs several experimental approaches to investigate gene functions. High-throughput techniques, such as loss-of-function screening and transcriptome profiling, allow to identify lists of genes potentially involved in biological processes of interest (so called hit list). Several computational methods exist to analyze and interpret such lists, the most widespread of which aim either at investigating of significantly enriched biological processes, or at extracting significantly represented subnetworks. Results Here we propose a novel network analysis method and corresponding computational software that employs the shortest path approach and centrality measure to discover members of molecular pathways leading to the studied phenotype, based on functional genomics screening data. The method works on integrated interactomes that consist of both directed and undirected networks – HIPPIE, SIGNOR, SignaLink, TFactS, KEGG, TransmiR, miRTarBase. The method finds nodes and short simple paths with significant high centrality in subnetworks induced by the hit genes and by so-called final implementers – the genes that are involved in molecular events responsible for final phenotypic realization of the biological processes of interest. We present the application of the method to the data from miRNA loss-of-function screen and transcriptome profiling of terminal human muscle differentiation process and to the gene loss-of-function screen exploring the genes that regulates human oxidative DNA damage recognition. The analysis highlighted the possible role of several known myogenesis regulatory miRNAs (miR-1, miR-125b, miR-216a) and their targets (AR, NR3C1, ARRB1, ITSN1, VAV3, TDGF1), as well as linked two major regulatory molecules of skeletal myogenesis, MYOD and SMAD3, to their previously known muscle-related targets (TGFB1, CDC42, CTCF) and also to a number of proteins such as C-KIT that have not been previously studied in the context of muscle differentiation. The analysis also showed the role of the interaction between H3 and SETDB1 proteins for oxidative DNA damage recognition. Conclusion The current work provides a systematic methodology to discover members of molecular pathways in integrated networks using functional genomics screening data. It also offers a valuable instrument to explain the appearance of a set of genes, previously not associated with the process of interest, in the hit list of each particular functional genomics screening.
Collapse
Affiliation(s)
- Natalia Rubanova
- Institut des Hautes Etudes Scientifiques, Le Bois-Marie 35 rte de Chartres, 91440, Bures-sur-Yvette, France. .,Université Paris Diderot, Paris, France. .,Skolkovo Institute of Science and Technology, Skolkovo, Russia.
| | - Guillaume Pinna
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Jeremie Kropp
- Institut des Hautes Etudes Scientifiques, Le Bois-Marie 35 rte de Chartres, 91440, Bures-sur-Yvette, France
| | - Anna Campalans
- Institute of Molecular and Cellular Radiobiology, Institut François Jacob, CEA, F-92265, Fontenay-aux-Roses, France.,INSERM, U967, bâtiment 56 PC 103 18 route du Panorama, BP6 92265, Fontenay-aux-Roses Cedex, France.,Université Paris Sud, U967, bâtiment 56 PC 103 18 route du Panorama, BP6 92265, Fontenay-aux-Roses Cedex, France
| | - Juan Pablo Radicella
- Institute of Molecular and Cellular Radiobiology, Institut François Jacob, CEA, F-92265, Fontenay-aux-Roses, France.,INSERM, U967, bâtiment 56 PC 103 18 route du Panorama, BP6 92265, Fontenay-aux-Roses Cedex, France.,Université Paris Sud, U967, bâtiment 56 PC 103 18 route du Panorama, BP6 92265, Fontenay-aux-Roses Cedex, France
| | - Anna Polesskaya
- Ecole Polytechnique, Université Paris-Saclay, CNRS UMR 7654, Laboratoire de Biochimie, Ecole Polytechnique, 91128, Palaiseau, France
| | - Annick Harel-Bellan
- Institut des Hautes Etudes Scientifiques, Le Bois-Marie 35 rte de Chartres, 91440, Bures-sur-Yvette, France
| | - Nadya Morozova
- Institut des Hautes Etudes Scientifiques, Le Bois-Marie 35 rte de Chartres, 91440, Bures-sur-Yvette, France.,Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| |
Collapse
|
6
|
Ahmad SS, Ahmad K, Lee EJ, Lee YH, Choi I. Implications of Insulin-Like Growth Factor-1 in Skeletal Muscle and Various Diseases. Cells 2020; 9:cells9081773. [PMID: 32722232 PMCID: PMC7465464 DOI: 10.3390/cells9081773] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is an essential tissue that attaches to bones and facilitates body movements. Insulin-like growth factor-1 (IGF-1) is a hormone found in blood that plays an important role in skeletal myogenesis and is importantly associated with muscle mass entity, strength development, and degeneration and increases the proliferative capacity of muscle satellite cells (MSCs). IGF-1R is an IGF-1 receptor with a transmembrane location that activates PI3K/Akt signaling and possesses tyrosine kinase activity, and its expression is significant in terms of myoblast proliferation and normal muscle mass maintenance. IGF-1 synthesis is elevated in MSCs of injured muscles and stimulates MSCs proliferation and myogenic differentiation. Mechanical loading also affects skeletal muscle production by IGF-1, and low IGF-1 levels are associated with low handgrip strength and poor physical performance. IGF-1 is potentially useful in the management of Duchenne muscular dystrophy, muscle atrophy, and promotes neurite development. This review highlights the role of IGF-1 in skeletal muscle, its importance during myogenesis, and its involvement in different disease conditions.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (K.A.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (K.A.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (K.A.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Yong-Ho Lee
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan 38430, Korea
- Correspondence: (Y.-H.L.); (I.C.); Fax: +82-53-810-4769
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (S.S.A.); (K.A.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
- Correspondence: (Y.-H.L.); (I.C.); Fax: +82-53-810-4769
| |
Collapse
|
7
|
Diao S, Yang H, Cao Y, Yang D, Fan Z. IGF2 enhanced the osteo-/dentinogenic and neurogenic differentiation potentials of stem cells from apical papilla. J Oral Rehabil 2019; 47 Suppl 1:55-65. [PMID: 31291686 DOI: 10.1111/joor.12859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES In dental tissue engineering, niche is important for maintaining stem cell function and regenerating the dental tissues. However, there is limited knowledge for the growth factors in niche to maintain the function of stem cells. In this study, we investigated the effect of IGF2, a growth factor in stem cells from apical papilla (SCAPs) niche, on differentiation and proliferation potentials of SCAPs. MATERIALS AND METHODS Recombinant human IGF2 protein (rhIGF2) was used. Cell counting kit-8 assay, Carboxyfluorescein succinimidyl ester assay, alkaline phosphatase (ALP) activity, Alizarin Red staining, quantitative calcium analysis, immunofluorescence staining and real-time RT-PCR were performed to investigate the cell proliferation and differentiation potentials of SCAPs. And proteomic analysis was used to identify the differential secreted proteins. RESULTS By ALP activity assay, we found that 5 ng/mL rhIGF2 might be the optimal concentration for treatment. Then, Alizarin Red staining, quantitative calcium analysis and osteogenesis-related gene expression results showed that 5 ng/mL rhIGF2 could enhance the osteo-/dentinogenic differentiation potentials in SCAPs. Immunofluorescence staining and real-time RT-PCR results showed that neurogenic markers were significantly induced by 5 ng/mL rhIGF2 in SCAPs. Then, CCK-8 assay and CFSE assay results showed that 5 ng/mL rhIGF2 could enhance the cell proliferation in SCAPs. Furthermore, proteomic analysis showed that IGF2 could induce some secreted proteins which function related to the osteogenesis, neurogenesis and cell proliferation. CONCLUSIONS Our results identified that IGF2 might be the potential mediator in niche to promote SCAP function and dental tissue regeneration.
Collapse
Affiliation(s)
- Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Dongmei Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
8
|
Baral K, Rotwein P. The insulin-like growth factor 2 gene in mammals: Organizational complexity within a conserved locus. PLoS One 2019; 14:e0219155. [PMID: 31251794 PMCID: PMC6599137 DOI: 10.1371/journal.pone.0219155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The secreted protein, insulin-like growth factor 2 (IGF2), plays a central role in fetal and prenatal growth and development, and is regulated at the genetic level by parental imprinting, being expressed predominantly from the paternally derived chromosome in mice and humans. Here, IGF2/Igf2 and its locus has been examined in 19 mammals from 13 orders spanning ~166 million years of evolutionary development. By using human or mouse DNA segments as queries in genome analyses, and by assessing gene expression using RNA-sequencing libraries, more complexity was identified within IGF2/Igf2 than was annotated previously. Multiple potential 5’ non-coding exons were mapped in most mammals and are presumably linked to distinct IGF2/Igf2 promoters, as shown for several species by interrogating RNA-sequencing libraries. DNA similarity was highest in IGF2/Igf2 coding exons; yet, even though the mature IGF2 protein was conserved, versions of 67 or 70 residues are produced secondary to species-specific maintenance of alternative RNA splicing at a variable intron-exon junction. Adjacent H19 was more divergent than IGF2/Igf2, as expected in a gene for a noncoding RNA, and was identified in only 10/19 species. These results show that common features, including those defining IGF2/Igf2 coding and several non-coding exons, were likely present at the onset of the mammalian radiation, but that others, such as a putative imprinting control region 5’ to H19 and potential enhancer elements 3’ to H19, diversified with speciation. This study also demonstrates that careful analysis of genomic and gene expression repositories can provide new insights into gene structure and regulation.
Collapse
Affiliation(s)
- Kabita Baral
- Graduate School, College of Science, University of Texas at El Paso, El Paso, Texas
| | - Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech Health University Health Sciences Center, El Paso, Texas
- * E-mail:
| |
Collapse
|
9
|
Liu Z, Li C, Li X, Yao Y, Ni W, Zhang X, Cao Y, Hazi W, Wang D, Quan R, Yu S, Wu Y, Niu S, Cui Y, Khan Y, Hu S. Expression profiles of microRNAs in skeletal muscle of sheep by deep sequencing. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2018; 32:757-766. [PMID: 30477295 PMCID: PMC6498074 DOI: 10.5713/ajas.18.0473] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/05/2018] [Indexed: 11/27/2022]
Abstract
Objective MicroRNAs are a class of endogenous small regulatory RNAs that regulate cell proliferation, differentiation and apoptosis. Recent studies on miRNAs are mainly focused on mice, human and pig. However, the studies on miRNAs in skeletal muscle of sheep are not comprehensive. Methods RNA-seq technology was used to perform genomic analysis of miRNAs in prenatal and postnatal skeletal muscle of sheep. Targeted genes were predicted using miRanda software and miRNA-mRNA interactions were verified by quantitative real-time polymerase chain reaction. To further investigate the function of miRNAs, candidate targeted genes were enriched for analysis using gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment. Results The results showed total of 1,086 known miRNAs and 40 new candidate miRNAs were detected in prenatal and postnatal skeletal muscle of sheep. In addition, 345 miRNAs (151 up-regulated, 94 down-regulated) were differentially expressed. Moreover, miRanda software was performed to predict targeted genes of miRNAs, resulting in a total of 2,833 predicted targets, especially miR-381 which targeted multiple muscle-related mRNAs. Furthermore, GO and KEGG pathway analysis confirmed that targeted genes of miRNAs were involved in development of skeletal muscles. Conclusion This study supplements the miRNA database of sheep, which provides valuable information for further study of the biological function of miRNAs in sheep skeletal muscle.
Collapse
Affiliation(s)
- Zhijin Liu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Cunyuan Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Xiaoyue Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yang Yao
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Wei Ni
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Xiangyu Zhang
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yang Cao
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Wureli Hazi
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, 832003, China
| | - Dawei Wang
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Renzhe Quan
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Shuting Yu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yuyu Wu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Songmin Niu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yulong Cui
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yaseen Khan
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Shengwei Hu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| |
Collapse
|
10
|
Ballarino M, Cipriano A, Tita R, Santini T, Desideri F, Morlando M, Colantoni A, Carrieri C, Nicoletti C, Musarò A, Carroll DO, Bozzoni I. Deficiency in the nuclear long noncoding RNA Charme causes myogenic defects and heart remodeling in mice. EMBO J 2018; 37:embj.201899697. [PMID: 30177572 PMCID: PMC6138438 DOI: 10.15252/embj.201899697] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 01/03/2023] Open
Abstract
Myogenesis is a highly regulated process that involves the conversion of progenitor cells into multinucleated myofibers. Besides proteins and miRNAs, long noncoding RNAs (lncRNAs) have been shown to participate in myogenic regulatory circuitries. Here, we characterize a murine chromatin‐associated muscle‐specific lncRNA, Charme, which contributes to the robustness of the myogenic program in vitro and in vivo. In myocytes, Charme depletion triggers the disassembly of a specific chromosomal domain and the downregulation of myogenic genes contained therein. Notably, several Charme‐sensitive genes are associated with human cardiomyopathies and Charme depletion in mice results in a peculiar cardiac remodeling phenotype with changes in size, structure, and shape of the heart. Moreover, the existence of an orthologous transcript in human, regulating the same subset of target genes, suggests an important and evolutionarily conserved function for Charme. Altogether, these data describe a new example of a chromatin‐associated lncRNA regulating the robustness of skeletal and cardiac myogenesis.
Collapse
Affiliation(s)
- Monica Ballarino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Andrea Cipriano
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Rossella Tita
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Tiziana Santini
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Fabio Desideri
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Mariangela Morlando
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Claudia Carrieri
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Carmine Nicoletti
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Antonio Musarò
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy.,DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Dònal O' Carroll
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Irene Bozzoni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy .,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy.,Institute Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy.,Institute of Molecular Biology and Pathology, CNR, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
11
|
Han X, Chen H, Huang D, Chen H, Fei L, Cheng C, Huang H, Yuan GC, Guo G. Mapping human pluripotent stem cell differentiation pathways using high throughput single-cell RNA-sequencing. Genome Biol 2018; 19:47. [PMID: 29622030 PMCID: PMC5887227 DOI: 10.1186/s13059-018-1426-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/21/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Human pluripotent stem cells (hPSCs) provide powerful models for studying cellular differentiations and unlimited sources of cells for regenerative medicine. However, a comprehensive single-cell level differentiation roadmap for hPSCs has not been achieved. RESULTS We use high throughput single-cell RNA-sequencing (scRNA-seq), based on optimized microfluidic circuits, to profile early differentiation lineages in the human embryoid body system. We present a cellular-state landscape for hPSC early differentiation that covers multiple cellular lineages, including neural, muscle, endothelial, stromal, liver, and epithelial cells. Through pseudotime analysis, we construct the developmental trajectories of these progenitor cells and reveal the gene expression dynamics in the process of cell differentiation. We further reprogram primed H9 cells into naïve-like H9 cells to study the cellular-state transition process. We find that genes related to hemogenic endothelium development are enriched in naïve-like H9. Functionally, naïve-like H9 show higher potency for differentiation into hematopoietic lineages than primed cells. CONCLUSIONS Our single-cell analysis reveals the cellular-state landscape of hPSC early differentiation, offering new insights that can be harnessed for optimization of differentiation protocols.
Collapse
Affiliation(s)
- Xiaoping Han
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Institute of Hematology, The 1st Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, 310058, China. .,College of Animal Science, Zhejiang University, Hangzhou, 310058, China.
| | - Daosheng Huang
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Huidong Chen
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Chan School of Public Health, Boston, MA, 02115, USA.,Department of Computer Science and Technology, Tongji University, Shanghai, 201804, China
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Chen Cheng
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - He Huang
- Institute of Hematology, The 1st Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Chan School of Public Health, Boston, MA, 02115, USA.
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Institute of Hematology, The 1st Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, 310058, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
12
|
Analysis of meat quality traits and gene expression profiling of pigs divergent in residual feed intake. Meat Sci 2018; 137:265-274. [DOI: 10.1016/j.meatsci.2017.11.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/08/2017] [Accepted: 11/16/2017] [Indexed: 11/19/2022]
|
13
|
Akiyama T, Sato S, Chikazawa-Nohtomi N, Soma A, Kimura H, Wakabayashi S, Ko SBH, Ko MSH. Efficient differentiation of human pluripotent stem cells into skeletal muscle cells by combining RNA-based MYOD1-expression and POU5F1-silencing. Sci Rep 2018; 8:1189. [PMID: 29352121 PMCID: PMC5775307 DOI: 10.1038/s41598-017-19114-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/20/2017] [Indexed: 01/22/2023] Open
Abstract
Direct generation of skeletal muscle cells from human pluripotent stem cells (hPSCs) would be beneficial for drug testing, drug discovery, and disease modelling in vitro. Here we show a rapid and robust method to induce myogenic differentiation of hPSCs by introducing mRNA encoding MYOD1 together with siRNA-mediated knockdown of POU5F1 (also known as OCT4 or OCT3/4). This integration-free approach generates functional skeletal myotubes with sarcomere-like structure and a fusion capacity in several days. The POU5F1 silencing facilitates MYOD1 recruitment to the target promoters, which results in the significant activation of myogenic genes in hPSCs. Furthermore, deep sequencing transcriptome analyses demonstrated that POU5F1-knockdown upregulates the genes associated with IGF- and FGF-signaling and extracellular matrix that may also support myogenic differentiation. This rapid and direct differentiation method may have potential applications in regenerative medicine and disease therapeutics for muscle disorders such as muscular dystrophy.
Collapse
Affiliation(s)
- Tomohiko Akiyama
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan
| | - Saeko Sato
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan
| | | | - Atsumi Soma
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan
| | - Hiromi Kimura
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan
| | - Shunichi Wakabayashi
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan
| | - Shigeru B H Ko
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan
| | - Minoru S H Ko
- Department of Systems Medicine, Keio University School of Medicine, Tokyo, 160, Japan.
| |
Collapse
|
14
|
Ben-Zaken S, Meckel Y, Nemet D, Eliakim A. High prevalence of the IGF2 rs680 GG polymorphism among top-level sprinters and jumpers. Growth Horm IGF Res 2017; 37:26-30. [PMID: 29107196 DOI: 10.1016/j.ghir.2017.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 09/25/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022]
Abstract
UNLABELLED Previous studies have shown that the IGF1 polymorphism is associated with greater muscle mass and improved power athletic ability, but very little is known about the IGF2 polymorphism and athletic performance. PURPOSE The aim of the present study was to assess the frequency distribution of the IGF2 rs680 polymorphism among Israeli athletes. METHODS 185 short- (n=72) and long-distance (n=113) runners, 94 short- (n=44) and long-distance (n=50) swimmers, 54 weight lifters and 111 controls participated in the study. Genomic DNA was extracted from peripheral EDTA treated anti-coagulated blood using a standard protocol. Genotyping of the IGF2 A/G polymorphism (rs680) was performed using allelic discrimination assay. RESULTS The frequency of IGF2 (rs680) G allele carriers was significantly greater among top compared to national-level track and field sprinters and jumpers (p<0.05). The IGF2 (rs680) GG genotype frequency was significantly greater among track and field sprinters and jumpers compared to weight lifters p<0.02), and among top-level sprinters and jumpers compared to top-level weight lifters p<0.01). There were no statistically significant differences in the IGF2 (rs680) GG genotype frequency among endurance athletes and between the swimmers and the other sports disciplines and the controls. CONCLUSIONS While a single polymorphism cannot determine athletic success or failure, the findings of the present study suggest a potential importance of the IGF2 polymorphism, mainly regarding speed sport performance.
Collapse
Affiliation(s)
- Sigal Ben-Zaken
- The Academic College of Physical Education and Sports Sciences at the Wingate Institute, Genetics and Molecular Biology Laboratory, Netanya 42902, Israel.
| | - Yoav Meckel
- The Academic College of Physical Education and Sports Sciences at the Wingate Institute, Genetics and Molecular Biology Laboratory, Netanya 42902, Israel
| | - Dan Nemet
- Meir Medical Center, Child Health and Sports Center, Pediatric Department, Sackler School of Medicine, Tel-Aviv University, Israel
| | - Alon Eliakim
- Meir Medical Center, Child Health and Sports Center, Pediatric Department, Sackler School of Medicine, Tel-Aviv University, Israel
| |
Collapse
|
15
|
Chen KW, Chang YJ, Yeh CM, Lian YL, Chan MWY, Kao CF, Chen L. SH2B1 modulates chromatin state and MyoD occupancy to enhance expressions of myogenic genes. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1860:270-281. [PMID: 28039048 DOI: 10.1016/j.bbagrm.2016.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/29/2016] [Accepted: 12/23/2016] [Indexed: 10/20/2022]
Abstract
As mesoderm-derived cell lineage commits to myogenesis, a spectrum of signaling molecules, including insulin growth factor (IGF), activate signaling pathways and ultimately instruct chromatin remodeling and the transcription of myogenic genes. MyoD is a key transcription factor during myogenesis. In this study, we have identified and characterized a novel myogenic regulator, SH2B1. Knocking down SH2B1 delays global chromatin condensation and decreases the formation of myotubes. SH2B1 interacts with histone H1 and is required for the removal of histone H1 from active transcription sites, allowing for the expressions of myogenic genes, IGF2 and MYOG. Chromatin immunoprecipitation assays suggest the requirement of SH2B1 for the induction of histone H3 lysine 4 trimethylation as well as the reduction of histone H3 lysine 9 trimethylation at the promoters and/or enhancers of IGF2 and MYOG genes during myogenesis. Furthermore, SH2B1 is required for the transcriptional activity of MyoD and MyoD occupancy at the enhancer/promoter regions of IGF2 and MYOG during myogenesis. Together, this study demonstrates that SH2B1 fine-tunes global-local chromatin states, expressions of myogenic genes and ultimately promotes myogenesis.
Collapse
Affiliation(s)
- Kuan-Wei Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, R.O.C
| | - Yu-Jung Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, R.O.C
| | - Chia-Ming Yeh
- Department of Life Science, National Chung Cheng University, Chia-yi, Taiwan, R.O.C
| | - Yen-Ling Lian
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, R.O.C
| | - Michael W Y Chan
- Department of Life Science, National Chung Cheng University, Chia-yi, Taiwan, R.O.C
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, R.O.C
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, R.O.C.; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan, R.O.C..
| |
Collapse
|
16
|
Barroca V, Lewandowski D, Jaracz-Ros A, Hardouin SN. Paternal Insulin-like Growth Factor 2 (Igf2) Regulates Stem Cell Activity During Adulthood. EBioMedicine 2016; 15:150-162. [PMID: 28007480 PMCID: PMC5233811 DOI: 10.1016/j.ebiom.2016.11.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 11/13/2016] [Accepted: 11/29/2016] [Indexed: 12/17/2022] Open
Abstract
Insulin-like Growth Factor 2 (IGF2) belongs to the IGF/Insulin pathway, a highly conserved evolutionarily network that regulates growth, aging and lifespan. Igf2 is highly expressed in the embryo and in cancer cells. During mouse development, Igf2 is expressed in all sites where hematopoietic stem cells (HSC) successively expand, then its expression drops at weaning and becomes undetectable when adult HSC have reached their niches in bones and start to self-renew. In the present study, we aim to discover the role of IGF2 during adulthood. We show that Igf2 is specifically expressed in adult HSC and we analyze HSC from adult mice deficient in Igf2 transcripts. We demonstrate that Igf2 deficiency avoids the age-related attrition of the HSC pool and that Igf2 is necessary for tissue homeostasis and regeneration. Our study reveals that the expression level of Igf2 is critical to maintain the balance between stem cell self-renewal and differentiation, presumably by regulating the interaction between HSC and their niche. Our data have major clinical interest for transplantation: understanding the changes in adult stem cells and their environments will improve the efficacy of regenerative medicine and impact health- and life-span. The imprinted gene Igf2 is expressed in adult tissue stem cells. Igf2 deficiency increases HSC (hematopoietic stem cells) self-renewal and avoids age-related attrition of the HSC pool. Igf2 deficiency decreases HSC differentiation and mobilization. Igf2 deficiency modifies the interaction between HSC and their environment.
IGF2 belongs to the IGF/Insulin family that regulates growth, aging and lifespan. This role is evolutionarily conserved from worms to mammals. IGF2 favors cell proliferation during embryonic development but its role in adulthood is unknown. To decipher its function we undertook a lifelong analysis of the consequences of Igf2 deficiency on hematopoiesis, in steady-state conditions and during bone marrow transplantation. We demonstrate that lowering Igf2 levels increases the pool of stem cells, without uncontrolled proliferation and migration of immature cells that would lead to cancer. This is a promising way to enhance the stem cells pool during aging that has major interest for transplantation.
Collapse
Affiliation(s)
- Vilma Barroca
- INSERM UMR 967, 92265 Fontenay-aux-roses cedex, France; CEA/DSV/iRCM, 92265 Fontenay-aux-roses cedex, France; Université Paris-Diderot, Paris 7, 92265 Fontenay-aux-roses cedex, France; Université Paris-Sud, Paris 11, 92265 Fontenay-aux-roses cedex, France
| | - Daniel Lewandowski
- INSERM UMR 967, 92265 Fontenay-aux-roses cedex, France; CEA/DSV/iRCM, 92265 Fontenay-aux-roses cedex, France; Université Paris-Diderot, Paris 7, 92265 Fontenay-aux-roses cedex, France; Université Paris-Sud, Paris 11, 92265 Fontenay-aux-roses cedex, France
| | - Agnieszka Jaracz-Ros
- INSERM UMR 967, 92265 Fontenay-aux-roses cedex, France; CEA/DSV/iRCM, 92265 Fontenay-aux-roses cedex, France; Université Paris-Diderot, Paris 7, 92265 Fontenay-aux-roses cedex, France; Université Paris-Sud, Paris 11, 92265 Fontenay-aux-roses cedex, France
| | - Sylvie-Nathalie Hardouin
- INSERM UMR 967, 92265 Fontenay-aux-roses cedex, France; CEA/DSV/iRCM, 92265 Fontenay-aux-roses cedex, France; Université Paris-Diderot, Paris 7, 92265 Fontenay-aux-roses cedex, France; Université Paris-Sud, Paris 11, 92265 Fontenay-aux-roses cedex, France.
| |
Collapse
|
17
|
Zhang J, Han B, Li X, Bies J, Jiang P, Koller RP, Wolff L. Distal regulation of c-myb expression during IL-6-induced differentiation in murine myeloid progenitor M1 cells. Cell Death Dis 2016; 7:e2364. [PMID: 27607579 PMCID: PMC5059869 DOI: 10.1038/cddis.2016.267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 12/31/2022]
Abstract
The c-Myb transcription factor is a major regulator that controls differentiation and proliferation of hematopoietic progenitor cells, which is frequently deregulated in hematological diseases, such as lymphoma and leukemia. Understanding of the mechanisms regulating the transcription of c-myb gene is challenging as it lacks a typical promoter and multiple factors are involved. Our previous studies identified some distal regulatory elements in the upstream regions of c-myb gene in murine myeloid progenitor M1 cells, but the detailed mechanisms still remain unclear. In the present study, we found that a cell differentiation-related DNase1 hypersensitive site is located at a -28k region upstream of c-myb gene and that transcription factors Hoxa9, Meis1 and PU.1 bind to the -28k region. Circular chromosome conformation capture (4C) assay confirmed the interaction between the -28k region and the c-myb promoter, which is supported by the enrichment of CTCF and Cohesin. Our analysis also points to a critical role for Hoxa9 and PU.1 in distal regulation of c-myb expression in murine myeloid cells and cell differentiation. Overexpression of Hoxa9 disrupted the IL-6-induced differentiation of M1 cells and upregulated c-myb expression through binding of the -28k region. Taken together, our results provide an evidence for critical role of the -28k region in distal regulatory mechanism for c-myb gene expression during differentiation of myeloid progenitor M1 cells.
Collapse
Affiliation(s)
- Junfang Zhang
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Bingshe Han
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Xiaoxia Li
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Juraj Bies
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Penglei Jiang
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Richard P Koller
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Linda Wolff
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Mukherjee A, Alzhanov D, Rotwein P. Defining human insulin-like growth factor I gene regulation. Am J Physiol Endocrinol Metab 2016; 311:E519-29. [PMID: 27406741 PMCID: PMC5005972 DOI: 10.1152/ajpendo.00212.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/08/2016] [Indexed: 11/22/2022]
Abstract
Growth hormone (GH) plays an essential role in controlling somatic growth and in regulating multiple physiological processes in humans and other species. Insulin-like growth factor I (IGF-I), a conserved, secreted 70-amino acid peptide, is a critical mediator of many of the biological effects of GH. Previous studies have demonstrated that GH rapidly and potently promotes IGF-I gene expression in rodents and in some other mammals through the transcription factor STAT5b, leading to accumulation of IGF-I mRNAs and production of IGF-I. Despite this progress, very little is known about how GH or other trophic factors control human IGF1 gene expression, in large part because of the absence of any cellular model systems that robustly express IGF-I. Here, we have addressed mechanisms of regulation of human IGF-I by GH after generating cells in which the IGF1 chromosomal locus has been incorporated into a mouse cell line. Using this model, we found that physiological levels of GH rapidly stimulate human IGF1 gene transcription and identify several potential transcriptional enhancers in chromatin that bind STAT5b in a GH-regulated way. Each of the putative enhancers also activates a human IGF1 gene promoter in reconstitution experiments in the presence of the GH receptor, STAT5b, and GH. Thus we have developed a novel experimental platform that now may be used to determine how human IGF1 gene expression is controlled under different physiological and pathological conditions.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon; and
| | - Damir Alzhanov
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon; and
| | - Peter Rotwein
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon; and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech Health University Health Sciences Center, El Paso, Texas
| |
Collapse
|
19
|
da Silva VH, Regitano LCDA, Geistlinger L, Pértille F, Giachetto PF, Brassaloti RA, Morosini NS, Zimmer R, Coutinho LL. Genome-Wide Detection of CNVs and Their Association with Meat Tenderness in Nelore Cattle. PLoS One 2016; 11:e0157711. [PMID: 27348523 PMCID: PMC4922624 DOI: 10.1371/journal.pone.0157711] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/03/2016] [Indexed: 12/20/2022] Open
Abstract
Brazil is one of the largest beef producers and exporters in the world with the Nelore breed representing the vast majority of Brazilian cattle (Bos taurus indicus). Despite the great adaptability of the Nelore breed to tropical climate, meat tenderness (MT) remains to be improved. Several factors including genetic composition can influence MT. In this article, we report a genome-wide analysis of copy number variation (CNV) inferred from Illumina® High Density SNP-chip data for a Nelore population of 723 males. We detected >2,600 CNV regions (CNVRs) representing ≈6.5% of the genome. Comparing our results with previous studies revealed an overlap in ≈1400 CNVRs (>50%). A total of 1,155 CNVRs (43.6%) overlapped 2,750 genes. They were enriched for processes involving guanosine triphosphate (GTP), previously reported to influence skeletal muscle physiology and morphology. Nelore CNVRs also overlapped QTLs for MT reported in other breeds (8.9%, 236 CNVRs) and from a previous study with this population (4.1%, 109 CNVRs). Two CNVRs were also proximal to glutathione metabolism genes that were previously associated with MT. Genome-wide association study of CN state with estimated breeding values derived from meat shear force identified 6 regions, including a region on BTA3 that contains genes of the cAMP and cGMP pathway. Ten CNVRs that overlapped regions associated with MT were successfully validated by qPCR. Our results represent the first comprehensive CNV study in Bos taurus indicus cattle and identify regions in which copy number changes are potentially of importance for the MT phenotype.
Collapse
Affiliation(s)
- Vinicius Henrique da Silva
- Animal Biotechnology Laboratory, Animal Science Department, University of São Paulo (USP)/Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, Brazil
- * E-mail: (LLC); (VHS)
| | | | - Ludwig Geistlinger
- Institute of Bioinformatics, Department of Informatics, Ludwig-Maximilians-Universität München (LMU), Amalienstrasse 17, 80333, München, Germany
| | - Fábio Pértille
- Animal Biotechnology Laboratory, Animal Science Department, University of São Paulo (USP)/Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, Brazil
| | | | - Ricardo Augusto Brassaloti
- Animal Biotechnology Laboratory, Animal Science Department, University of São Paulo (USP)/Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, Brazil
| | - Natália Silva Morosini
- Animal Biotechnology Laboratory, Animal Science Department, University of São Paulo (USP)/Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, Brazil
| | - Ralf Zimmer
- Institute of Bioinformatics, Department of Informatics, Ludwig-Maximilians-Universität München (LMU), Amalienstrasse 17, 80333, München, Germany
| | - Luiz Lehmann Coutinho
- Animal Biotechnology Laboratory, Animal Science Department, University of São Paulo (USP)/Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, Brazil
- * E-mail: (LLC); (VHS)
| |
Collapse
|
20
|
Alzhanov D, Rotwein P. Characterizing a distal muscle enhancer in the mouse Igf2 locus. Physiol Genomics 2015; 48:167-72. [PMID: 26645089 DOI: 10.1152/physiolgenomics.00095.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/03/2015] [Indexed: 11/22/2022] Open
Abstract
Insulin-like growth factor-2 (IGF2) is highly expressed in skeletal muscle and was identified as a quantitative trait locus for muscle mass. Yet little is known about mechanisms of its regulation in muscle. Recently, a DNA segment found ∼100 kb from the Igf2 gene was identified as a possible muscle transcriptional control element. Here we have developed an in vivo reporter system to assess this putative enhancer by substituting nuclear (n) EGFP for Igf2 coding exons in a bacterial artificial chromosome containing the mouse Igf2 - H19 chromosomal locus. After stable transfection into a mesenchymal stem cell line, individual clones were converted to myoblasts and underwent progressive muscle-specific gene expression and myotube formation in differentiation medium. Transgenic mRNA and nuclear-targeted enhanced green fluorescent protein were produced coincident with endogenous Igf2 mRNA, but only in lines containing an intact distal conserved DNA element. Our results show that a 294 bp DNA fragment containing two E-boxes is a necessary and sufficient long-range enhancer for induction of Igf2 gene transcription during skeletal muscle differentiation and provides a robust experimental platform for its further functional dissection.
Collapse
Affiliation(s)
- Damir Alzhanov
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon; and
| | - Peter Rotwein
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon; and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas
| |
Collapse
|
21
|
Hunt LC, Xu B, Finkelstein D, Fan Y, Carroll PA, Cheng PF, Eisenman RN, Demontis F. The glucose-sensing transcription factor MLX promotes myogenesis via myokine signaling. Genes Dev 2015; 29:2475-89. [PMID: 26584623 PMCID: PMC4691951 DOI: 10.1101/gad.267419.115] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/26/2015] [Indexed: 11/27/2022]
Abstract
Metabolic stress and changes in nutrient levels modulate many aspects of skeletal muscle function during aging and disease. Growth factors and cytokines secreted by skeletal muscle, known as myokines, are important signaling factors, but it is largely unknown whether they modulate muscle growth and differentiation in response to nutrients. Here, we found that changes in glucose levels increase the activity of the glucose-responsive transcription factor MLX (Max-like protein X), which promotes and is necessary for myoblast fusion. MLX promotes myogenesis not via an adjustment of glucose metabolism but rather by inducing the expression of several myokines, including insulin-like growth factor 2 (IGF2), whereas RNAi and dominant-negative MLX reduce IGF2 expression and block myogenesis. This phenotype is rescued by conditioned medium from control muscle cells and by recombinant IGF2, which activates the myogenic kinase Akt. Importantly, MLX-null mice display decreased IGF2 induction and diminished muscle regeneration in response to injury, indicating that the myogenic function of MLX is manifested in vivo. Thus, glucose is a signaling molecule that regulates myogenesis and muscle regeneration via MLX/IGF2/Akt signaling.
Collapse
Affiliation(s)
- Liam C Hunt
- Department of Developmental Neurobiology, Division of Developmental Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Patrick A Carroll
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Pei-Feng Cheng
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Robert N Eisenman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, Division of Developmental Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
22
|
Alzhanov D, Mukherjee A, Rotwein P. Identifying growth hormone-regulated enhancers in the Igf1 locus. Physiol Genomics 2015; 47:559-68. [PMID: 26330488 DOI: 10.1152/physiolgenomics.00062.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/27/2015] [Indexed: 11/22/2022] Open
Abstract
Growth hormone (GH) plays a central role in regulating somatic growth and in controlling multiple physiological processes in humans and other vertebrates. A key agent in many GH actions is the secreted peptide, IGF-I. As established previously, GH stimulates IGF-I gene expression via the Stat5b transcription factor, leading to production of IGF-I mRNAs and proteins. However, the precise mechanisms by which GH-activated Stat5b promotes IGF-I gene transcription have not been defined. Unlike other GH-regulated genes, there are no Stat5b sites near either of the two IGF-I gene promoters. Although dispersed GH-activated Stat5b binding elements have been mapped in rodent Igf1 gene chromatin, it is unknown how these distal sites might function as potential transcriptional enhancers. Here we have addressed mechanisms of regulation of IGF-I gene transcription by GH by generating cell lines in which the rat Igf1 chromosomal locus has been incorporated into the mouse genome. Using these cells we find that physiological levels of GH rapidly and potently activate Igf1 gene transcription while stimulating physical interactions in chromatin between inducible Stat5b-binding elements and the Igf1 promoters. We have thus developed a robust experimental platform for elucidating how dispersed transcriptional enhancers control Igf1 gene expression under different biological conditions.
Collapse
Affiliation(s)
- Damir Alzhanov
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon; and
| | - Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon; and
| | - Peter Rotwein
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas
| |
Collapse
|
23
|
Lee KY, Singh MK, Ussar S, Wetzel P, Hirshman MF, Goodyear LJ, Kispert A, Kahn CR. Tbx15 controls skeletal muscle fibre-type determination and muscle metabolism. Nat Commun 2015; 6:8054. [PMID: 26299309 PMCID: PMC4552045 DOI: 10.1038/ncomms9054] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/13/2015] [Indexed: 01/15/2023] Open
Abstract
Skeletal muscle is composed of both slow-twitch oxidative myofibers and fast-twitch glycolytic myofibers that differentially impact muscle metabolism, function and eventually whole-body physiology. Here we show that the mesodermal transcription factor T-box 15 (Tbx15) is highly and specifically expressed in glycolytic myofibers. Ablation of Tbx15 in vivo leads to a decrease in muscle size due to a decrease in the number of glycolytic fibres, associated with a small increase in the number of oxidative fibres. This shift in fibre composition results in muscles with slower myofiber contraction and relaxation, and also decreases whole-body oxygen consumption, reduces spontaneous activity, increases adiposity and glucose intolerance. Mechanistically, ablation of Tbx15 leads to activation of AMPK signalling and a decrease in Igf2 expression. Thus, Tbx15 is one of a limited number of transcription factors to be identified with a critical role in regulating glycolytic fibre identity and muscle metabolism. The transcriptional regulator Tbx15 has a role in organ development. Here Lee et al. show that Tbx15 influences fibre-type determination in murine skeletal muscles, explaining local and systemic metabolic derangements in heterozygous Tbx15 knockout mice.
Collapse
Affiliation(s)
- Kevin Y Lee
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| | - Manvendra K Singh
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.,Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School Singapore, National Heart Centre Singapore, 8 College Road, Singapore 169857, Singapore
| | - Siegfried Ussar
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA.,Institute for Diabetes and Obesity, Helmholtz Center, Parkring, 1385748 Munich/Garching, Germany
| | - Petra Wetzel
- Zentrum Physiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Plaza, Boston, Massachusetts 02215, USA
| |
Collapse
|
24
|
Gonzalez JM, Camacho LE, Ebarb SM, Swanson KC, Vonnahme KA, Stelzleni AM, Johnson SE. Realimentation of nutrient restricted pregnant beef cows supports compensatory fetal muscle growth1. J Anim Sci 2013; 91:4797-806. [DOI: 10.2527/jas.2013-6704] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- J. M. Gonzalez
- Dept. of Animal Sciences and Industry, Kansas State University, Manhattan 66506
| | - L. E. Camacho
- Dept. of Animal Sciences, North Dakota State University, Fargo 58108
| | - S. M. Ebarb
- Dept. of Animal Sciences and Industry, Kansas State University, Manhattan 66506
| | - K. C. Swanson
- Dept. of Animal Sciences, North Dakota State University, Fargo 58108
| | - K. A. Vonnahme
- Dept. of Animal Sciences, North Dakota State University, Fargo 58108
| | - A. M. Stelzleni
- Dept. of Animal and Dairy Science, University of Georgia, Athens 30602
| | - S. E. Johnson
- Dept. of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg 24061
| |
Collapse
|
25
|
Eun B, Sampley ML, Van Winkle MT, Good AL, Kachman MM, Pfeifer K. The Igf2/H19 muscle enhancer is an active transcriptional complex. Nucleic Acids Res 2013; 41:8126-34. [PMID: 23842673 PMCID: PMC3783178 DOI: 10.1093/nar/gkt597] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 06/13/2013] [Accepted: 06/15/2013] [Indexed: 02/06/2023] Open
Abstract
In eukaryotic cells, gene expression is mediated by enhancer activation of RNA polymerase at distant promoters. Recently, distinctions between enhancers and promoters have been blurred by the discovery that enhancers are associated with RNA polymerase and are sites of RNA synthesis. Here, we present an analysis of the insulin-like growth factor 2/H19 muscle enhancer. This enhancer includes a short conserved core element that is organized into chromatin typical of mammalian enhancers, binds tissue-specific transcription factors and functions on its own in vitro to activate promoter transcription. However, in a chromosomal context, this element is not sufficient to activate distant promoters. Instead, enhancer function also requires transcription in cis of a long non-coding RNA, Nctc1. Thus, the insulin-like growth factor 2/H19 enhancer is an active transcriptional complex whose own transcription is essential to its function.
Collapse
Affiliation(s)
- Bokkee Eun
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA and Core-Laboratory, College of Medicine, Korea University, Seoul 136-701, Republic of Korea
| | - Megan L. Sampley
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA and Core-Laboratory, College of Medicine, Korea University, Seoul 136-701, Republic of Korea
| | - Matthew T. Van Winkle
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA and Core-Laboratory, College of Medicine, Korea University, Seoul 136-701, Republic of Korea
| | - Austin L. Good
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA and Core-Laboratory, College of Medicine, Korea University, Seoul 136-701, Republic of Korea
| | - Marika M. Kachman
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA and Core-Laboratory, College of Medicine, Korea University, Seoul 136-701, Republic of Korea
| | - Karl Pfeifer
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA and Core-Laboratory, College of Medicine, Korea University, Seoul 136-701, Republic of Korea
| |
Collapse
|
26
|
Börnigen D, Pers TH, Thorrez L, Huttenhower C, Moreau Y, Brunak S. Concordance of gene expression in human protein complexes reveals tissue specificity and pathology. Nucleic Acids Res 2013; 41:e171. [PMID: 23921638 PMCID: PMC3794609 DOI: 10.1093/nar/gkt661] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Disease-causing variants in human genes usually lead to phenotypes specific to only a few tissues. Here, we present a method for predicting tissue specificity based on quantitative deregulation of protein complexes. The underlying assumption is that the degree of coordinated expression among proteins in a complex within a given tissue may pinpoint tissues that will be affected by a mutation in the complex and coordinated expression may reveal the complex to be active in the tissue. We identified known disease genes and their protein complex partners in a high-quality human interactome. Each susceptibility gene's tissue involvement was ranked based on coordinated expression with its interaction partners in a non-disease global map of human tissue-specific expression. The approach demonstrated high overall area under the curve (0.78) and was very successfully benchmarked against a random model and an approach not using protein complexes. This was illustrated by correct tissue predictions for three case studies on leptin, insulin-like-growth-factor 2 and the inhibitor of NF-κB kinase subunit gamma that show high concordant expression in biologically relevant tissues. Our method identifies novel gene-phenotype associations in human diseases and predicts the tissues where associated phenotypic effects may arise.
Collapse
Affiliation(s)
- Daniela Börnigen
- Department of Electrical Engineering, ESAT-SCD, IBBT-KU Leuven Future Health Department, KU Leuven, 3001 Leuven, Belgium, Biostatistics Department, Harvard School of Public Health, Harvard University, Boston, 02115 MA, USA, Broad Institute of MIT and Harvard, Cambridge, 02142 MA, USA, Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, DK-2800 Lyngby, Denmark, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, 02142 MA, USA, Division of Endocrinology and Center for Basic and Translational Obesity Research, Children's Hospital Boston, Boston, 02115 MA, USA, Department of Development and Regeneration @ Kulak, KU Leuven, E. Sabbelaan 53, 8500 Kortrijk, Belgium, and NNF Center for Protein Research, Health Sciences Faculty, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
27
|
Song YH, Song JL, Delafontaine P, Godard MP. The therapeutic potential of IGF-I in skeletal muscle repair. Trends Endocrinol Metab 2013; 24:310-9. [PMID: 23628587 PMCID: PMC3732824 DOI: 10.1016/j.tem.2013.03.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/22/2013] [Accepted: 03/22/2013] [Indexed: 12/30/2022]
Abstract
Skeletal muscle loss due to aging, motor-neuron degeneration, cancer, heart failure, and ischemia is a serious condition for which currently there is no effective treatment. Insulin-like growth factor 1 (IGF-I) plays an important role in muscle maintenance and repair. Preclinical studies have shown that IGF-I is involved in increasing muscle mass and strength, reducing degeneration, inhibiting the prolonged and excessive inflammatory process due to toxin injury, and increasing the proliferation potential of satellite cells. However, clinical trials have not been successful due to ineffective delivery methods. Choosing the appropriate isoforms or peptides and developing targeted delivery techniques can resolve this issue. Here we discuss the latest development in the field with special emphasis on novel therapeutic approaches.
Collapse
Affiliation(s)
- Yao-Hua Song
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China
- Corresponding authors: Yao-Hua Song, M.D. Ph.D., Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China, Phone: 86-512-65880899/626, Fax: 86-512-65880929,
| | - Jenny L. Song
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China
| | - Patrice Delafontaine
- Tulane University Heart and Vascular Institute, Tulane University School of Medicine
- Corresponding authors: Yao-Hua Song, M.D. Ph.D., Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China, Phone: 86-512-65880899/626, Fax: 86-512-65880929,
| | - Michael P. Godard
- Department of Nutrition and Kinesiology, University of Central Missouri, Warrensburg, MO
- Corresponding authors: Yao-Hua Song, M.D. Ph.D., Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China, Phone: 86-512-65880899/626, Fax: 86-512-65880929,
| |
Collapse
|
28
|
Borensztein M, Monnier P, Court F, Louault Y, Ripoche MA, Tiret L, Yao Z, Tapscott SJ, Forné T, Montarras D, Dandolo L. Myod and H19-Igf2 locus interactions are required for diaphragm formation in the mouse. Development 2013; 140:1231-9. [PMID: 23406902 DOI: 10.1242/dev.084665] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The myogenic regulatory factor Myod and insulin-like growth factor 2 (Igf2) have been shown to interact in vitro during myogenic differentiation. In order to understand how they interact in vivo, we produced double-mutant mice lacking both the Myod and Igf2 genes. Surprisingly, these mice display neonatal lethality due to severe diaphragm atrophy. Alteration of diaphragm muscle development occurs as early as 15.5 days post-coitum in the double-mutant embryos and leads to a defect in the terminal differentiation of muscle progenitor cells. A negative-feedback loop was detected between Myod and Igf2 in embryonic muscles. Igf2 belongs to the imprinted H19-Igf2 locus. Molecular analyses show binding of Myod on a mesodermal enhancer (CS9) of the H19 gene. Chromatin conformation capture experiments reveal direct interaction of CS9 with the H19 promoter, leading to increased H19 expression in the presence of Myod. In turn, the non-coding H19 RNA represses Igf2 expression in trans. In addition, Igf2 also negatively regulates Myod expression, possibly by reducing the expression of the Srf transcription factor, a known Myod activator. In conclusion, Igf2 and Myod are tightly co-regulated in skeletal muscles and act in parallel pathways in the diaphragm, where they affect the progression of myogenic differentiation. Igf2 is therefore an essential player in the formation of a functional diaphragm in the absence of Myod.
Collapse
Affiliation(s)
- Maud Borensztein
- Genetics and Development Department, Inserm U1016, CNRS UMR 8104, University of Paris Descartes, Institut Cochin, 75014 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Eun B, Sampley ML, Good AL, Gebert CM, Pfeifer K. Promoter cross-talk via a shared enhancer explains paternally biased expression of Nctc1 at the Igf2/H19/Nctc1 imprinted locus. Nucleic Acids Res 2012; 41:817-26. [PMID: 23221643 PMCID: PMC3553941 DOI: 10.1093/nar/gks1182] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Developmentally regulated transcription often depends on physical interactions between distal enhancers and their cognate promoters. Recent genomic analyses suggest that promoter–promoter interactions might play a similarly critical role in organizing the genome and establishing cell-type-specific gene expression. The Igf2/H19 locus has been a valuable model for clarifying the role of long-range interactions between cis-regulatory elements. Imprinted expression of the linked, reciprocally imprinted genes is explained by parent-of-origin-specific chromosomal loop structures between the paternal Igf2 or maternal H19 promoters and their shared tissue-specific enhancer elements. Here, we further analyze these loop structures for their composition and their impact on expression of the linked long non-coding RNA, Nctc1. We show that Nctc1 is co-regulated with Igf2 and H19 and physically interacts with the shared muscle enhancer. In fact, all three co-regulated genes have the potential to interact not only with the shared enhancer but also with each other via their enhancer interactions. Furthermore, developmental and genetic analyses indicate functional significance for these promoter–promoter interactions. Altogether, we present a novel mechanism to explain developmental specific imprinting of Nctc1 and provide new information about enhancer mechanisms and about the role of chromatin domains in establishing gene expression patterns.
Collapse
Affiliation(s)
- Bokkee Eun
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
30
|
van de Corput MPC, de Boer E, Knoch TA, van Cappellen WA, Quintanilla A, Ferrand L, Grosveld FG. Super-resolution imaging reveals three-dimensional folding dynamics of the β-globin locus upon gene activation. J Cell Sci 2012; 125:4630-9. [PMID: 22767512 DOI: 10.1242/jcs.108522] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chromatin architecture is constantly changing because of cellular processes such as proliferation, differentiation and changes in the expression profile during gene activation or silencing. Unravelling the changes that occur in the chromatin structure during these processes has been a topic of interest for many years. It is known that gene activation of large gene loci is thought to occur by means of an active looping mechanism. It was also shown for the β-globin locus that the gene promoter interacts with an active chromatin hub by means of an active looping mechanism. This means that the locus changes in three-dimensional (3D) nuclear volume and chromatin shape. As a means of visualizing and measuring these dynamic changes in chromatin structure of the β-globin locus, we used a 3D DNA-FISH method in combination with 3D image acquisition to volume render fluorescent signals into 3D objects. These 3D chromatin structures were geometrically analysed, and results prior to and after gene activation were quantitatively compared. Confocal and super-resolution imaging revealed that the inactive locus occurs in several different conformations. These conformations change in shape and surface structure upon cell differentiation into a more folded and rounded structure that has a substantially smaller size and volume. These physical measurements represent the first non-biochemical evidence that, upon gene activation, an actively transcribing chromatin hub is formed by means of additional chromatin looping.
Collapse
Affiliation(s)
- Mariëtte P C van de Corput
- Department of Cell Biology and Genetics and Center for Biomedical Genetics, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
31
|
Sodium arsenite represses the expression of myogenin in C2C12 mouse myoblast cells through histone modifications and altered expression of Ezh2, Glp, and Igf-1. Toxicol Appl Pharmacol 2012; 260:250-9. [PMID: 22426358 DOI: 10.1016/j.taap.2012.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 12/22/2022]
Abstract
Arsenic is a toxicant commonly found in water systems and chronic exposure can result in adverse developmental effects including increased neonatal death, stillbirths, and miscarriages, low birth weight, and altered locomotor activity. Previous studies indicate that 20 nM sodium arsenite exposure to C2C12 mouse myocyte cells delayed myoblast differentiation due to reduced myogenin expression, the transcription factor that differentiates myoblasts into myotubes. In this study, several mechanisms by which arsenic could alter myogenin expression were examined. Exposing differentiating C2C12 cells to 20 nM arsenic increased H3K9 dimethylation (H3K9me2) and H3K9 trimethylation (H3K9me3) by 3-fold near the transcription start site of myogenin, which is indicative of increased repressive marks, and reduced H3K9 acetylation (H3K9Ac) by 0.5-fold, indicative of reduced permissive marks. Protein expression of Glp or Ehmt1, a H3-K9 methyltransferase, was also increased by 1.6-fold in arsenic-exposed cells. In addition to the altered histone remodeling status on the myogenin promoter, protein and mRNA levels of Igf-1, a myogenic growth factor, were significantly repressed by arsenic exposure. Moreover, a 2-fold induction of Ezh2 expression, and an increased recruitment of Ezh2 (3.3-fold) and Dnmt3a (~2-fold) to the myogenin promoter at the transcription start site (-40 to +42), were detected in the arsenic-treated cells. Together, we conclude that the repressed myogenin expression in arsenic-exposed C2C12 cells was likely due to a combination of reduced expression of Igf-1, enhanced nuclear expression and promoter recruitment of Ezh2, and altered histone remodeling status on myogenin promoter (-40 to +42).
Collapse
|
32
|
Cianfarani S. Insulin-like growth factor-II: new roles for an old actor. Front Endocrinol (Lausanne) 2012; 3:118. [PMID: 23060858 PMCID: PMC3462314 DOI: 10.3389/fendo.2012.00118] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 09/14/2012] [Indexed: 11/22/2022] Open
Abstract
Insulin-like growth factor-II (IGF-II), traditionally considered as a growth factor implicated in growth of fetal tissues and cancer cells, is now emerging as a potential metabolic regulator. The aim of this overview is to provide the available evidence, obtained in both experimental conditions and in humans, for a role of IGF-II in the fine-tuning of metabolism and body composition. The underlying mechanisms and the potential clinical implications are discussed.
Collapse
Affiliation(s)
- Stefano Cianfarani
- Department of Systems Medicine, Tor Vergata UniversityRome, Italy
- Molecular Endocrinology Unit, Bambino Gesù Children’s HospitalRome, Italy
- *Correspondence: Stefano Cianfarani, Molecular Endocrinology Unit, Bambino Gesù Children’s Hospital, Piazza Sant’Onofrio 4, 00165 Rome, Italy. e-mail:
| |
Collapse
|
33
|
Activation of hypoxia-inducible factor 1 in skeletal muscle cells after exposure to damaged muscle cell debris. Shock 2011; 35:632-8. [PMID: 21283061 DOI: 10.1097/shk.0b013e3182111f3d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Skeletal muscle damage provokes complex repair mechanisms including recruitment of leukocytes as well as activation of myogenic precursor cells such as satellite cells. To study muscle cell repair mechanisms after muscle fiber damage, we used an in vitro model of scrape-injured myotubes. Exposing vital C2C12 myoblasts and myotubes to cell debris of damaged myotubes revealed mRNA upregulation of adrenomedullin (ADM), insulin-like growth factors 1 and 2, metallopeptidase 9, and monocyte chemoattractant protein11. When cell debris was treated with ultrasound, frozen in liquid nitrogen, or heat inactivated before addition to C2C12 cells, gene expression was drastically reduced or completely absent. Moreover, incubations of myoblasts with debris separated by transwell inserts indicated that direct cell contact is required for gene induction. Incubation with albumin and PolyIC ruled out that ADM induction by cell debris simply results from increased protein or nucleic acid concentrations in the supernatant. Because the genes, which were upregulated by cell debris, are potential target genes of hypoxia-inducible factor (HIF), cells were analyzed for HIF-1α expression. Western blot analysis showed accumulation of the α-subunit upon contact to cell debris. Knockdown of HIF-1α in C2C12 cells proved that activation of HIF-1 in response to cell debris was responsible for upregulating ADM and monocyte chemoattractant protein 1. Furthermore, by incubating cells on gas-permeable culture dishes, we excluded a reduced pericellular pO2 induced by cell debris as the cause for ADM upregulation. Our data suggest that damaged myofibers activate HIF-1 in neighboring myotubes and precursor myoblasts by direct contact, concomitantly upregulating factors necessary for angiogenesis, tissue regeneration, and phagocyte recruitment.
Collapse
|
34
|
Novitskaya T, Baserga M, de Caestecker MP. Organ-specific defects in insulin-like growth factor and insulin receptor signaling in late gestational asymmetric intrauterine growth restriction in Cited1 mutant mice. Endocrinology 2011; 152:2503-16. [PMID: 21486933 PMCID: PMC3100618 DOI: 10.1210/en.2010-1385] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Late gestational placental insufficiency resulting in asymmetric intrauterine organ growth restriction (IUGR) is associated with an increased incidence of diabetes, cardiovascular and renal disease in adults. The molecular mechanisms mediating these defects are poorly understood. To explore this, we investigated the mechanisms leading to IUGR in Cited1 knockout mice, a genetic model of late gestational placental insufficiency. We show that loss of placental Cited1 leads to asymmetric IUGR with decreased liver, lung, and kidney sizes and preservation of fetal brain weight. IGF and insulin signaling regulate embryonic organ growth. IGF-I and IGF-II protein and mRNA expression are reduced in livers, lungs, and kidneys of embryonic d 18.5 embryos with IUGR. Decreased IGF-I is associated with reduced activating phosphorylation of the type 1 IGF receptor (pIGF-IR) in the kidney, whereas reduced IGF-II is associated with decreased phosphorylation of the insulin receptor (pIR) in the lung. In contrast, decreased pIR is associated with reduced IGF-I but not IGF-II in the liver. However, pancreatic β-cell mass and serum insulin levels are also decreased in mice with IUGR, suggesting that hepatic IR signaling may be regulated by alterations in fetal insulin production. These findings contrast with observations in IUGR fetal brains in which there is no change in IGF-IR/IR phosphorylation, and IGF-I and IGF-II expression is actually increased. In conclusion, IUGR disrupts normal fetal IGF and insulin production and is associated with organ-specific defects in IGF-IR and IR signaling that may regulate asymmetric IUGR in late gestational placental insufficiency.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
35
|
Esposito D, Rathinasabapathy T, Poulev A, Komarnytsky S, Raskin I. Akt-dependent anabolic activity of natural and synthetic brassinosteroids in rat skeletal muscle cells. J Med Chem 2011; 54:4057-66. [PMID: 21491949 DOI: 10.1021/jm200028h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Brassinosteroids are plant-derived polyhydroxylated derivatives of 5α-cholestane, structurally similar to cholesterol-derived animal steroid hormones and insect ecdysteroids. In this study, we synthesized a set of brassinosteroid analogues of a natural brassinosteroid (22S,23S)-homobrassinolide (HB, 1), including (22S,23S)-homocastasterone (2), (22S,23S)-3α-fluoro-homobrasinolide (3), (22S,23S)-3α-fluoro-homocastasterone (4), (22S,23S)-7-aza-homobrassinolide (5), and (22S,23S)-6-aza-homobrassinolide (6) and studied their anabolic efficacy in the L6 rat skeletal muscle cells in comparison to other synthetic and naturally occurring brassinosteroids (22R,23R)-homobrassinolide (7), (22S,23S)-epibrassinolide (8), and (22R,23R)-epibrassinolide (9). Presence of the 6-keto group in the B ring and stereochemistry of 22α,23α-vicinal hydroxyl groups in the side chain were critical for the anabolic activity, possibly due to higher cytotoxicity of the 22β,23β-hydroxylated brassinosteroids. All anabolic brassinosteroids tested in this study selectively activated PI3K/Akt signaling pathway as evident by increased Akt phosphorylation in vitro. Plant brassinosteroids and their synthetic derivatives may offer a novel therapeutic strategy for promoting growth, repair, and maintenance of skeletal muscles.
Collapse
Affiliation(s)
- Debora Esposito
- Biotech Center, SEBS, Rutgers University, 59 Dudley Road, New Brunswick, New Jersey 08901, United States
| | | | | | | | | |
Collapse
|
36
|
Gardner S, Alzhanov D, Knollman P, Kuninger D, Rotwein P. TGF-β inhibits muscle differentiation by blocking autocrine signaling pathways initiated by IGF-II. Mol Endocrinol 2010; 25:128-37. [PMID: 21106882 DOI: 10.1210/me.2010-0292] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle differentiation and regeneration are regulated by interactions between exogenous hormone- and growth factor-activated signaling cascades and endogenous muscle-specific transcriptional programs. IGF-I and IGF-II can promote muscle differentiation in vitro and can enhance muscle maintenance and repair in vivo. In contrast, members of the TGF-β superfamily prominently inhibit muscle differentiation and regeneration. In this study, we have evaluated functional interactions between IGF- and TGF-β-regulated signaling pathways during skeletal muscle differentiation. In the mouse C2 muscle cell line and in human myoblasts in primary culture, addition of TGF-β1 blocked differentiation in a dose-dependent way, inhibited expression of muscle-specific mRNAs and proteins, and impaired myotube formation. TGF-β1 also diminished stimulation of IGF-II gene expression in myoblasts, decreased IGF-II secretion, and reduced IGF-I receptor activation. To test the hypothesis that TGF-β1 prevents muscle differentiation primarily by blocking IGF-II production, we examined effects of IGF analogues on TGF-β actions in myoblasts. Although both IGF-I and IGF-II restored muscle gene and protein expression, and stimulated myotube formation in the presence of TGF-β1, they did not reduce TGF-β1-stimulated signaling, as measured by no decline in phosphorylation of SMA and mothers against decapentaplegic homolog (Smad)3, or in induction of TGF-β-activated target genes, including a Smad-dependent promoter-reporter plasmid. Our results demonstrate that TGF-β disrupts an IGF-II-stimulated autocrine amplification cascade that is necessary for muscle differentiation in vitro. Because this inhibitory pathway can be overcome by exogenous IGFs, our observations point toward potential strategies to counteract disorders that reduce muscle mass and strength.
Collapse
Affiliation(s)
- Samantha Gardner
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | | | | | | | | |
Collapse
|