1
|
Farr JN, Saul D, Doolittle ML, Kaur J, Rowsey JL, Vos SJ, Froemming MN, Lagnado AB, Zhu Y, Weivoda M, Ikeno Y, Pignolo RJ, Niedernhofer LJ, Robbins PD, Jurk D, Passos JF, LeBrasseur NK, Tchkonia T, Kirkland JL, Monroe DG, Khosla S. Local senolysis in aged mice only partially replicates the benefits of systemic senolysis. J Clin Invest 2023; 133:e162519. [PMID: 36809340 PMCID: PMC10104901 DOI: 10.1172/jci162519] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Clearance of senescent cells (SnCs) can prevent several age-related pathologies, including bone loss. However, the local versus systemic roles of SnCs in mediating tissue dysfunction remain unclear. Thus, we developed a mouse model (p16-LOX-ATTAC) that allowed for inducible SnC elimination (senolysis) in a cell-specific manner and compared the effects of local versus systemic senolysis during aging using bone as a prototype tissue. Specific removal of Sn osteocytes prevented age-related bone loss at the spine, but not the femur, by improving bone formation without affecting osteoclasts or marrow adipocytes. By contrast, systemic senolysis prevented bone loss at the spine and femur and not only improved bone formation, but also reduced osteoclast and marrow adipocyte numbers. Transplantation of SnCs into the peritoneal cavity of young mice caused bone loss and also induced senescence in distant host osteocytes. Collectively, our findings provide proof-of-concept evidence that local senolysis has health benefits in the context of aging, but, importantly, that local senolysis only partially replicates the benefits of systemic senolysis. Furthermore, we establish that SnCs, through their senescence-associated secretory phenotype (SASP), lead to senescence in distant cells. Therefore, our study indicates that optimizing senolytic drugs may require systemic instead of local SnC targeting to extend healthy aging.
Collapse
Affiliation(s)
- Joshua N. Farr
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
- Department of Physiology and Biomedical Engineering, and
| | - Dominik Saul
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | | | - Japneet Kaur
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | | | - Stephanie J. Vos
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | | | - Anthony B. Lagnado
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - Megan Weivoda
- Department of Hematology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Yuji Ikeno
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Robert J. Pignolo
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - João F. Passos
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | | - David G. Monroe
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
- Department of Physiology and Biomedical Engineering, and
| |
Collapse
|
2
|
Yao Z, Lin M, Lin T, Gong X, Qin P, Li H, Kang T, Ye J, Zhu Y, Hong Q, Liu Y, Li Y, Wang J, Fang F. The expression of IGFBP-5 in the reproductive axis and effect on the onset of puberty in female rats. Reprod Biol Endocrinol 2022; 20:100. [PMID: 35821045 PMCID: PMC9277959 DOI: 10.1186/s12958-022-00966-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/18/2022] [Indexed: 11/21/2022] Open
Abstract
Insulin-like growth factor-binding protein-5 (IGFBP-5) has recently been shown to alter the reproductive capacity by regulating insulin-like growth factor (IGF) bioavailability or IGF-independent effects. The present study aimed to investigate the effect and mechanism of IGFBP-5 on the onset of puberty in female rats. Immunofluorescence and real-time quantitative PCR were used to determine the expression and location of IGFBP-5 mRNA and protein distribution in the infant's hypothalamus-pituitary-ovary (HPO) axis prepuberty, peripuberty, puberty and adult female rats. Prepubertal rats with IGFBP-5 intracerebroventricular (ICV) were injected to determine the puberty-related genes expression and the concentrations of reproductive hormones. Primary hypothalamic cells were treated with IGFBP-5 to determine the expression of puberty-related genes and the Akt and mTOR proteins. Results showed that Igfbp-5 mRNA and protein were present on the HPO axis. The addition of IGFBP-5 to primary hypothalamic cells inhibited the expression of Gnrh and Igf-1 mRNAs (P < 0.05) and increased the expression of AKT and mTOR protein (P < 0.01). IGFBP-5 ICV-injection delayed the onset of puberty, reduced Gnrh, Igf-1, and Fshβ mRNAs, and decreased the concentrations of E2, P4, FSH,serum LH levels and the ovaries weight (P < 0.05). More corpus luteum and fewer primary follicles were found after IGFBP-5 injection (P < 0.05).
Collapse
Affiliation(s)
- Zhiqiu Yao
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Maosen Lin
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Tao Lin
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Xinbao Gong
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Pin Qin
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Hailing Li
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Tiezhu Kang
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Jing Ye
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Yanyun Zhu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Qiwen Hong
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Ya Liu
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Yunsheng Li
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Juhua Wang
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China
| | - Fugui Fang
- Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, 230036, Anhui, China.
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.
| |
Collapse
|
3
|
Identification of Impacted Pathways and Transcriptomic Markers as Potential Mediators of Pulmonary Fibrosis in Transgenic Mice Expressing Human IGFBP5. Int J Mol Sci 2021; 22:ijms222212609. [PMID: 34830489 PMCID: PMC8619832 DOI: 10.3390/ijms222212609] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022] Open
Abstract
Pulmonary fibrosis is a serious disease characterized by extracellular matrix (ECM) component overproduction and remodeling. Insulin-like growth factor-binding protein 5 (IGFBP5) is a conserved member of the IGFBP family of proteins that is overexpressed in fibrotic tissues and promotes fibrosis. We used RNA sequencing (RNAseq) to identify differentially expressed genes (DEGs) between primary lung fibroblasts (pFBs) of homozygous (HOMO) transgenic mice expressing human IGFBP5 (hIGFBP5) and wild type mice (WT). The results of the differential expression analysis showed 2819 DEGs in hIGFBP5 pFBs. Functional enrichment analysis confirmed the pro-fibrotic character of IGFBP5 and revealed its impact on fundamental signaling pathways, including cytokine–cytokine receptor interaction, focal adhesion, AGE-RAGE signaling, calcium signaling, and neuroactive ligand-receptor interactions, to name a few. Noticeably, 7% of the DEGs in hIGFBP5-expressing pFBs are receptors and integrins. Furthermore, hub gene analysis revealed 12 hub genes including Fpr1, Bdkrb2, Mchr1, Nmur1, Cnr2, P2ry14, and Ptger3. Validation assays were performed to complement the RNAseq data. They confirmed significant differences in the levels of the corresponding proteins in cultured pFBs. Our study provides new insights into the molecular mechanism(s) of IGFBP5-associated pulmonary fibrosis through possible receptor interactions that drive fibrosis and tissue remodeling.
Collapse
|
4
|
Rogowska MD, Pena UNV, Binning N, Christians JK. Recovery of the maternal skeleton after lactation is impaired by advanced maternal age but not by reduced IGF availability in the mouse. PLoS One 2021; 16:e0256906. [PMID: 34469481 PMCID: PMC8409645 DOI: 10.1371/journal.pone.0256906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/17/2021] [Indexed: 01/23/2023] Open
Abstract
Background Lactation results in substantial maternal bone loss that is recovered following weaning. However, the mechanisms underlying this recovery, and in particular the role of insulin-like growth factor 1 (IGF-I), is not clear. Furthermore, there is little data regarding whether recovery is affected by advanced maternal age. Methods Using micro-computed tomography, we studied bone recovery following lactation in mice at 2, 5 and 7 months of age. We also investigated the effects of reduced IGF-I availability using mice lacking PAPP-A2, a protease of insulin-like growth factor binding protein 5 (IGFBP-5). Results In 2 month old mice, lactation affected femoral trabecular and cortical bone, but only cortical bone showed recovery 3 weeks after weaning. This recovery was not affected by deletion of the Pappa2 gene. The amount of trabecular bone was reduced in 5 and 7 month old mice, and was not further reduced by lactation. However, the recovery of cortical bone was impaired at 5 and 7 months compared with at 2 months. Conclusions Recovery of the maternal skeleton after lactation is impaired in moderately-aged mice compared with younger mice. Our results may be relevant to the long-term effects of breastfeeding on the maternal skeleton in humans, particularly given the increasing median maternal age at childbearing.
Collapse
Affiliation(s)
- Monika D Rogowska
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Uriel N V Pena
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Nimrat Binning
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian K Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada.,Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, Canada.,British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,BC Women's Hospital and Health Centre, Women's Health Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
5
|
Feng J, Meng Z. Insulin growth factor-1 promotes the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells through the Wnt/β-catenin pathway. Exp Ther Med 2021; 22:891. [PMID: 34194569 PMCID: PMC8237273 DOI: 10.3892/etm.2021.10323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are stem cells that exist in bone marrow tissue and have osteogenic differentiation potential. Insulin growth factor-1 (IGF-1) plays a key role in the proliferation and osteogenic differentiation of BMSCs. However, the specific mechanism of IGF-1 in cell proliferation and osteogenic differentiation remains unclear. In the present study, BMSCs were transfected with lentivirus carrying the siRNA-Wnt3a gene, and the Wnt3a level in BMSCs was revealed to be reduced by western blotting, real-time quantitative polymerase chain reaction and immunofluorescence detection. Then, BMSCs were treated with 80 ng/ml IGF-1 in complete medium for 5 days. CCK-8 and cell cycle assays revealed that cell proliferation was significantly decreased in the siRNA-Wnt3a group than in the control group. The protein and mRNA levels of β-catenin and cyclin D1 were significantly downregulated in the siRNA-Wnt3a group compared with the control group. In addition, BMSCs were treated with IGF-1 in osteogenic differentiation medium for 7 and 21 days, and alkaline phosphatase staining and Alizarin Red staining demonstrated significantly reduced osteogenic differentiation ability in the siRNA-Wnt3a group compared with the control group. Furthermore, the protein and mRNA levels of β-catenin, RUNX2, and OPN were downregulated compared with the control group. Our findings revealed that IGF-1 promoted the proliferation and differentiation of BMSCs at least partially through the Wnt/β-catenin pathway. These findings provided new insight into the clinical treatment of bone disease.
Collapse
Affiliation(s)
- Jing Feng
- Jiaozuo Coal Industry (Group) Co. Ltd. Central Hospital, Jiaozuo, Henan 454000, P.R. China
| | - Zhiqiang Meng
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
6
|
Recombinant IGF-1 Induces Sex-Specific Changes in Bone Composition and Remodeling in Adult Mice with Pappa2 Deficiency. Int J Mol Sci 2021; 22:ijms22084048. [PMID: 33919940 PMCID: PMC8070906 DOI: 10.3390/ijms22084048] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/04/2021] [Accepted: 04/11/2021] [Indexed: 12/11/2022] Open
Abstract
Deficiency of pregnancy-associated plasma protein-A2 (PAPP-A2), an IGF-1 availability regulator, causes postnatal growth failure and dysregulation of bone size and density. The present study aimed to determine the effects of recombinant murine IGF-1 (rmIGF-1) on bone composition and remodeling in constitutive Pappa2 knock-out (ko/ko) mice. To address this challenge, X-ray diffraction (XRD), attenuated total reflection-fourier transform infra-red (ATR-FTIR) spectroscopy and gene expression analysis of members of the IGF-1 system and bone resorption/formation were performed. Pappa2ko/ko mice (both sexes) had reduced body and bone length. Male Pappa2ko/ko mice had specific alterations in bone composition (mineral-to-matrix ratio, carbonate substitution and mineral crystallinity), but not in bone remodeling. In contrast, decreases in collagen maturity and increases in Igfbp3, osteopontin (resorption) and osteocalcin (formation) characterized the bone of Pappa2ko/ko females. A single rmIGF-1 administration (0.3 mg/kg) induced short-term changes in bone composition in Pappa2ko/ko mice (both sexes). rmIGF-1 treatment in Pappa2ko/ko females also increased collagen maturity, and Igfbp3, Igfbp5, Col1a1 and osteopontin expression. In summary, acute IGF-1 treatment modifies bone composition and local IGF-1 response to bone remodeling in mice with Pappa2 deficiency. These effects depend on sex and provide important insights into potential IGF-1 therapy for growth failure and bone loss and repair.
Collapse
|
7
|
Bahamonde M, Misra M. Potential applications for rhIGF-I: Bone disease and IGFI. Growth Horm IGF Res 2020; 52:101317. [PMID: 32252004 PMCID: PMC7231643 DOI: 10.1016/j.ghir.2020.101317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/09/2020] [Accepted: 03/21/2020] [Indexed: 12/18/2022]
Abstract
Growth hormone (GH) and insulin like growth factor-I (IGFI) are key bone trophic hormones, whose rising levels during puberty are critical for pubertal bone accrual. Conditions of GH deficiency and genetic resistance impact cortical and trabecular bone deleteriously with reduced estimates of bone strength. In humans, conditions of undernutrition (as in anorexia nervosa (AN), or subsequent to chronic illnesses) are associated with low IGF-I levels, which correlate with disease severity, and also with lower bone mineral density (BMD), impaired bone structure and lower strength estimates. In adolescents and adults with AN, studies have demonstrated a nutritionally acquired GH resistance with low IGF-I levels despite high concentrations of GH. IGF-I levels go up with increasing body weight, and are associated with rising levels of bone turnover markers. In short-term studies lasting 6-10 days, recombinant human IGF-I (rhIGF-I) administration in physiologic replacement doses normalized IGF-I levels and increased levels of bone formation markers in both adults and adolescents with AN. In a randomized controlled trial in adults with AN in which participants were randomized to one of four arms: (i) rhIGF-I with oral estrogen-progesterone (EP), (ii) rhIGF-I alone, (iii) EP alone, or (iv) neither for 9 months, a significant increase in bone formation markers was noted in the groups that received rhIGF-I, and a significant decrease in bone resorption markers in the groups that received EP. The group that received both rhIGF-I and EP had a significant increase in bone density at the spine and hip compared to the group that received neither. Side effects were minimal, with no documented fingerstick glucose of <50 mg/dl. These data thus suggest a potential role for rhIGF-I administration in optimizing bone accrual in states of undernutrition associated with low IGF-I.
Collapse
Affiliation(s)
- Marisol Bahamonde
- Department of Pediatrics, Universidad San Francisco de Quito (USFQ), Cumbayá, Ecuador
| | - Madhusmita Misra
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
8
|
Duan C, Allard JB. Insulin-Like Growth Factor Binding Protein-5 in Physiology and Disease. Front Endocrinol (Lausanne) 2020; 11:100. [PMID: 32194505 PMCID: PMC7063065 DOI: 10.3389/fendo.2020.00100] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/17/2020] [Indexed: 12/25/2022] Open
Abstract
Insulin-like growth factor (IGF) signaling is regulated by a conserved family of IGF binding proteins (IGFBPs) in vertebrates. Among the six distinct types of IGFBPs, IGFBP-5 is the most highly conserved across species and has the broadest range of biological activities. IGFBP-5 is expressed in diverse cell types, and its expression level is regulated by a variety of signaling pathways in different contexts. IGFBP-5 can exert a range of biological actions including prolonging the half-life of IGFs in the circulation, inhibition of IGF signaling by competing with the IGF-1 receptor for ligand binding, concentrating IGFs in certain cells and tissues, and potentiation of IGF signaling by delivery of IGFs to the IGF-1 receptor. IGFBP-5 also has IGF-independent activities and is even detected in the nucleus. Its broad biological activities make IGFBP-5 an excellent representative for understanding IGFBP functions. Despite its evolutionary conservation and numerous biological activities, knockout of IGFBP-5 in mice produced only a negligible phenotype. Recent research has begun to explain this paradox by demonstrating cell type-specific and physiological/pathological context-dependent roles for IGFBP-5. In this review, we survey and discuss what is currently known about IGFBP-5 in normal physiology and human disease. Based on recent in vivo genetic evidence, we suggest that IGFBP-5 is a multifunctional protein with the ability to act as a molecular switch to conditionally regulate IGF signaling.
Collapse
|
9
|
McCallum-Loudeac J, Anderson G, Wilson MJ. Age and Sex-Related Changes to Gene Expression in the Mouse Spinal Cord. J Mol Neurosci 2019; 69:419-432. [PMID: 31267314 DOI: 10.1007/s12031-019-01371-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
The spinal cord is essential for neuronal communication between the brain and rest of the body. To gain further insight into the molecular changes underpinning maturation of the mouse spinal cord, we analysed gene expression differences between 4 weeks of age (prior to puberty onset) and adulthood (8 weeks). We found 800 genes were significantly differentially expressed between juvenile and adult spinal cords. Gene ontology analysis revealed an overrepresentation of genes with roles in myelination and signal transduction among others. The expression of a further 19 genes was sexually dimorphic; these included both autosomal and sex-linked genes. Given the presence of steroid hormone receptors in the spinal cord, we also looked at the impact of two major steroid hormones, oestradiol and dihydrotestosterone (DHT) on spinal cord gene expression for selected genes. In gonadectomised male animals, implants with oestradiol and DHT produced significant changes to spinal cord gene expression. This study provides an overview of the global gene expression changes that occur as the spinal cord matures, over a key period of maturation. This confirms that both age and sex are important considerations in studies involving the spinal cord.
Collapse
Affiliation(s)
- Jeremy McCallum-Loudeac
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Greg Anderson
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Megan J Wilson
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
10
|
Fujimoto M, Andrew M, Liao L, Zhang D, Yildirim G, Sluss P, Kalra B, Kumar A, Yakar S, Hwa V, Dauber A. Low IGF-I Bioavailability Impairs Growth and Glucose Metabolism in a Mouse Model of Human PAPPA2 p.Ala1033Val Mutation. Endocrinology 2019; 160:1363-1376. [PMID: 30977789 PMCID: PMC6507901 DOI: 10.1210/en.2018-00755] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/05/2019] [Indexed: 02/03/2023]
Abstract
Bioactive free IGF-I is critically important for growth. The bioavailability of IGF-I is modulated by the IGF-binding proteins (IGFBPs) and their proteases, such as pregnancy-associated plasma protein-A2 (PAPP-A2). We have created a mouse model with a specific mutation in PAPPA2 identified in a human with PAPP-A2 deficiency. The human mutation was introduced to the mouse genome via a knock-in strategy, creating knock-in mice with detectable protein levels of Papp-a2 but without protease activities. We found that the Pappa2 mutation led to significant reductions in body length (10%), body weight (10% and 20% in males and females, respectively), and relative lean mass in mice. Micro-CT analyses of Pappa2 knock-in femurs from adult mice showed inhibited periosteal bone expansion leading to more slender bones in both male and female mice. Furthermore, in the Pappa2 knock-in mice, insulin resistance correlated with decreased serum free IGF-I and increased intact IGFBP-3 concentrations. Interestingly, mice heterozygous for the knock-in mutation demonstrated a growth rate for body weight and length as well as a biochemical phenotype that was intermediate between wild-type and homozygous mice. This study models a human PAPPA2 mutation in mice. The mouse phenotype closely resembles that of the human patients, and it provides further evidence that the regulation of IGF-I bioavailability by PAPP-A2 is critical for human growth and for glucose and bone metabolism.
Collapse
Affiliation(s)
- Masanobu Fujimoto
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Melissa Andrew
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Endocrinology, Children’s National Medical Center, Washington, DC
| | - Lihong Liao
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Dongsheng Zhang
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Gozde Yildirim
- Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | | | | | | | - Shoshana Yakar
- Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Vivian Hwa
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Correspondence: Andrew Dauber, MD, Children’s National Medical Center, 111 Michigan Avenue NW, WW3.5, Suite 200, Room 1215, Washington, DC 20010. E-mail: ; or Vivian Hwa, PhD, Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, 240 Albert Sabin Way, T5.605, Cincinnati, Ohio 45229. E-mail:
| | - Andrew Dauber
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Endocrinology, Children’s National Medical Center, Washington, DC
- Correspondence: Andrew Dauber, MD, Children’s National Medical Center, 111 Michigan Avenue NW, WW3.5, Suite 200, Room 1215, Washington, DC 20010. E-mail: ; or Vivian Hwa, PhD, Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, 240 Albert Sabin Way, T5.605, Cincinnati, Ohio 45229. E-mail:
| |
Collapse
|
11
|
Christians JK, Amiri N, Schipilow JD, Zhang SW, May-Rashke KI. Pappa2 deletion has sex- and age-specific effects on bone in mice. Growth Horm IGF Res 2019; 44:6-10. [PMID: 30445407 DOI: 10.1016/j.ghir.2018.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE In humans, loss-of-function mutations in the gene encoding pregnancy-associated pregnancy protein-A2 cause short stature and slightly reduced bone density. The goal of this study was to determine the effects of Pappa2 deletion on bone in mice. DESIGN Pappa2 deletion mice and littermate controls were culled at 10, 19 or 30 weeks of age and femurs were analysed by micro-computed tomography. Serum markers of bone turnover and insulin-like growth factor binding protein 5 (IGFBP-5), a proteolytic target of PAPP-A2, were measured by ELISA. RESULTS At 10 and 19 weeks of age, Pappa2 deletion mice had slightly reduced trabecular parameters, but by 19 weeks of age, female deletion mice had increased cortical tissue mineral density, and this trait was increased by a small amount in deletion mice of both sexes at 30 weeks. Cortical area fraction was increased in Pappa2 deletion mice at all ages. Deletion of Pappa2 increased circulating IGFBP-5 levels and reduced markers of bone turnover (PINP and TRACP 5b). CONCLUSIONS PAPP-A2 contributes to the regulation of bone structure and mass in mice, likely through control of IGFBP-5 levels. The net effect of changes in bone formation and resorption depend on sex and age, and differ between trabecular and cortical bone.
Collapse
Affiliation(s)
- Julian K Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada.
| | - Neilab Amiri
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada.
| | - John D Schipilow
- Centre for High-Throughput Phenogenomics, Oral Biological and Medical Sciences, University of British Columbia, Vancouver, Canada.
| | - Steven W Zhang
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada.
| | | |
Collapse
|
12
|
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) 1-6 bind IGFs but not insulin with high affinity. They were initially identified as serum carriers and passive inhibitors of IGF actions. However, subsequent studies showed that, although IGFBPs inhibit IGF actions in many circumstances, they may also potentiate these actions. IGFBPs are widely expressed in most tissues, and they are flexible endocrine and autocrine/paracrine regulators of IGF activity, which is essential for this important physiological system. More recently, individual IGFBPs have been shown to have IGF-independent actions. Mechanisms underlying these actions include (i) interaction with non-IGF proteins in compartments including the extracellular space and matrix, the cell surface and intracellular space, (ii) interaction with and modulation of other growth factor pathways including EGF, TGF-β and VEGF, and (iii) direct or indirect transcriptional effects following nuclear entry of IGFBPs. Through these IGF-dependent and IGF-independent actions, IGFBPs modulate essential cellular processes including proliferation, survival, migration, senescence, autophagy and angiogenesis. They have been implicated in a range of disorders including malignant, metabolic, neurological and immune diseases. A more complete understanding of their cellular roles may lead to the development of novel IGFBP-based therapeutic opportunities.
Collapse
Affiliation(s)
- L A Bach
- Department of Medicine (Alfred)Monash University, Melbourne, Australia
- Department of Endocrinology and DiabetesAlfred Hospital, Melbourne, Australia
| |
Collapse
|
13
|
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
|
14
|
Yakar S, Werner H, Rosen CJ. Insulin-like growth factors: actions on the skeleton. J Mol Endocrinol 2018; 61:T115-T137. [PMID: 29626053 PMCID: PMC5966339 DOI: 10.1530/jme-17-0298] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022]
Abstract
The discovery of the growth hormone (GH)-mediated somatic factors (somatomedins), insulin-like growth factor (IGF)-I and -II, has elicited an enormous interest primarily among endocrinologists who study growth and metabolism. The advancement of molecular endocrinology over the past four decades enables investigators to re-examine and refine the established somatomedin hypothesis. Specifically, gene deletions, transgene overexpression or more recently, cell-specific gene-ablations, have enabled investigators to study the effects of the Igf1 and Igf2 genes in temporal and spatial manners. The GH/IGF axis, acting in an endocrine and autocrine/paracrine fashion, is the major axis controlling skeletal growth. Studies in rodents have clearly shown that IGFs regulate bone length of the appendicular skeleton evidenced by changes in chondrocytes of the proliferative and hypertrophic zones of the growth plate. IGFs affect radial bone growth and regulate cortical and trabecular bone properties via their effects on osteoblast, osteocyte and osteoclast function. Interactions of the IGFs with sex steroid hormones and the parathyroid hormone demonstrate the significance and complexity of the IGF axis in the skeleton. Finally, IGFs have been implicated in skeletal aging. Decreases in serum IGFs during aging have been correlated with reductions in bone mineral density and increased fracture risk. This review highlights many of the most relevant studies in the IGF research landscape, focusing in particular on IGFs effects on the skeleton.
Collapse
Affiliation(s)
- Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
| |
Collapse
|
15
|
The Wilms tumor protein WT1 stimulates transcription of the gene encoding insulin-like growth factor binding protein 5 (IGFBP5). Gene 2017; 619:21-29. [DOI: 10.1016/j.gene.2017.03.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 03/02/2017] [Accepted: 03/25/2017] [Indexed: 11/24/2022]
|
16
|
Yakar S, Isaksson O. Regulation of skeletal growth and mineral acquisition by the GH/IGF-1 axis: Lessons from mouse models. Growth Horm IGF Res 2016; 28:26-42. [PMID: 26432542 PMCID: PMC4809789 DOI: 10.1016/j.ghir.2015.09.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/16/2015] [Accepted: 09/24/2015] [Indexed: 12/31/2022]
Abstract
The growth hormone (GH) and its downstream mediator, the insulin-like growth factor-1 (IGF-1), construct a pleotropic axis affecting growth, metabolism, and organ function. Serum levels of GH/IGF-1 rise during pubertal growth and associate with peak bone acquisition, while during aging their levels decline and associate with bone loss. The GH/IGF-1 axis was extensively studied in numerous biological systems including rodent models and cell cultures. Both hormones act in an endocrine and autocrine/paracrine fashion and understanding their distinct and overlapping contributions to skeletal acquisition is still a matter of debate. GH and IGF-1 exert their effects on osteogenic cells via binding to their cognate receptor, leading to activation of an array of genes that mediate cellular differentiation and function. Both hormones interact with other skeletal regulators, such as sex-steroids, thyroid hormone, and parathyroid hormone, to facilitate skeletal growth and metabolism. In this review we summarized several rodent models of the GH/IGF-1 axis and described key experiments that shed new light on the regulation of skeletal growth by the GH/IGF-1 axis.
Collapse
Affiliation(s)
- Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology New York University College of Dentistry New York, NY 10010-408
| | - Olle Isaksson
- Institute of Medicine, Sahlgrenska University Hospital, University of Gothenburg, SE-41345 Gothenburg, Sweden
| |
Collapse
|
17
|
Liu Z, Mohan S, Yakar S. Does the GH/IGF-1 axis contribute to skeletal sexual dimorphism? Evidence from mouse studies. Growth Horm IGF Res 2016; 27:7-17. [PMID: 26843472 PMCID: PMC5488285 DOI: 10.1016/j.ghir.2015.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/24/2015] [Accepted: 12/03/2015] [Indexed: 11/22/2022]
Abstract
The contribution of the gonadotropic axis to skeletal sexual dimorphism (SSD) was clarified in recent years. Studies with animal models of estrogen receptor (ER) or androgen receptor (AR) null mice, as well as mice with bone cell-specific ablation of ER or AR, revealed that both hormones play major roles in skeletal acquisition, and that estrogen regulates skeletal accrual in both sexes. The growth hormone (GH) and its downstream effector, the insulin-like growth factor-1 (IGF-1) are also major determinants of peak bone mass during puberty and young adulthood, and play important roles in maintaining bone integrity during aging. A few studies in both humans and animal models suggest that in addition to the differences in sex steroid actions on bone, sex-specific effects of GH and IGF-1 play essential roles in SSD. However, the contributions of the somatotropic (GH/IGF-1) axis to SSD are controversial and data is difficult to interpret. GH/IGF-1 are pleotropic hormones that act in an endocrine and autocrine/paracrine fashion on multiple tissues, affecting body composition as well as metabolism. Thus, understanding the contribution of the somatotropic axis to SSD requires the use of mouse models that will differentiate between these two modes of action. Elucidation of the relative contribution of GH/IGF-1 axis to SSD is significant because GH is approved for the treatment of normal children with short stature and children with congenital growth disorders. Thus, if the GH/IGF-1 axis determines SSD, treatment with GH may be tailored according to sex. In the following review, we give an overview of the roles of sex steroids in determining SSD and how they may interact with the GH/IGF-1 axis in bone. We summarize several mouse models with impaired somatotropic axis and speculate on the possible contribution of that axis to SSD.
Collapse
Affiliation(s)
- Zhongbo Liu
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology New York University College of Dentistry New York, NY 10010-408, US
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare Systems, Loma Linda, CA 92357
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology New York University College of Dentistry New York, NY 10010-408, US.
| |
Collapse
|
18
|
Frenkel B, White W, Tuckermann J. Glucocorticoid-Induced Osteoporosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215995 DOI: 10.1007/978-1-4939-2895-8_8] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Osteoporosis is among the most devastating side effects of glucocorticoid (GC) therapy for the management of inflammatory and auto-immune diseases. Evidence from both humans and mice indicate deleterious skeletal effects within weeks of pharmacological GC administration, both related and unrelated to a decrease in bone mineral density (BMD). Osteoclast numbers and bone resorption are also rapidly increased, and together with osteoblast inactivation and decreased bone formation, these changes lead the fastest loss in BMD during the initial disease phase. Bone resorption then decreases to sub-physiological levels, but persistent and severe inhibition of bone formation leads to further bone loss and progressively increased fracture risk, up to an order of magnitude higher than that observed in untreated individuals. Bone forming osteoblasts are thus considered the main culprits in GC-induced osteoporosis (GIO). Accordingly, we focus this review primarily on deleterious effects on osteoblasts: inhibition of cell replication and function and acceleration of apoptosis. Mediating these adverse effects, GCs target pivotal regulatory mechanisms that govern osteoblast growth, differentiation and survival. Specifically, GCs inhibit growth factor pathways, including Insulin Growth Factors, Growth Hormone, Hepatocyte Growth/Scatter Factor and IL6-type cytokines. They also inhibit downstream kinases, including PI3-kinase and the MAP kinase ERK, the latter attributable in part to direct transcriptional stimulation of MAP kinase phosphatase 1. Most importantly, however, GCs inhibit the Wnt signaling pathway, which plays a pivotal role in osteoblast replication, function and survival. They transcriptionally stimulate expression of Wnt inhibitors of both the Dkk and Sfrp families, and they induce reactive oxygen species (ROS), which result in loss of ß-catenin to ROS-activated FoxO transcription factors. Identification of dissociated GCs, which would suppress the immune system without causing osteoporosis, is proving more challenging than initially thought, and GIO is currently managed by co-treatment with bisphosphonates or PTH. These drugs, however, are not ideally suited for GIO. Future therapeutic approaches may aim at GC targets such as those mentioned above, or newly identified targets including the Notch pathway, the AP-1/Il11 axis and the osteoblast master regulator RUNX2.
Collapse
Affiliation(s)
- Baruch Frenkel
- Department of Orthopaedic Surgery, Keck School of Medicine, Institute for Genetic Medicine, University of Southern California, 2250 Alcazar Street, CSC-240, Los Angeles, CA, 90033, USA,
| | | | | |
Collapse
|
19
|
Abstract
Insulin-like growth factor binding proteins (IGFBPs) 4-6 have important roles as modulators of IGF actions. IGFBP-4 and IGFBP-6 predominantly inhibit IGF actions, whereas IGFBP-5 may enhance these actions under some circumstances. IGFBP-6 is unique among the IGFBPs for its marked IGF-II binding preference. IGFBPs 4-6 are found in the circulation as binary complexes with IGFs that can enter tissues. Additionally, about half of the circulating IGFBP-5 is found in ternary complexes with IGFs and an acid labile subunit; this high molecular complex cannot leave the circulation and acts as an IGF reservoir. IGFBPs 4-6 also have IGF-independent actions. These IGFBPs are regulated in a cell-specific manner and their dysregulation may play a role in a range of diseases including cancer. However, there is no clear clinical indication for measuring serum levels of these IGFBPs at present.
Collapse
Affiliation(s)
- Leon A Bach
- Department of Medicine (Alfred), Monash University, Prahran, 3181, Australia; Department of Endocrinology and Diabetes, Alfred Hospital, Melbourne, 3004, Australia.
| |
Collapse
|
20
|
Anderson NG, Butler AP. Clinical applications of spectral molecular imaging: potential and challenges. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 9:3-12. [PMID: 24470290 DOI: 10.1002/cmmi.1550] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/20/2013] [Accepted: 05/28/2013] [Indexed: 12/22/2022]
Abstract
Spectral molecular imaging is a new X-ray-based imaging technology providing highly specific 3D imaging at high spatial resolution that has the potential to measure disease activity and response to treatment noninvasively. The ability to identify and quantify components of tissue and biomarkers of disease activity derive from the properties of the photon-processing detector. Multiple narrow sections of the energy spectrum are sampled simultaneously, providing a range of energy dependent Hounsfield units. As each material has a specific measurable X-ray spectrum, spectroscopic imaging allows for multiple materials to be quantified and differentiated from each other simultaneously. The technology, currently in its infancy, is set to grow rapidly, much as magnetic resonance did. The critical clinical applications have not yet been established, but it is likely to play a major role in identifying and directing treatment for unstable atherosclerotic plaque, assessing activity and response to treatment of a range of inflammatory diseases, and monitoring biomarkers of cancer and its treatment. If combined with Positron-emission tomography (PET), spectral molecular imaging could have a far greater effective role in cancer diagnosis and treatment monitoring than PET-CT does at present. It is currently used for small animal and specimen imaging. There are many challenges to be overcome before spectral imaging can be introduced into clinical medicine - these include technological improvements to detector design, bonding to the semiconductor layer, image reconstruction and display software, identifying which biomarkers are of most relevance to the disease in question, and accelerating drug discovery enabled by the new capabilities provided by spectral imaging.
Collapse
Affiliation(s)
- Nigel G Anderson
- Academic Radiology and Centre for Bioengineering, University of Otago, Christchurch, New Zealand; Medical Imaging, Royal Hobart Hospital, Hobart, Australia
| | | |
Collapse
|
21
|
Rodríguez-de la Rosa L, López-Herradón A, Portal-Núñez S, Murillo-Cuesta S, Lozano D, Cediel R, Varela-Nieto I, Esbrit P. Treatment with N- and C-terminal peptides of parathyroid hormone-related protein partly compensate the skeletal abnormalities in IGF-I deficient mice. PLoS One 2014; 9:e87536. [PMID: 24503961 PMCID: PMC3913635 DOI: 10.1371/journal.pone.0087536] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 12/31/2013] [Indexed: 11/18/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) deficiency causes growth delay, and IGF-I has been shown to partially mediate bone anabolism by parathyroid hormone (PTH). PTH-related protein (PTHrP) is abundant in bone, and has osteogenic features by poorly defined mechanisms. We here examined the capacity of PTHrP (1-36) and PTHrP (107-111) (osteostatin) to reverse the skeletal alterations associated with IGF-I deficiency. Igf1-null mice and their wild type littermates were treated with each PTHrP peptide (80 µg/Kg/every other day/2 weeks; 2 males and 4 females for each genotype) or saline vehicle (3 males and 3 females for each genotype). We found that treatment with either PTHrP peptide ameliorated trabecular structure in the femur in both genotypes. However, these peptides were ineffective in normalizing the altered cortical structure at this bone site in Igf1-null mice. An aberrant gene expression of factors associated with osteoblast differentiation and function, namely runx2, osteoprotegerin/receptor activator of NF-κB ligand ratio, Wnt3a , cyclin D1, connexin 43, catalase and Gadd45, as well as in osteocyte sclerostin, was found in the long bones of Igf1-null mice. These mice also displayed a lower amount of trabecular osteoblasts and osteoclasts in the tibial metaphysis than those in wild type mice. These alterations in Igf1-null mice were only partially corrected by each PTHrP peptide treatment. The skeletal expression of Igf2, Igf1 receptor and Irs2 was increased in Igf1-null mice, and this compensatory profile was further improved by treatment with each PTHrP peptide related to ERK1/2 and FoxM1 activation. In vitro, PTHrP (1-36) and osteostatin were effective in promoting bone marrow stromal cell mineralization in normal mice but not in IGF-I-deficient mice. Collectively, these findings indicate that PTHrP (1-36) and osteostatin can exert several osteogenic actions even in the absence of IGF-I in the mouse bone.
Collapse
Affiliation(s)
- Lourdes Rodríguez-de la Rosa
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Ana López-Herradón
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Portal-Núñez
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Daniel Lozano
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Cediel
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
22
|
Kjaer-Sorensen K, Engholm DH, Jepsen MR, Morch MG, Weyer K, Hefting LL, Skov LL, Laursen LS, Oxvig C. Pregnancy-associated plasma protein-A2 modulates development of cranial cartilage and angiogenesis in zebrafish embryos. J Cell Sci 2014; 127:5027-37. [DOI: 10.1242/jcs.152587] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Pregnancy-associated plasma protein-A2 (PAPP-A2, pappalysin-2) is a large metalloproteinase, known to be required for normal postnatal growth and bone development in mice. We here report the detection of zebrafish papp-a2 mRNA in chordamesoderm, notochord, and lower jaw of zebrafish (Danio rerio) embryos, and that papp-a2 knockdown embryos display broadened axial mesoderm, notochord bends, and severely reduced cranial cartilages. Genetic data link these phenotypes to insulin-like growth factor binding protein-3 (Igfbp-3) and Bmp signaling, and biochemical analysis show specific Igfbp-3 proteolysis by Papp-a2, implicating Papp-a2 in the modulation of Bmp signaling by Igfbp-3 proteolysis. Knockdown of papp-a2 additionally resulted in angiogenesis defects, strikingly similar to previous observations in embryos with mutations in components of the Notch system. Concordantly, we find that Notch signaling is modulated by Papp-a2 in vivo, and, furthermore, that PAPP-A2 is capable of modulating Notch signaling independently of its proteolytic activity in cell culture. Based on these results, we conclude that Papp-a2 modulates Bmp and Notch signaling by independent mechanisms in zebrafish embryos. In conclusion, these data link pappalysin function in zebrafish to two different signaling pathways outside the IGF system.
Collapse
|
23
|
Guerra-Menéndez L, Sádaba MC, Puche JE, Lavandera JL, de Castro LF, de Gortázar AR, Castilla-Cortázar I. IGF-I increases markers of osteoblastic activity and reduces bone resorption via osteoprotegerin and RANK-ligand. J Transl Med 2013; 11:271. [PMID: 24161214 PMCID: PMC4231608 DOI: 10.1186/1479-5876-11-271] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/16/2013] [Indexed: 02/06/2023] Open
Abstract
Background Bone is one of the major target tissues for Insulin-like Growth Factor I (IGF-I). Low doses of IGF-I were able to improve liver-associated osteopenia. In the present work, a model of partial IGF-I deficiency was used in order to provide insight into the mechanisms of the beneficial actions of IGF-I replacement therapy in bone. Methods Several proteins involved in osteoblastic/osteocyte and osteoclastic differentiation and activity were studied in the three experimental groups: control (CO) group (wild type mice, Igf+/+, n = 10), heterozygous Igf+/- group with partial IGF-I deficiency (Hz, n = 10), and heterozygous Igf+/- mice treated with IGF-I for 10 days (Hz + IGF-I, n = 10). Results Data in this paper confirm that the simple partial IGF-I deficiency is responsible for osteopenia, determined by densitometry and histopathology. These findings are associated with a reduced gene expression of osteoprotegerin, sclerostin, calcitonin receptor (CTR), insulin-like growth factor binding protein 5 and RUNX2. IGF-I replacement therapy normalized CTR gene expression and reduced markers of osteoclastic activity. Conclusions Low doses of IGF-I constituted a real replacement therapy that normalized IGF-I serum levels improving the expression of most of these proteins closely involved in bone-forming, and reducing bone resorption by mechanisms related to osteoprotegerin, RANKL and PTH receptor.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Inma Castilla-Cortázar
- Department of Medical Physiology, Universidad CEU San Pablo, Institute of Applied Molecular Medicine (IMMA), School of Medicine, Room D-201, C/ Boadilla del Monte s/n, km 5,3, 28668 Madrid, Spain.
| |
Collapse
|
24
|
Torres M, Montserrat JM, Pavía J, Dalmases M, Ros D, Fernandez Y, Barbé F, Navajas D, Farré R. Chronic intermittent hypoxia preserves bone density in a mouse model of sleep apnea. Respir Physiol Neurobiol 2013; 189:646-8. [PMID: 23994179 DOI: 10.1016/j.resp.2013.08.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 01/12/2023]
Abstract
Very recent clinical research has investigated whether obstructive sleep apnea (OSA) may modulate bone homeostasis but the few data available are conflicting. Here we report novel data obtained in a mouse study specifically designed to determine whether chronic intermittent hypoxia realistically mimicking OSA modifies bone mineral density (BMD). Normal male and female mice and orchidectomized mice (N=10 each group) were subjected to a pattern of high-frequency intermittent hypoxia (20s at 5% and 40s at 21%, 60 cycles/h) for 6h/day. Identical groups breathing room air (normoxia) were the controls. After 32 days of intermittent hypoxia/normoxia the trabecular bone mineral density (BMD) in the peripheral femora were measured by micro-CT scanning. When compared with normoxia (two-way ANOVA), intermittent hypoxia did not significantly modify BMD in the three animal groups tested. Data in this study suggest that the type of intermittent hypoxia characterizing OSA, applied as a single challenge, preserves bone homeostasis.
Collapse
Affiliation(s)
- Marta Torres
- CIBER Enfermedades Respiratorias, Bunyola, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Doorn J, Roberts SJ, Hilderink J, Groen N, van Apeldoorn A, van Blitterswijk C, Schrooten J, de Boer J. Insulin-like growth factor-I enhances proliferation and differentiation of human mesenchymal stromal cells in vitro. Tissue Eng Part A 2013; 19:1817-28. [PMID: 23530894 DOI: 10.1089/ten.tea.2012.0522] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human mesenchymal stromal cells (hMSCs) offer great potential for bone tissue engineering applications, but their in vivo performance remains limited. Preconditioning of these cells with small molecules to improve their differentiation before implantation, or incorporation of growth factors are possible solutions. Insulin-like growth factor-1 (IGF-1) is one of the most abundant growth factors in bone, involved in growth, development, and metabolism, but its effects on hMSCs are still subject of debate. Here we examined the effects of IGF-1 on proliferation and differentiation of hMSCs in vitro and we found that serum abolished the effects of IGF-1. Only in the absence of serum, IGF-1 increased proliferation, alkaline phosphatase expression, and osteogenic gene expression of hMSCs. Furthermore, we examined synergistic effects of bone morphogenetic protein-2 (BMP-2) and IGF-1 and, although IGF-1 enhanced BMP-2-induced mineralization, IGF-1 only slightly affected in vivo bone formation.
Collapse
Affiliation(s)
- Joyce Doorn
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Strohbach CA, Scofield DE, Nindl BC, Centi AJ, Yanovich R, Evans RK, Moran DS. Female recruits sustaining stress fractures during military basic training demonstrate differential concentrations of circulating IGF-I system components: a preliminary study. Growth Horm IGF Res 2012; 22:151-157. [PMID: 22704365 DOI: 10.1016/j.ghir.2012.04.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 04/24/2012] [Accepted: 04/25/2012] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Stress fracture injuries sustained during military basic combat training (BT) are a significant problem and occur at a higher rate in female recruits than male recruits. Insulin-like growth factor-I (IGF-I) is an easily measured biomarker that is involved in bone formation and positively correlated with bone mineral density, especially in women. This study examined the response of the IGF-I system between female soldiers that sustained a stress fracture (SFX, n=13) during BT and female soldiers who did not (NSFX, n=49). DESIGN Female soldiers (n=62, 18.8 ± 0.6 yr) from 2 companies of a gender-integrated combat battalion in the Israeli Defense Forces participated in this study. Height, weight and blood draws were taken upon entry to BT (preBT) and after a four-month BT program (postBT). Stress fractures were diagnosed by bone scan. Serum was analyzed for total IGF-I, free IGF-I, IGF binding proteins (IGFBP)1-6, BAP, calcium, CTx, IL1β, IL6, PINP, PTH, TNFα, TRAP, and 25(OH)D. Statistical differences between SFX and NSFX groups and time points were assessed by RM ANOVA with Fisher post-hoc (p≤0.05). RESULTS The SFX group was significantly taller and had lower BMI than NSFX (p≤0.05). Serum concentrations of total IGF-I, bioavailable IGF-I, other bone biomarkers, and cytokines were not significantly different between SFX and NSFX preBT. Serum IGFBP-2 and IGFBP-5 were significantly higher in the SFX compared to the NSFX preBT (p≤0.05). In both groups, total IGF-I increased pre to postBT (p≤0.05). Additionally, a significant difference was observed in the bioavailable IGF-I response pre to postBT for both groups. The SFX group demonstrated a significant decrease in bioavailable IGF-I pre to postBT (preBT: 0.58 ± 0.58 ng/mL; postBT 0.39 ± 0.48; p≤0.05) whereas the NSFX group demonstrated a significant increase in bioavailable IGF-I pre to postBT (preBT: 0.53 ± 0.37 ng/mL; postBT: 0.63 ± 0.45; p≤0.05). CONCLUSIONS Our study demonstrated that serum IGF-I changes during basic training and that women sustaining stress fractures during BT significantly decreased bioavailable IGF-I, whereas their uninjured counter parts increased bioavailable IGF-I. These results suggest that stress fracture susceptibility may be related to differential IGF-I system concentrations and response to physical training.
Collapse
Affiliation(s)
- C A Strohbach
- Military Performance Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA 01760, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
The Lsktm1 locus modulates lung and skin tumorigenesis in the mouse. G3-GENES GENOMES GENETICS 2012; 2:1041-6. [PMID: 22973541 PMCID: PMC3429918 DOI: 10.1534/g3.112.003525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 06/27/2012] [Indexed: 02/06/2023]
Abstract
Alleles derived from skin tumor−resistant Car-R mice provide resistance to both skin and lung tumorigenesis over the susceptibility of the SWR/J strain. In an effort to map tumor modifier loci affecting both tumor types, we carried out a genetic linkage analysis in backcross SWR/J x (SWR/J x Car-R) mice and identified a locus (Lsktm1) on chromosome 1 linked to both skin (LOD score = 3.93) and lung (LOD score = 8.74) tumorigenesis. Two genes, Igfbp5 and Igfbp2, residing in this locus and belonging to the insulin-like growth factor binding protein family were expressed at significantly greater levels in normal lung tissue from cancer-resistant Car-R mice than in cancer-susceptible SWR/J mice. Overexpression of the recombinant Igfbp5 and Igfbp2 genes in two lung cancer cell lines significantly inhibited clonogenicity (P < 0.0001). Collectively, we have identified a single polymorphic locus that affects skin and lung tumorigenesis and identify Igfbp5 and Igfbp2 as candidate modifier genes of lung tumorigenesis.
Collapse
|
28
|
Bikle DD, Wang Y. Insulin like growth factor-I: a critical mediator of the skeletal response to parathyroid hormone. Curr Mol Pharmacol 2012; 5:135-142. [PMID: 21787292 PMCID: PMC3690777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 08/16/2010] [Accepted: 03/03/2011] [Indexed: 05/31/2023]
Abstract
This review focuses on the mechanisms by which PTH stimulates both osteoblast and osteoclast function, emphasizing the critical role that IGF-I plays in these processes. After reviewing the current literature on the skeletal actions of PTH and the modulation of IGF action on bone by the different IGF-binding proteins, the review then examines studies from mouse models in which IGF-I or its receptor have been selectively deleted in different cells of the skeletal system, in particular osteoprogenitors, mature osteoblasts, and osteoclasts. Mice in which IGF-I production has been deleted from all cells are deficient in both bone formation and bone resorption with few osteoblasts or osteoclasts in bone in vivo, reduced osteoblast colony forming units, and an inability of either the osteoblasts or osteoclast precursors to support osteoclastogenesis in vitro. Mice in which the IGF-I receptor is specifically deleted in mature osteoblasts have a mineralization defect in vivo, and bone marrow stromal cells from these mice fail to mineralize in vitro. Mice in which the IGF-I receptor is deleted in osteoprogenitor cells have a marked reduction in osteoblast proliferation and differentiation leading to osteopenia. Finally mice lacking the IGF-I receptor in their osteoclasts have increased bone and decreased osteoclast formation. PTH fails to stimulate bone formation in the mice lacking IGF-I or its receptor in osteoblasts or enhance the signaling between osteoblasts and osteoclasts through RANKL/RANK and Ephrin B2/Eph B4, emphasizing the role IGF-I signaling plays in cell-communication per se and as stimulated by PTH.
Collapse
Affiliation(s)
- Daniel D Bikle
- Veterans Affairs Medical Center and University of California, San Francisco, San Francisco, CA 94121, USA.
| | | |
Collapse
|
29
|
Abstract
Insulin-like growth factor 1 (IGF-1) is a pleiotropic polypeptide. Its expression is tightly regulated and it plays significant roles during early development, maturation, and adulthood. This article discusses the roles of IGF-1 in determination of body size, skeletal acquisition, muscle growth, carbohydrate metabolism, and longevity, as learned from mouse models.
Collapse
Affiliation(s)
- Shoshana Yakar
- Department of Basic Science and Craniofacial Biology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA.
| | | |
Collapse
|
30
|
Jia J, Li L, Zhao Q, Zhang L, Ru J, Liu X, Li Q, Shi L. Association of a single nucleotide polymorphism in pregnancy-associated plasma protein-A2 with developmental dysplasia of the hip: a case-control study. Osteoarthritis Cartilage 2012; 20:60-3. [PMID: 22037112 DOI: 10.1016/j.joca.2011.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/21/2011] [Accepted: 10/03/2011] [Indexed: 02/02/2023]
Abstract
Developmental dysplasia of the hip (DDH), previously known as congenital hip dislocation, is a frequently disabling condition characterized by premature arthritis in later life. Genetic influence on DDH has been long known, but is still poorly understood. Previously, we have performed a genome-wide linkage scan with Affymetrix 10K genechip for a four-generation Chinese family, which included 19 healthy members and five patients with DDH. Parametric and nonparametric multipoint linkage analyses were carried out with Genespring GT v.2.0 software, and the logarithm of odds (LOD) score and nonparametric linkage (NPL) score were calculated. For parametric linkage analysis, an assuming autosomal recessive trait was used with full penetrance, and Affymetrix "Asian" allele frequencies. The NPL score of 2.698 (P=0.0156) and LOD score of 2.119 (θ=0) were obtained on chromosome 1q25.2a for one marker (rs726252). The single nucleotide polymorphism (SNP) rs726252 locates in the region of fifth intron of pregnancy-associated plasma protein-A2 (PAPPA2). Although neither LOD nor NPL scores of rs726252 has exceeded 3.0, several researches have demonstrated that PAPPA2 have important consequences for the development of the fetus and normal postnatal growth. To further evaluate this possible association, in the present study, we examined the genetic association of rs726252 in PAPPA2 gene with sporadic DDH in Han Chinese population using case-control study, including 310 patients with sporadic DDH and 487 control subjects, and found a significant association between PAPPA2 and DDH.
Collapse
Affiliation(s)
- Jingyu Jia
- Department of Paediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
|
33
|
Olson LE, Ohlsson C, Mohan S. The role of GH/IGF-I-mediated mechanisms in sex differences in cortical bone size in mice. Calcif Tissue Int 2011; 88:1-8. [PMID: 21113585 PMCID: PMC3042806 DOI: 10.1007/s00223-010-9436-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/31/2010] [Indexed: 12/20/2022]
Abstract
Cortical bone dimensions are important determinants of bone strength. Gender differences in cortical bone size caused by greater periosteal expansion in males than in females during the pubertal growth spurt are well established both in humans and in experimental animal models. However, the mechanism by which gender influences cortical bone size is still a matter of investigation. The role of androgens and estrogen in pubertal bone growth has been examined in human disorders as well as animal models, such as gonadectomized or sex steroid receptor knockout mice. Based on the findings that growth hormone (GH) and insulin-like growth factor I (IGF-I) are major regulators of postnatal skeletal growth, we and others have predicted that sex hormones interact with the GH/IGF-I axis to regulate cortical bone size. However, studies conflict as to whether estrogen and androgens impact cortical bone size through the canonical pathway, through GH without IGF-I mediation, through IGF-I without GH stimulation, or independent of GH/IGF-I. We review recent data on the impact of sex steroids and components of the GH/IGF axis on sexual dimorphism in bone size. While the GH/IGF-I axis is a major player in regulating peak bone size, the relative contribution of GH/IGF-dependent mechanisms to sex differences in cortical bone size remains to be established.
Collapse
Affiliation(s)
- Lisa E Olson
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center and Loma Linda University, Loma Linda, CA 92357, USA
| | | | | |
Collapse
|
34
|
Abstract
Insulin-like growth factor-1 (IGF-1) plays a central role in cellular growth, differentiation, survival, and cell cycle progression. It is expressed early during development and its effects are mediated through binding to a tyrosine kinase receptor, the insulin-like growth factor-1 receptor (IGF-1R). In the circulation, the IGFs bind to IGF-binding proteins (IGFBPs), which determine their bioavailability and regulate the interaction between the IGFs and IGF-1R. Studies in animal models and in humans have established critical roles for IGFs in skeletal growth and development. In this review we present new and old findings from mouse models of the IGF system and discuss their clinical relevance to normal and pathological skeletal physiology.
Collapse
Affiliation(s)
- Shoshana Yakar
- Division of Endocrinology, University of North Carolina, Chapel Hill, NC, USA.
| | | | | |
Collapse
|
35
|
Chen L, Jiang W, Huang J, He BC, Zuo GW, Zhang W, Luo Q, Shi Q, Zhang BQ, Wagner ER, Luo J, Tang M, Wietholt C, Luo X, Bi Y, Su Y, Liu B, Kim SH, He CJ, Hu Y, Shen J, Rastegar F, Huang E, Gao Y, Gao JL, Zhou JZ, Reid RR, Luu HH, Haydon RC, He TC, Deng ZL. Insulin-like growth factor 2 (IGF-2) potentiates BMP-9-induced osteogenic differentiation and bone formation. J Bone Miner Res 2010; 25:2447-2459. [PMID: 20499340 PMCID: PMC3179288 DOI: 10.1002/jbmr.133] [Citation(s) in RCA: 211] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2009] [Revised: 03/28/2010] [Accepted: 05/05/2010] [Indexed: 12/23/2022]
Abstract
Efficient osteogenic differentiation and bone formation from mesenchymal stem cells (MSCs) should have clinical applications in treating nonunion fracture healing. MSCs are adherent bone marrow stromal cells that can self-renew and differentiate into osteogenic, chondrogenic, adipogenic, and myogenic lineages. We have identified bone morphogenetic protein 9 (BMP-9) as one of the most osteogenic BMPs. Here we investigate the effect of insulin-like growth factor 2 (IGF-2) on BMP-9-induced bone formation. We have found that endogenous IGF-2 expression is low in MSCs. Expression of IGF-2 can potentiate BMP-9-induced early osteogenic marker alkaline phosphatase (ALP) activity and the expression of later markers. IGF-2 has been shown to augment BMP-9-induced ectopic bone formation in the stem cell implantation assay. In perinatal limb explant culture assay, IGF-2 enhances BMP-9-induced endochondral ossification, whereas IGF-2 itself can promote the expansion of the hypertropic chondrocyte zone of the cultured limb explants. Expression of the IGF antagonists IGFBP3 and IGFBP4 leads to inhibition of the IGF-2 effect on BMP-9-induced ALP activity and matrix mineralization. Mechanistically, IGF-2 is further shown to enhance the BMP-9-induced BMPR-Smad reporter activity and Smad1/5/8 nuclear translocation. PI3-kinase (PI3K) inhibitor LY294002 abolishes the IGF-2 potentiation effect on BMP-9-mediated osteogenic signaling and can directly inhibit BMP-9 activity. These results demonstrate that BMP-9 crosstalks with IGF-2 through PI3K/AKT signaling pathway during osteogenic differentiation of MSCs. Taken together, our findings suggest that a combination of BMP-9 and IGF-2 may be explored as an effective bone-regeneration agent to treat large segmental bony defects, nonunion fracture, and/or osteoporotic fracture.
Collapse
Affiliation(s)
- Liang Chen
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical UniversityChongqing, People's Republic of China
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Wei Jiang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Jiayi Huang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Bai-Cheng He
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Guo-Wei Zuo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Wenli Zhang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Department of Orthopaedic Surgery, West China Hospital, Sichuan UniversityChengdu, Sichuan, People's Republic of China
| | - Qing Luo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Qiong Shi
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Bing-Qiang Zhang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Eric R Wagner
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Jinyong Luo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Min Tang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | | | - Xiaoji Luo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Yang Bi
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Yuxi Su
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Bo Liu
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Stephanie H Kim
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Connie J He
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Yawen Hu
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Jikun Shen
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Farbod Rastegar
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Enyi Huang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- School of Bioengineering, Chongqing UniversityChongqig, People's Republic of China
| | - Yanhong Gao
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Department of Geriatrics, Xinhua Hospital of Shanghai Jiatong UniversityShanghai, People's Republic of China
| | - Jian-Li Gao
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Jian-Zhong Zhou
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| | - Zhong-Liang Deng
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical UniversityChongqing, People's Republic of China
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical CenterChicago, IL, USA
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and the Affiliated Hospitals, Chongqing Medical UniversityChongqing, People's Republic of China
| |
Collapse
|
36
|
Okudaira S, Shimizu M, Otsuki B, Nakanishi R, Ohta A, Higuchi K, Hosokawa M, Tsuboyama T, Nakamura T. Quantitative trait locus on chromosome X affects bone loss after maturation in mice. J Bone Miner Metab 2010; 28:520-31. [PMID: 20354743 DOI: 10.1007/s00774-010-0168-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 02/08/2010] [Indexed: 12/24/2022]
Abstract
Genetic programming is known to affect the peak bone mass and bone loss after maturation. However, little is known about how polymorphic genes on chromosome X (Chr X) modulate bone loss after maturation. We previously reported a quantitative trait locus (QTL) on Chr X, designated Pbd3, which had a suggestive linkage to bone mass, in male SAMP2 and SAMP6 mice. In this study, we aimed to clarify the effects of Pbd3 on the skeletal phenotype. We generated a congenic strain, P2.P6-X, carrying a 45.6-cM SAMP6-derived Chr X interval on a SAMP2 genetic background. The effects of Pbd3 on the bone phenotype were determined by microcomputed tomography (microCT), whole-body dual-energy X-ray absorptiometry (DXA), serum bone turnover markers, and histomorphometric parameters. Both the bone area fraction (BA/TA) on microCT and whole-body DXA revealed reduced bone loss in P2.P6-X compared with that in SAMP2. The serum concentrations of bone turnover markers at 4 months of age were significantly lower in P2.P6-X than in SAMP2, but did not differ at 8 months of age. These results were observed in female mice, but not in male mice. In conclusion, a QTL within a segregated 45.6-cM interval on Chr X is sex-specifically related to the rate of bone loss after maturation.
Collapse
Affiliation(s)
- Shuzo Okudaira
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Phang D, Rehage M, Bonafede B, Hou D, Xing W, Mohan S, Wergedal JE, Qin X. Inactivation of insulin-like-growth factors diminished the anabolic effects of pregnancy-associated plasma protein-A (PAPP-A) on bone in mice. Growth Horm IGF Res 2010; 20:192-200. [PMID: 20144555 DOI: 10.1016/j.ghir.2010.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 01/06/2010] [Accepted: 01/12/2010] [Indexed: 11/16/2022]
Abstract
In vivo studies have provided ubiquitous evidence that pregnancy-associated plasma protein-A (PAPP-A) functions as a potent anabolic factor. While some evidence supports the prediction that increasing IGF bioavailability contributes to the anabolic effects of PAPP-A, definitive evidence has been lacking. This important issue has been addressed in this study using a unique mouse model in which PAPP-A was overexpressed in bone either alone or together with a protease-resistant IGFBP-4 analog (PRBP-4) which serves as an IGF inhibitor. PAPP-A transgenic mice exhibited a 25% increase in skull bone mineral density (BMD) whereas PRBP-4 transgenic mice showed a 20-25% decrease in this parameter at an age of 3months. Femur/tibia size-related parameters were significantly increased in PAPP-A transgenic mice but decreased in PRBP-4 transgenic mice. This data clearly demonstrates that PAPP-A transgenic mice exhibit opposite phenotypes in both flat bone and long bone compared to PRBP-4 transgenic mice which have reduced IGF bioavailability in bone. Importantly, PRBP-4 and PRBP-4/PAPP-A double transgenic mice shared essentially identical phenotypes in both flat and long bones. Calvarial thickness, skull BMD and long bone parameters were reduced to similar degrees in PRBP-4 and PRBP-4/PAPP-A transgenic mice relative to wild-type littermates. Our findings provide compelling evidence that PAPP-A increases bone formation primarily by increasing IGF bioavailability and that other alternative pathways may play a negligible role in mediating the anabolic effect of PAPPA in bone. This clear definition of PAPP-A's mechanism of action is critical for future translational studies on the therapeutic application of PAPP-A.
Collapse
Affiliation(s)
- David Phang
- Musculoskeletal Disease Center, Loma Linda University, Loma Linda, CA 92354, United States
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Xiong Q, Jiao Y, Hasty KA, Canale ST, Stuart JM, Beamer WG, Deng HW, Baylink D, Gu W. Quantitative trait loci, genes, and polymorphisms that regulate bone mineral density in mouse. Genomics 2009; 93:401-14. [PMID: 19150398 DOI: 10.1016/j.ygeno.2008.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 11/26/2008] [Accepted: 12/15/2008] [Indexed: 01/23/2023]
Abstract
This is an in silico analysis of data available from genome-wide scans. Through analysis of QTL, genes and polymorphisms that regulate BMD, we identified 82 BMD QTL, 191 BMD-associated (BMDA) genes, and 83 genes containing known BMD-associated polymorphisms (BMDAP). The catalogue of all BMDA/BMDAP genes and relevant literatures are provided. In total, there are substantially more BMDA/BMDAP genes in regions of the genome where QTL have been identified than in non-QTL regions. Among 191 BMDA genes and 83 BMDAP genes, 133 and 58 are localized in QTL regions, respectively. The difference was still noticeable for the chromosome distribution of these genes between QTL and non-QTL regions. These results have allowed us to generate an integrative profile of QTL, genes, polymorphisms that determine BMD. These data could facilitate more rapid and comprehensive identification of causal genes underlying the determination of BMD in mouse and provide new insights into how BMD is regulated in humans.
Collapse
Affiliation(s)
- Qing Xiong
- Department of Orthopaedic Surgery - Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Flint DJ, Tonner E, Beattie J, Allan GJ. Role of insulin-like growth factor binding proteins in mammary gland development. J Mammary Gland Biol Neoplasia 2008; 13:443-53. [PMID: 18998203 DOI: 10.1007/s10911-008-9095-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 10/28/2008] [Indexed: 01/08/2023] Open
Abstract
Insulin-like growth factors (IGFs) play an important role in mammary gland development and their effects are, in turn, influenced by a family of 6 IGF-binding proteins (IGFBPs). The IGFBPs are expressed in time- and tissue-specific fashion during the periods of rapid growth and involution of the mammary gland. The precise roles of these proteins in vivo have, however, been difficult to determine. This review examines the indirect evidence (evolution, chromosomal location and roles in lower life-forms) the evidence from in vitro studies and the attempts to examine their roles in vivo, using IGFBP-deficient and over-expression models. Evidence exists for a role of the IGFBPs in inhibition of the survival effects of IGFs as well as in IGF-enhancing effects from in vitro studies. The location of the IGFBPs, often associated with the extracellular matrix, suggests roles as a reservoir of IGFs or as a potential barrier, restricting access of IGFs to distinct cellular compartments. We also discuss the relative importance of IGF-dependent versus IGF-independent effects. IGF-independent effects include nuclear localization, activation of proteases and interaction with a variety of extracellular matrix and cell surface proteins. Finally, we examine the increasing evidence for the IGFBPs to be considered as part of a larger family of extracellular matrix proteins involved in morphogenesis and tissue re-modeling.
Collapse
Affiliation(s)
- D J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0NR, UK.
| | | | | | | |
Collapse
|
40
|
Govoni KE, Wergedal JE, Chadwick RB, Srivastava AK, Mohan S. Prepubertal OVX increases IGF-I expression and bone accretion in C57BL/6J mice. Am J Physiol Endocrinol Metab 2008; 295:E1172-80. [PMID: 18812464 PMCID: PMC2584807 DOI: 10.1152/ajpendo.90507.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is generally well accepted that the pubertal surge in estrogen is responsible for the rapid bone accretion that occurs during puberty and that this effect is mediated by an estrogen-induced increase in growth hormone (GH)/insulin-like growth factor (IGF) action. To test the cause and effect relationship between estrogen and GH/IGF, we evaluated the consequence of ovariectomy (OVX) in prepubertal mice (C57BL/6J mice at 3 wk of age) on skeletal changes and the GH/IGF axis during puberty. Contrary to our expectations, OVX increased body weight (12-18%), bone mineral content (11%), bone length (4%), bone size (3%), and serum, liver, and bone IGF-I (30-50%) and decreased total body fat (18%) at 3 wk postsurgery. To determine whether estrogen is the key ovarian factor responsible for these changes, we performed a second experiment in which OVX mice were treated with placebo or estrogen implants. In addition to observing similar results compared with our first experiment, estrogen treatment partially rescued the increased body weight and bone size and completely rescued body fat and IGF-I levels. The increased bone accretion in OVX mice was due to increased bone formation rate (as determined by bone histomorphometry) and increased serum procollagen peptide. In conclusion, contrary to the known estrogen effect as an initiator of GH/IGF surge and thereby pubertal growth spurt, our findings demonstrate that loss of estrogen and/or other hormones during the prepubertal growth period effect leads to an increase in IGF-I production and bone accretion in mice.
Collapse
Affiliation(s)
- Kristen E Govoni
- Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California 92357, USA
| | | | | | | | | |
Collapse
|
41
|
Pérez-Casellas LA, Wang X, Howard KD, Rehage MW, Strong DD, Linkhart TA. Nuclear factor I transcription factors regulate IGF binding protein 5 gene transcription in human osteoblasts. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1789:78-87. [PMID: 18809517 DOI: 10.1016/j.bbagrm.2008.08.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 08/11/2008] [Accepted: 08/26/2008] [Indexed: 11/17/2022]
Abstract
Insulin-like growth factor binding protein 5 (IGFBP5) is expressed in many cell types including osteoblasts and modulates IGF activities. IGFBP5 may affect osteoblasts and bone formation, in part by mechanisms independent of binding IGFs. The highly conserved IGFBP5 proximal promoter within 100 nucleotides of the start of transcription contains functional cis regulatory elements for C/EBP, Myb and AP-2. We report evidence for a functional Nuclear Factor I (NFI) cis element that mediates activation or repression of IGFBP5 transcription by the NFI gene family. All four NFI genes were expressed in human osteoblast cultures and osteosarcoma cell lines. Co-transfection with human IGFBP5 promoter luciferase reporter and murine Nfi expression vectors showed that Nfib was the most active in stimulating transcription. Nfix was less active and Nfia and Nfic were inhibitory. Knockdown of NFIB and NFIC expression using siRNA decreased and increased IGFBP5 expression, respectively. Analysis of IGFBP5 promoter deletion and mutation reporter constructs identified a functional NFI cis element. All four NFI proteins bound the NFI site in electrophoretic mobility shift experiments and NFIB bound in chromatin immunoprecipitation assays. Results suggest that NFI proteins are important regulators of IGFBP5 expression in human osteoblasts and thus in modulating IGFBP5 functions in bone.
Collapse
|
42
|
Hoeflich A. Contrasting bone effects of temporary versus permanent IGFBP administration in rodents. Growth Horm IGF Res 2008; 18:181-187. [PMID: 18308605 DOI: 10.1016/j.ghir.2008.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 01/11/2008] [Accepted: 01/14/2008] [Indexed: 01/14/2023]
Abstract
Transgenic animal technology has tremendously improved our current comprehension of IGFBP biology. The high potential of IGFBP transgenic mouse models is due to the fact that they mimic elevated serum IGFBP levels, which are diagnosed under the conditions of impaired growth or critical illness. In general, long term elevated levels of IGFBPs in transgenic mouse models almost exclusively resulted in inhibitory phenotypes e.g. of body or organ growth, indicating specific effects in different cell types. This holds especially for the distinct cellular populations present in the bone environment. After establishing transgenic mouse lines modelling permanent increases of IGFBPs, a second question now poses challenge to current functional genome analysis: what is the function of temporary exposure of a certain cell type to isolated IGFBPs? This question is particularly important due to the fact that elevated IGFBP expression is often found in a conditional fashion and in line with the contradictory findings after long or short term IGFBP exposure in rodent models. In order to understand the potential roles of the conditional increases of IGFBP expression, e.g. during illness, and to further study the adaptive or even therapeutic potential of IGFBPs for certain applications like osteoporosis, it is imperative to take a closer look also to the acute effects of the IGFBPs.
Collapse
Affiliation(s)
- Andreas Hoeflich
- Laboratory of Mouse Genetics, Research Unit of Genetics and Biometry, Research Institute for the Biology of Farm Animals Dummerstorf (FBN), Wilhelm Stahl Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
43
|
Guo Y, Yang TL, Pan F, Xu XH, Dong SS, Deng HW. Molecular genetic studies of gene identification for osteoporosis. Expert Rev Endocrinol Metab 2008; 3:223-267. [PMID: 30764094 DOI: 10.1586/17446651.3.2.223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This review comprehensively summarizes the most important and representative molecular genetics studies of gene identification for osteoporosis published up to the end of September 2007. It is intended to constitute a sequential update of our previously published reviews covering the available data up to the end of 2004. Evidence from candidate gene-association studies, genome-wide linkage and association studies, as well as functional genomic studies (including gene-expression microarray and proteomics) on osteogenesis and osteoporosis, are reviewed separately. Studies of transgenic and knockout mice models relevant to osteoporosis are summarized. The major results of all studies are tabulated for comparison and ease of reference. Comments are made on the most notable findings and representative studies for their potential influence and implications on our present understanding of genetics of osteoporosis. The format adopted by this review should be ideal for accommodating future new advances and studies.
Collapse
Affiliation(s)
- Yan Guo
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Tie-Lin Yang
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Pan
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Xiang-Hong Xu
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Shan-Shan Dong
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Hong-Wen Deng
- b The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China and Departments of Orthopedic Surgery and Basic Medical Sciences, University of Missouri - Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
44
|
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: 46] [Impact Index Per Article: 2.7] [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
|
45
|
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) are important regulators of bone metabolism. However, their precise roles are not fully understood, since IGFBPs can have both enhancing and inhibiting effects on IGF action, depending on context and posttranslational modifications, as well as IGF-independent effects. This review focuses on recent findings from cell culture, rodent models, and clinical studies concerning local IGFBP-2, IGFBP-4, and IGFBP-5 action in bone.
Collapse
Affiliation(s)
- Cheryl A Conover
- Endocrine Research Unit, Division of Endocrinology and Metabolosm, Department of Medicine, Mayo Clinic, 200 First St. SW, 5-194 Joseph, Rochester, MN 55905, USA.
| |
Collapse
|
46
|
Govoni KE, Wergedal JE, Florin L, Angel P, Baylink DJ, Mohan S. Conditional deletion of insulin-like growth factor-I in collagen type 1alpha2-expressing cells results in postnatal lethality and a dramatic reduction in bone accretion. Endocrinology 2007; 148:5706-15. [PMID: 17717052 PMCID: PMC2916650 DOI: 10.1210/en.2007-0608] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IGF-I acts through endocrine and local, autocrine/paracrine routes. Disruption of both endocrine and local IGF-I action leads to neonatal lethality and impaired growth in various tissues including bone; however, the severity of growth and skeletal phenotype caused by disruption of endocrine IGF-I action is far less than with total IGF-I disruption. Based on these data and the fact that bone cells express IGF-I in high abundance, we and others predicted that locally produced IGF-I is also critical in regulating growth and bone accretion. To determine the role of local IGF-I, type 1alpha2 collagen-Cre mice were crossed with IGF-I loxP mice to generate Cre+ (conditional mutant) and Cre- (control) loxP homozygous mice. Surprisingly, approximately 40-50% of the conditional mutants died at birth, which is similar to total IGF-I disruption, but not observed in mice lacking circulating IGF-I. Expression of IGF-I in bone and muscle but not liver and brain was significantly decreased in the conditional mutant. Accordingly, circulating levels of serum IGF-I were also not affected. Disruption of local IGF-I dramatically reduced body weight 28-37%, femur areal bone mineral density 10-25%, and femur bone size 18-24% in growing mice. In addition, mineralization was reduced as early as during embryonic development. Consistently, histomorphometric analysis determined impaired osteoblast function as demonstrated by reduced mineral apposition rate (14-30%) and bone formation rate (35-57%). In conclusion, both local and endocrine IGF-I actions are involved in regulating growth of various tissues including bone, but they act via different mechanisms.
Collapse
Affiliation(s)
- Kristen E Govoni
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, 11201 Benton Street, Loma Linda, CA 92357, USA
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
The insulin-like growth factors (IGFs) play a central role in controlling somatic growth in mammals and exert anabolic effects on most tissues, including bone. IGF action is mediated by the IGF-I receptor and additionally is regulated by six high-affinity IGF binding proteins (IGFBP-1 through IGFBP-6), of which IGFBP-4 and IGFBP-5 are most abundant in bone. The focus of this brief review is on the role of IGFBP-5 in bone biology. IGFBP-5 has been implicated as a pro-osteogenic factor in several studies but conversely has been shown to act as an inhibitor of bone formation, primarily by interfering with IGF actions on osteoblasts. These potentially contradictory effects of IGFBP-5 in bone are further complicated by observations indicating that IGFBP-5 additionally may function in an IGF-independent way, and may have been accentuated by differences in both experimental design and methodology among published studies. Suggestions are made for a more systematic approach to help discern the true roles of IGFBP-5 in bone physiology.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | | |
Collapse
|
48
|
Hoeflich A, Götz W, Lichanska AM, Bielohuby M, Tönshoff B, Kiepe D. Effects of insulin-like growth factor binding proteins in bone -- a matter of cell and site. Arch Physiol Biochem 2007; 113:142-53. [PMID: 17922310 DOI: 10.1080/13813450701531193] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The actions of the insulin-like growth factor (IGF)-system are controlled by six IGF-binding proteins (IGFBPs). The IGFBPs are thought to affect local effects of IGF-I and IGF-II due to higher affinity if compared to IGF-I receptors and due to cell-type specific IGFBP expression patterns. It was found in IGFBP knockout models that the IGFBP family is functionally redundant. Thus, functional analysis of potential effects of IGFBPs is dependent on descriptive studies and models of IGFBP overexposure in vitro and in vivo. In the literature, the role of the IGFBPs for bone growth is highly controversial and, to date, no systematic look has been taken at IGFBPs resolving functional aspects of IGFBPs at levels of cell types and specific locations within bones. Since IGFBPs are thought to represent local modulators of the IGF actions and also exert IGF-independent effects, this approach is particularly reasonable on a physiological level. By sorting the huge number of in part controversial results on IGFBP effects in bone present in the literature for distinct cell types and bone sites it is possible to generate a focused, more specific and a less controversial picture of IGFBP functions in bone.
Collapse
Affiliation(s)
- A Hoeflich
- Laboratory of Mouse Genetics, Research Unit of Genetics and Biometry, Research Institute for the Biology of Farm Animals Dummerstorf (FBN), Germany.
| | | | | | | | | | | |
Collapse
|
49
|
Govoni KE, Lee SK, Chung YS, Behringer RR, Wergedal JE, Baylink DJ, Mohan S. Disruption of insulin-like growth factor-I expression in type IIalphaI collagen-expressing cells reduces bone length and width in mice. Physiol Genomics 2007; 30:354-62. [PMID: 17519362 PMCID: PMC2925693 DOI: 10.1152/physiolgenomics.00022.2007] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
It is well established that insulin-like growth factor (IGF)-I is critical for the regulation of peak bone mineral density (BMD) and bone width. However, the role of systemic vs. local IGF-I is not well understood. To determine the role local IGF-I plays in regulating BMD and bone width, we crossed IGF-I flox/flox mice with procollagen, typeIIalphaI-Cre mice to generate conditional mutants in which chondrocyte-derived IGF-I was disrupted. Bone parameters were measured by dual X-ray absorptiometry at 2, 4, 8, and 12 wk of age and peripheral quantitative computed tomography at 12 wk of age. Body length, areal BMD, and bone mineral content (BMC) were reduced (P < 0.05) between 4 and 12 wk in the conditional mutant mice. Bone width was reduced 7% in the vertebrae and femur (P < 0.05) of conditional mutant mice at 12 wk. Gains in body length and total body BMC and BMD were reduced by 27, 22, and 18%, respectively (P < 0.05) in conditional mutant mice between 2 and 4 wk of age. Expression of parathyroid hormone related protein, parathyroid hormone receptor, distal-less homeobox (Dlx)-5, SRY-box containing gene-9, and IGF binding protein (IGFBP)-5 were reduced 27, 36, 45, 33, and 45%, respectively, in the conditional mutant cartilage (P < 0.05); however, no changes in Indian hedgehog, Dlx-3, growth hormone receptor, IGF-I receptor, and IGFBP-3 expression were observed (P > or = 0.20). In conclusion, IGF-I from cells expressing procollagen type IIalphaI regulates bone accretion that occurs during postnatal growth period.
Collapse
Affiliation(s)
- Kristen E Govoni
- Jerry L. Pettis Veterans Affairs Medical Center and Loma Linda University, Loma Linda, California 92357, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Yasuoka H, Zhou Z, Pilewski JM, Oury TD, Choi AMK, Feghali-Bostwick CA. Insulin-like growth factor-binding protein-5 induces pulmonary fibrosis and triggers mononuclear cellular infiltration. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:1633-42. [PMID: 17071587 PMCID: PMC1780193 DOI: 10.2353/ajpath.2006.060501] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have recently shown that insulin-like growth factor-binding protein (IGFBP)-5 is overexpressed in idiopathic pulmonary fibrosis lung tissues and increases collagen and fibronectin deposition. Here, we further examined the effect of IGFBP-5 in vivo by intratracheal administration of replication-deficient adenovirus expressing human IGFBP-5 (Ad5), IGFBP-3 (Ad3), or no cDNA (cAd) to wild-type mice. Increased cellular infiltration and extracellular matrix deposition were observed in mice after Ad5 administration compared with Ad3 and cAd. Mononuclear cell infiltration consisted predominantly of T lymphocytes at day 8. By day 14, the number of infiltrating T cells decreased, whereas that of B cells and monocytes/macrophages increased. IGFBP-5 also induced migration of peripheral blood mononuclear cells in vitro, suggesting that in vivo mononuclear cell infiltration may be the direct result of IGFBP-5 expression. alpha-Smooth muscle actin and Mucin-1 co-localized in cells of mice treated with Ad5, suggesting that IGFBP-5 induced epithelial-mesenchymal transition. In addition, exogenous IGFBP-5 induced alpha-smooth muscle actin expression in primary fibroblasts and epithelial-mesenchymal transition of pulmonary epithelial cells in vitro. In conclusion, our results suggest that overexpression of IGFBP-5 in mouse lung results in fibroblast activation, increased extracellular matrix deposition, and myofibroblastic changes. Thus, the IGFBP-5-induced fibrotic phenotype in vivo may represent a novel model to better understand the pathogenesis of fibrosis.
Collapse
Affiliation(s)
- Hidekata Yasuoka
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|