1
|
Zhang Y, Katkhada K, Meng LZ, Zhao B, Tong S, Chaabane W, Kallai A, Tobin NP, Östman A, Mega A, Ehnman M. Myogenic IGFBP5 levels in rhabdomyosarcoma are nourished by mesenchymal stromal cells and regulate growth arrest and apoptosis. Cell Commun Signal 2025; 23:184. [PMID: 40234830 PMCID: PMC12001570 DOI: 10.1186/s12964-025-02171-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Mesenchymal stromal cells belong to a diverse collection of cells in different states that are poorly characterized in soft-tissue sarcomas. In this study, we explored tumor growth-regulatory signaling between differentially educated non-malignant mesenchymal stromal cells and malignant cells in pediatric rhabdomyosarcoma (RMS). METHODS Xenograft experiments demonstrated that non-malignant stromal cells influence tumor behavior. Gene expression analysis identified deregulated genes, which were further studied using cell culture assays and patient data. Clinicopathological correlations were made in a discovery cohort (N = 147) and a validation cohort (N = 101). RESULTS The results revealed transiently suppressive paracrine effects of orthotopic stromal cells derived from skeletal muscle. These effects were lost when the stromal cells were exposed to RMS cells, either short-term in vitro, or long-term in hindlimb muscle in vivo. High resolution microarray-based Clariom D gene expression analysis identified insulin-like growth factor binding protein 5 (IGFBP5) as the top upregulated gene in RMS cells exposed to naïve stromal cells, and effects on growth arrest, caspase 3/7 activation, and myogenic cell identity were demonstrated in functional assays. Furthermore, IGFBP5 associated with the caspase 3 substrate growth arrest specific protein 2 (GAS2), lower disease stage and favorable survival in patient cohorts. CONCLUSIONS This study uses functional modeling and omics approaches to identify IGFBP5 as a candidate mediator of anti-tumor growth mechanisms originating from tumor-neighboring mesenchymal stromal cells. Tumors of mesenchymal origin, such as RMS, are known for their heterogeneity, and this could potentially pose a limitation to the study. However, a clinical relevance is emphasized by consistent findings across patient cohorts. These insights pave the way for novel therapeutic strategies modulating activities of stromal cell subsets at primary and metastatic sites in RMS.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karim Katkhada
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Liu Zhen Meng
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Binbin Zhao
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Shanlin Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Wiem Chaabane
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Aditi Kallai
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas P Tobin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Centre for Cancer Biomarkers CCBIO, University of Bergen, 5021, Bergen, Norway
| | - Alessandro Mega
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Ehnman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- PO Bröst- och endokrina tumörer och sarkom, Tema Cancer, Karolinska University Hospital, Visionsgatan 4, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
2
|
Vaccaro LA, Herring K, Wilson A, England E, Smith AL, Ellestad LE. Dynamic changes in insulin-like growth factor binding protein expression occur between embryonic and early post-hatch development in broiler chickens. Poult Sci 2024; 103:104174. [PMID: 39197338 PMCID: PMC11398635 DOI: 10.1016/j.psj.2024.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Somatotropic gene expression has been altered by genetic selection, and developmental changes in insulin-like growth factor (IGF) and IGF binding protein (IGFBP) expression may contribute to rapid growth and muscle accretion in commercial broilers. The objective of this study was to evaluate changes in somatotropic axis activity between embryonic day (e) 12 and post-hatch day (d) 21. Liver and breast muscle (pectoralis major) were collected to measure gene expression, and blood was collected post-hatch to measure circulating IGFs. Liver IGF1 rose rapidly post-hatch and, in muscle, IGF1 exhibited a dynamic expression pattern. Levels decreased from e14 to e20, returned to e14 levels at d3, decreased again at d10, and stayed low thereafter. In both tissues, mRNA levels of several IGFBPs changed between embryogenesis and post-hatch. Liver IGFBP2 increased between e12 and e20, returned to e12 levels on d1, and remained low. Conversely, liver IGFBP4 expression was greater post-hatch than during embryogenesis. Expression of select IGFBPs was depressed in liver during the peri-hatch period. Liver IGFBP1, IGFBP3, IGFBP5, and IGFBP7 mRNA levels all decreased around this time and returned to embryonic levels by d3. In breast muscle, expression of both IGFBP2 and IGFBP4 was reduced after hatch. Circulating insulin-like growth factor IGF1 and IGF2 levels did not change between hatch and d21. These data suggest that post-hatch IGF effects are likely modulated by target tissue IGFR1 and IGFBP expression rather than changes in circulating hormone levels, with promotion or restriction of IGF-receptor binding regulating growth. Downregulation of several IGFBPs synthesized in the liver may facilitate the metabolic transition from utilizing yolk lipids to dietary carbohydrates. Several IGFBPs produced in breast muscle appear to have growth-promotive effects during embryogenesis but restrict growth of this tissue after hatch, as their post-hatch downregulation could facilitate local IGF signaling. These developmental gene expression patterns suggest that somatotropic hormonal signaling regulating growth and muscle accretion might be controlled through differential actions of IGFBPs and provide a basis for future functional studies.
Collapse
Affiliation(s)
- Lauren A Vaccaro
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Kyle Herring
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Abigail Wilson
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Emma England
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Addison L Smith
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Laura E Ellestad
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
3
|
Akhmetshina A, Bianco V, Bradić I, Korbelius M, Pirchheim A, Kuentzel KB, Eichmann TO, Hinteregger H, Kolb D, Habisch H, Liesinger L, Madl T, Sattler W, Radović B, Sedej S, Birner-Gruenberger R, Vujić N, Kratky D. Loss of lysosomal acid lipase results in mitochondrial dysfunction and fiber switch in skeletal muscles of mice. Mol Metab 2024; 79:101869. [PMID: 38160938 PMCID: PMC7615526 DOI: 10.1016/j.molmet.2023.101869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE Lysosomal acid lipase (LAL) is the only enzyme known to hydrolyze cholesteryl esters (CE) and triacylglycerols in lysosomes at an acidic pH. Despite the importance of lysosomal hydrolysis in skeletal muscle (SM), research in this area is limited. We hypothesized that LAL may play an important role in SM development, function, and metabolism as a result of lipid and/or carbohydrate metabolism disruptions. RESULTS Mice with systemic LAL deficiency (Lal-/-) had markedly lower SM mass, cross-sectional area, and Feret diameter despite unchanged proteolysis or protein synthesis markers in all SM examined. In addition, Lal-/- SM showed increased total cholesterol and CE concentrations, especially during fasting and maturation. Regardless of increased glucose uptake, expression of the slow oxidative fiber marker MYH7 was markedly increased in Lal-/-SM, indicating a fiber switch from glycolytic, fast-twitch fibers to oxidative, slow-twitch fibers. Proteomic analysis of the oxidative and glycolytic parts of the SM confirmed the transition between fast- and slow-twitch fibers, consistent with the decreased Lal-/- muscle size due to the "fiber paradox". Decreased oxidative capacity and ATP concentration were associated with reduced mitochondrial function of Lal-/- SM, particularly affecting oxidative phosphorylation, despite unchanged structure and number of mitochondria. Impairment in muscle function was reflected by increased exhaustion in the treadmill peak effort test in vivo. CONCLUSION We conclude that whole-body loss of LAL is associated with a profound remodeling of the muscular phenotype, manifested by fiber type switch and a decline in muscle mass, most likely due to dysfunctional mitochondria and impaired energy metabolism, at least in mice.
Collapse
Affiliation(s)
- Alena Akhmetshina
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Ivan Bradić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Melanie Korbelius
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Katharina B Kuentzel
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Helga Hinteregger
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Dagmar Kolb
- BioTechMed-Graz, Graz, Austria; Core Facility Ultrastructural Analysis, Medical University of Graz, Graz, Austria; Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Hansjoerg Habisch
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Laura Liesinger
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Wolfgang Sattler
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Branislav Radović
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Simon Sedej
- BioTechMed-Graz, Graz, Austria; Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria; Institute of Physiology, Faculty of Medicine, University of Maribor, Slovenia
| | - Ruth Birner-Gruenberger
- BioTechMed-Graz, Graz, Austria; Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria; Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
4
|
Wei Y, Guo D, Bai Y, Liu Z, Li J, Chen Z, Shi B, Zhao Z, Hu J, Han X, Wang J, Liu X, Li S, Zhao F. Transcriptome Analysis of mRNA and lncRNA Related to Muscle Growth and Development in Gannan Yak and Jeryak. Int J Mol Sci 2023; 24:16991. [PMID: 38069312 PMCID: PMC10707067 DOI: 10.3390/ijms242316991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
The production performance of Jeryak, resulting from the F1 generation of the cross between Gannan yak and Jersey cattle, exhibits a significantly superior outcome compared with that of Gannan yak. Therefore, we used an RNA-seq approach to identify differentially expressed mRNAs (DEMs) and differentially expressed lncRNAs (DELs) influencing muscle growth and development in Gannan yaks and Jeryaks. A total of 304 differentially expressed lncRNAs and 1819 differentially expressed mRNAs were identified based on the screening criteria of |log 2 FC| > 1 and FDR < 0.05. Among these, 132 lncRNAs and 1081 mRNAs were found to be down-regulated, while 172 lncRNAs and 738 mRNAs were up-regulated. GO and KEGG analyses showed that the identified DELs and DEMs were enriched in the entries of pathways associated with muscle growth and development. On this basis, we constructed an lncRNA-mRNA interaction network. Interestingly, two candidate DELs (MSTRG.16260.9 and MSTRG.22127.1) had targeting relationships with 16 (MYC, IGFBP5, IGFBP2, MYH4, FGF6, etc.) genes related to muscle growth and development. These results could provide a basis for further studies on the roles of lncRNAs and mRNAs in muscle growth in Gannan yaks and Jeryak breeds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.W.); (D.G.); (B.S.)
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.W.); (D.G.); (B.S.)
| | | | | | | | | | | |
Collapse
|
5
|
Chaves-Coira I, García-Magro N, Zegarra-Valdivia J, Torres-Alemán I, Núñez Á. Cognitive Deficits in Aging Related to Changes in Basal Forebrain Neuronal Activity. Cells 2023; 12:1477. [PMID: 37296598 PMCID: PMC10252596 DOI: 10.3390/cells12111477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Aging is a physiological process accompanied by a decline in cognitive performance. The cholinergic neurons of the basal forebrain provide projections to the cortex that are directly engaged in many cognitive processes in mammals. In addition, basal forebrain neurons contribute to the generation of different rhythms in the EEG along the sleep/wakefulness cycle. The aim of this review is to provide an overview of recent advances grouped around the changes in basal forebrain activity during healthy aging. Elucidating the underlying mechanisms of brain function and their decline is especially relevant in today's society as an increasingly aged population faces higher risks of developing neurodegenerative diseases such as Alzheimer's disease. The profound age-related cognitive deficits and neurodegenerative diseases associated with basal forebrain dysfunction highlight the importance of investigating the aging of this brain region.
Collapse
Affiliation(s)
- Irene Chaves-Coira
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - Nuria García-Magro
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain;
| | - Jonathan Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (J.Z.-V.); (I.T.-A.)
- Facultad de Ciencias de la Salud, Universidad Señor de Sipán, Chiclayo 02001, Peru
| | - Ignacio Torres-Alemán
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (J.Z.-V.); (I.T.-A.)
- Ikerbasque Science Foundation, 48009 Bilbao, Spain
| | - Ángel Núñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| |
Collapse
|
6
|
Shi B, Shi X, Zuo Z, Zhao S, Zhao Z, Wang J, Zhou H, Luo Y, Hu J, Hickford JGH. Identification of differentially expressed genes at different post-natal development stages of longissimus dorsi muscle in Tianzhu white yak. Gene X 2022; 823:146356. [PMID: 35227854 DOI: 10.1016/j.gene.2022.146356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
The regulatory mechanisms controlling post-natal muscle development in the yak (Bos grunniens) are still largely unknown, yet the growth and development of muscle is a complex process that plays a crucial role in determining the yield and quality of an animal's meat. In this study, we performed a transcriptome analysis based on the RNA sequencing (RNA-Seq) of yak longissimus dorsi muscle tissue obtained from calves (6 months of age; 6 M), young adults (30 months of age; 30 M) and adult (54 months of age; 54 M) to identify which genes are differentially expressed and to investigate their temporal expression profiles. In total, 1788 differentially expressed genes (DEGs) (|log2FC| ≥ 1, P-adjusted < 0.05) were detected by pairwise comparisons between the different age groups. The expression levels of 10 of the DEGs were confirmed using reverse transcription-quantitative PCR (RT-qPCR), and the results were consistent with the transcriptome profile. A time-series expression profile analysis clustered the DEGs into four groups that could be divided into two classes (P < 0.05): class 1 profiles, which had up-regulated patterns of gene expression and class 2 profiles, which featured down-regulated patterns. Based on that cluster analysis, GO enrichment analysis revealed 1073, 127, and 184 terms as significantly enriched in biological process (BP), cellular component (CC), and molecular function (MF) categories in the class 1 profiles, while 714, 66, and 206 terms were significantly enriched in BP, CC, and MF in the class 2 profiles. A KEGG pathway analysis revealed that DEGs from the class 1 profiles were enriched in 62 pathways, with the most enriched being the phosphoinositide 3-kinase (PI3K) - protein kinase B (Akt)-signaling pathway. The DEGs from the class 2 profiles were enriched in 16 pathways, of which forkhead box protein O (FoxO) - signaling was the most enriched. Taken together, these results provide insight into the mechanisms of skeletal muscle development, as well suggesting some potential genes of importance for yak meat production.
Collapse
Affiliation(s)
- Bingang Shi
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xuehong Shi
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Zhi Zuo
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shijie Zhao
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Zhidong Zhao
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiqing Wang
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Huitong Zhou
- Gene-Marker Laboratory, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Yuzhu Luo
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiang Hu
- Faculty of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Jon G H Hickford
- Gene-Marker Laboratory, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand.
| |
Collapse
|
7
|
Differential Expression of IGF1, IGFBP5, MSTN, and MYH1 Across Different Age Classes in American Quarter Horses. J Equine Vet Sci 2020; 94:103226. [PMID: 33077104 DOI: 10.1016/j.jevs.2020.103226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 11/22/2022]
Abstract
The objective of this study was to determine the influence of age on expression of insulin-like growth factor 1 (IGF1), insulin-like growth factor binding protein (IGFBP5), myostatin (MSTN), and myosin (MYH1) genes which are related to growth and muscle development in the American Quarter Horse. Thus, horses (n = 10) from weanling, yearling, 2-, 3-, and 10-year-old age classes were sampled and gene expression was assessed by RT-qPCR. ΔCT was calculated using the hypoxanthine-guanine phosphoribosyltransferase gene as an internal normalizer. The generalized linear model was used to determine differentially expressed genes, by pairwise comparison between ages. Among technical replicates, the coefficient of variation ranged from 1.0 to 5.2% and was lower than the variation observed between biological replicates (2.1-12.9%). IGF1 demonstrated significantly lower expression in the 3-year-old age class than in weanlings and yearlings, but the 10-year-old age class displayed a significantly higher level than 2- and 3-year-old age classes. Expression of IGFBP5 was highest in weanlings compared with all other age classes. Expression of MSTN was significantly higher in weanlings than in other age classes, whereas 10-year-old horses had an intermediate level of expression, but significantly different from yearlings, 2- and 3-year-old fillies. Finally, expression of MYH1 was lower in 2- and 10-year-old horses than in weanlings and yearlings, whereas 3-year-old fillies demonstrated an intermediate level of expression. Differential expression patterns observed in this preliminary study provide insight into the physiological changes occurring throughout the life span of horses. These patterns could also help explain the variation in performance and endurance between individuals at different developmental stages.
Collapse
|
8
|
Hunt ER, Villasanta-Tezanos AG, Butterfield TA, Lattermann C, Jacobs CA. Upregulation of Systemic Inflammatory Pathways Following Anterior Cruciate Ligament Injury Relates to Both Cartilage and Muscular Changes: A Pilot Study. J Orthop Res 2020; 38:387-392. [PMID: 31517396 DOI: 10.1002/jor.24467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/03/2019] [Indexed: 02/04/2023]
Abstract
In conjunction with cartilage breakdown, muscle maladaptation including atrophy and increased fibrosis have been observed in the quadriceps following anterior cruciate ligament (ACL) injury. Previously observed upregulated muscle-related proteins in the synovial fluid following ACL rupture allude to cellular communication between the joint and muscle. Therefore, the purpose of this study was to determine whether muscle-related analytes are differentially expressed in the serum. Sixteen patients with an acute ACL tear participated in this IRB-approved study. Serum was obtained at two different time points at a mean of 6 and 14 days post-injury, and serum was analyzed by a highly multiplexed assay of 1,300 proteins. Pathway analysis using DAVID was performed; genes included met three criteria: significant change between the two study time points using a paired t test, significant change between the two study time points using a Mann-Whitney non-parametric test, and significant Benjamini post hoc analysis. Twelve analytes significantly increased between time points. Proteins chitinase-3-like protein 1 (p = 0.01), insulin-like growth factor binding protein 1 (p = 0.01), insulin-like growth factor binding protein 5 (p = 0.02), renin (p = 0.004), and lymphotoxin alpha 1: beta 2 (p = 0.03) were significantly upregulated in serum following acute ACL injury. The current results confirm the inflammatory pattern previously seen in the synovial fluid thought to play a role in the progression of post-traumatic osteoarthritis after ACL injury, and this data also provides further insights into important communication between the joint and quadriceps group, whose function is important in long term health. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:387-392, 2020.
Collapse
Affiliation(s)
- Emily R Hunt
- Department of Orthopedic Surgery, University of Kentucky, 740 S Limestone, Suite K401, Lexington, Kentucky, 40536-0284
| | | | - Timothy A Butterfield
- College of Health Sciences, Rehabilitation Science PhD Program, University of Kentucky, Lexington, Kentucky
| | - Christian Lattermann
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cale A Jacobs
- Department of Orthopedic Surgery, University of Kentucky, 740 S Limestone, Suite K401, Lexington, Kentucky, 40536-0284
| |
Collapse
|
9
|
Nindl BC, Ahtiainen J, Gagnon SS, Taipale RS, Pierce JR, Martin BJ, Beckner ME, Lehti M, Häkkinen K, Kyröläinen H. Microdialysis-Assessed Exercised Muscle Reveals Localized and Differential IGFBP Responses to Unilateral Stretch Shortening Cycle Exercise. Front Endocrinol (Lausanne) 2020; 11:315. [PMID: 32547489 PMCID: PMC7272679 DOI: 10.3389/fendo.2020.00315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023] Open
Abstract
Microdialysis allows for a preview into local muscle metabolism and can provide physiological insight that blood measurements cannot. Purpose: To examine the potential differential IGF-I system regulation in interstitial fluid during unilateral stretch shortening cycle exercise. Methods: 10 men (26 ± 7 year) performed unilateral jumping [stretch shortening cycle (SSC) exercise at 50% of optimal jump height] until volitional fatigue on a sled apparatus. Biological sampling took place using a catheter inserted into an antecubital vein (serum), and 100 kDa microdialysis probes inserted into the thigh muscle of each exercise/control leg (dialysate). Serum was drawn before (Pre; -3 h) and after SSC [Post I (+0 h), II (+3 h), or III (+20 h)]; dialysate was sampled for 2 h before (Pre), during/immediately after (Ex), and 3 h into recovery (Rec) following SSC. IGF-I system parameters (free/total IGF-I and IGFBPs 1-6) were measured with immunoassays. Interstitial free IGF-I was estimated from dialysate IGF-I and relative recovery (ethanol) correction. Data were analyzed with repeated measures ANOVA. Results: Serum total IGF-I remained elevated +3 h (Post II: 182.8 ± 37.6 vs. Pre: 168.3 ± 35.0 ng/mL, p < 0.01), but returned to baseline by +20 h (Post III vs. Pre, p = 0.31). No changes in serum free IGF-I were noted. Serum BP-1 and -3 increased over baseline, but not until + 20 h after SSC (Post III vs. Pre: 7.6 ± 4.9 vs. 3.7 ± 2.3 and 1,048.6 ± 269.2 vs. 891.4 ± 171.2 ng/mL, respectively). We observed a decreased serum BP-6 +3 h after SSC (p < 0.01), followed by a return to baseline at +20 h (p = 0.64 vs. Pre). There were no exercise-induced changes in serum BP-2, -4, or -5. Unlike serum, there were no changes in dialysate or interstitial free IGF-I in either leg (p > 0.05). Dialysate BP-1 remained increased in both exercise and control legs through 3 h into recovery (Rec vs. Pre, p < 0.01). Dialysate BP-3 also demonstrated a prolonged elevation over Pre SSC concentrations, but in the exercise leg only (Ex and Rec vs. Pre, p < 0.04). We observed a prolonged decrease in dialysate BP-5 (Ex and Rec vs. Pre, p < 0.03) and an increase in BP-4 IP in the exercise leg only. There were no changes relative to Pre SSC in dialysate BP-2 or -6. Conclusions: Unilateral exercise drives differential regulation of the IGF-I system at both local and systemic levels. More specifically, this is the first study to demonstrate that localized exercise increases IGFBP-3, IGFBP-4 and decreases in IGFBP-5 in muscle interstitial fluid.
Collapse
Affiliation(s)
- Bradley C. Nindl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
- Army Public Health Center, Aberdeen Proving Ground, MD, United States
- *Correspondence: Bradley C. Nindl
| | - Juha Ahtiainen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Sheila S. Gagnon
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Ritva S. Taipale
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Joseph R. Pierce
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
- Army Public Health Center, Aberdeen Proving Ground, MD, United States
| | - Brian J. Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Meaghan E. Beckner
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - M. Lehti
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Keijo Häkkinen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Heikki Kyröläinen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| |
Collapse
|
10
|
Clemmons DR. Role of IGF-binding proteins in regulating IGF responses to changes in metabolism. J Mol Endocrinol 2018; 61:T139-T169. [PMID: 29563157 DOI: 10.1530/jme-18-0016] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022]
Abstract
The IGF-binding protein family contains six members that share significant structural homology. Their principal function is to regulate the actions of IGF1 and IGF2. These proteins are present in plasma and extracellular fluids and regulate access of both IGF1 and II to the type I IGF receptor. Additionally, they have functions that are independent of their ability to bind IGFs. Each protein is regulated independently of IGF1 and IGF2, and this provides an important mechanism by which other hormones and physiologic variables can regulate IGF actions indirectly. Several members of the family are sensitive to changes in intermediary metabolism. Specifically the presence of obesity/insulin resistance can significantly alter the expression of these proteins. Similarly changes in nutrition or catabolism can alter their synthesis and degradation. Multiple hormones such as glucocorticoids, androgens, estrogen and insulin regulate IGFBP synthesis and bioavailability. In addition to their ability to regulate IGF access to receptors these proteins can bind to distinct cell surface proteins or proteins in extracellular matrix and several cellular functions are influenced by these interactions. IGFBPs can be transported intracellularly and interact with nuclear proteins to alter cellular physiology. In pathophysiologic states, there is significant dysregulation between the changes in IGFBP synthesis and bioavailability and changes in IGF1 and IGF2. These discordant changes can lead to marked alterations in IGF action. Although binding protein physiology and pathophysiology are complex, experimental results have provided an important avenue for understanding how IGF actions are regulated in a variety of physiologic and pathophysiologic conditions.
Collapse
Affiliation(s)
- David R Clemmons
- Department of MedicineUNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
Jia J, Ahmed I, Liu L, Liu Y, Xu Z, Duan X, Li Q, Dou T, Gu D, Rong H, Wang K, Li Z, Talpur MZ, Huang Y, Wang S, Yan S, Tong H, Zhao S, Zhao G, te Pas MFW, Su Z, Ge C. Selection for growth rate and body size have altered the expression profiles of somatotropic axis genes in chickens. PLoS One 2018; 13:e0195378. [PMID: 29630644 PMCID: PMC5891002 DOI: 10.1371/journal.pone.0195378] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 03/21/2018] [Indexed: 11/18/2022] Open
Abstract
The growth hormone / insulin-like growth factor-1 (GH/IGF-1) pathway of the somatotropic axis is the major controller for growth rate and body size in vertebrates, but the effect of selection on the expression of GH/IGF-1 somatotropic axis genes and their association with body size and growth performance in farm animals is not fully understood. We analyzed a time series of expression profiles of GH/IGF-1 somatotropic axis genes in two chicken breeds, the Daweishan mini chickens and Wuding chickens, and the commercial Avian broilers hybrid exhibiting markedly different body sizes and growth rates. We found that growth rate and feed conversion efficiency in Daweishan mini chickens were significantly lower than those in Wuding chickens and Avian broilers. The Wuding and Daweishan mini chickens showed higher levels of plasma GH, pituitary GH mRNA but lower levels of hepatic growth hormone receptor (GHR) mRNA than in Avian broilers. Daweishan mini chickens showed significantly lower levels of plasma IGF-1, thigh muscle and hepatic IGF-1 mRNA than did Avian broilers and Wuding chickens. These results suggest that the GH part of the somatotropic axis is the main regulator of growth rate, while IGF-1 may regulate both growth rate and body weight. Selection for growth performance and body size have altered the expression profiles of somatotropic axis genes in a breed-, age-, and tissue-specific manner, and manner, and alteration of regulatory mechanisms of these genes might play an important role in the developmental characteristics of chickens.
Collapse
Affiliation(s)
- Junjing Jia
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Irfan Ahmed
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Lixian Liu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Yong Liu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Zhiqiang Xu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Xiaohua Duan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Qihua Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Tengfei Dou
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Dahai Gu
- Department of Food Science, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Hua Rong
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Kun Wang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Zhengtian Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Mir Zulqarnain Talpur
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Ying Huang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Shanrong Wang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Shixiong Yan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Huiquan Tong
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Sumei Zhao
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
| | - Guiping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Marinus F. W. te Pas
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
- Animal Breeding and Genetics, Wageningen UR Livestock Science, Wageningen, The Netherlands
- Dali University, Dali, Yunnan Province, People’s Republic of China
- * E-mail: (MFWP); (ZS); (CG)
| | - Zhengchang Su
- Department of Bioinformatics and Genomics, College of Computing and Informatics, the University of North Carolina at Charlotte, Charlotte, NC, United States of America
- * E-mail: (MFWP); (ZS); (CG)
| | - Changrong Ge
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
- * E-mail: (MFWP); (ZS); (CG)
| |
Collapse
|
12
|
Comprehensive analysis of lncRNAs and mRNAs in skeletal muscle of rainbow trout (Oncorhynchus mykiss) exposed to estradiol. Sci Rep 2017; 7:11780. [PMID: 28924252 PMCID: PMC5603547 DOI: 10.1038/s41598-017-12136-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/05/2017] [Indexed: 02/01/2023] Open
Abstract
Estradiol (E2) is a steroid hormone that negatively affects muscle growth in rainbow trout (Oncorhynchus mykiss), but the mechanisms directing with this response are not fully understood. To better characterize the effects of E2 in muscle, we identified differentially regulated mRNAs and lncRNAs in juvenile rainbow trout exposed to E2. Here, we performed next-generation RNA sequencing and comprehensive bioinformatics analyses to characterize the transcriptome profiles, including mRNAs and long noncoding RNAs (lncRNAs), in skeletal muscle of rainbow trout injected with E2. A total of 226 lncRNAs and 253 mRNAs were identified as differentially regulated. We identified crucial pathways, including several signal transduction pathways, hormone response, oxidative response and protein, carbon and fatty acid metabolism pathways. Subsequently, a functional lncRNA-mRNA co-expression network was constructed, which consisted of 681 co-expression relationships between 164 lncRNAs and 201 mRNAs. Moreover, a lncRNA-pathway network was constructed. A total of 65 key lncRNAs were identified that regulate 20 significantly enriched pathways. Overall, our analysis provides insights into mRNA and lncRNA networks in rainbow trout skeletal muscle and their regulation by E2 while understanding the molecular mechanism of lncRNAs.
Collapse
|
13
|
Kalbe C, Lösel D, Block J, Lefaucheur L, Brüssow KP, Bellmann O, Pfuhl R, Puppe B, Otten W, Metges CC, Rehfeldt C. Moderate high or low maternal protein diets change gene expression but not the phenotype of skeletal muscle from porcine fetuses. Domest Anim Endocrinol 2017; 58:63-75. [PMID: 27664381 DOI: 10.1016/j.domaniend.2016.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 08/03/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
The aim of our study was to characterize the immediate phenotypic and adaptive regulatory responses of fetuses to different in utero conditions reflecting inadequate maternal protein supply during gestation. The gilts fed high- (250% above control) or low- (50% under control) protein diets isoenergetically adjusted at the expense of carbohydrates from the day of insemination until the fetuses were collected at day 64 or 94 of gestation. We analyzed body composition, histomorphology, biochemistry, and messenger RNA (mRNA) expression of fetal skeletal muscle. Both diets had only marginal effects on body composition and muscular cellularity of fetuses including an unchanged total number of myofibers. However, mRNA expression of myogenic regulatory factors (MYOG, MRF4, P ≤ 0.1), IGF system (IGF1, IGF1R, P ≤ 0.05) and myostatin antagonist FST (P = 0.6, in males only) was reduced in the fetal muscle exposed to a maternal low-protein diet. As a result of excess protein, MYOD, MYOG, IGF1R, and IGFBP5 mRNA expression (P ≤ 0.05) was upregulated in fetal muscle. Differences in muscular mRNA expression indicate in utero regulatory adaptive responses to maternal diet. Modulation of gene expression immediately contributes to the maintenance of an appropriate fetal phenotype that would be similar to that observed in the control fetuses. Moreover, we suggest that the modified gene expression in fetal skeletal muscle can be viewed as the origin of developmental muscular plasticity involved in the concept of fetal programming.
Collapse
Affiliation(s)
- C Kalbe
- Institute of Muscle Biology & Growth, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| | - D Lösel
- Institute of Muscle Biology & Growth, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - J Block
- Institute of Muscle Biology & Growth, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - L Lefaucheur
- Institut National de la Recherche Agronomique (INRA), Unité Mixte de Recherche 1348 sur la Physiologie, l'Environnement et la Génétique pour l'Animal et les Systèmes d'Elevage, F-35590 Saint-Gilles, France
| | - K-P Brüssow
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - O Bellmann
- Institutional Veterinarian of the Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - R Pfuhl
- Institute of Muscle Biology & Growth, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - B Puppe
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - W Otten
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - C C Metges
- Institute of Nutritional Physiology 'Oskar Kellner', Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - C Rehfeldt
- Institute of Muscle Biology & Growth, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| |
Collapse
|
14
|
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
|
15
|
Haramizu S, Mori T, Yano M, Ota N, Hashizume K, Otsuka A, Hase T, Shimotoyodome A. Habitual exercise plus dietary supplementation with milk fat globule membrane improves muscle function deficits via neuromuscular development in senescence-accelerated mice. SPRINGERPLUS 2014; 3:339. [PMID: 25110626 PMCID: PMC4125610 DOI: 10.1186/2193-1801-3-339] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/26/2014] [Indexed: 01/11/2023]
Abstract
We examined the effects of habitual exercise plus nutritional intervention through consumption of milk fat globule membrane (MFGM), a milk component, on aging-related deficits in muscle mass and function in senescence-accelerated P1 mice. Combining wheel-running and MFGM (MFGMEx) intake significantly attenuated age-related declines in quadriceps muscle mass (control: 318 ± 6 mg; MFGMEx: 356 ± 9 mg; P < 0.05) and in contractile force (1.4-fold and 1.5-fold higher in the soleus and extensor digitorum longus muscles, respectively). Microarray analysis of genes in the quadriceps muscle revealed that MFGMEx stimulated neuromuscular development; this was supported by significantly increased docking protein-7 (Dok-7) and myogenin mRNA expression. Treatment of differentiating myoblasts with MFGM-derived phospholipid or sphingolipid fractions plus mechanical stretching also significantly increased Dok-7 mRNA expression. These findings suggest that habitual exercise plus dietary MFGM improves muscle function deficits through neuromuscular development, and that phospholipid and sphingolipid in MFGM contribute to its physiological actions.
Collapse
Affiliation(s)
- Satoshi Haramizu
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Takuya Mori
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Michiko Yano
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Noriyasu Ota
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | | | - Atsuko Otsuka
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Tadashi Hase
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | | |
Collapse
|
16
|
Mukherjee A, Larson EA, Klein RF, Rotwein P. Distinct actions of akt1 on skeletal architecture and function. PLoS One 2014; 9:e93040. [PMID: 24663486 PMCID: PMC3963959 DOI: 10.1371/journal.pone.0093040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/28/2014] [Indexed: 11/24/2022] Open
Abstract
Skeletal integrity is dependent on the coordinated actions of bone-forming osteoblasts and bone-resorbing osteoclasts, which recognize and respond to multiple environmental inputs. Here we have studied the roles in bone development and growth of Akt1 and Akt2, two closely related signaling proteins, by evaluating mice lacking either of these enzymes. Global deficiency of Akt1 but not Akt2 caused a reduction in whole body and femoral bone mineral density, in femoral cortical thickness and volume, and in trabecular thickness in both males and females when measured at 20-weeks of age, which was reflected in diminished femoral resistance to fracture. Haplo-deficiency of Akt1 in male mice also decreased femoral cortical and trabecular skeletal parameters, and reduced bone strength. Cell-based studies showed that genetic Akt1 deficiency diminished the rate of proliferation of osteoblast progenitors and impaired osteoclast differentiation in primary culture but that loss of Akt2 did not. Our results demonstrate differential effects of Akt1 and Akt2 on skeletal maturation and architecture through actions on both osteoblast and osteoclast precursors.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Emily A. Larson
- Bone and Mineral Research Unit, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Robert F. Klein
- Bone and Mineral Research Unit, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
- Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Peter Rotwein
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
17
|
IGF-1 antibody prolongs the effective duration time of botulinum toxin in decreasing muscle strength. Int J Mol Sci 2013; 14:9051-61. [PMID: 23698763 PMCID: PMC3676772 DOI: 10.3390/ijms14059051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 03/28/2013] [Accepted: 04/15/2013] [Indexed: 12/12/2022] Open
Abstract
Botulinum toxin type-A (Btx-A), a powerful therapeutic tool in various medical specialties, requires repeated injections to maintain its effect. Therefore, novel methods to prolong the effective duration time of Btx-A are highly needed. Rats were assigned to three major groups: control group (n = 30), Btx-A group (n = 30), and IGF-1 Ab groups. IGF-1 Ab groups were composed by sub-groups A1-A5 (each has 25 rats) for the subsequent IGF-1Ab dose-effect study. Muscle strength was determined by a survey system for rat lower limbs nerve and muscle function. Muscle-specific receptor tyrosine kinase (MuSK), Insulin-like growth factor binding protein-5 (IGFBP5), and growth-associated protein, 43-kDa (GAP43) were determined by real-time polymerase chain reactions (PCRs) and Western blot. We found that Btx-A decreased the muscle strength, with a paralysis maintained for 70 days. IGF-1Ab prolonged the effective duration time of Btx-A. Real-time PCRs and Western blot showed that IGF-1Ab delayed the increase of MuSK and IGFBP5 after Btx-A injection, without affecting GAP43. These results indicate that IGF-1Ab might prolong the effective duration time of Btx-A on muscle strength through delaying the increase of MuSK. It would be interesting to determine whether IGF-1Ab can be used as an auxiliary measure to the Btx-A treatment in the future.
Collapse
|
18
|
Sharples AP, Al-Shanti N, Lewis MP, Stewart CE. Reduction of myoblast differentiation following multiple population doublings in mouse C2 C12 cells: a model to investigate ageing? J Cell Biochem 2012; 112:3773-85. [PMID: 21826704 DOI: 10.1002/jcb.23308] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ageing skeletal muscle displays declines in size, strength, and functional capacity. Given the acknowledged role that the systemic environment plays in reduced regeneration (Conboy et al. [2005] Nature 433: 760-764), the role of resident satellite cells (termed myoblasts upon activation) is relatively dismissed, where, multiple cellular divisions in-vivo throughout the lifespan could also impact on muscular deterioration. Using a model of multiple population doublings (MPD) in-vitro thus provided a system in which to investigate the direct impact of extensive cell duplications on muscle cell behavior. C(2) C(12) mouse skeletal myoblasts (CON) were used fresh or following 58 population doublings (MPD). As a result of multiple divisions, reduced morphological and biochemical (creatine kinase, CK) differentiation were observed. Furthermore, MPD cells had significantly increased cells in the S and decreased cells in the G1 phases of the cell cycle versus CON, following serum withdrawal. These results suggest continued cycling rather than G1 exit and thus reduced differentiation (myotube atrophy) occurs in MPD muscle cells. These changes were underpinned by significant reductions in transcript expression of: IGF-I and myogenic regulatory factors (myoD and myogenin) together with elevated IGFBP5. Signaling studies showed that decreased differentiation in MPD was associated with decreased phosphorylation of Akt, and with later increased phosphorylation of JNK1/2. Chemical inhibition of JNK1/2 (SP600125) in MPD cells increased IGF-I expression (non-significantly), however, did not enhance differentiation. This study provides a potential model and molecular mechanisms for deterioration in differentiation capacity in skeletal muscle cells as a consequence of multiple population doublings that would potentially contribute to the ageing process.
Collapse
Affiliation(s)
- Adam P Sharples
- Faculty of Science and Engineering, Institute for Biomedical Research into Human Movement and Health (IRM), Manchester Metropolitan University, John Dalton Building, Oxford Road, Manchester, UK.
| | | | | | | |
Collapse
|
19
|
Güllü G, Karabulut S, Akkiprik M. Functional roles and clinical values of insulin-like growth factor-binding protein-5 in different types of cancers. CHINESE JOURNAL OF CANCER 2012; 31:266-80. [PMID: 22313597 PMCID: PMC3777492 DOI: 10.5732/cjc.011.10405] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) are critical regulators of the mitogenic activity of insulin-like growth factors (IGFs). IGFBP5, one of these IGFBPs, has special structural features, including a nuclear transport domain, heparin-binding motif, and IGF/extracellular matrix/acid-labile subunit-binding sites. Furthermore, IGFBP5 has several functional effects on carcinogenesis and even normal cell processes, such as cell growth, death, motility, and tissue remodeling. These biological effects are sometimes related with IGF (IGF-dependent effects) and sometimes not (IGF-independent effects). The functional role of IGFBP5 is most likely determined in a cell-type and tissue-type specific manner but also depends on cell context, especially in terms of the diversity of interacting proteins and the potential for nuclear localization. Clinical findings show that IGFBP5 has the potential to be a useful clinical biomarker for predicting response to therapy and clinical outcome of cancer patients. In this review, we summarize the functional diversity and clinical importance of IGFBP5 in different types of cancers.
Collapse
Affiliation(s)
- Gökçe Güllü
- Department of Medical Biology, School of Medicine, DMarmara University, Istanbul 34468, Turkey
| | | | | |
Collapse
|
20
|
López-Menduiña M, Martín AI, Castillero E, Villanúa MA, López-Calderón A. Short-term growth hormone or IGF-I administration improves the IGF-IGFBP system in arthritic rats. Growth Horm IGF Res 2012; 22:22-29. [PMID: 22244673 DOI: 10.1016/j.ghir.2011.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 12/08/2011] [Accepted: 12/14/2011] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Adjuvant-induced arthritis is an experimental model of rheumatoid arthritis that inhibits the GH-IGF-I axis and decreases body weight gain and muscle mass. Although chronic GH or IGF-I treatment increases body weight gain in arthritic rats, muscle resistance to GH and IGF-I is a very common complication in inflammatory diseases. In this study we examine the effect of short-term administration of rhGH and rhIGF-I on liver and muscle IGF-I, IGFBP-3 and -5 as well as on the ubiquitin-ligases MuRF1 and atrogin-1 in the muscle of arthritic rats. DESIGN Arthritis was induced in adult male Wistar rats by an intradermal injection of 4 mg of Freund's adjuvant. Fifteen days after adjuvant injection, 300 μg/kg of rhGH or 200 μg/kg of rhIGF or saline was administrated 18 and 3h before decapitation. A pair-fed group injected with saline was included in order to discard a possible effect of decreased food intake. Gene expression of IGF-I, GHR, IGFBP-3, IGFBP-5, atrogin-1 and MuRF1 were quantified using RT-PCR. In serum, IGF-I was measured by radioimmunoassay (RIA) and IGFBP-3 by ligand blot. RESULTS Arthritis decreased serum IGF-I and IGF mRNA in liver (P<0.05), but not in skeletal muscle. In arthritic rats, rhGH increased serum IGF-I and liver IGF-I mRNA similar to the levels of pair-fed rats. Arthritis increased atrogin-1, MuRF1, IGFBP-3 and IGFBP-5 mRNA in muscle (P<0.01). IGFBP-3 mRNA was downregulated by rhIGF-I, but not by rhGH, administration in control and arthritic rats (P<0.05). Administration of rhGH and rhIGF-I increased IGFBP-5 in the gastrocnemius of arthritic rats. CONCLUSIONS Short-term rhGH and rhIGF-I administration was found to increase muscle IGFBP-5 mRNA, whereas only rhIGF-I administration decreased muscle IGFBP-3 mRNA in control and arthritic rats. These data suggest that arthritis does not induce GH or IGF-I resistance in skeletal muscle.
Collapse
Affiliation(s)
- M López-Menduiña
- Department of Physiology, Faculty of Medicine, Complutense University, Avda. Complutense s/n. 28040 Madrid, Spain
| | | | | | | | | |
Collapse
|
21
|
Nili M, Mukherjee A, Shinde U, David L, Rotwein P. Defining the disulfide bonds of insulin-like growth factor-binding protein-5 by tandem mass spectrometry with electron transfer dissociation and collision-induced dissociation. J Biol Chem 2011; 287:1510-9. [PMID: 22117064 DOI: 10.1074/jbc.m111.285528] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The six high-affinity insulin-like growth factor-binding proteins (IGFBPs) comprise a conserved family of secreted molecules that modulate IGF actions by regulating their half-life and access to signaling receptors, and also exert biological effects that are independent of IGF binding. IGFBPs are composed of cysteine-rich amino- (N-) and carboxyl- (C-) terminal domains, along with a cysteine-poor central linker segment. IGFBP-5 is the most conserved IGFBP, and contains 18 cysteines, but only 2 of 9 putative disulfide bonds have been mapped to date. Using a mass spectrometry (MS)-based strategy combining sequential electron transfer dissociation (ETD) and collision-induced dissociation (CID) steps, in which ETD fragmentation preferentially induces cleavage of disulfide bonds, and CID provides exact disulfide linkage assignments between liberated peptides, we now have definitively mapped 5 disulfide bonds in IGFBP-5. In addition, in conjunction with ab initio molecular modeling we are able to assign the other 4 disulfide linkages to within a GCGCCXXC motif that is conserved in five IGFBPs. Because of the nature of ETD fragmentation MS experiments were performed without chemical reduction of IGFBP-5. Our results not only establish a disulfide bond map of IGFBP-5 but also define a general approach that takes advantage of the specificity of ETD and the scalability of tandem MS, and the predictive power of ab initio molecular modeling to characterize unknown disulfide linkages in proteins.
Collapse
Affiliation(s)
- Mahta Nili
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon 97239-3098, USA
| | | | | | | | | |
Collapse
|
22
|
Castillero E, López-Menduiña M, Martín AI, Villanúa MÁ, López-Calderón A. Comparison of the effects of the n-3 polyunsaturated fatty acid eicosapentaenoic and fenofibrate on the inhibitory effect of arthritis on IGF1. J Endocrinol 2011; 210:361-8. [PMID: 21715432 DOI: 10.1530/joe-11-0170] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Adjuvant-induced arthritis is a chronic inflammatory illness that induces muscle wasting and decreases circulating IGF1. Eicosapentaenoic acid (EPA) and fenofibrate, a peroxisome proliferator-activated receptors α agonist, have anti-inflammatory actions and ameliorate muscle wasting in arthritic rats. The aim of this work was to elucidate whether EPA and fenofibrate administration are able to prevent the effect of arthritis on the IGF1-IGFBP system. On day 4 after adjuvant injection control, arthritic rats were gavaged with EPA (1 g/kg) or fenofibrate (300 mg/kg) until day 15 when all rats were killed. Arthritis decreased body weight gain, serum IGF1, and liver Igf1 mRNA, whereas it increased gastrocnemius Igfbp3 mRNA. EPA, but not fenofibrate, administration prevented arthritis-induced decrease in serum IGF1 and liver Igf1 mRNA. In the rats treated with EPA arthritis increased Igfbp5 mRNA in the gastrocnemius. Fenofibrate treatment decreased IGF1 and Igf1 mRNA in the liver and gastrocnemius. In arthritic rats, fenofibrate increased body weight gain and decreased gastrocnemius Igfbp3 and Igfbp5 mRNA. These data suggest that the mechanisms through which EPA and fenofibrate act on the IGF1 system and ameliorate muscle wasting in arthritic rats are different. EPA administration increased circulating levels of IGF1, whereas fenofibrate decreased the Igfbp3 and Igfbp5 in the gastrocnemius muscle.
Collapse
Affiliation(s)
- Estíbaliz Castillero
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
23
|
Tinoco AD, Saghatelian A. Investigating endogenous peptides and peptidases using peptidomics. Biochemistry 2011; 50:7447-61. [PMID: 21786763 DOI: 10.1021/bi200417k] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rather than simply being protein degradation products, peptides have proven to be important bioactive molecules. Bioactive peptides act as hormones, neurotransmitters, and antimicrobial agents in vivo. The dysregulation of bioactive peptide signaling is also known to be involved in disease, and targeting peptide hormone pathways has been a successful strategy in the development of novel therapeutics. The importance of bioactive peptides in biology has spurred research to elucidate the function and regulation of these molecules. Classical methods for peptide analysis have relied on targeted immunoassays, but certain scientific questions necessitated a broader and more detailed view of the peptidome--all the peptides in a cell, tissue, or organism. In this review we discuss how peptidomics has emerged to fill this need through the application of advanced liquid chromatography--tandem mass spectrometry (LC-MS/MS) methods that provide unique insights into peptide activity and regulation.
Collapse
Affiliation(s)
- Arthur D Tinoco
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | | |
Collapse
|
24
|
Keady SM, Kenny DA, Keane MG, Waters SM. Effect of sire breed and genetic merit for carcass weight on the transcriptional regulation of the somatotropic axis in longissimus dorsi of crossbred steers. J Anim Sci 2011; 89:4007-16. [PMID: 21724946 DOI: 10.2527/jas.2011-4032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The somatotropic axis plays an important role in postnatal growth, development, and differentiation of skeletal muscle. The aim of this study was to examine the effect of sire breed and sire EPD for carcass weight (EPD(cwt)) on the expression of components of the somatotropic axis in LM of beef cattle at slaughter. Crossbred Aberdeen Angus (AA; n = 17) and Belgian Blue (BB; n = 16) steers born to Holstein-Friesian dams and sired by bulls with either high (H) or low (L) EPD(cwt) were used in the study. Thus, there were 4 genetic groups [i.e., BBH (n = 8), BBL (n = 8), AAH (n = 8), and AAL (n = 9)]. Blood samples were collected via jugular venipuncture at regular intervals for analysis of plasma concentrations of IGF-1 and insulin. Total RNA was isolated from LM collected at slaughter, and the mRNA expression of IGF-1, IGF-2, their receptors (IGF-1R; IGF-2R), 6 IGFBP, acid labile subunit (ALS), and GH receptor (GHR) was measured by real-time reverse-transcription quantitative PCR. There was no effect of either sire breed or EPD(cwt) on concentrations of circulating IGF or insulin (P > 0.05). Gene expression of IGF-1R and IGFBP3 was upregulated in AA (P < 0.001) compared with BB, whereas IGF-1 was upregulated in H compared with L animals (P < 0.01). Correlation analysis indicated moderate positive associations between gene expression of IGFBP3 and IGF-1 (r = 0.54; P < 0.001) and IGF-1R (r = 0.48; P < 0.01). In addition, correlation analysis revealed that mRNA expression of IGFBP3 was moderately negatively associated with LM area per kilogram of carcass weight (r = -0.40; P < 0.05). Greater gene expression of IGF-1 and reduced transcript abundance of IGFBP3 in muscle may have a role in increased muscle growth potential in steers during the finishing period. These data will contribute to a better understanding of the molecular control of muscle growth at a tissue level in cattle.
Collapse
Affiliation(s)
- S M Keady
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, Co. Meath, Ireland
| | | | | | | |
Collapse
|
25
|
Stevens-Lapsley JE, Ye F, Liu M, Borst SE, Conover C, Yarasheski KE, Walter GA, Sweeney HL, Vandenborne K. Impact of viral-mediated IGF-I gene transfer on skeletal muscle following cast immobilization. Am J Physiol Endocrinol Metab 2010; 299:E730-40. [PMID: 20739512 PMCID: PMC2980362 DOI: 10.1152/ajpendo.00230.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 08/17/2010] [Indexed: 11/22/2022]
Abstract
Insulin-like growth factor I (IGF-I) is a potent myogenic factor that plays a critical role in muscle regeneration and muscle hypertrophy. The purpose of this study was to evaluate the effect of IGF-I overexpression on the recovery of muscle size and function during reloading/reambulation after a period of cast immobilization in predominantly fast twitch muscles. In addition, we investigated concomitant molecular responses in IGF-I receptor and binding proteins (BPs). Recombinant adeno-associated virus vector for IGF-I (rAAV-IGF-IA) was injected into the anterior compartment of one of the hindlimbs of young (3 wk) C57BL6 female mice. At 20 wk of age, both hindlimbs were cast immobilized in a shortened position for 2 wk to unload the tibialis anterior (TA) and extensor longus digitorum (EDL) muscles. The TA and EDL muscles were removed bilaterally after 2 wk of cast immobilization and after 1 and 3 wk of free cage reambulation. Increases in IGF-I mRNA and protein levels with IGF-I overexpression were associated with significant increases in muscle wet weight, fiber size, and tetanic force, although overexpression did not protect against cast immobilization-induced muscle atrophy. After 1 wk of reambulation, evidence of enhanced muscle regeneration was noted in IGF-I-overexpressing muscles with an increased prevalence of central nuclei, embryonic myosin, and Pax7 positive fibers. We also observed larger relative gains in muscle size (wet weight and fiber area), but not force, during the 3-wk reambulation period in hindlimb muscles overexpressing IGF-I compared with contralateral control legs. Changes in IGFBP-5 mRNA expression during cast immobilization and reambulation paralleled those of IGF-I, whereas IGFBP-3 expression changed inversely to IGFBP-5.
Collapse
|
26
|
Velloso CP, Harridge SDR. Insulin-like growth factor-I E peptides: implications for aging skeletal muscle. Scand J Med Sci Sports 2010; 20:20-7. [PMID: 19883387 DOI: 10.1111/j.1600-0838.2009.00997.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In skeletal muscle there is good evidence to suggest that locally produced insulin-like growth factor-1 (IGF-I), rather than circulating IGF-I, is important in regard to muscle mass maintenance, repair and hypertrophy. This "mature" IGF-I comprises exons 3 and 4 of the IGF-I gene, but during processing the full length gene (which contains six exons) is subject to a process of alternative splicing. As a result smaller peptides (E peptides) are believed to be cleaved from the mature IGF-I peptide during processing of the prohormone and the likelihood is that they have different biological roles. In human skeletal muscle three transcripts encoding for these splice variants (IGF-IEa, IGF-IEb and IGF-IEc, also known as MGF) can be identified. When studied at the mRNA level these three transcripts are known to be upregulated in the muscles of elderly people following high resistance exercise, albeit with different time courses. However, compared with mature IGF-I relatively little is known about the mechanism of action of the different E peptides.
Collapse
Affiliation(s)
- Cristina P Velloso
- Division of Applied Biomedical Research, King's College London, London, UK
| | | |
Collapse
|
27
|
Samuel W, Kutty RK, Vijayasarathy C, Pascual I, Duncan T, Redmond TM. Decreased expression of insulin-like growth factor binding protein-5 during N-(4-hydroxyphenyl)retinamide-induced neuronal differentiation of ARPE-19 human retinal pigment epithelial cells: regulation by CCAAT/enhancer-binding protein. J Cell Physiol 2010; 224:827-36. [PMID: 20583135 PMCID: PMC2946194 DOI: 10.1002/jcp.22191] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Insulin-like growth factor (IGF)-binding protein-5 (IGFBP5), an important member of the IGF axis involved in regulating cell growth and differentiation, acts by modulating IGF signaling and also by IGF-independent mechanisms. We identified IGFBP5 by microarray analysis as a gene differentially regulated during N-(4-hydroxyphenyl)retinamide (4HPR)-induced neuronal differentiation of human retinal pigment epithelial (RPE) cells. IGFBP5 is expressed in human RPE cells, and its expression, mRNA as well as protein, is greatly decreased during the 4HPR-induced neuronal differentiation. Exogenous IGFBP5 does not block the neuronal differentiation indicating that IGFBP5 down-regulation may not be a prerequisite for the neuronal differentiation. IGFBP5 down-regulation, similar to neuronal differentiation, is mediated by the MAPK pathway since U0126, an inhibitor of MEK1/2, effectively blocked it. The overexpression of transcription factor CCAAT/enhancer binding protein-beta (C/EBPbeta) inhibited the 4HPR-induced down-regulation of IGFBP5 expression and the neuronal differentiation of RPE cells. Interestingly, the binding of C/EBPbeta to the IGFBP5 promoter was decreased by the 4HPR treatment as indicated by gel shift and chromatin immunoprecipitation analyses. Further, the deletion of C/EBP response element from IGFBP5 promoter markedly decreased the basal promoter activity and abolished its responsiveness to 4HPR treatment in reporter assays, suggesting that the expression of IGFBP5 is regulated by C/EBP. Thus, our results clearly demonstrate that the IGFBP5 expression is down-regulated during 4HPR-induced neuronal differentiation of human RPE cells through a MAPK signal transduction pathway involving C/EBPbeta.
Collapse
Affiliation(s)
- William Samuel
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-0706, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Henningsen J, Rigbolt KTG, Blagoev B, Pedersen BK, Kratchmarova I. Dynamics of the skeletal muscle secretome during myoblast differentiation. Mol Cell Proteomics 2010; 9:2482-96. [PMID: 20631206 DOI: 10.1074/mcp.m110.002113] [Citation(s) in RCA: 229] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During recent years, increased efforts have focused on elucidating the secretory function of skeletal muscle. Through secreted molecules, skeletal muscle affects local muscle biology in an auto/paracrine manner as well as having systemic effects on other tissues. Here we used a quantitative proteomics platform to investigate the factors secreted during the differentiation of murine C2C12 skeletal muscle cells. Using triple encoding stable isotope labeling by amino acids in cell culture, we compared the secretomes at three different time points of muscle differentiation and followed the dynamics of protein secretion. We identified and quantitatively analyzed 635 secreted proteins, including 35 growth factors, 40 cytokines, and 36 metallopeptidases. The extensive presence of these proteins that can act as potent signaling mediators to other cells and tissues strongly highlights the important role of the skeletal muscle as a prominent secretory organ. In addition to previously reported molecules, we identified many secreted proteins that have not previously been shown to be released from skeletal muscle cells nor shown to be differentially released during the process of myogenesis. We found 188 of these secreted proteins to be significantly regulated during the process of myogenesis. Comparative analyses of selected secreted proteins revealed little correlation between their mRNA and protein levels, indicating pronounced regulation by posttranscriptional mechanisms. Furthermore, analyses of the intracellular levels of members of the semaphorin family and their corresponding secretion dynamics demonstrated that the release of secreted proteins is tightly regulated by the secretory pathway, the stability of the protein, and/or the processing of secreted proteins. Finally, we provide 299 unique hydroxyproline sites mapping to 48 distinct secreted proteins and have discovered a novel hydroxyproline motif.
Collapse
Affiliation(s)
- Jeanette Henningsen
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, Denmark
| | | | | | | | | |
Collapse
|
29
|
Castigliego L, Armani A, Grifoni G, Rosati R, Mazzi M, Gianfaldoni D, Guidi A. Effects of growth hormone treatment on the expression of somatotropic axis genes in the skeletal muscle of lactating Holstein cows. Domest Anim Endocrinol 2010; 39:40-53. [PMID: 20399067 DOI: 10.1016/j.domaniend.2010.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 02/01/2010] [Accepted: 02/01/2010] [Indexed: 11/30/2022]
Abstract
This study focused on the expression of somatotropic axis genes in the skeletal muscle of dairy cattle. A slow-release recombinant bovine growth hormone (GH) (rbGH) formulation was administered to 5 cows, and saline solution (control) was administered to another 5 cows every 2 wk for a total of 10 wk, starting from the peak of lactation. Tissue and blood samples were collected on days 2 and 14 after each rbGH injection. As target genes insulin-like growth factor (IGF)-1, IGF-2, IGFBPs (1, 2, 3, 4, 5, 6), acute labile subunit (ALS), IGF-1 receptor (IGF-1R), GH receptor (GHR), and the known GHR 5'-UTR variants were selected as target genes, and their relative expression was measured using real-time polymerase chain reaction. In GH-treated cows, an increase in expression was observed for GHR 5'-UTR variant 1I on day 14 (P < 0.05), whereas a significant down-regulation of GHR (P < 0.05) was found after comparing values of treated cows between day 2 and day 14. However, only IGF binding proteins (BP)-5 was found to be appreciably up-regulated in GH-treated cows (P < 0.001), which may indicate the importance of this gene in the overall molecular response to GH administration. Our study indicated that GH treatment did not affect the expression of most somatotropic axis genes, despite the marked increase in GH and IGF-1 in blood (P < 0.001). Nor did it have a large impact on the proportion of GHR 5'-UTR variants in the skeletal muscle of lactating cows. Finally, although we observed a significant variation in the expression of some genes, it would appear that the differences between GH-treated cows and controls were not great enough to be considered as reliable indirect indicators of GH treatment in dairy cattle.
Collapse
Affiliation(s)
- L Castigliego
- Department of Animal Pathology, Prophylaxis and Food Hygiene, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
Duan C, Ren H, Gao S. Insulin-like growth factors (IGFs), IGF receptors, and IGF-binding proteins: roles in skeletal muscle growth and differentiation. Gen Comp Endocrinol 2010; 167:344-51. [PMID: 20403355 DOI: 10.1016/j.ygcen.2010.04.009] [Citation(s) in RCA: 326] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 04/07/2010] [Accepted: 04/14/2010] [Indexed: 12/22/2022]
Abstract
The insulin-like growth factor (IGF) signaling pathway consists of multiple IGF ligands, IGF receptors, and IGF-binding proteins (IGFBPs). Studies in a variety of animal and cellular systems suggest that the IGF signaling pathway plays a key role in regulating skeletal muscle growth, differentiation, and in maintaining homeostasis of the adult muscle tissues. Intriguingly, IGFs stimulate both myoblast proliferation and differentiation, which are two mutually exclusive biological events during myogenesis. Both of these actions are mediated through the same IGF-1 receptor. Recent studies have shed new insights into the molecular mechanisms underlying these paradoxical actions of IGFs in muscle cells. In this article, we provide a brief review of our current understanding of the IGF signaling system and discuss recent findings on how local oxygen availability and IGFBPs act to specify IGF actions in muscle cells.
Collapse
Affiliation(s)
- Cunming Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
31
|
Baertling F, Kokozidou M, Pufe T, Clarner T, Windoffer R, Wruck CJ, Brandenburg LO, Beyer C, Kipp M. ADAM12 is expressed by astrocytes during experimental demyelination. Brain Res 2010; 1326:1-14. [PMID: 20176000 DOI: 10.1016/j.brainres.2010.02.049] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 02/07/2010] [Accepted: 02/14/2010] [Indexed: 12/18/2022]
Abstract
A disintegrin and metalloproteinase (ADAM) 12 represents a member of a large family of similarly structured multi-domain proteins. In the central nervous system (CNS), ADAM12 has been suggested to play a role in brain development, glioblastoma cell proliferation, and in experimental autoimmune encephalomyelitis. Furthermore, ADAM12 was reported to be almost exclusively expressed by oligodendrocytes and could, therefore, be considered as suitable marker for this cell type. In the present study, we investigated ADAM12 expression in the healthy and pathologically altered murine CNS. As pathological paradigm, we used the cuprizone demyelination model in which myelin loss during multiple sclerosis is imitated. Besides APC(+) oligodendrocytes, SMI311(+) neurons and GFAP(+) astrocytes express ADAM12 in the adult mouse brain. ADAM12 expression was further analyzed in vitro. After the induction of demyelination, we observed that activated astrocytes are the main source of ADAM12 in brain regions affected by oligodendrocyte loss. Exposure of astrocytes in vitro to either lipopolysaccharides (LPS), tumor necrosis factor alpha (TNFalpha), glutamate, or hydrogen peroxide revealed a highly stimulus-specific regulation of ADAM12 expression which was not seen in microglial BV2 cells. It appears that LPS- and TNFalpha-induced ADAM12 expression is mediated via the classic NFkappaB pathway. In summary, we demonstrated that ADAM12 is not a suitable marker for oligodendrocytes. Our results further suggest that ADAM12 might be implicated in the course of distinct CNS diseases such as demyelinating disorders.
Collapse
Affiliation(s)
- Fabian Baertling
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abrass CK, Hansen KM. Insulin-like growth factor-binding protein-5-induced laminin gamma1 transcription requires filamin A. J Biol Chem 2010; 285:12925-34. [PMID: 20167606 DOI: 10.1074/jbc.m109.061754] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin-like growth factor-binding protein-5 (IGFBP-5) has IGF-1-independent intranuclear effects that are poorly defined. Treatment of cells with IGFBP-5 induces migration, prevents apoptosis, and leads to increased laminin subunit transcription. Similarly, filamin A (FLNa), an actin-binding protein that participates in cell attachment, plays important additional roles in signal transduction and modulation of transcriptional responses. In this report, we show that IGFBP-5 leads to dephosphorylation of FLNa with subsequent FLNa cleavage. Following cleavage, there is enhanced recruitment of Smad3/4 to a C-terminal FLNa fragment with nuclear translocation and subsequent binding to the promoter region of the laminin gamma1 (lamc1) gene. FLNa knockdown prevents IGFBP-5-mediated increases in lamc1 transcription. These data indicate that IGFBP-5 induces formation of a FLNa-based nuclear shuttle that recruits transcription factors and regulates transcription of IGFBP-5 target genes. These studies provide new insights into the mechanisms whereby IGFBP-5 and FLNa exert intranuclear effects.
Collapse
Affiliation(s)
- Christine K Abrass
- Department of Medicine, Allergy & Inflammation Program, University of Washington School of Medicine, Seattle, Washington 98109, USA.
| | | |
Collapse
|
33
|
Selective signaling by Akt2 promotes bone morphogenetic protein 2-mediated osteoblast differentiation. Mol Cell Biol 2009; 30:1018-27. [PMID: 19995912 DOI: 10.1128/mcb.01401-09] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal stem cells are essential for repair of bone and other supporting tissues. Bone morphogenetic proteins (BMPs) promote commitment of these progenitors toward an osteoblast fate via functional interactions with osteogenic transcription factors, including Dlx3, Dlx5, and Runx2, and also can direct their differentiation into bone-forming cells. BMP-2-stimulated osteoblast differentiation additionally requires continual signaling from insulin-like growth factor (IGF)-activated pathways. Here we identify Akt2 as a critical mediator of IGF-regulated osteogenesis. Targeted knockdown of Akt2 in mouse primary bone marrow stromal cells or in a mesenchymal stem cell line, or genetic knockout of Akt2, did not interfere with BMP-2-mediated signaling but resulted in inhibition of osteoblast differentiation at an early step that preceded production of Runx2. In contrast, Akt1-deficient cells differentiated normally. Complete biochemical and morphological osteoblast differentiation was restored in cells lacking Akt2 by adenoviral delivery of Runx2 or by a recombinant lentivirus encoding wild-type Akt2. In contrast, lentiviral Akt1 was ineffective. Taken together, these observations define a specific role for Akt2 as a gatekeeper of osteogenic differentiation through regulation of Runx2 gene expression and indicate that the closely related Akt1 and Akt2 exert distinct effects on the differentiation of mesenchymal precursors.
Collapse
|
34
|
Developmental expression changes of insulin-like growth factors (IGFs) system genes in longissimus dorsi muscle of two pig breeds. YI CHUAN = HEREDITAS 2009; 31:837-43. [DOI: 10.3724/sp.j.1005.2009.00837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Clemmons DR. Role of IGF-I in skeletal muscle mass maintenance. Trends Endocrinol Metab 2009; 20:349-56. [PMID: 19729319 DOI: 10.1016/j.tem.2009.04.002] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 04/02/2009] [Accepted: 04/02/2009] [Indexed: 01/24/2023]
Abstract
The recent identification of signaling elements that regulate skeletal muscle protein balance has provided the opportunity to determine how IGF-I alters these processes. Animal studies have revealed the important role of IGF-I in preventing muscle atrophy and enabled investigators to determine the hierarchy of signaling pathways and events within each pathway that are modulated by IGF-I. These discoveries provide opportunity for future studies to target these important signaling events and develop strategies to reverse loss of muscle mass that accompanies these catabolic states. Because there are no approved medical therapies that will reverse catabolism at present, this represents an opportunity to fulfill a major unmet medical need.
Collapse
Affiliation(s)
- David R Clemmons
- Division of Endocrinology, University of North Carolina School of Medicine, hapel Hill, NC 27599-7170, USA.
| |
Collapse
|
36
|
Mukherjee A, Rotwein P. Akt promotes BMP2-mediated osteoblast differentiation and bone development. J Cell Sci 2009; 122:716-26. [PMID: 19208758 DOI: 10.1242/jcs.042770] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Signaling through the IGF-I receptor by locally synthesized IGF-I or IGF-II is crucial for normal skeletal development and for bone remodeling. Osteogenesis is primarily regulated by bone morphogenetic proteins (BMPs), which activate gene expression programs driven by bone-specific transcription factors. In a mesenchymal stem cell model of osteoblast commitment and differentiation controlled by BMP2, we show that an inhibitor of PI3-kinase or a dominant-negative Akt were as potent in preventing osteoblast differentiation as the IGF binding protein IGFBP5, whereas a Mek inhibitor was ineffective. Conversely, an adenovirus encoding an inducible-active Akt was able to overcome the blockade of differentiation caused by IGFBP5 or the PI3-kinase inhibitor, and could restore normal osteogenesis. Inhibition of PI3-kinase or Akt did not block BMP2-mediated signaling, because the Smad-responsive genes Sox9 and JunB were induced normally under all experimental conditions. When activated during different stages of osteoblast maturation, dominant-negative Akt prevented accumulation of bone-specific alkaline phosphatase and reduced mineralization, and more significantly inhibited the longitudinal growth of metatarsal bones in primary culture by interfering with both chondrocyte and osteoblast development and function. We conclude that an intact IGF-induced PI3-kinase-Akt signaling cascade is essential for BMP2-activated osteoblast differentiation and maturation, bone development and growth, and suggest that manipulation of this pathway could facilitate bone remodeling and fracture repair.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | | |
Collapse
|
37
|
Gardan D, Mourot J, Louveau I. Decreased expression of the IGF-II gene during porcine adipose cell differentiation. Mol Cell Endocrinol 2008; 292:63-8. [PMID: 18640238 DOI: 10.1016/j.mce.2008.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Revised: 04/24/2008] [Accepted: 06/23/2008] [Indexed: 10/21/2022]
Abstract
IGF-I and IGF-II are known to regulate cell development and recent data suggest a possible role of IGF-II on adipose tissue development. This study was undertaken to examine the IGF system gene expression in porcine differentiating adipocytes. Both adipocytes and stromal-vascular (s/v) cells were isolated from subcutaneous adipose tissue collected from 7-day-old piglets. s/v cells were cultured in chemically defined medium. Compared with isolated adipocytes, IGF-II and IGFBP-5 mRNA levels were very high in freshly isolated s/v cells, whereas IGF-I mRNA levels were lower in s/v cells than in adipocytes. Between day 0 and day 6 of culture, IGF-II and IGFBP-5 gene expression decreased whereas expression levels of late markers of adipocyte differentiation were up-regulated. Cell differentiation was also associated with an increase in the expression of IGF-I, insulin and IGF receptor genes. The current findings suggest that IGF-I and IGF-II have different effects on porcine adipose cell development.
Collapse
|
38
|
Ren H, Yin P, Duan C. IGFBP-5 regulates muscle cell differentiation by binding to IGF-II and switching on the IGF-II auto-regulation loop. ACTA ACUST UNITED AC 2008; 182:979-91. [PMID: 18762576 PMCID: PMC2528583 DOI: 10.1083/jcb.200712110] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
IGF-II stimulates both mitogenesis and myogenesis through its binding and activation of the IGF-I receptor (IGF-IR). How this growth factor pathway promotes these two opposite cellular responses is not well understood. We investigate whether local IGF binding protein-5 (IGFBP-5) promotes the myogenic action of IGF-II. IGFBP-5 is induced before the elevation of IGF-II expression during myogenesis. Knockdown of IGFBP-5 impairs myogenesis and suppresses IGF-II gene expression. IGF-II up-regulates its own gene expression via the PI3K-Akt signaling pathway. Adding IGF-II or constitutively activating Akt rescues the IGFBP-5 knockdown-caused defects. However, an IGF analogue that binds to the IGF-IR but not IGFBP has only a limited effect. When added with low concentrations of IGF-II, IGFBP-5 restores IGF-II expression and myogenic differentiation, whereas an IGF binding–deficient IGFBP-5 mutant has no effect. These findings suggest that IGFBP-5 promotes muscle cell differentiation by binding to and switching on the IGF-II auto-regulation loop.
Collapse
Affiliation(s)
- Hongxia Ren
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
39
|
Mukherjee A, Rotwein P. Insulin-like growth factor-binding protein-5 inhibits osteoblast differentiation and skeletal growth by blocking insulin-like growth factor actions. Mol Endocrinol 2008; 22:1238-50. [PMID: 18292241 DOI: 10.1210/me.2008-0001] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Signaling through the IGF-I receptor by locally synthesized IGF-I or IGF-II is critical for normal skeletal development and for bone remodeling and repair throughout the lifespan. In most tissues, IGF actions are modulated by IGF-binding proteins (IGFBPs). IGFBP-5 is the most abundant IGFBP in bone, and previous studies have suggested that it may either enhance or inhibit osteoblast differentiation in culture and may facilitate or block bone growth in vivo. To resolve these contradictory observations and discern the mechanisms of action of IGFBP-5 in bone, we studied its effects in differentiating osteoblasts and in primary bone cultures. Purified wild-type (WT) mouse IGFBP-5 or a recombinant adenovirus expressing IGFBP-5WT each prevented osteogenic differentiation induced by the cytokine bone morphogenetic protein (BMP)-2 at its earliest stages without interfering with BMP-mediated signaling, whereas an analog with reduced IGF binding (N domain mutant) was ineffective. When added at later phases of bone cell maturation, IGFBP-5WT but not IGFBP-5N blocked mineralization, prevented longitudinal growth of mouse metatarsal bones in short-term primary culture, and inhibited their endochondral ossification. Because an IGF-I variant (R3IGF-I) with diminished affinity for IGFBPs promoted full osteogenic differentiation in the presence of IGFBP-5WT, our results show that IGFBP-5 interferes with IGF action in osteoblasts and provides a framework for discerning mechanisms of collaboration between signal transduction pathways activated by BMPs and IGFs in bone.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | |
Collapse
|
40
|
Kuns-Hashimoto R, Kuninger D, Nili M, Rotwein P. Selective binding of RGMc/hemojuvelin, a key protein in systemic iron metabolism, to BMP-2 and neogenin. Am J Physiol Cell Physiol 2008; 294:C994-C1003. [PMID: 18287331 DOI: 10.1152/ajpcell.00563.2007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Juvenile hemochromatosis is a severe and rapidly progressing hereditary disorder of iron overload, and it is caused primarily by defects in the gene encoding repulsive guidance molecule c/hemojuvelin (RGMc/HJV), a recently identified protein that undergoes a complicated biosynthetic pathway in muscle and liver, leading to cell membrane-linked single-chain and heterodimeric species, and two secreted single-chain isoforms. RGMc modulates expression of the hepatic iron regulatory factor, hepcidin, potentially through effects on signaling by the bone morphogenetic protein (BMP) family of soluble growth factors. To date, little is known about specific pathogenic defects in disease-causing RGMc/HJV proteins. Here we identify functional abnormalities in three juvenile hemochromatosis-linked mutants. Using a combination of approaches, we first show that BMP-2 could interact in biochemical assays with single-chain RGMc species, and also could bind to cell-associated RGMc. Two mouse RGMc amino acid substitution mutants, D165E and G313V (corresponding to human D172E and G320V), also could bind BMP-2, but less effectively than wild-type RGMc, while G92V (human G99V) could not. In contrast, the membrane-spanning protein, neogenin, a receptor for the related molecule, RGMa, preferentially bound membrane-associated heterodimeric RGMc and was able to interact on cells only with wild-type RGMc and G92V. Our results show that different isoforms of RGMc/HJV may play unique physiological roles through defined interactions with distinct signaling proteins and demonstrate that, in some disease-linked RGMc mutants, these interactions are defective.
Collapse
Affiliation(s)
- Robin Kuns-Hashimoto
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | | | | | | |
Collapse
|