1
|
Vahidi G, Moody M, Welhaven HD, Davidson L, Rezaee T, Behzad R, Karim L, Roggenbeck BA, Walk ST, Martin SA, June RK, Heveran CM. Germ-Free C57BL/6 Mice Have Increased Bone Mass and Altered Matrix Properties but Not Decreased Bone Fracture Resistance. J Bone Miner Res 2023; 38:1154-1174. [PMID: 37221143 PMCID: PMC10530360 DOI: 10.1002/jbmr.4835] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023]
Abstract
The gut microbiome impacts bone mass, which implies a disruption to bone homeostasis. However, it is not yet clear how the gut microbiome affects the regulation of bone mass and bone quality. We hypothesized that germ-free (GF) mice have increased bone mass and decreased bone toughness compared with conventionally housed mice. We tested this hypothesis using adult (20- to 21-week-old) C57BL/6J GF and conventionally raised female and male mice (n = 6-10/group). Trabecular microarchitecture and cortical geometry were measured from micro-CT of the femur distal metaphysis and cortical midshaft. Whole-femur strength and estimated material properties were measured using three-point bending and notched fracture toughness. Bone matrix properties were measured for the cortical femur by quantitative back-scattered electron imaging and nanoindentation, and, for the humerus, by Raman spectroscopy and fluorescent advanced glycation end product (fAGE) assay. Shifts in cortical tissue metabolism were measured from the contralateral humerus. GF mice had reduced bone resorption, increased trabecular bone microarchitecture, increased tissue strength and decreased whole-bone strength that was not explained by differences in bone size, increased tissue mineralization and fAGEs, and altered collagen structure that did not decrease fracture toughness. We observed several sex differences in GF mice, most notably for bone tissue metabolism. Male GF mice had a greater signature of amino acid metabolism, and female GF mice had a greater signature of lipid metabolism, exceeding the metabolic sex differences of the conventional mice. Together, these data demonstrate that the GF state in C57BL/6J mice alters bone mass and matrix properties but does not decrease bone fracture resistance. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Ghazal Vahidi
- Department of Mechanical & Industrial Engineering; Montana State University, Bozeman MT 59717
| | - Maya Moody
- Department of Chemistry & Biochemistry; Montana State University, Bozeman MT 59717
| | - Hope D. Welhaven
- Department of Chemistry & Biochemistry; Montana State University, Bozeman MT 59717
| | - Leah Davidson
- Department of Chemical and Biological Engineering; University of Idaho, Moscow ID 83844
| | - Taraneh Rezaee
- Department of Bioengineering; University of Massachusetts, Dartmouth, MA 02747
| | - Ramina Behzad
- Department of Bioengineering; University of Massachusetts, Dartmouth, MA 02747
| | - Lamya Karim
- Department of Bioengineering; University of Massachusetts, Dartmouth, MA 02747
| | - Barbara A. Roggenbeck
- Department of Microbiology & Cell Biology, Montana State University; Bozeman MT 59717
| | - Seth T. Walk
- Department of Microbiology & Cell Biology, Montana State University; Bozeman MT 59717
| | - Stephan A. Martin
- Translational Biomarkers Core Laboratory; Center for American Indian and Rural Health Equity; Montana State University, Bozeman MT 59717
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering; Montana State University, Bozeman MT 59717
| | - Chelsea M. Heveran
- Department of Mechanical & Industrial Engineering; Montana State University, Bozeman MT 59717
| |
Collapse
|
2
|
Mun SH, Jastrzebski S, Kalinowski J, Zeng S, Oh B, Bae S, Eugenia G, Khan NM, Drissi H, Zhou P, Shin B, Lee S, Lorenzo J, Park‐Min K. Sexual Dimorphism in Differentiating Osteoclast Precursors Demonstrates Enhanced Inflammatory Pathway Activation in Female Cells. J Bone Miner Res 2021; 36:1104-1116. [PMID: 33567098 PMCID: PMC11140852 DOI: 10.1002/jbmr.4270] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Sexual dimorphism of the skeleton is well documented. At maturity, the male skeleton is typically larger and has a higher bone density than the female skeleton. However, the underlying mechanisms for these differences are not completely understood. In this study, we examined sexual dimorphism in the formation of osteoclasts between cells from female and male mice. We found that the number of osteoclasts in bones was greater in females. Similarly, in vitro osteoclast differentiation was accelerated in female osteoclast precursor (OCP) cells. To further characterize sex differences between female and male osteoclasts, we performed gene expression profiling of cultured, highly purified, murine bone marrow OCPs that had been treated for 3 days with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). We found that 125 genes were differentially regulated in a sex-dependent manner. In addition to genes that are contained on sex chromosomes, transcriptional sexual dimorphism was found to be mediated by genes involved in innate immune and inflammatory response pathways. Furthermore, the NF-κB-NFATc1 axis was activated earlier in female differentiating OCPs, which partially explains the differences in transcriptomic sexual dimorphism in these cells. Collectively, these findings identify multigenic sex-dependent intrinsic difference in differentiating OCPs, which results from an altered response to osteoclastogenic stimulation. In humans, these differences could contribute to the lower peak bone mass and increased risk of osteoporosis that females demonstrate relative to males. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Se Hwan Mun
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Sandra Jastrzebski
- Department of Medicine University of Connecticut Health Farmington CT USA
| | - Judy Kalinowski
- Department of Medicine University of Connecticut Health Farmington CT USA
| | - Steven Zeng
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Brian Oh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Seyeon Bae
- Department of Medicine Weill Cornell Medical College New York NY USA
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Giannopoulou Eugenia
- Biological Sciences Department New York City College of Technology, City University of New York Brooklyn NY USA
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Nazir M Khan
- Department of Orthopaedics School of Medicine, Emory University Atlanta GA USA
| | - Hicham Drissi
- Department of Orthopaedics School of Medicine, Emory University Atlanta GA USA
| | - Ping Zhou
- Feil Family Brain & Mind Research Institute (BMRI), Weill Cornell Medical College New York NY USA
| | - Bongjin Shin
- Center on Aging University of Connecticut Health Farmington CT USA
| | - Sun‐Kyeong Lee
- Center on Aging University of Connecticut Health Farmington CT USA
| | - Joseph Lorenzo
- Department of Orthopaedic Surgery University of Connecticut Health Farmington CT USA
- Department of Medicine University of Connecticut Health Farmington CT USA
| | - Kyung‐Hyun Park‐Min
- BCMB Allied Program Weill Cornell Graduate School of Medical Sciences New York NY USA
- Department of Medicine Weill Cornell Medical College New York NY USA
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| |
Collapse
|
3
|
Xi G, Demambro VE, D’Costa S, Xia SK, Cox ZC, Rosen CJ, Clemmons DR. Estrogen Stimulation of Pleiotrophin Enhances Osteoblast Differentiation and Maintains Bone Mass in IGFBP-2 Null Mice. Endocrinology 2020; 161:5805123. [PMID: 32168373 PMCID: PMC7069688 DOI: 10.1210/endocr/bqz007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 03/12/2020] [Indexed: 12/22/2022]
Abstract
Insulin-like growth factor binding protein-2 (IGFBP-2) stimulates osteoblast differentiation but only male Igfbp2 null mice have a skeletal phenotype. The trophic actions of IGFBP-2 in bone are mediated through its binding to receptor tyrosine phosphatase beta (RPTPβ). Another important ligand for RPTPβ is pleiotrophin (PTN), which also stimulates osteoblast differentiation. We determined the change in PTN and RPTPβ in Igfbp2-/- mice. Analysis of whole bone mRNA in wild-type and knockout mice revealed increased expression of Ptn. Rptpβ increased in gene-deleted animals with females having greater expression than males. Knockdown of PTN expression in osteoblasts in vitro inhibited differentiation, and addition of PTN to the incubation medium rescued the response. Estradiol stimulated PTN secretion and PTN knockdown blocked estradiol-stimulated differentiation. PTN addition to IGFBP-2 silenced osteoblast stimulated differentiation, and an anti-fibronectin-3 antibody, which inhibits PTN binding to RPTPβ, inhibited this response. Estrogen stimulated PTN secretion and downstream signaling in the IGFBP-2 silenced osteoblasts and these effects were inhibited with anti-fibronectin-3. Administration of estrogen to wild-type and Igfbp2-/- male mice stimulated an increase in both areal bone mineral density and trabecular bone volume fraction but the increase was significantly greater in the Igfbp2-/- animals. Estrogen also stimulated RPTPβ expression in the null mice. We conclude that loss of IGFBP-2 expression is accompanied by upregulation of PTN and RPTPβ expression in osteoblasts, that the degree of increase is greater in females due to estrogen secretion, and that this compensatory change may account for some component of the maintenance of normal bone mass in female mice.
Collapse
Affiliation(s)
- Gang Xi
- Department of Medicine UNC School of Medicine Chapel Hill, North Carolina
| | | | - Susan D’Costa
- Department of Medicine UNC School of Medicine Chapel Hill, North Carolina
| | - Shalier K Xia
- Department of Medicine UNC School of Medicine Chapel Hill, North Carolina
| | - Zach C Cox
- Department of Medicine UNC School of Medicine Chapel Hill, North Carolina
| | | | - David R Clemmons
- Department of Medicine UNC School of Medicine Chapel Hill, North Carolina
- Correspondence: David R. Clemmons, MD, CB#7170, 8024 Burnett-Womack, Division of Endocrinology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7170. E-mail:
| |
Collapse
|
4
|
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
|
5
|
Oh HS, Oh SK, Lee JS, Wu C, Lee SJ. Effects of l-arginine on growth hormone and insulin-like growth factor 1. Food Sci Biotechnol 2017; 26:1749-1754. [PMID: 30263714 DOI: 10.1007/s10068-017-0236-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 10/18/2022] Open
Abstract
l-Arginine has been reported to promote cellular and organismal growth. In this study, the effects of l-arginine on the expression of growth hormone (GH) and insulin-like growth factor 1 (IGF-1), the two key growth factors, are investigated in cultured GH3 pituitary epithelium and HepG2 cells, respectively. l-Arginine significantly induced the gene expression of GH and IGF-1 in GH3 pituitary epithelium and HepG2 hepatocytes respectively, and reduced IGF binding protein-1 gene expression in HepG2 cells assessed via quantitative polymerase chain reaction analysis. l-Arginine also significantly induced GH and IGF-1 hormone secretion from GH3 and HepG2 cells, respectively. In addition, the multi-target ELISA analysis conducted revealed that phosphorylation of p-38 MAPK, MEK, and JNK were significantly increased in HepG2 cells, suggesting l-arginine-induced activation of the MAPK signaling pathway. These results suggest that l-arginine promotes the synthesis and secretion of GH and IGF-1 in vitro and induces the MAPK signaling cascade in cultured hepatocytes.
Collapse
Affiliation(s)
- Hyun-Seok Oh
- 1Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Korea University, Seoul, 02841 Korea
| | - Se Kwan Oh
- 2Rice Research Division, National Institute of Crop Science, Rural Development Administration, Suwon, 441-857 Korea
| | - Jum Seek Lee
- 2Rice Research Division, National Institute of Crop Science, Rural Development Administration, Suwon, 441-857 Korea
| | - Chunyan Wu
- 1Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Korea University, Seoul, 02841 Korea
| | - Sung-Joon Lee
- 1Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Korea University, Seoul, 02841 Korea
| |
Collapse
|
6
|
Bornstein S, Moschetta M, Kawano Y, Sacco A, Huynh D, Brooks D, Manier S, Fairfield H, Falank C, Roccaro AM, Nagano K, Baron R, Bouxein M, Vary C, Ghobrial IM, Rosen CJ, Reagan MR. Metformin Affects Cortical Bone Mass and Marrow Adiposity in Diet-Induced Obesity in Male Mice. Endocrinology 2017; 158:3369-3385. [PMID: 28977604 PMCID: PMC5659683 DOI: 10.1210/en.2017-00299] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/21/2017] [Indexed: 01/15/2023]
Abstract
Obesity during maturation can affect the growing skeleton directly and indirectly, although these effects and the mechanisms behind them are not fully understood. Our objective was to determine how a high-fat diet with or without metformin treatment affects skeletal development. We also sought to characterize changes that occur in white adipose tissue, circulating metabolites, lipids, and gut microbiota. A diet-induced obesity C57BL/6J mouse model was used to test the effects of obesity and metformin on bone using bone histomorphometry and microcomputed tomography. Bone marrow adipose tissue was quantified with osmium tetroxide microcomputed tomography and histology. Dual-energy x-ray absorptiometry was used to analyze body composition. Hematoxylin and eosin staining was used to assess changes in white adipose depots, mass spectrometry was used for circulating lipids and protein metabolite analysis, and ribosomal RNA sequencing was used for gut microbiome analysis. Mice fed a high fat-diet since wean displayed increased medullary areas and decreased osteoblast numbers in the long bones; this phenotype was partially normalized by metformin. Marrow and inguinal adipose expansion was also noted in obese mice, and this was partially normalized by metformin. A drug-by-diet interaction was noted for circulating lipid molecules, protein metabolites, and gut microbiome taxonomical units. Obesity was not detrimental to trabecular bone in growing mice, but bone marrow medullary expansion was observed, likely resulting from inhibition of osteoblastogenesis, and this was partially reversed by metformin treatment.
Collapse
Affiliation(s)
- Sheila Bornstein
- Maine Medical Center Research Institute, Scarborough, Maine 04074
| | | | - Yawara Kawano
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | - Antonio Sacco
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115
- Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Progettazione Ricerca Clinica e Studi di Fase I, Laboratorio Centro Ricerca oncoEmatologica AIL, Brescia, BS, Italy
| | - Daisy Huynh
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | - Daniel Brooks
- Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
- Center for Skeletal Research, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Salomon Manier
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | - Heather Fairfield
- Maine Medical Center Research Institute, Scarborough, Maine 04074
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine 04469
- Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, Maine 04074
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine 04469
- Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Aldo M. Roccaro
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115
- Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Progettazione Ricerca Clinica e Studi di Fase I, Laboratorio Centro Ricerca oncoEmatologica AIL, Brescia, BS, Italy
| | - Kenichi Nagano
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard Medical School, Boston, Massachusetts 02115
| | - Roland Baron
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard Medical School, Boston, Massachusetts 02115
| | - Mary Bouxein
- Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
- Center for Skeletal Research, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Calvin Vary
- Maine Medical Center Research Institute, Scarborough, Maine 04074
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine 04469
- Tufts University School of Medicine, Boston, Massachusetts 02111
| | | | - Clifford J. Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 04074
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine 04469
- Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Michaela R. Reagan
- Maine Medical Center Research Institute, Scarborough, Maine 04074
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine 04469
- Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
7
|
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
|
8
|
Kristianto J, Litscher SJ, Johnson MG, Patel F, Patel M, Fisher J, Zastrow RK, Radcliff AB, Blank RD. Congenic Strains Confirm the Pleiotropic Effect of Chromosome 4 QTL on Mouse Femoral Geometry and Biomechanical Performance. PLoS One 2016; 11:e0148571. [PMID: 26849124 PMCID: PMC4743951 DOI: 10.1371/journal.pone.0148571] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/19/2016] [Indexed: 11/19/2022] Open
Abstract
A pleiotropic quantitative trait locus (QTL) for bone geometry and mechanical performance in mice was mapped to distal chromosome 4 via an intercross of recombinant congenic mice HcB-8 and HcB-23. To study the QTL in isolation, we have generated C3H.B10-(rs6355453-rs13478087) (C.B.4.3) and C3H.B10-(rs6369860-D4Mit170) (C.B.4.2) congenic strains that harbor ~20 Mb and ~3 Mb, respectively, of chromosome 4 overlapping segments from C57BL/10ScSnA (B10) within the locus on a C3H/DiSnA (C3H) background. Using 3-point bend testing and standard beam equations, we phenotyped these mice for femoral mid-diaphyseal geometry and biomechanical performance. We analyzed the results via 2-way ANOVA, using sex and genotype as factors. In the C.B.4.3 strain, we found that homozygous B10/B10 male mice had smaller cross sectional area (CSA) and reduced total displacement than homozygous C3H/C3H mice. Sex by genotype interaction was also observed for maximum load and stiffness for C3H/C3H and B10/B10 mice, respectively. In C.B.4.2 strain, we found that homozygous B10/B10 mice had lower total displacement, post-yield displacement (PYD), stiffness, yield load and maximum load than mice harboring C3H allele. Sex by genotype interaction was observed in B10/B10 mice for perimeter, outer minor axis (OMA) and CSA. There were no significant differences in tissue level mechanical performance, which suggest that the QTL acts primarily on circumferential bone size. These data confirm the prior QTL mapping data and support other work demonstrating the importance of chromosome 4 QTL on bone modeling and bone responses to mechanical loading.
Collapse
Affiliation(s)
- Jasmin Kristianto
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| | - Suzanne J. Litscher
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michael G. Johnson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Forum Patel
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mital Patel
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jacqueline Fisher
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ryley K. Zastrow
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail B. Radcliff
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Robert D. Blank
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
- Milwaukee VA Medical Center, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
9
|
DeMambro VE, Le PT, Guntur AR, Maridas DE, Canalis E, Nagano K, Baron R, Clemmons DR, Rosen CJ. Igfbp2 Deletion in Ovariectomized Mice Enhances Energy Expenditure but Accelerates Bone Loss. Endocrinology 2015; 156:4129-40. [PMID: 26230658 PMCID: PMC4606757 DOI: 10.1210/en.2014-1452] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Previously, we reported sexually dimorphic bone mass and body composition phenotypes in Igfbp2(-/-) mice (-/-), where male mice exhibited decreased bone and increased fat mass, whereas female mice displayed increased bone but no changes in fat mass. To investigate the interaction between IGF-binding protein (IGFBP)-2 and estrogen, we subjected Igfbp2 -/- and +/+ female mice to ovariectomy (OVX) or sham surgery at 8 weeks of age. At 20 weeks of age, mice underwent metabolic cage analysis and insulin tolerance tests before killing. At harvest, femurs were collected for microcomputed tomography, serum for protein levels, brown adipose tissue (BAT) and inguinal white adipose tissue (IWAT) adipose depots for histology, gene expression, and mitochondrial respiration analysis of whole tissue. In +/+ mice, serum IGFBP-2 dropped 30% with OVX. In the absence of IGFBP-2, OVX had no effect on preformed BAT; however, there was significant "browning" of the IWAT depot coinciding with less weight gain, increased insulin sensitivity, lower intraabdominal fat, and increased bone loss due to higher resorption and lower formation. Likewise, after OVX, energy expenditure, physical activity and BAT mitochondrial respiration were decreased less in the OVX-/- compared with OVX+/+. Mitochondrial respiration of IWAT was reduced in OVX+/+ yet remained unchanged in OVX-/- mice. These changes were associated with significant increases in Fgf21 and Foxc2 expression, 2 proteins known for their insulin sensitizing and browning of WAT effects. We conclude that estrogen deficiency has a profound effect on body and bone composition in the absence of IGFBP-2 and may be related to changes in fibroblast growth factor 21.
Collapse
Affiliation(s)
- Victoria E DeMambro
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Phuong T Le
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Anyonya R Guntur
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - David E Maridas
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Ernesto Canalis
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Kenichi Nagano
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Roland Baron
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - David R Clemmons
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| | - Clifford J Rosen
- Maine Medical Center Research Institute (V.E.M., P.T.L., A.R.G., D.E.M., C.J.R.), Scarborough, Maine 04074; Departments of Orthopedic Surgery and Medicine (E.C.), University of Connecticut Health Center, Farmington, Connecticut 06032; Department of Medicine (K.N., R.B.), Harvard Medical School and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, Massachusetts 02115; and University of North Carolina (D.R.C.), Chapel Hill, North Carolina 27514
| |
Collapse
|
10
|
Stohn JP, Wang Q, Siviski ME, Kennedy K, Jin YR, Kacer D, DeMambro V, Liaw L, Vary CP, Rosen CJ, Prudovsky I, Lindner V. Cthrc1 controls adipose tissue formation, body composition, and physical activity. Obesity (Silver Spring) 2015; 23:1633-42. [PMID: 26148471 PMCID: PMC4509980 DOI: 10.1002/oby.21144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE This study investigated the effects of loss of Cthrc1 on adipogenesis, body composition, metabolism, physical activity, and muscle physiology. METHODS Complete metabolic and activity monitoring as well as grip strength measurements and muscle myography was performed in Cthrc1 null and wildtype mice. RESULTS Compared to wildtypes, Cthrc1 null mice had similar body weights but significantly reduced energy expenditure, decreased lean mass, and increased fat mass, especially visceral fat. In vitro studies demonstrated that Cthrc1 inhibited adipocyte differentiation as well as PPAR and CREB reporter activity, while preadipocytes isolated from Cthrc1 null mice exhibited enhanced adipogenic differentiation. Voluntary physical activity in Cthrc1 null mice as assessed by wheel running was reduced to approximately half the distance covered by wildtypes. Reduced grip strength was observed in Cthrc1 null mice at the age of 15 weeks or older with reduced performance and mass of hyphenate muscle. In the brain, Cthrc1 expression was most prominent in neurons of thalamic and hypothalamic nuclei with evidence for secretion into the circulation in the median eminence. CONCLUSIONS Our data indicate that Cthrc1 regulates body composition through inhibition of adipogenesis. In addition, central Cthrc1 may be a mediator of muscle function and physical activity.
Collapse
Affiliation(s)
- J Patrizia Stohn
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Qiaozeng Wang
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Matthew E Siviski
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Kevin Kennedy
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Yong-Ri Jin
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Doreen Kacer
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Victoria DeMambro
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Lucy Liaw
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Calvin P Vary
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Clifford J Rosen
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Igor Prudovsky
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Volkhard Lindner
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| |
Collapse
|
11
|
White JR, Confides AL, Moore-Reed S, Hoch JM, Dupont-Versteegden EE. Regrowth after skeletal muscle atrophy is impaired in aged rats, despite similar responses in signaling pathways. Exp Gerontol 2015; 64:17-32. [PMID: 25681639 DOI: 10.1016/j.exger.2015.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/14/2015] [Accepted: 02/10/2015] [Indexed: 01/07/2023]
Abstract
Skeletal muscle regrowth after atrophy is impaired in the aged and in this study we hypothesized that this can be explained by a blunted response of signaling pathways and cellular processes during reloading after hind limb suspension in muscles from old rats. Male Brown Norway Fisher 344 rats at 6 (young) and 32 (old) months of age were subjected to normal ambulatory conditions (amb), hind limb suspension for 14 days (HS), and HS followed by reloading through normal ambulation for 14 days (RE); soleus muscles were used for analysis of intracellular signaling pathways and cellular processes. Soleus muscle regrowth was blunted in old compared to young rats which coincided with a recovery of serum IGF-1 and IGFBP-3 levels in young but not old. However, the response to reloading for p-Akt, p-p70s6k and p-GSK3β protein abundance was similar between muscles from young and old rats, even though main effects for age indicate an increase in activation of this protein synthesis pathway in the aged. Similarly, MAFbx mRNA levels in soleus muscle from old rats recovered to the same extent as in the young, while Murf-1 was unchanged. mRNA abundance of autophagy markers Atg5 and Atg7 showed an identical response in muscle from old compared to young rats, but beclin did not. Autophagic flux was not changed at either age at the measured time point. Apoptosis was elevated in soleus muscle from old rats particularly with HS, but recovered in HSRE and these changes were not associated with differences in caspase-3, -8 or -9 activity in any group. Protein abundance of apoptosis repressor with caspase-recruitment domain (ARC), cytosolic EndoG, as well as cytosolic and nuclear apoptosis inducing factor (AIF) were lower in muscle from old rats, and there was no age-related difference in the response to atrophy or regrowth. Soleus muscles from old rats had a higher number of ED2 positive macrophages in all groups and these decreased with HS, but recovered in HSRE in the old, while no changes were observed in the young. Pro-inflammatory cytokines in serum did not show a differential response with age to different loading conditions. Results indicate that at the measured time point the impaired skeletal muscle regrowth after atrophy in aged animals is not associated with a general lack of responsiveness to changes in loading conditions.
Collapse
Affiliation(s)
- Jena R White
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S Limestone, Lexington, KY 40536-0200, USA
| | - Amy L Confides
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S Limestone, Lexington, KY 40536-0200, USA
| | - Stephanie Moore-Reed
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S Limestone, Lexington, KY 40536-0200, USA
| | - Johanna M Hoch
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S Limestone, Lexington, KY 40536-0200, USA
| | - Esther E Dupont-Versteegden
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S Limestone, Lexington, KY 40536-0200, USA.
| |
Collapse
|
12
|
Smith SS, Kessler CB, Shenoy V, Rosen CJ, Delany AM. IGF-I 3' untranslated region: strain-specific polymorphisms and motifs regulating IGF-I in osteoblasts. Endocrinology 2013; 154:253-62. [PMID: 23183171 PMCID: PMC3529377 DOI: 10.1210/en.2012-1476] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 11/06/2012] [Indexed: 12/12/2022]
Abstract
Reduced IGF-I is associated with low bone mass in humans and mice. C3H/He/J (C3H) mice have higher skeletal IGF-I and greater bone mass than C57BL/6J (B6). We hypothesized that strain-related genotypic differences in Igf1 affected skeletal function. The Igf1 coding region is nonpolymorphic, but its 3' untranslated region (UTR) is polymorphic between C3H and B6. Luciferase-Igf1 3' UTR reporter constructs showed that these polymorphic regions did not affect UTR function. IGF-I splice variants give rise to a common mature IGF-I peptide, but different E peptides. We identified two splice products, exon 4+6 (Ea) and exon 4+5+6 (Eb, mechano-growth factor) and found that their abundance was unchanged during osteoblastic differentiation. The Igf1 3' UTR encoded by exon 6 contains alternative polyadenylation sites. Proximal site use produces a short 3' UTR of approximately 195 bases, whereas distal site usage results in an approximately 6300-base UTR. Although Igf1 mRNA levels did not change during osteoblastic differentiation, distal polyadenylation site usage was increased in B6 cells but not in C3H. The resulting long Igf1 RNA isoform is less stable and has decreased translation efficiency, which may be one mechanism contributing to decreased IGF-I in B6 vs. C3H mice. Although the long UTR contains a conserved [GU](18) repeat, which is a positive regulator of UTR activity, it is also targeted by negative regulators, miR-29 and miR-365. These microRNAs are increased in B6 and C3H cells during osteoblastic differentiation. Differential expression of the long Igf1 3' UTR isoform may be a possible mechanism for enhanced IGF-I regulation in B6 vs. C3H mice.
Collapse
Affiliation(s)
- Spenser S Smith
- Center for Molecular Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | | | | | | | | |
Collapse
|
13
|
Le P, Kawai M, Bornstein S, DeMambro VE, Horowitz MC, Rosen CJ. A high-fat diet induces bone loss in mice lacking the Alox5 gene. Endocrinology 2012; 153:6-16. [PMID: 22128029 PMCID: PMC3249675 DOI: 10.1210/en.2011-0082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
5-Lipoxygenase catalyzes leukotriene generation from arachidonic acid. The gene that encodes 5-lipoxygenase, Alox5, has been identified in genome-wide association and mouse Quantitative Trait Locus studies as a candidate gene for obesity and low bone mass. Thus, we tested the hypothesis that Alox5(-/-) mice would exhibit metabolic and skeletal changes when challenged by a high-fat diet (HFD). On a regular diet, Alox5(-/-) mice did not differ in total body weight, percent fat mass, or bone mineral density compared with wild-type (WT) controls (P < 0.05). However, when placed on a HFD, Alox5(-/-) gained more fat mass and lost greater areal bone mass vs. WT (P < 0.05). Microarchitectural analyses revealed that on a HFD, WT showed increases in cortical area (P < 0.01) and trabecular thickness (P < 0.01), whereas Alox5(-/-) showed no change in cortical parameters but a decrease in trabecular number (P < 0.05) and bone volume fraction compared with WT controls (P < 0.05). By histomorphometry, a HFD did not change bone formation rates of either strain but produced an increase in osteoclast number per bone perimeter in Alox5(-/-) mice (P < 0.03). In vitro, osteoclastogenesis of marrow stromal cells was enhanced in mutant but not WT mice fed a HFD. Gene expression for Rankl, Pparg, and Cox-2 was greater in the femur of Alox5(-/-) than WT mice on a HFD (P < 0.01), but these increases were suppressed in the Alox5(-/-) mice after 8 wk of treatment with celecoxib, a cyclooxygenase-2 inhibitor. In sum, there is a strong gene by environmental interaction for bone mass when mice lacking the Alox5 gene are fed a HFD.
Collapse
Affiliation(s)
- Phuong Le
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074-7205, USA
| | | | | | | | | | | |
Collapse
|
14
|
Bokov AF, Garg N, Ikeno Y, Thakur S, Musi N, DeFronzo RA, Zhang N, Erickson RC, Gelfond J, Hubbard GB, Adamo ML, Richardson A. Does reduced IGF-1R signaling in Igf1r+/- mice alter aging? PLoS One 2011; 6:e26891. [PMID: 22132081 PMCID: PMC3223158 DOI: 10.1371/journal.pone.0026891] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 10/05/2011] [Indexed: 12/21/2022] Open
Abstract
Mutations in insulin/IGF-1 signaling pathway have been shown to lead to increased longevity in various invertebrate models. Therefore, the effect of the haplo- insufficiency of the IGF-1 receptor (Igf1r+/−) on longevity/aging was evaluated in C57Bl/6 mice using rigorous criteria where lifespan and end-of-life pathology were measured under optimal husbandry conditions using large sample sizes. Igf1r+/− mice exhibited reductions in IGF-1 receptor levels and the activation of Akt by IGF-1, with no compensatory increases in serum IGF-1 or tissue IGF-1 mRNA levels, indicating that the Igf1r+/− mice show reduced IGF-1 signaling. Aged male, but not female Igf1r+/− mice were glucose intolerant, and both genders developed insulin resistance as they aged. Female, but not male Igf1r+/− mice survived longer than wild type mice after lethal paraquat and diquat exposure, and female Igf1r+/− mice also exhibited less diquat-induced liver damage. However, no significant difference between the lifespans of the male Igf1r+/− and wild type mice was observed; and the mean lifespan of the Igf1r+/− females was increased only slightly (less than 5%) compared to wild type mice. A comprehensive pathological analysis showed no significant difference in end-of-life pathological lesions between the Igf1r+/− and wild type mice. These data show that the Igf1r+/− mouse is not a model of increased longevity and delayed aging as predicted by invertebrate models with mutations in the insulin/IGF-1 signaling pathway.
Collapse
Affiliation(s)
- Alex F. Bokov
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Neha Garg
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yuji Ikeno
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Geriatric Research, Education, and Clinical Center (GRECC), South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Sachin Thakur
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Geriatric Research, Education, and Clinical Center (GRECC), South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Ralph A. DeFronzo
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Ning Zhang
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Rebecca C. Erickson
- College of Natural Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Jon Gelfond
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Gene B. Hubbard
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Martin L. Adamo
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Arlan Richardson
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Geriatric Research, Education, and Clinical Center (GRECC), South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Mahalingam CD, Datta T, Patil RV, Kreider J, Bonfil RD, Kirkwood KL, Goldstein SA, Abou-Samra AB, Datta NS. Mitogen-activated protein kinase phosphatase 1 regulates bone mass, osteoblast gene expression, and responsiveness to parathyroid hormone. J Endocrinol 2011; 211:145-56. [PMID: 21852324 PMCID: PMC3783352 DOI: 10.1530/joe-11-0144] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parathyroid hormone (PTH) signaling via PTH 1 receptor (PTH1R) involves mitogen-activated protein kinase (MAPK) pathways. MAPK phosphatase 1 (MKP1) dephosphorylates and inactivates MAPKs in osteoblasts, the bone-forming cells. We previously showed that PTH1R activation in differentiated osteoblasts upregulates MKP1 and downregulates pERK1/2-MAPK and cyclin D1. In this study, we evaluated the skeletal phenotype of Mkp1 knockout (KO) mice and the effects of PTH in vivo and in vitro. Microcomputed tomography analysis of proximal tibiae and distal femora from 12-week-old Mkp1 KO female mice revealed osteopenic phenotype with significant reduction (8-46%) in bone parameters compared with wild-type (WT) controls. Histomorphometric analysis showed decreased trabecular bone area in KO females. Levels of serum osteocalcin (OCN) were lower and serum tartrate-resistant acid phosphatase 5b (TRAP5b) was higher in KO animals. Treatment of neonatal mice with hPTH (1-34) for 3 weeks showed attenuated anabolic responses in the distal femora of KO mice compared with WT mice. Primary osteoblasts derived from KO mice displayed delayed differentiation determined by alkaline phosphatase activity, and reduced expressions of Ocn and Runx2 genes associated with osteoblast maturation and function. Cells from KO females exhibited attenuated PTH response in mineralized nodule formation in vitro. Remarkably, this observation was correlated with decreased PTH response of matrix Gla protein expression. Expressions of pERK1/2 and cyclin D1 were inhibited dramatically by PTH in differentiated osteoblasts from WT mice but much less in osteoblasts from Mkp1 KO mice. In conclusion, MKP1 is important for bone homeostasis, osteoblast differentiation and skeletal responsiveness to PTH.
Collapse
Affiliation(s)
- Chandrika D Mahalingam
- Division of Endocrinology, Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Maile LA, DeMambro VE, Wai C, Aday AW, Capps BE, Beamer WG, Rosen CJ, Clemmons DR. An essential role for the association of CD47 to SHPS-1 in skeletal remodeling. J Bone Miner Res 2011; 26:2068-81. [PMID: 21638321 PMCID: PMC3383326 DOI: 10.1002/jbmr.441] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Integrin-associated protein (IAP/CD47) has been implicated in macrophage-macrophage fusion. To understand the actions of CD47 on skeletal remodeling, we compared Cd47(-/-) mice with Cd47(+/+) controls. Cd47(-/-) mice weighed less and had decreased areal bone mineral density compared with controls. Cd47(-/-) femurs were shorter in length with thinner cortices and exhibited lower trabecular bone volume owing to decreased trabecular number and thickness. Histomorphometry revealed reduced bone-formation and mineral apposition rates, accompanied by decreased osteoblast numbers. No differences in osteoclast number were observed despite a nonsignificant but 40% decrease in eroded surface/bone surface in Cd47(-/-) mice. In vitro, the number of functional osteoclasts formed by differentiating Cd47(-/-) bone marrow cells was significantly decreased compared with wild-type cultures and was associated with a decrease in bone-resorption capacity. Furthermore, by disrupting the CD47-SHPS-1 association, we found that osteoclastogenesis was markedly impaired. Assays for markers of osteoclast maturation suggested that the defect was at the point of fusion and not differentiation and was associated with a lack of SHPS-1 phosphorylation, SHP-1 phosphatase recruitment, and subsequent dephosphorylation of non-muscle cell myosin IIA. We also demonstrated a significant decrease in osteoblastogenesis in bone marrow stromal cells derived from Cd47(-/-) mice. Our finding of cell-autonomous defects in Cd47(-/-) osteoblast and osteoclast differentiation coupled with the pronounced skeletal phenotype of Cd47(-/-) mice support the conclusion that CD47 plays an important role in regulating skeletal acquisition and maintenance through its actions on both bone formation and bone resorption.
Collapse
Affiliation(s)
- Laura A Maile
- University of North Carolina at Chapel Hill, Division of Endocrinology
| | | | - Christine Wai
- University of North Carolina at Chapel Hill, Division of Endocrinology
| | - Ariel W Aday
- University of North Carolina at Chapel Hill, Division of Endocrinology
| | - Byron E Capps
- University of North Carolina at Chapel Hill, Division of Endocrinology
| | | | | | - David R Clemmons
- University of North Carolina at Chapel Hill, Division of Endocrinology
| |
Collapse
|
17
|
JIAO FENG, CHIU HANK, JIAO YAN, DE RIJK WALDEMARG, LI XINMIN, ECKSTEIN EUGENEC, BEAMER WESLEYG, GU WEIKUAN. Quantitative trait loci for tibial bone strength in C57BL/6J and C3H/HeJ inbred strains of mice. J Genet 2010; 89:21-7. [PMID: 20505243 PMCID: PMC3162481 DOI: 10.1007/s12041-010-0007-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Three-point bending technology has been widely used in the measurement of bone strength. Quantitative trait loci (QTLs) for bone strength have been identified using mouse femurs. In this study, we investigate the use of mouse tibiae in identification of QTLs that regulate bone strength. Mouse tibiae were from a F(2) population derived from C57BL/6J (B6) and C3H/HeJ (C3H). Three-point bending was measured using ISO 4049, with the support width adjustable to accommodate specimen sizes outside the scope of ISO 4049. The strain rate is selectable from 0.05 to 500 mm per min. All stress strain diagrams are recorded and retrieved in digital electronic form. Genome scan was performed in The Jackson Laboratory (TJL). QTL mapping was conducted using Map Manager QTX software. Data show that (i) both elastic modulus (stiffness) and maximum loading (strength) value appear as normal distributions, suggesting that multiple genetic factors control the bone strength; (ii) 11 QTLs, accounting for 90% of variation for strength, have been detected. More than half QTLs of three-point bending are located on the same locations of bone density earlier identified from mouse femurs; (iii) a major QTL of femoral and vertebral bone mineral density (BMD) was not detected for bone strength of tibiae; (iv) the QTL on chromosome 4 has extremely high LOD score of 31.8 and represents 60% of the variation of bone strength; and (v) four QTLs of stiffness (chromosomes 2, 11, 15 and 19) have been identified.
Collapse
Affiliation(s)
- FENG JIAO
- Department of Orthopedic Surgery-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - HANK CHIU
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA
| | - YAN JIAO
- Department of Orthopedic Surgery-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - WALDEMAR G. DE RIJK
- Department of Bioengineering, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - XINMIN LI
- Functional Genomics Facility, University of Chicago, Chicago, IL 60637, USA
| | - EUGENE C. ECKSTEIN
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA
| | | | - WEIKUAN GU
- Department of Orthopedic Surgery-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
18
|
Abstract
Cancellous bone decreases and bone marrow fat content increases with age. Osteoblasts and adipocytes are derived from a common precursor, and growth hormone (GH), a key hormone in integration of energy metabolism, regulates the differentiation and function of both cell lineages. Since an age-related decline in GH is associated with bone loss, we investigated the relationship between GH and bone marrow adiposity in hypophysectomized (HYPOX) rats and in mice with defects in GH signaling. HYPOX dramatically reduced body weight gain, bone growth and mineralizing perimeter, serum insulin-like growth factor 1 (IGF-1) levels, and mRNA levels for IGF-1 in liver and bone. Despite reduced body mass and adipocyte precursor pool size, HYPOX resulted in a dramatic increase in bone lipid levels, as reflected by increased bone marrow adiposity and bone triglyceride and cholesterol content. GH replacement normalized bone marrow adiposity and precursor pool size, as well as mineralizing perimeter in HYPOX rats. In contrast, 17beta -estradiol, IGF-1, thyroxine, and cortisone were ineffective. Parathyroid hormone (PTH) reversed the inhibitory effects of HYPOX on mineralizing perimeter but had no effect on adiposity. Finally, bone marrow adiposity was increased in mice deficient in GH and IGF-1 but not in mice deficient in serum IGF-1. Taken together, our findings indicate that the reciprocal changes in bone and fat mass in GH signaling-deficient rodents are not directly coupled with one another. Rather, GH enhances adipocyte as well as osteoblast precursor pool size. However, GH increases osteoblast differentiation while suppressing bone marrow lipid accumulation.
Collapse
|
19
|
DeMambro VE, Kawai M, Clemens TL, Fulzele K, Maynard JA, Marín de Evsikova C, Johnson KR, Canalis E, Beamer WG, Rosen CJ, Donahue LR. A novel spontaneous mutation of Irs1 in mice results in hyperinsulinemia, reduced growth, low bone mass and impaired adipogenesis. J Endocrinol 2010; 204:241-53. [PMID: 20032200 PMCID: PMC3033737 DOI: 10.1677/joe-09-0328] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A spontaneous mouse mutant, designated 'small' (sml), was recognized by reduced body size suggesting a defect in the IGF1/GH axis. The mutation was mapped to the chromosome 1 region containing Irs1, a viable candidate gene whose sequence revealed a single nucleotide deletion resulting in a premature stop codon. Despite normal mRNA levels in mutant and control littermate livers, western blot analysis revealed no detectable protein in mutant liver lysates. When compared with the control littermates, Irs1(sml)/Irs1(sml) (Irs1(sml/sml)) mice were small, lean, hearing impaired; had 20% less serum IGF1; were hyperinsulinemic; and were mildly insulin resistant. Irs1(sml/sml) mice had low bone mineral density, reduced trabecular and cortical thicknesses, and low bone formation rates, while osteoblast and osteoclast numbers were increased in the females but not different in the males compared with the Irs1(+/+) controls. In vitro, Irs1(sml/sml) bone marrow stromal cell cultures showed decreased alkaline phosphatase-positive colony forming units (pre-osteoblasts; CFU-AP+) and normal numbers of tartrate-resistant acid phosphatase-positive osteoclasts. Irs1(sml/sml) stromal cells treated with IGF1 exhibited a 50% decrease in AKT phosphorylation, indicative of defective downstream signaling. Similarities between engineered knockouts and the spontaneous mutation of Irs1(sml) were identified as well as significant differences with respect to heterozygosity and gender. In sum, we have identified a spontaneous mutation in the Irs1 gene associated with a major skeletal phenotype. Changes in the heterozygous Irs1(+)(/sml) mice raise the possibility that similar mutations in humans are associated with short stature or osteoporosis.
Collapse
|
20
|
Increased physical activity cosegregates with higher intake of carbohydrate and total calories in a subcongenic mouse strain. Mamm Genome 2009; 21:52-63. [PMID: 20033694 DOI: 10.1007/s00335-009-9243-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 12/02/2009] [Indexed: 12/30/2022]
Abstract
C57BL/6 J (B6) and CAST/EiJ (CAST), the inbred strain derived from M. musculus castaneus, differ in nutrient intake behaviors, including dietary fat and carbohydrate consumption in a two-diet-choice paradigm. Significant quantitative trait loci (QTLs) for carbohydrate (Mnic1) and total energy intake (Kcal2) are present between these strains on chromosome (Chr) 17. Here we report the refinement of the Chr 17 QTL in a subcongenic strain of the B6.CAST-( D17Mit19-D17Mit91 ) congenic mice described previously. This new subcongenic strain possesses CAST Chr 17 donor alleles from 4.8 to 45.4 Mb on a B6 background. Similar to CAST, the subcongenic mice exhibit increased carbohydrate and total calorie intake per body weight, while fat intake remains equivalent. Unexpectedly, this CAST genomic segment also confers two new physical activity phenotypes: 22% higher spontaneous physical activity levels and significantly increased voluntary wheel-running activity compared with the parental B6 strain. Overall, these data suggest that gene(s) involved in carbohydrate preference and increased physical activity are contained within the proximal region of Chr 17. Interval-specific microarray analysis in hypothalamus and skeletal muscle revealed differentially expressed genes within the subcongenic region, including neuropeptide W (Npw); glyoxalase I (Glo1); cytochrome P450, family 4, subfamily f, polypeptide 1 (Cyp4f15); phospholipase A2, group VII (Pla2g7); and phosphodiesterase 9a (Pde9a). This subcongenic strain offers a unique model for dissecting the contributions and possible interactions among genes controlling food intake and physical activity, key components of energy balance.
Collapse
|
21
|
Maddalozzo GF, Turner RT, Edwards CHT, Howe KS, Widrick JJ, Rosen CJ, Iwaniec UT. Alcohol alters whole body composition, inhibits bone formation, and increases bone marrow adiposity in rats. Osteoporos Int 2009; 20:1529-38. [PMID: 19238309 DOI: 10.1007/s00198-009-0836-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Accepted: 11/17/2008] [Indexed: 12/29/2022]
Abstract
UNLABELLED Chronic alcohol abuse is a risk factor for osteoporosis and sarcopenia, but the long-term effects of alcohol on the immature musculoskeletal system are less clear. The present investigation in growing rats was designed to determine the effects of alcohol consumption on body composition, muscle mass, and bone mass, architecture, and turnover. INTRODUCTION Few studies have focused on the long-term effects of drinking on bone and muscle during skeletal maturation. METHODS Alcohol was included in the diet of 4-week-old male Sprague-Dawley rats (35% caloric intake) for 3 months. The controls were fed an isocaloric alcohol-free liquid diet ad libitum. A second study was performed in which the controls were pair-fed to the alcohol-fed animals. RESULTS Compared to ad libitum-fed age-matched controls, alcohol-fed rats weighed less and had lower lean mass, fat mass, and percent body fat. In addition, they had lower slow- and fast-twitch muscle mass, lower total body bone mineral content and bone mineral density, and lower cancellous bone volume in the lumbar vertebra and proximal tibia. The effects of alcohol consumption on body composition were reduced when compared to the pair-fed control diet, indicating that caloric restriction was a comorbidity factor. In contrast, the effects of alcohol to decrease bone formation and serum leptin and IGF-I levels and to increase bone marrow adiposity appeared independent of caloric restriction. CONCLUSIONS The skeletal abnormalities in growing alcohol-fed rats were due to a combination of effects specific to alcohol consumption and alcohol-induced caloric restriction.
Collapse
Affiliation(s)
- G F Maddalozzo
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Mårtensson UEA, Salehi SA, Windahl S, Gomez MF, Swärd K, Daszkiewicz-Nilsson J, Wendt A, Andersson N, Hellstrand P, Grände PO, Owman C, Rosen CJ, Adamo ML, Lundquist I, Rorsman P, Nilsson BO, Ohlsson C, Olde B, Leeb-Lundberg LMF. Deletion of the G protein-coupled receptor 30 impairs glucose tolerance, reduces bone growth, increases blood pressure, and eliminates estradiol-stimulated insulin release in female mice. Endocrinology 2009; 150:687-98. [PMID: 18845638 DOI: 10.1210/en.2008-0623] [Citation(s) in RCA: 311] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In vitro studies suggest that the G protein-coupled receptor (GPR) 30 is a functional estrogen receptor. However, the physiological role of GPR30 in vivo is unknown, and it remains to be determined whether GPR30 is an estrogen receptor also in vivo. To this end, we studied the effects of disrupting the GPR30 gene in female and male mice. Female GPR30((-/-)) mice had hyperglycemia and impaired glucose tolerance, reduced body growth, increased blood pressure, and reduced serum IGF-I levels. The reduced growth correlated with a proportional decrease in skeletal development. The elevated blood pressure was associated with an increased vascular resistance manifested as an increased media to lumen ratio of the resistance arteries. The hyperglycemia and impaired glucose tolerance in vivo were associated with decreased insulin expression and release in vivo and in vitro in isolated pancreatic islets. GPR30 is expressed in islets, and GPR30 deletion abolished estradiol-stimulated insulin release both in vivo in ovariectomized adult mice and in vitro in isolated islets. Our findings show that GPR30 is important for several metabolic functions in female mice, including estradiol-stimulated insulin release.
Collapse
Affiliation(s)
- Ulrika E A Mårtensson
- Units of Drug Target Discovery, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ackert-Bicknell CL, Demissie S, Marín de Evsikova C, Hsu YH, DeMambro VE, Karasik D, Cupples LA, Ordovas JM, Tucker KL, Cho K, Canalis E, Paigen B, Churchill GA, Forejt J, Beamer WG, Ferrari S, Bouxsein ML, Kiel DP, Rosen CJ. PPARG by dietary fat interaction influences bone mass in mice and humans. J Bone Miner Res 2008; 23:1398-408. [PMID: 18707223 PMCID: PMC2683155 DOI: 10.1359/jbmr.080419] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 04/09/2008] [Accepted: 04/17/2008] [Indexed: 01/19/2023]
Abstract
Adult BMD, an important risk factor for fracture, is the result of genetic and environmental interactions. A quantitative trait locus (QTL) for the phenotype of volumetric BMD (vBMD), named Bmd8, was found on mid-distal chromosome (Chr) 6 in mice. This region is homologous to human Chr 3p25. The B6.C3H-6T (6T) congenic mouse was previously created to study this QTL. Using block haplotyping of the 6T congenic region, expression analysis in the mouse, and examination of nonsynonymous SNPs, peroxisome proliferator activated receptor gamma (Pparg) was determined to be the most likely candidate gene for the Bmd8 QTL of the 630 genes located in the congenic region. Furthermore, in the C3H/HeJ (C3H) strain, which is the donor strain for the 6T congenic, several polymorphisms were found in the Pparg gene. On challenge with a high-fat diet, we found that the 6T mouse has a lower areal BMD (aBMD) and volume fraction of trabecular bone (BV/TV%) of the distal femur compared with B6 mice. Interactions between SNPs in the PPARG gene and dietary fat for the phenotype of BMD were examined in the Framingham Offspring Cohort. This analysis showed that there was a similar interaction of the PPARG gene and diet (fat intake) on aBMD in both men and women. These findings suggest that dietary fat has a significant influence on BMD that is dependent on the alleles present for the PPARG gene.
Collapse
Affiliation(s)
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | | | - Yi-Hsiang Hsu
- Program for Population Genetics, Harvard School of Public Health, Boston, Massachusetts, USA
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Jose M Ordovas
- Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Katherine L Tucker
- Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Kelly Cho
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut, USA
- The University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | | | | | - Jiri Forejt
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Serge Ferrari
- Service of Bone Diseases, Department of Rehabilitation and Geriatrics, WHO Center for Osteoporosis Prevention, Geneva University Hospital, Geneva, Switzerland
| | - Mary L Bouxsein
- Orthopedic Biomechanics Laboratory, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Douglas P Kiel
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
24
|
Govoni KE, Donahue LR, Marden C, Mohan S. Complex genetic regulation of bone mineral density and insulin-like growth factor-I in C57BL/6J-Chr #A/J/NaJ chromosome substitution strains. Physiol Genomics 2008; 35:159-64. [PMID: 18682576 DOI: 10.1152/physiolgenomics.90203.2008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Low bone mineral density (BMD) is a phenotype associated with osteoporosis and increased risk of fracture. Since 60-80% of variation in BMD is associated with genetic factors, we used the novel approach of chromosome substitution strains (CSS) to identify chromosomes that harbor genes that regulate BMD. Specifically, we evaluated 24 wk old C57BL/6J-Chr #(A/J)/NaJ CSS (n = 7 to 18) in which each chromosome in the host C57BL/6J strain is replaced by a corresponding chromosome from the donor A/J strain (19 autosomes, X, Y). We determined several A/J chromosomes contribute to body weight (BW), percent body fat (BF), whole body areal BMD (aBMD), and serum insulin-like growth factor (IGF)-I in both a positive and negative manner when compared with C57BL/6J. Specifically, C57BL/6J-Chr 5(A/J)/NaJ (B.A-5) (males) and B.A-13 (females) contributed to increased BW, whereas B.A-3, 4, 8, 9, 12, and 18 (males) and B.A-3, 4, and 11 (females) contributed to reduced BW. B.A-5 (males) and B.A-13 (females) contributed to increased BF, whereas B.A-12 (males) and B.A-3, 4, 10, and 11 (females) contributed to reduced BF. B.A-14 (females) contributed to increased aBMD and B.A-1, 2, 6, 9, 10, and 18 (males) and B.A-8, 9, and 10 (females) contributed to reduced aBMD. To determine if similar chromosomes regulate aBMD and IGF-I, we determined serum concentrations of IGF-I. B.A-14 and Y (males) and B.A-6 (females) contributed to increased IGF-I and male B.A-3 and female B.A-8 contributed to reduced IGF-I. Overall, we identified several sex-dependent and -independent chromosomes that regulate aBMD and IGF-I in adult mice.
Collapse
Affiliation(s)
- K E Govoni
- Jerry L Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California 92357, USA
| | | | | | | |
Collapse
|
25
|
Garrett AJ, Rincon G, Medrano JF, Elzo MA, Silver GA, Thomas MG. Promoter region of the bovine growth hormone receptor gene: single nucleotide polymorphism discovery in cattle and association with performance in Brangus bulls. J Anim Sci 2008; 86:3315-23. [PMID: 18676722 DOI: 10.2527/jas.2008-0990] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Expression of the GH receptor (GHR) gene and its binding with GH is essential for growth and fat metabolism. A GT microsatellite exists in the promoter of bovine GHR segregating short (11 bp) and long (16 to 20 bp) allele sequences. To detect SNP and complete an association study of genotype to phenotype, we resequenced a 1,195-bp fragment of DNA including the GT microsatellite and exon 1A. Resequencing was completed in 48 familialy unrelated Holstein, Jersey, Brown Swiss, Simmental, Angus, Brahman, and Brangus cattle. Nine SNP were identified. Phylogeny analyses revealed minor distance (i.e., <5%) in DNA sequence among the 5 Bos taurus breeds; however, sequence from Brahman cattle averaged 27.4 +/- 0.07% divergence from the Bos taurus breeds, whereas divergence of Brangus was intermediate. An association study of genotype to phenotype was completed with data from growing Brangus bulls (n = 553 from 96 sires) and data from 4 of the SNP flanking the GT microsatellite. These SNP were found to be in Hardy-Weinberg equilibrium and in phase based on linkage disequilibrium analyses (r(2) = 0.84 and D'= 0.92). An A/G tag SNP was identified (ss86273136) and was located in exon 1A, which began 88 bp downstream from the GT microsatellite. Minor allele frequency of the tag SNP was greater than 10%, and Mendelian segregation was verified in 3 generation pedigrees. The A allele was derived from Brahman, and the G allele was derived from Angus. This tag SNP genotype was a significant effect in analyses of rib fat data collected with ultrasound when bulls were ~365 d of age. Specifically, bulls of the GG genotype had 6.1% more (P = 0.0204) rib fat than bulls of the AA and AG genotypes, respectively. Tag SNP (ss86273136), located in the promoter of GHR, appears to be associated with a measure of corporal fat in Bos taurus x Bos indicus composite cattle.
Collapse
Affiliation(s)
- A J Garrett
- New Mexico State University, Las Cruces 88003, USA
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
INTRODUCTION The metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A) functions to enhance local insulin-like growth factor (IGF)-I bioavailability through cleavage of inhibitory IGF binding proteins. Because IGF-I is an important regulator of skeletal growth and remodeling and PAPP-A is highly expressed by osteoblastic cells, we hypothesized that, in the absence of PAPP-A, bone physiology would be compromised because of a blunting of local IGF-I action even in the presence of normal circulating IGF-I levels. MATERIALS AND METHODS pQCT, muCT, histomorphometry, and mechanical strength testing were performed on bones from PAPP-A knockout (KO) mice and wildtype (WT) littermates at 2-12 mo of age. IGF-I levels and bone formation and resorption markers were determined in sera from these animals. RESULTS Volumetric BMD in PAPP-A KO mice measured by pQCT at the femoral midshaft, which is primarily cortical bone, was 10% less than WT at 2 mo. This difference was maintained at 4, 6, and 12 mo. Cortical thickness at this site was similarly decreased. On the other hand, trabecular bone at the distal femur (pQCT) and in the tibia (muCT) showed age-progressive decreases in bone volume fraction in PAPP-A KO compared with WT mice. Tibial muCT indicated a 46% relative decrease in trabecular bone volume/total volume (BV/TV) and a 28% relative decrease in trabecular thickness in PAPP-A KO compared with WT mice at 6 mo. These trabecular deficiencies in PAPP-A KO mice corresponded to a weakening of the bone. Serum markers and bone histomorphometry indicated that the primary impact of PAPP-A is on skeletal remodeling resulting in a state of low-turnover osteopenia in adult PAPP-A KO mice. Circulating IGF-I levels were not altered in PAPP-A KO mice. CONCLUSIONS PAPP-A is a bone growth regulatory factor in vivo and, in its absence, mice show skeletal insufficiency in mass, density, architecture, and strength. The data suggest a primary role for PAPP-A in modulating local IGF bioavailability for trabecular bone remodeling.
Collapse
|
27
|
DeMambro VE, Clemmons DR, Horton LG, Bouxsein ML, Wood TL, Beamer WG, Canalis E, Rosen CJ. Gender-specific changes in bone turnover and skeletal architecture in igfbp-2-null mice. Endocrinology 2008; 149:2051-61. [PMID: 18276763 PMCID: PMC2329262 DOI: 10.1210/en.2007-1068] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
IGF-binding protein-2 (IGFBP-2) is a 36-kDa protein that binds to the IGFs with high affinity. To determine its role in bone turnover, we compared Igfbp2(-/-) mice with Igfbp2(+/+) colony controls. Igfbp2(-/-) males had shorter femurs and were heavier than controls but were not insulin resistant. Serum IGF-I levels in Igfbp2(-/-) mice were 10% higher than Igfbp2(+/+) controls at 8 wk of age; in males, this was accompanied by a 3-fold increase in hepatic Igfbp3 and Igfbp5 mRNA transcripts compared with Igfbp2(+/+) controls. The skeletal phenotype of the Igfbp2(-/-) mice was gender and compartment specific; Igfbp2(-/-) females had increased cortical thickness with a greater periosteal circumference compared with controls, whereas male Igfbp2(-/-) males had reduced cortical bone area and a 20% reduction in the trabecular bone volume fraction due to thinner trabeculae than Igfbp2(+/+) controls. Serum osteocalcin levels were reduced by nearly 40% in Igfbp2(-/-) males, and in vitro, both CFU-ALP(+) preosteoblasts, and tartrate-resistant acid phosphatase-positive osteoclasts were significantly less abundant than in Igfbp2(+/+) male mice. Histomorphometry confirmed fewer osteoblasts and osteoclasts per bone perimeter and reduced bone formation in the Igfbp2(-/-) males. Lysates from both osteoblasts and osteoclasts in the Igfbp2(-/-) males had phosphatase and tensin homolog (PTEN) levels that were significantly higher than Igfbp2(+/+) controls and were suppressed by addition of exogenous IGFBP-2. In summary, there are gender- and compartment-specific changes in Igfbp2(-/-) mice. IGFBP-2 may regulate bone turnover in both an IGF-I-dependent and -independent manner.
Collapse
Affiliation(s)
- V E DeMambro
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Haston CK, Li W, Li A, Lafleur M, Henderson JE. Persistent Osteopenia in Adult Cystic Fibrosis Transmembrane Conductance Regulator–deficient Mice. Am J Respir Crit Care Med 2008; 177:309-15. [DOI: 10.1164/rccm.200705-659oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
29
|
Bibliography. Current world literature. Parathyroids, bone and mineral metabolism. Curr Opin Endocrinol Diabetes Obes 2007; 14:494-501. [PMID: 17982358 DOI: 10.1097/med.0b013e3282f315ef] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Léger J, Mercat I, Alberti C, Chevenne D, Armoogum P, Tichet J, Czernichow P. The relationship between the GH/IGF-I axis and serum markers of bone turnover metabolism in healthy children. Eur J Endocrinol 2007; 157:685-92. [PMID: 17984250 DOI: 10.1530/eje-07-0402] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
CONTEXT There is evidence to suggest that IGF-I plays a role in regulating bone turnover. OBJECTIVE To evaluate the relationships between serum concentrations of IGF-I and IGF-binding protein-3 (IGFBP-3), and bone metabolism markers in healthy children. DESIGN AND SETTING Prospective cross-sectional study. SUBJECTS AND METHODS A cohort of 579 boys and 540 girls, all healthy Caucasian, were included in this study. Serum IGF-I and IGFBP-3 concentrations, bone alkaline phosphatase (BAP) and CrossLaps (markers of bone formation and bone resorption respectively) levels were evaluated as a function of age, gender, pubertal stage and body mass index. RESULTS Serum IGF-I SDS levels were positively correlated with BAP and CrossLaps SDS levels before and after puberty, and also with CrossLaps during puberty (weak correlation). Serum IGFBP-3 SDS levels were positively correlated with BAP and CrossLaps levels before, during (weak correlation) and after puberty (for BAP levels only). CONCLUSIONS This study demonstrated the independent association between serum IGF-I and IGFBP-3 concentrations with both serum bone formation and resorption markers in healthy children. Physiological differences before, during and after puberty in the association of serum IGF-I and IGFBP-3 levels with the serum bone metabolism markers were found. These differences may be related to differences in interactions between sex steroid hormones and the GH/IGF-I system, bone metabolism and growth during the pubertal transition. Improvements in our understanding of life course determinants of the IGF-I system and bone metabolism are required to shed further light on the role of the GH/IGF-I axis in bone remodelling.
Collapse
Affiliation(s)
- Juliane Léger
- Pediatric Endocrinology Department, Centre de Référence des Maladies Endocriniennes Rares de la Croissance and INSERM Unit 690, Assistance Publique-Hôpitaux de Paris, Université Paris VII, Robert Debré Hospital, France.
| | | | | | | | | | | | | |
Collapse
|
31
|
Cao JJ, Kurimoto P, Boudignon B, Rosen C, Lima F, Halloran BP. Aging impairs IGF-I receptor activation and induces skeletal resistance to IGF-I. J Bone Miner Res 2007; 22:1271-9. [PMID: 17488198 DOI: 10.1359/jbmr.070506] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
UNLABELLED IGF-I plays an important anabolic role in stimulating bone formation and maintaining bone mass. We show that the pro-proliferative, anti-apoptotic, and functional responses to IGF-I in bone and BMSCs decrease with aging. These changes are associated with impaired receptor activation and signal transduction through the MAPK and PI3K pathways. INTRODUCTION IGF-I is a potent anabolic agent having effects across diverse tissues and cell types. With aging, bone becomes resistant to the anabolic actions of IGF-I. To examine the effects of aging on bone responsiveness to IGF-I, we measured the pro-proliferative, anti-apoptotic, and functional responses of bone and bone marrow stromal cells (BMSCs) to IGF-I and evaluated IGF-I signal transduction in young, adult, and old mice. MATERIALS AND METHODS Male C57BL/6 mice 6 wk (young), 6 mo (adult), and 24 mo (old) were treated with IGF-I for 2 wk using osmotic minipumps, and osteoblast proliferation (BrdU labeling) in vivo, and osteoprogenitor number (BMSC culture and calcium nodule formation) were measured. Proliferation, apoptosis, and expression of key osteoblast factors (alkaline phosphatase, collagen, osteocalcin, RANKL, osteoprotegerin (OPG), macrophage-colony stimulating factor [M-CSF]) and IGF-I signaling elements and their activation in IGF-I-treated cells were studied using QRT-PCR and Western blot analysis. Data were analyzed using ANOVA. RESULTS Aging decreased the basal and IGF-I-stimulated number of BrdU-labeled osteoblasts and reduced the ability of IGF-I to stimulate osteoprogenitor formation (calcium nodule number) by 50%. The pro-proliferative and anti-apoptotic actions of IGF-I were blunted in cells from old animals. These changes were accompanied by age-related alterations in the ability of IGF-I to regulate alkaline phosphatase, collagen, osteocalcin, RANKL, OPG, and M-CSF expression. IGF-I binding was normal, but IGF-I receptor mRNA and protein expression was increased in aged animals by 2- and 10-fold, respectively. The age-related changes in proliferation, apoptosis, and function were accompanied by loss of IGF-I-induced signaling at the receptor level and at key regulatory sites along the MAPK (ERK1/2) and PI3K (AKT) pathways. CONCLUSIONS Our data show that aging is accompanied by loss of bone and BMSC/osteoblast responsiveness to IGF-I and that these changes are associated with resistance to IGF-I signaling that involve receptor activation and downstream signaling events.
Collapse
Affiliation(s)
- Jay J Cao
- Division of Endocrinology, Veterans Affairs Medical Center, and Department of Medicine, University of California, San Francisco, California 94121, USA
| | | | | | | | | | | |
Collapse
|
32
|
Bibliography. Current world literature. Diabetes and the endocrine pancreas II. Curr Opin Endocrinol Diabetes Obes 2007; 14:329-57. [PMID: 17940461 DOI: 10.1097/med.0b013e3282c3a898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Jiao Y, Chiu H, Fan Z, Jiao F, Eckstein EC, Beamer WG, Gu W. Quantitative trait loci that determine mouse tibial nanoindentation properties in an F2 population derived from C57BL/6J x C3H/HeJ. Calcif Tissue Int 2007; 80:383-90. [PMID: 17551771 DOI: 10.1007/s00223-007-9030-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 03/29/2007] [Accepted: 03/29/2007] [Indexed: 01/17/2023]
Abstract
Use of nanoindentation technology to identify quantitative trait loci (QTL) that regulate bone properties represents a novel approach to improving our understanding of molecular mechanisms that control bone matrix properties. Tibiae for QTL mapping were from an F2 population derived from C57BL/6J and C3H/HeJ. A nanoindenter (Triboindenter; Hysitron, Minneapolis, MN) was used to conduct indentation tests on transverse sections. Genotyping was performed in The Jackson Laboratory. QTL mapping was conducted using software. We found that (1) tibiae from mice at 16 weeks of age were mature and suitable for measurement by a nanoindentor; (2) both stiffness modulus and hardness modulus in the F2 population appeared to have normal distributions, which suggested that multiple genetic factors control the bone properties; and (3) QTL for hardness were identified from five chromosomes (Chr 8, 12, 13, 17, and 19) and for stiffness, from four chromosomes (Chr 3, 8, 12, and 13). Among all detected QTL, one at the same location on Chr 12 was detected for both hardness and stiffness data. It explained the highest percentage of phenotypic variation in bone properties. Using nanoindentation technology to identify QTL that regulate bone properties yielded as many as six different chromosomal regions. Although the actual genes remain to be identified, nanoindentation will contribute to our understanding of molecular mechanisms and normal development processes that control the matrix properties of bone.
Collapse
Affiliation(s)
- Yan Jiao
- Department of Orthopedic Surgery-Campbell Clinic, University of Tennessee Health Science Center, A331 Coleman Building, 956 Court Avenue, Memphis, TN 38163, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Lecka-Czernik B, Ackert-Bicknell C, Adamo ML, Marmolejos V, Churchill GA, Shockley KR, Reid IR, Grey A, Rosen CJ. Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) by rosiglitazone suppresses components of the insulin-like growth factor regulatory system in vitro and in vivo. Endocrinology 2007; 148:903-11. [PMID: 17122083 PMCID: PMC1851001 DOI: 10.1210/en.2006-1121] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rosiglitazone (Rosi) belongs to the class of thiazolidinediones (TZDs) that are ligands for peroxisome proliferator-activated receptor gamma (PPARgamma). Stimulation of PPARgamma suppresses bone formation and enhances marrow adipogenesis. We hypothesized that activation of PPARgamma down-regulates components of the IGF regulatory system, leading to impaired osteoblast function. Rosi treatment (1 microm) of a marrow stromal cell line (UAMS-33) transfected with empty vector (U-33/c) or with PPARgamma2 (U-33/gamma2) were analyzed by microarray. Rosi reduced IGF-I, IGF-II, IGFBP-4, and the type I and II IGF receptor (IGF1R and IGF2R) expression at 72 h in U-33/gamma2 compared with U-33/c cells (P < 0.01); these findings were confirmed by RT-PCR. Rosi reduced secreted IGF-I from U-33/gamma2 cells by 75% (P < 0.05). Primary marrow stromal cells (MSCs) extracted from adult (8 months) and old (24 months) C57BL/6J (B6) mice were treated with Rosi (1 microm) for 48 h. IGF-I, IGFBP-4, and IGF1R transcripts were reduced in Rosi-treated MSCs compared with vehicle (P < 0.01) and secreted IGF-I was also suppressed (P < 0.05). B6 mice treated with Rosi (20 mg/kg.d) for short duration (i.e. 4 d), and long term (i.e. 7 wk) had reduced serum IGF-I; this was accompanied by markedly suppressed IGF-I transcripts in the liver and peripheral fat of treated animals. To determine whether Rosi affected circulating IGF-I in humans, we measured serum IGF-I, IGFBP-2, and IGFBP-3 at four time points in 50 postmenopausal women randomized to either Rosi (8 mg/d) or placebo. Rosi-treated subjects had significantly lower IGF-I at 8 wk than baseline (-25%, P < 0.05), and at 16 wk their levels were reduced 14% vs. placebo (P = 0.15). We conclude that Rosi suppresses IGF-I expression in bone and liver; these changes could affect skeletal acquisition through endocrine and paracrine pathways.
Collapse
Affiliation(s)
- B Lecka-Czernik
- St. Joseph Hospital, Maine Center for Osteoporosis Research and Education, 360 Broadway, Bangor, Maine 04401, USA
| | | | | | | | | | | | | | | | | |
Collapse
|