1
|
Parlato MB, Lee JS, Belair DG, Fontana G, Leiferman E, Hanna R, Chamberlain C, Ranheim EA, Murphy WL, Halanski MA. Subperiosteal delivery of transforming growth factor beta 1 and human growth hormone from mineralized PCL films. J Biomed Mater Res A 2024; 112:1578-1593. [PMID: 38530161 DOI: 10.1002/jbm.a.37684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/15/2024] [Accepted: 01/31/2024] [Indexed: 03/27/2024]
Abstract
The ability to locally deliver bioactive molecules to distinct regions of the skeleton may provide a novel means by which to improve fracture healing, treat neoplasms or infections, or modulate growth. In this study, we constructed single-sided mineral-coated poly-ε-caprolactone membranes capable of binding and releasing transforming growth factor beta 1 (TGF-β1) and human growth hormone (hGH). After demonstrating biological activity in vitro and characterization of their release, these thin bioabsorbable membranes were surgically implanted using an immature rabbit model. Membranes were circumferentially wrapped under the periosteum, thus placed in direct contact with the proximal metaphysis to assess its bioactivity in vivo. The direct effects on the metaphyseal bone, bone marrow, and overlying periosteum were assessed using radiography and histology. Effects of membrane placement at the tibial growth plate were assessed via physeal heights, tibial growth rates (pulsed fluorochrome labeling), and tibial lengths. Subperiosteal placement of the mineralized membranes induced greater local chondrogenesis in the plain mineral and TGF-β1 samples than the hGH. More exuberant and circumferential ossification was seen in the TGF-β1 treated tibiae. The TGF-β1 membranes also induced hypocellularity of the bone marrow with characteristics of gelatinous degeneration not seen in the other groups. While the proximal tibial growth plates were taller in the hGH treated than TGF-β1, no differences in growth rates or overall tibial lengths were found. In conclusion, these data demonstrate the feasibility of using bioabsorbable mineral coated membranes to deliver biologically active compounds subperiosteally in a sustained fashion to affect cells at the insertion site, bone marrow, and even growth plate.
Collapse
|
2
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
3
|
Wang Z, Zhu P, Liao B, You H, Cai Y. Effects and action mechanisms of individual cytokines contained in PRP on osteoarthritis. J Orthop Surg Res 2023; 18:713. [PMID: 37735688 PMCID: PMC10515001 DOI: 10.1186/s13018-023-04119-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Osteoarthritis (OA) is defined as a degenerative joint disease that can affect all tissues of the joint, including the articular cartilage, subchondral bone, ligaments capsule, and synovial membrane. The conventional nonoperative treatments are ineffective for cartilage repair and induce only symptomatic relief. Platelet-rich plasma (PRP) is a platelet concentrate derived from autologous whole blood with a high concentration of platelets, which can exert anti-inflammatory and regenerative effects by releasing multiple growth factors and cytokines. Recent studies have shown that PRP exhibits clinical benefits in patients with OA. However, high operational and equipment requirements greatly limit the application of PRP to OA treatment. Past studies have indicated that high-concentration PRP growth factors and cytokines may be applied as a commercial replacement for PRP. We reviewed the relevant articles to summarize the feasibility and mechanisms of PRP-based growth factors in OA. The available evidence suggests that transforming growth factor-α and β, platelet-derived growth factors, epidermal growth factor, insulin-like growth factor-1, and connective tissue growth factors might benefit OA, while vascular endothelial growth factor, tumor necrosis factor-α, angiopoietin-1, and stromal cell derived factor-1α might induce negative effects on OA. The effects of fibroblast growth factor, hepatocyte growth factor, platelet factor 4, and keratinocyte growth factor on OA remain uncertain. Thus, it can be concluded that not all cytokines released by PRP are beneficial, although the therapeutic action of PRP has a valuable potential to improve.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan, China
| | - Pengfei Zhu
- Department of Cardiovascular, Wuhan Fourth Hospital, Wuhan, China
- Department of Cardiovascular, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bokai Liao
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University and Technology, Jiefang Avenue No.1095, Qiaokou District, Wuhan, 430030, Hubei Province, China.
| | - Yu Cai
- Department of Rehabilitation, Wuhan Fourth Hospital, Hanzheng Street No.473, Qiaokou District, Wuhan, 430000, Hubei Province, China.
| |
Collapse
|
4
|
Barffour MA, Bernstein RM, Hinnouho GM, Wessells KR, Arnold CD, Kounnavong S, Hess SY. Insulin-like Growth Factor 1 (IGF1), IGF Binding Protein-3 (IGFBP3) and Growth Response to Daily Zinc Supplementation: A Randomized Trial in Rural Laotian Children. Nutrients 2023; 15:nu15112590. [PMID: 37299552 DOI: 10.3390/nu15112590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVES To assess (a) the impact of daily preventive zinc tablets (7 mg; PZ), zinc-containing multiple micronutrient powder (10 mg zinc, and 13 other micronutrients; MNP) or placebo, delivered for 9 months, on Insulin-like Growth Factor 1 (IGF1) and IGF Binding Protein 3 (IGFBP3) among Laotian children 6-23 months, and (b) whether the effects of PZ and MNP on length-for-age z-scores (LAZ) and weight-for-age z-scores (WAZ) are modified by baseline IGF1 and IGFBP3. DESIGN A double-blind, placebo-controlled trial (N = 419). METHODS Plasma IGF1 and IGFBP3 concentrations at baseline and 36 weeks were analyzed by automated chemiluminescent assay. Anthropometry was assessed at baseline, at 18 and 36 weeks. Intervention effects were estimated using ANCOVA. RESULTS At 36 weeks, geometric mean IGF1 (~39.0-39.2 ng/mL; p = 0.99) and IGFBP3 (2038-2076 ng/mL; p = 0.83) did not differ by group. At 18 weeks (but not at 36 weeks), LAZ in the PZ group (-1.45) was higher than the MNP (-1.70) and control (-1.55) groups (p = 0.01) among children in the highest baseline IGF1 tertile (p for interaction = 0.006). At 36 weeks (but not at 18 weeks), WAZ in the PZ group (-1.55) was significantly higher than the MNP (-1.75) and control (-1.65) groups (p = 0.03), among children in the lowest baseline IGFBP3 tertile (p for interactions = 0.06). CONCLUSIONS Although IGF1 and IGFBP3 did not respond to PZ and MNP, baseline IGF1 and IGFBP3 significantly modified the impact of PZ on linear and ponderal growth, suggesting that IGF1 bioavailability may drive catch-up growth in zinc-supplemented children.
Collapse
Affiliation(s)
- Maxwell A Barffour
- Department of Nutrition and Institute for Global Nutrition, University of California, Davis, CA 95616, USA
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Public Health Program, McQueary College of Health and Human Services, Missouri State University, Springfield, MO 65897, USA
| | - Robin M Bernstein
- Department of Anthropology, University of Colorado, Boulder, CO 80309, USA
- Health and Society Program, Institute for Behavioral Science, University of Colorado, Boulder, CO 80309, USA
| | - Guy-Marino Hinnouho
- Department of Nutrition and Institute for Global Nutrition, University of California, Davis, CA 95616, USA
- Helen Keller International, Washington, DC 20006, USA
| | - K Ryan Wessells
- Department of Nutrition and Institute for Global Nutrition, University of California, Davis, CA 95616, USA
| | - Charles D Arnold
- Department of Nutrition and Institute for Global Nutrition, University of California, Davis, CA 95616, USA
| | - Sengchanh Kounnavong
- Lao Tropical and Public Health Institute, Ban Kaognot, Sisattanack District, Vientiane 01030, Laos
| | - Sonja Y Hess
- Department of Nutrition and Institute for Global Nutrition, University of California, Davis, CA 95616, USA
| |
Collapse
|
5
|
Lubis AMT, Wijaya MT, Priosoeryanto BP, Saleh RF, Farqani S. Comparison of weekly and single dose intraarticular recombinant human growth hormone injection on cartilage degeneration in osteoarthritic model of white New Zealand rabbits. J Exp Orthop 2022; 9:19. [PMID: 35190919 PMCID: PMC8861207 DOI: 10.1186/s40634-022-00458-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
|
6
|
PCOS and Depression: Common Links and Potential Targets. Reprod Sci 2021; 29:3106-3123. [PMID: 34642910 DOI: 10.1007/s43032-021-00765-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
PCOS or polycystic ovary syndrome is a common endocrine disorder that occurs during the reproductive age in females. It manifests in the form of a wide range of symptoms including (but not limited to) hirsutism, amenorrhea, oligomenorrhea, obesity, acne vulgaris, infertility, alopecia, and insulin resistance. The incidence of depression in PCOS population is increasing as compared to the general population. Increased depression in PCOS significantly alters the quality of life (QOL) of affected females. Also, self-esteem is found to be low in both depression and PCOS. The loss in self-esteem in such patients can be largely attributed to the associated factors including (but not limited to) obesity, acne, androgenic alopecia, and hirsutism. The reason behind the occurrence of depression in PCOS remains elusive to date. Literature suggests that there is an overlap of clinical symptoms between depression and PCOS. As the symptoms overlap, there is a possibility of common associations between depression, PCOS, and PCOS-associated abnormalities including insulin resistance (IR), obesity, CVD, and androgen excess. Studies demonstrate that depression is an inflammatory disorder marked with increased levels of inflammatory markers. On the other hand, PCOS is also regarded as a pro-inflammatory state that is characterized by increased levels of pro-inflammatory markers. Thus, there is a possibility of an inflammatory relationship existing between depression and PCOS. It is also possible that the inflammatory markers in PCOS can cross the blood-brain barrier (BBB) leading to the development of depression. Through the present review, we have attempted to shed light on common associations/shared links between depression and PCOS with respect to the levels of cortisol, androgen, vitamin D, neurotransmitters, monoaminoxidase (MAO), and insulin-like growth factor-1 (IGF-1). Tracking down common associations between depression and PCOS will help find potential drug therapies and improve the QOL of females with depression in PCOS.
Collapse
|
7
|
Gv V, Tripathi T. Non-invasive methods for the assessment of biomarkers and their correlation with radiographic maturity indicators - a scoping review. Prog Orthod 2021; 22:26. [PMID: 34486079 PMCID: PMC8419155 DOI: 10.1186/s40510-021-00372-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Detection of skeletal maturity is vital in orthodontic treatment timing and planning. Traditional methods include hand-wrist radiography and cervical vertebral maturation index (CVMI). Though the radiographic methods are well established and routinely used to assess skeletal maturation, they carry the drawback of subjective perception and low reproducibility. With evolving concepts, skeletal maturation has been assessed quantitatively through biomarkers obtained from saliva, gingival crevicular fluid (GCF), and urine. The scoping review aims to explore the various biomarkers assessed through non-invasive methods and their correlation with radiographic skeletal maturity. METHODOLOGY The literature search was carried out on MEDLINE via Pubmed, Cochrane Library (Cochrane database of systematic reviews), Cochrane central register of controlled trials (CENTRAL), Google Scholar, Semantic Scholar, ScienceDirect, and Opengrey.eu for articles up to and including November 2020. Pertinent articles were selected based on inclusion and exclusion criteria. The results were tabulated based on the type of sample collected, the biomarker assessed, method of sample collection, and the radiographic method used. RESULTS The literature search resulted in 12 relevant articles. Among all the studies, 10 studies showed that the concentration of biomarkers increases during the pubertal growth peak. On the contrary, 2 articles showed no significant difference between the levels of biomarkers and pubertal growth peak. CONCLUSION It can be concluded that the level of biomarkers increases during the pubertal growth spurt and can provide a quantitative way of assessing skeletal maturity.
Collapse
Affiliation(s)
- Veena Gv
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah Zafar Marg, New Delhi, 110002, India
| | - Tulika Tripathi
- Department of Orthodontics and Dentofacial Orthopaedics, Maulana Azad Institute of Dental Sciences, Bahadur Shah Zafar Marg, New Delhi, 110002, India.
| |
Collapse
|
8
|
Hartley A, Sanderson E, Paternoster L, Teumer A, Kaplan RC, Tobias JH, Gregson CL. Mendelian randomization provides evidence for a causal effect of higher serum IGF-1 concentration on risk of hip and knee osteoarthritis. Rheumatology (Oxford) 2021; 60:1676-1686. [PMID: 33027520 PMCID: PMC8023994 DOI: 10.1093/rheumatology/keaa597] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/12/2020] [Indexed: 11/25/2022] Open
Abstract
Objectives How insulin-like growth factor-1 (IGF-1) is related to OA is not well understood. We determined relationships between IGF-1 and hospital-diagnosed hand, hip and knee OA in UK Biobank, using Mendelian randomization (MR) to determine causality. Methods Serum IGF-1 was assessed by chemiluminescent immunoassay. OA was determined using Hospital Episode Statistics. One-sample MR (1SMR) was performed using two-stage least-squares regression, with an unweighted IGF-1 genetic risk score as an instrument. Multivariable MR included BMI as an additional exposure (instrumented by BMI genetic risk score). MR analyses were adjusted for sex, genotyping chip and principal components. We then performed two-sample MR (2SMR) using summary statistics from Cohorts for Heart and Aging Research in Genetic Epidemiology (CHARGE) (IGF-1, N = 30 884) and the recent genome-wide association study meta-analysis (N = 455 221) of UK Biobank and Arthritis Research UK OA Genetics (arcOGEN). Results A total of 332 092 adults in UK Biobank had complete data. Their mean (s.d.) age was 56.5 (8.0) years and 54% were female. IGF-1 was observationally related to a reduced odds of hand OA [odds ratio per doubling = 0.87 (95% CI 0.82, 0.93)], and an increased odds of hip OA [1.04 (1.01, 1.07)], but was unrelated to knee OA [0.99 (0.96, 1.01)]. Using 1SMR, we found strong evidence for an increased risk of hip [odds ratio per s.d. increase = 1.57 (1.21, 2.01)] and knee [1.30 (1.07, 1.58)] OA with increasing IGF-1 concentration. By contrast, we found no evidence for a causal effect of IGF-1 concentration on hand OA [0.98 (0.57, 1.70)]. Results were consistent when estimated using 2SMR and in multivariable MR analyses accounting for BMI. Conclusion We have found evidence that increased serum IGF-1 is causally related to higher risk of hip and knee OA.
Collapse
Affiliation(s)
- April Hartley
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Robert C Kaplan
- Department of Epidemiology and Public Health, Albert Einstein College of Medicine, New York, NY, USA
| | - Jon H Tobias
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Celia L Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
9
|
Li W, Guo R, Song Y, Jiang Z. Erythroblastic Island Macrophages Shape Normal Erythropoiesis and Drive Associated Disorders in Erythroid Hematopoietic Diseases. Front Cell Dev Biol 2021; 8:613885. [PMID: 33644032 PMCID: PMC7907436 DOI: 10.3389/fcell.2020.613885] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/22/2020] [Indexed: 01/13/2023] Open
Abstract
Erythroblastic islands (EBIs), discovered more than 60 years ago, are specialized microenvironments for erythropoiesis. This island consists of a central macrophage with surrounding developing erythroid cells. EBI macrophages have received intense interest in the verifications of the supporting erythropoiesis hypothesis. Most of these investigations have focused on the identification and functional analyses of EBI macrophages, yielding significant progresses in identifying and isolating EBI macrophages, as well as verifying the potential roles of EBI macrophages in erythropoiesis. EBI macrophages express erythropoietin receptor (Epor) both in mouse and human, and Epo acts on both erythroid cells and EBI macrophages simultaneously in the niche, thereby promoting erythropoiesis. Impaired Epor signaling in splenic niche macrophages significantly inhibit the differentiation of stress erythroid progenitors. Moreover, accumulating evidence suggests that EBI macrophage dysfunction may lead to certain erythroid hematological disorders. In this review, the heterogeneity, identification, and functions of EBI macrophages during erythropoiesis under both steady-state and stress conditions are outlined. By reviewing the historical data, we discuss the influence of EBI macrophages on erythroid hematopoietic disorders and propose a new hypothesis that erythroid hematopoietic disorders are driven by EBI macrophages.
Collapse
Affiliation(s)
- Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxin Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Dixit M, Poudel SB, Yakar S. Effects of GH/IGF axis on bone and cartilage. Mol Cell Endocrinol 2021; 519:111052. [PMID: 33068640 PMCID: PMC7736189 DOI: 10.1016/j.mce.2020.111052] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Growth hormone (GH) and its mediator, the insulin-like growth factor-1 (IGF-1) regulate somatic growth, metabolism and many aspects of aging. As such, actions of GH/IGF have been studied in many tissues and organs over decades. GH and IGF-1 are part of the hypothalamic/pituitary somatotrophic axis that consists of many other regulatory hormones, receptors, binding proteins, and proteases. In humans, GH/IGF actions peak during pubertal growth and regulate skeletal acquisition through stimulation of extracellular matrix production and increases in bone mineral density. During aging the activity of these hormones declines, a state called somatopaguss, which associates with deleterious effects on the musculoskeletal system. In this review, we will focus on GH/IGF-1 action in bone and cartilage. We will cover many studies that have utilized congenital ablation or overexpression of members of this axis, as well as cell-specific gene-targeting approaches used to unravel the nature of the GH/IGF-1 actions in the skeleton in vivo.
Collapse
Affiliation(s)
- Manisha Dixit
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, NY, 10010, USA
| | - Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, NY, 10010, USA
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, NY, 10010, USA.
| |
Collapse
|
11
|
Sharma R, Kopchick JJ, Puri V, Sharma VM. Effect of growth hormone on insulin signaling. Mol Cell Endocrinol 2020; 518:111038. [PMID: 32966863 PMCID: PMC7606590 DOI: 10.1016/j.mce.2020.111038] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
Growth hormone (GH) is a pleiotropic hormone that coordinates an array of physiological processes, including effects on bone, muscle, and fat, ultimately resulting in growth. Metabolically, GH promotes anabolic action in most tissues except adipose, where its catabolic action causes the breakdown of stored triglycerides into free fatty acids (FFA). GH antagonizes insulin action via various molecular pathways. Chronic GH secretion suppresses the anti-lipolytic action of insulin and increases FFA flux into the systemic circulation; thus, promoting lipotoxicity, which causes pathophysiological problems, including insulin resistance. In this review, we will provide an update on GH-stimulated adipose lipolysis and its consequences on insulin signaling in liver, skeletal muscle, and adipose tissue. Furthermore, we will discuss the mechanisms that contribute to the diabetogenic action of GH.
Collapse
Affiliation(s)
- Rita Sharma
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA
| | - John J Kopchick
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA; Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Diabetes Institute, Ohio University, Athens, OH, 45701, USA
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA; Diabetes Institute, Ohio University, Athens, OH, 45701, USA
| | - Vishva M Sharma
- Department of Biomedical Sciences, Ohio University, Athens, OH, 45701, USA; Diabetes Institute, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
12
|
Endo K, Fujita N, Nakagawa T, Nishimura R. Comparison of the effect of growth factors on chondrogenesis of canine mesenchymal stem cells. J Vet Med Sci 2019; 81:1211-1218. [PMID: 31167981 PMCID: PMC6715918 DOI: 10.1292/jvms.18-0551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are proposed to be useful in cartilage regenerative
medicine, however, canine MSCs have been reported to show poor chondrogenic capacity.
Therefore, optimal conditions for chondrogenic differentiation should be determined by
mimicking the developmental process. We have previously established novel and superior
canine MSCs named bone marrow peri-adipocyte cells (BM-PACs) and the objective of this
study was to evaluate the effects of growth factors required for in vivo
chondrogenesis using canine BM-PACs. Spheroids of BM-PACs were cultured in chondrogenic
medium containing 10 ng/ml transforming growth factor-β1
(TGF-β1) with or without 100 ng/ml bone morphogenetic
protein-2 (BMP-2), 100 ng/ml growth differentiation
factor-5 (GDF-5) or 100 ng/ml insulin-like growth
factor-1 (IGF-1). Chondrogenic differentiation was evaluated by the quantification of
glycosaminoglycan and Safranin O staining for proteoglycan production. The expression of
cartilage matrix or hypertrophic gene/protein was also evaluated by qPCR and
immunohistochemistry. Spheroids in all groups were strongly stained with Safranin O.
Although BMP-2 significantly increased glycosaminoglycan production, Safranin O-negative
outer layer was formed and the mRNA expression of COL10 relating to cartilage hypertrophy
was also significantly upregulated (P<0.05). GDF-5 promoted the
production of glycosaminoglycan and type II collagen without increasing COL10 mRNA
expression. The supplementation of IGF-1 did not significantly affect cartilaginous and
hypertrophic differentiation. Our results indicate that GDF-5 is a useful growth factor
for the generation of articular cartilage from canine MSCs.
Collapse
Affiliation(s)
- Kentaro Endo
- Laboratory of Veterinary Surgery, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Naoki Fujita
- Laboratory of Veterinary Surgery, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryohei Nishimura
- Laboratory of Veterinary Surgery, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
13
|
Storr HL, Chatterjee S, Metherell LA, Foley C, Rosenfeld RG, Backeljauw PF, Dauber A, Savage MO, Hwa V. Nonclassical GH Insensitivity: Characterization of Mild Abnormalities of GH Action. Endocr Rev 2019; 40:476-505. [PMID: 30265312 PMCID: PMC6607971 DOI: 10.1210/er.2018-00146] [Citation(s) in RCA: 30] [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: 06/01/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022]
Abstract
GH insensitivity (GHI) presents in childhood with growth failure and in its severe form is associated with extreme short stature and dysmorphic and metabolic abnormalities. In recent years, the clinical, biochemical, and genetic characteristics of GHI and other overlapping short stature syndromes have rapidly expanded. This can be attributed to advancing genetic techniques and a greater awareness of this group of disorders. We review this important spectrum of defects, which present with phenotypes at the milder end of the GHI continuum. We discuss their clinical, biochemical, and genetic characteristics. The objective of this review is to clarify the definition, identification, and investigation of this clinically relevant group of growth defects. We also review the therapeutic challenges of mild GHI.
Collapse
Affiliation(s)
- Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Sumana Chatterjee
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Louise A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Corinne Foley
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ron G Rosenfeld
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| | - Philippe F Backeljauw
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Andrew Dauber
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Martin O Savage
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Vivian Hwa
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
14
|
Yakar S, Werner H, Rosen CJ. Insulin-like growth factors: actions on the skeleton. J Mol Endocrinol 2018; 61:T115-T137. [PMID: 29626053 PMCID: PMC5966339 DOI: 10.1530/jme-17-0298] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022]
Abstract
The discovery of the growth hormone (GH)-mediated somatic factors (somatomedins), insulin-like growth factor (IGF)-I and -II, has elicited an enormous interest primarily among endocrinologists who study growth and metabolism. The advancement of molecular endocrinology over the past four decades enables investigators to re-examine and refine the established somatomedin hypothesis. Specifically, gene deletions, transgene overexpression or more recently, cell-specific gene-ablations, have enabled investigators to study the effects of the Igf1 and Igf2 genes in temporal and spatial manners. The GH/IGF axis, acting in an endocrine and autocrine/paracrine fashion, is the major axis controlling skeletal growth. Studies in rodents have clearly shown that IGFs regulate bone length of the appendicular skeleton evidenced by changes in chondrocytes of the proliferative and hypertrophic zones of the growth plate. IGFs affect radial bone growth and regulate cortical and trabecular bone properties via their effects on osteoblast, osteocyte and osteoclast function. Interactions of the IGFs with sex steroid hormones and the parathyroid hormone demonstrate the significance and complexity of the IGF axis in the skeleton. Finally, IGFs have been implicated in skeletal aging. Decreases in serum IGFs during aging have been correlated with reductions in bone mineral density and increased fracture risk. This review highlights many of the most relevant studies in the IGF research landscape, focusing in particular on IGFs effects on the skeleton.
Collapse
Affiliation(s)
- Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
| |
Collapse
|
15
|
Possible effects of an early diagnosis and treatment in patients with growth hormone deficiency: the state of art. Ital J Pediatr 2017; 43:81. [PMID: 28915901 PMCID: PMC5603037 DOI: 10.1186/s13052-017-0402-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/11/2017] [Indexed: 01/16/2023] Open
Abstract
Growth hormone deficiency (GHD) is a relatively uncommon and heterogeneous endocrine disorder presenting in childhood with short stature. However, during the neonatal period, the metabolic effects of GHD may to require prompt replacement therapy to avoid possible life-threatening complications. An increasing amount of data suggests the importance of an early diagnosis and treatment of GHD because of its auxological, metabolic, and neurodevelopmental features with respect to the patients diagnosed and treated later in life. The available results show favourable auxological outcomes for patients with GHD diagnosed and treated with r-hGH early in life compared with those from patients with GHD who do not receive this early diagnosis and treatment. Because delayed referral for GHD diagnosis and treatment is still frequent, these results highlight the need for more attention in the diagnosis and treatment of GHD. Despite these very encouraging data regarding metabolic and neurodevelopmental features, further studies are needed to better characterize these findings. Overall, the importance of early diagnosis and treatment of GHD needs to be addressed.
Collapse
|
16
|
Troike KM, Henry BE, Jensen EA, Young JA, List EO, Kopchick JJ, Berryman DE. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 2017. [PMID: 28640444 DOI: 10.1002/cphy.c160027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increasing prevalence of obesity and obesity-related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot-dependent. © 2017 American Physiological Society. Compr Physiol 7:819-840, 2017.
Collapse
Affiliation(s)
- Katie M Troike
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Brooke E Henry
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Elizabeth A Jensen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Jonathan A Young
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
17
|
Pereira AM. Long-term effects of treatment of pituitary adenomas. HANDBOOK OF CLINICAL NEUROLOGY 2016; 124:361-71. [PMID: 25248599 DOI: 10.1016/b978-0-444-59602-4.00024-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Pituitary adenomas can be treated effectively in the vast majority of cases. After successful treatment for pituitary disease, many patients still report reduced quality of life in the presence of persistent morbidity and (slightly) increased mortality. At present, there is an increasing awareness that in many cases long-term remission of functioning or nonfunctioning adenomas does not equal cure. The causes are most probably multifactorial. Hypopituitarism, intrinsic imperfections of surgical or endocrine replacement therapy, but also persistent effects of treatment and of previous hormone excess on the central nervous system all affect long-term morbidity, general well-being, and mortality. This implies that treatment goals for patients with pituitary adenomas will shift from long-term cure to long-term care. Further research is therefore needed to get more insight into each of these factors of influence, such as the extent of reversibility of hormone excess syndromes on cardiovascular risk and behavior. The fact that coping strategies, despite long-term remission, are altered and illness perceptions are affected strongly suggests that long-term care should incorporate self-management interventions that might help to improve quality of life for patients.
Collapse
Affiliation(s)
- Alberto M Pereira
- Department of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
18
|
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: 90] [Impact Index Per Article: 11.3] [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
|
19
|
Santa Maria C, Cheng Z, Li A, Wang J, Shoback D, Tu CL, Chang W. Interplay between CaSR and PTH1R signaling in skeletal development and osteoanabolism. Semin Cell Dev Biol 2016; 49:11-23. [PMID: 26688334 PMCID: PMC4761456 DOI: 10.1016/j.semcdb.2015.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/05/2015] [Indexed: 12/01/2022]
Abstract
Parathyroid hormone (PTH)-related peptide (PTHrP) controls the pace of pre- and post-natal growth plate development by activating the PTH1R in chondrocytes, while PTH maintains mineral and skeletal homeostasis by modulating calciotropic activities in kidneys, gut, and bone. The extracellular calcium-sensing receptor (CaSR) is a member of family C, G protein-coupled receptor, which regulates mineral and skeletal homeostasis by controlling PTH secretion in parathyroid glands and Ca(2+) excretion in kidneys. Recent studies showed the expression of CaSR in chondrocytes, osteoblasts, and osteoclasts and confirmed its non-redundant roles in modulating the recruitment, proliferation, survival, and differentiation of the cells. This review emphasizes the actions of CaSR and PTH1R signaling responses in cartilage and bone and discusses how these two signaling cascades interact to control growth plate development and maintain skeletal metabolism in physiological and pathological conditions. Lastly, novel therapeutic regimens that exploit interrelationship between the CaSR and PTH1R are proposed to produce more robust osteoanabolism.
Collapse
Affiliation(s)
- Christian Santa Maria
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Zhiqiang Cheng
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Alfred Li
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Jiali Wang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Dolores Shoback
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Chia-Ling Tu
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Wenhan Chang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA.
| |
Collapse
|
20
|
Abstract
The regulation of organ size is essential to human health and has fascinated biologists for centuries. Key to the growth process is the ability of most organs to integrate organ-extrinsic cues (eg, nutritional status, inflammatory processes) with organ-intrinsic information (eg, genetic programs, local signals) into a growth response that adapts to changing environmental conditions and ensures that the size of an organ is coordinated with the rest of the body. Paired organs such as the vertebrate limbs and the long bones within them are excellent models for studying this type of regulation because it is possible to manipulate one member of the pair and leave the other as an internal control. During development, growth plates at the end of each long bone produce a transient cartilage model that is progressively replaced by bone. Here, we review how proliferation and differentiation of cells within each growth plate are tightly controlled mainly by growth plate-intrinsic mechanisms that are additionally modulated by extrinsic signals. We also discuss the involvement of several signaling hubs in the integration and modulation of growth-related signals and how they could confer remarkable plasticity to the growth plate. Indeed, long bones have a significant ability for "catch-up growth" to attain normal size after a transient growth delay. We propose that the characterization of catch-up growth, in light of recent advances in physiology and cell biology, will provide long sought clues into the molecular mechanisms that underlie organ growth regulation. Importantly, catch-up growth early in life is commonly associated with metabolic disorders in adulthood, and this association is not completely understood. Further elucidation of the molecules and cellular interactions that influence organ size coordination should allow development of novel therapies for human growth disorders that are noninvasive and have minimal side effects.
Collapse
Affiliation(s)
- Alberto Roselló-Díez
- Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065
| | - Alexandra L Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065
| |
Collapse
|
21
|
Desai NA, Patel SS. Increased insulin-like growth factor-1 in relation to cardiovascular function in polycystic ovary syndrome: friend or foe? Gynecol Endocrinol 2015; 31:801-7. [PMID: 26288196 DOI: 10.3109/09513590.2015.1075497] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The incidence of cardiovascular disease (CVD) in patients with polycystic ovary syndrome (PCOS) is very high and conventional risk factors only partially explain excessive risk of developing CVD in patients of PCOS. The pathophysiology of PCOS is very unique, and several hormonal and metabolic changes occur. Several observations suggest that serum IGF-1 levels decrease in insulin resistance, which results in IGF-1 deficiency. In patient of PCOS, close relationships have been demonstrated between insulin resistance and serum IGF-1 levels. Hyperinsulinemic insulin resistance results in a general augmentation of steroidogenesis and LH release in PCOS. The action of IGF-1 varies in different tissues possibly via autocrine or paracrine mechanisms. The increase or decrease in IGF-1 in different tissues results in differential outcomes. Several studies suggest that lowered circulating IGF-1 levels play important role in the initiation of the cardiac hypertrophic response which results in the risk of cardiovascular disease. While recent results suggests that individual with elevated IGF-1 is protected against cardiovascular disease. Thus IGF-1 shows versatile pleiotropic actions. This review provides a current perspective on increased level of IGF-1 in PCOS and also adds to the current controversy regarding the roles of IGF-1 in cardiovascular disease.
Collapse
Affiliation(s)
- Namrata Ajaykumar Desai
- a Department of Pharmacology , Institute of Pharmacy, Nirma University , Ahmedabad , Gujarat , India
| | - Snehal S Patel
- a Department of Pharmacology , Institute of Pharmacy, Nirma University , Ahmedabad , Gujarat , India
| |
Collapse
|
22
|
Sederquist B, Fernandez-Vojvodich P, Zaman F, Sävendahl L. Recent research on the growth plate: Impact of inflammatory cytokines on longitudinal bone growth. J Mol Endocrinol 2014; 53:T35-44. [PMID: 24711646 DOI: 10.1530/jme-14-0006] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Children with inflammatory diseases usually display abnormal growth patterns as well as delayed puberty. This is a result of several factors related to the disease itself, such as malnutrition, hypercortisolism, and elevated levels of pro-inflammatory cytokines. These factors in combination with glucocorticoid treatment contribute to growth retardation during chronic inflammation by systemically affecting the major regulator of growth, the GH/IGF1 axis. However, recent studies have also shown evidence of a direct effect of these factors at the growth plate level. In conditions of chronic inflammation, pro-inflammatory cytokines are upregulated and released into the circulation. The most abundant of these, tumor necrosis factor α, interleukin 1β (IL1β), and IL6, are all known to directly act on growth plate cartilage to induce apoptosis and thereby suppress bone growth. Both clinical and experimental studies have shown that growth retardation can partly be rescued when these cytokines are blocked. Therefore, therapy modulating the local actions of these cytokines may be effective for preventing growth failure in patients with chronic inflammatory disorders. In this review, we report the current knowledge of inflammatory cytokines and their role in regulating bone growth.
Collapse
Affiliation(s)
- Bettina Sederquist
- Pediatric Endocrinology Unit Q2:08Department of Women's and Children's Health, Karolinska University Hospital, SE-171 76 Stockholm, SwedenDevelopmental and Stem Cell BiologyThe Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Paola Fernandez-Vojvodich
- Pediatric Endocrinology Unit Q2:08Department of Women's and Children's Health, Karolinska University Hospital, SE-171 76 Stockholm, SwedenDevelopmental and Stem Cell BiologyThe Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Farasat Zaman
- Pediatric Endocrinology Unit Q2:08Department of Women's and Children's Health, Karolinska University Hospital, SE-171 76 Stockholm, SwedenDevelopmental and Stem Cell BiologyThe Hospital for Sick Children, University of Toronto, Toronto, Ontario, CanadaPediatric Endocrinology Unit Q2:08Department of Women's and Children's Health, Karolinska University Hospital, SE-171 76 Stockholm, SwedenDevelopmental and Stem Cell BiologyThe Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Lars Sävendahl
- Pediatric Endocrinology Unit Q2:08Department of Women's and Children's Health, Karolinska University Hospital, SE-171 76 Stockholm, SwedenDevelopmental and Stem Cell BiologyThe Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Chia DJ. Minireview: mechanisms of growth hormone-mediated gene regulation. Mol Endocrinol 2014; 28:1012-25. [PMID: 24825400 DOI: 10.1210/me.2014-1099] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GH exerts a diverse array of physiological actions that include prominent roles in growth and metabolism, with a major contribution via stimulating IGF-1 synthesis. GH achieves its effects by influencing gene expression profiles, and Igf1 is a key transcriptional target of GH signaling in liver and other tissues. This review examines the mechanisms of GH-mediated gene regulation that begin with signal transduction pathways activated downstream of the GH receptor and continue with chromatin events at target genes and additionally encompasses the topics of negative regulation and cross talk with other cellular inputs. The transcription factor, signal transducer and activator of transcription 5b, is regarded as the major signaling pathway by which GH achieves its physiological effects, including in stimulating Igf1 gene transcription in liver. Recent studies exploring the mechanisms of how activated signal transducer and activator of transcription 5b accomplishes this are highlighted, which begin to characterize epigenetic features at regulatory domains of the Igf1 locus. Further research in this field offers promise to better understand the GH-IGF-1 axis in normal physiology and disease and to identify strategies to manipulate the axis to improve human health.
Collapse
Affiliation(s)
- Dennis J Chia
- Department of Pediatrics, Icahn School of Medicine at Mt Sinai, New York, New York 10029
| |
Collapse
|
24
|
Oran PE, Trenchevska O, Nedelkov D, Borges CR, Schaab MR, Rehder DS, Jarvis JW, Sherma ND, Shen L, Krastins B, Schwenke DC, Reaven PD, Nelson RW. Parallel workflow for high-throughput (>1,000 samples/day) quantitative analysis of human insulin-like growth factor 1 using mass spectrometric immunoassay. PLoS One 2014; 9:e92801. [PMID: 24664114 PMCID: PMC3963945 DOI: 10.1371/journal.pone.0092801] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/26/2014] [Indexed: 12/11/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1) is an important biomarker for the management of growth hormone disorders. Recently there has been rising interest in deploying mass spectrometric (MS) methods of detection for measuring IGF1. However, widespread clinical adoption of any MS-based IGF1 assay will require increased throughput and speed to justify the costs of analyses, and robust industrial platforms that are reproducible across laboratories. Presented here is an MS-based quantitative IGF1 assay with performance rating of >1,000 samples/day, and a capability of quantifying IGF1 point mutations and posttranslational modifications. The throughput of the IGF1 mass spectrometric immunoassay (MSIA) benefited from a simplified sample preparation step, IGF1 immunocapture in a tip format, and high-throughput MALDI-TOF MS analysis. The Limit of Detection and Limit of Quantification of the resulting assay were 1.5 μg/L and 5 μg/L, respectively, with intra- and inter-assay precision CVs of less than 10%, and good linearity and recovery characteristics. The IGF1 MSIA was benchmarked against commercially available IGF1 ELISA via Bland-Altman method comparison test, resulting in a slight positive bias of 16%. The IGF1 MSIA was employed in an optimized parallel workflow utilizing two pipetting robots and MALDI-TOF-MS instruments synced into one-hour phases of sample preparation, extraction and MSIA pipette tip elution, MS data collection, and data processing. Using this workflow, high-throughput IGF1 quantification of 1,054 human samples was achieved in approximately 9 hours. This rate of assaying is a significant improvement over existing MS-based IGF1 assays, and is on par with that of the enzyme-based immunoassays. Furthermore, a mutation was detected in ∼1% of the samples (SNP: rs17884626, creating an A→T substitution at position 67 of the IGF1), demonstrating the capability of IGF1 MSIA to detect point mutations and posttranslational modifications.
Collapse
Affiliation(s)
- Paul E Oran
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Olgica Trenchevska
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Dobrin Nedelkov
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Chad R Borges
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Matthew R Schaab
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Douglas S Rehder
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jason W Jarvis
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Nisha D Sherma
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Luhui Shen
- Center for Innovations in Medicine, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Bryan Krastins
- Thermo Fisher Scientific, The Biomarkers Research Initiatives in Mass Spectrometry Center, Cambridge, Massachusetts, United States of America
| | - Dawn C Schwenke
- Phoenix VA Health Care System, Phoenix, Arizona, United States of America; College of Nursing & Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, Arizona, United States of America
| | - Randall W Nelson
- Molecular Biomarkers Laboratory, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
25
|
Jiang H, Ge X. MEAT SCIENCE AND MUSCLE BIOLOGY SYMPOSIUM--mechanism of growth hormone stimulation of skeletal muscle growth in cattle. J Anim Sci 2013; 92:21-9. [PMID: 24166991 DOI: 10.2527/jas.2013-7095] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Growth hormone, also called somatotropin (ST), is a polypeptide hormone produced by the anterior pituitary. The major functions of GH include stimulating bone and skeletal muscle growth, lipolysis, milk production, and expression of the IGF-I gene in the liver. Based on these functions, recombinant bovine ST (bST) and recombinant porcine ST (pST) have been used to improve milk production in dairy cows and lean tissue growth in pigs, respectively. However, despite these applications, the mechanisms of action of GH are not fully understood. Indeed, there has been a lot of controversy over the role of liver-derived circulating IGF-I and locally produced IGF-I in mediating the growth-stimulatory effect of GH during the last 15 yr. It is in this context that we have conducted studies to further understand how GH stimulates skeletal muscle growth in cattle. Our results do not support a role of skeletal muscle-derived IGF-I in GH-stimulated skeletal muscle growth in cattle. Our results indicate that GH stimulates skeletal muscle growth in cattle, in part, by stimulating protein synthesis in muscle through a GH receptor-mediated, IGF-I-independent mechanism. In this review, besides discussing these results, we also argue that liver-derived circulating IGF-I should be still considered as the major mechanism that mediates the growth-stimulatory effect of GH on skeletal muscle in cattle and other domestic animals.
Collapse
Affiliation(s)
- H Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg 24060
| | | |
Collapse
|
26
|
Farquharson C, Ahmed SF. Inflammation and linear bone growth: the inhibitory role of SOCS2 on GH/IGF-1 signaling. Pediatr Nephrol 2013; 28:547-56. [PMID: 22886280 DOI: 10.1007/s00467-012-2271-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/16/2012] [Accepted: 07/11/2012] [Indexed: 12/29/2022]
Abstract
Linear bone growth is widely recognized to be adversely affected in children with chronic kidney disease (CKD) and other chronic inflammatory disorders. The growth hormone (GH)/insulin-like growth factor-1 (IGF-1) pathway is anabolic to the skeleton and inflammatory cytokines compromise bone growth through a number of different mechanisms, which include interference with the systemic as well as the tissue-level GH/IGF-1 axis. Despite attempts to promote growth and control disease, there are an increasing number of reports of the persistence of poor growth in a substantial proportion of patients receiving rhGH and/or drugs that block cytokine action. Thus, there is an urgent need to consider better and alternative forms of therapy that are directed specifically at the mechanism of the insult which leads to abnormal bone health. Suppressor of cytokine signaling 2 (SOCS2) expression is increased in inflammatory conditions including CKD, and is a recognized inhibitor of GH signaling. Therefore, in this review, we will focus on the premise that SOCS2 signaling represents a critical pathway in growth plate chondrocytes through which pro-inflammatory cytokines alter both GH/IGF-1 signaling and cellular function.
Collapse
Affiliation(s)
- Colin Farquharson
- Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| | | |
Collapse
|
27
|
Panzani S, Castagnetti C, Prandi A, Faustini M, Zamboni A, Veronesi MC. Insulin-like growth factor I: could it be a marker of prematurity in the foal? Theriogenology 2012; 79:495-501. [PMID: 23218397 DOI: 10.1016/j.theriogenology.2012.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 10/29/2012] [Accepted: 11/01/2012] [Indexed: 10/27/2022]
Abstract
Insulin-like growth factor (IGF)-I represents one of the most important growth regulators, playing a central role in fetal and neonatal growth. Plasma IGF-I levels increase rapidly after birth, and they are influenced by numerous factors, including sex, age, nutritional state, and premature birth. The aims of this study were: (1) to evaluate the IGF-I plasma profile in healthy newborn foals during the first 2 weeks of life; (2) to assess the possible influence of sex and birth weight on this hormone; (3) to analyze the percentage increment of IGF-I values in healthy foals; (4) to evaluate the influence of prematurity on IGF-I profile; (5) to verify the role of IGF-I as a diagnostic marker of prematurity; and (6) to analyze the percentage increment of IGF-I in premature foals. Thirty-four healthy term foals were enrolled as the control group and from each foal plasma was collected within 6 hours from birth, at 12 hours, daily from Day 1 to Day 7, and at Days 10 and 14 after birth. Eleven foals aged younger than 1 week and diagnosed as premature and hospitalized at a Equine Perinatology Unit were also enrolled; from each foal plasma was collected daily from the day of admission to discharge or death. Insulin-like growth factor I was analyzed by RIA. In the control group, an increasing trend of IGF-I concentrations was found, with higher values from Day 4 to 10 compared with data obtained at less than 6 hours of life, and from Day 5 to 10 compared with 12 and 24 hours and 3 days. No differences were found in healthy foals analyzed in relation to birth weight and sex. In premature foals an increasing trend was observed but no statistical differences were found among sampling times, and no differences were found between healthy and premature foals. The IGF value in premature foals at admission was always higher compared with the lowest recorded level in healthy age-matched foals, thus this parameter does not seem to have a diagnostic role for prematurity in foals. Finally, the evaluation of the percentage increment of IGF-I concentrations showed a significant increase in full-term foals on Day 5, 6, 7, and 10 compared with 12 and 24 hours, and no differences were observed in premature foals. In conclusion, prematurity in newborn foals seems to affect only partially IGF-I plasma concentrations and it does not seem to be a reliable marker for this pathological condition.
Collapse
Affiliation(s)
- S Panzani
- Department of Health, Animal Science and Food Safety, Faculty of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | |
Collapse
|
28
|
Pass C, MacRae VE, Huesa C, Faisal Ahmed S, Farquharson C. SOCS2 is the critical regulator of GH action in murine growth plate chondrogenesis. J Bone Miner Res 2012; 27:1055-66. [PMID: 22228213 DOI: 10.1002/jbmr.1544] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Suppressor of Cytokine Signaling-2 (SOCS2) is a negative regulator of growth hormone (GH) signaling and bone growth via inhibition of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. This has been classically demonstrated by the overgrowth phenotype of SOCS2(-/-) mice, which has normal systemic insulin-like growth factor 1 (IGF-1) levels. The local effects of GH on bone growth are equivocal, and therefore this study aimed to understand better the SOCS2 signaling mechanisms mediating the local actions of GH on epiphyseal chondrocytes and bone growth. SOCS2, in contrast to SOCS1 and SOCS3 expression, was increased in cultured chondrocytes after GH challenge. Gain- and loss-of-function studies indicated that GH-stimulated chondrocyte STATs-1, -3, and -5 phosphorylation was increased in SOCS2(-/-) chondrocytes but not in cells overexpressing SOCS2. This increased chondrocyte STAT signaling in the absence of SOCS2 is likely to explain the observed GH stimulation of longitudinal growth of cultured SOCS2(-/-) embryonic metatarsals and the proliferation of chondrocytes within. Consistent with this metatarsal data, bone growth rates, growth plate widths, and chondrocyte proliferation were all increased in SOCS2(-/-) 6-week-old mice as was the number of phosphorylated STAT-5-positive hypertrophic chondrocytes. The SOCS2(-/-) mouse represents a valid model for studying the local effects of GH on bone growth.
Collapse
Affiliation(s)
- Chloe Pass
- Bone Biology Group, Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Roslin, Midlothian, UK
| | | | | | | | | |
Collapse
|
29
|
Savage MO, Hwa V, David A, Rosenfeld RG, Metherell LA. Genetic Defects in the Growth Hormone-IGF-I Axis Causing Growth Hormone Insensitivity and Impaired Linear Growth. Front Endocrinol (Lausanne) 2011; 2:95. [PMID: 22654835 PMCID: PMC3356141 DOI: 10.3389/fendo.2011.00095] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 11/21/2011] [Indexed: 11/13/2022] Open
Abstract
Human genetic defects in the growth hormone (GH)-IGF-I axis affecting the IGF system present with growth failure as their principal clinical feature. This is usually associated with GH insensitivity (GHI) presenting in childhood as severe or mild short stature. Dysmorphic features and metabolic abnormalities may also be present. The field of GHI due to mutations affecting GH action has evolved rapidly since the first description of the extreme phenotype related to homozygous GH receptor (GHR) mutations in 1966. A continuum of genetic, phenotypic, and biochemical abnormalities can be defined associated with clinically relevant defects in linear growth. The mechanisms of the GH-IGF-I axis in the regulation of normal human growth is discussed followed by descriptions of mutations in GHR, STAT5B, IGF-I, IGFALS, IGF1R, and GH1 defects causing bio-inactive GH or anti-GH antibodies. These GH-IGF-I axis defects are associated with a range of clinical, and hormonal characteristics. An up-dated approach to the clinical assessment of the patient with GHI focusing on investigation of the GH-IGF-I axis and relevant molecular studies contributing to the identification of causative genetic defects is also discussed.
Collapse
Affiliation(s)
- Martin O. Savage
- Department of Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryLondon, UK
| | - Vivian Hwa
- Department of Pediatrics, Oregon Health and Science UniversityPortland, OR, USA
| | - Alessia David
- Department of Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryLondon, UK
| | - Ron G. Rosenfeld
- Department of Pediatrics, Oregon Health and Science UniversityPortland, OR, USA
| | - Louise A. Metherell
- Department of Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryLondon, UK
| |
Collapse
|
30
|
Modi HN, Suh SW, Prjvc B, Hong JY, Yang JH, Park YH, Lee JM, Kwon YH. Bone quality and growth characteristics of growth plates following limb transplantation between animals of different ages--results of an experimental study in male syngeneic rats. J Orthop Surg Res 2011; 6:53. [PMID: 21999811 PMCID: PMC3207911 DOI: 10.1186/1749-799x-6-53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 10/14/2011] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The purpose of this study was to identify graft osteoporosis post transplantation by micro-CT analysis, and the growth potential of growth plates in the transplanted limb. METHODS Ten juvenile to juvenile and five juvenile to adult hind limb transplants were performed in male syngeneic Lewis rats. Upper tibial bone density in isochronograft and heterochronograft limbs was measured by 3D micro-CT and compared with that of the opposite non-operated limbs. RESULTS We observed inferior bone quality (p < 0.05) in heterochronografts compared to isochronografts. After transplantation, isochronografts did not exhibit increases in tibial lengths compared to opposite juvenile non-operated tibias (p = 0.66) or heterochronograft tibias (p = 0.61). However, significant differences were observed between heterochrongraft tibial lengths when and opposite adult non operated tibial lengths (p < 0.001). CONCLUSIONS Age dependent alterations affect bone quality, resulting in post transplantation osteoporosis in heterochronografts, but not isochronografts. However, the growth plates of transplanted limbs retain their properties of longitudinal growth and continue to grow at the same rate.
Collapse
Affiliation(s)
- Hitesh N Modi
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Seung Woo Suh
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Boopalan Prjvc
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Jae-Young Hong
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Jae-Hyuk Yang
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Young-Hwan Park
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Jae-Moon Lee
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| | - Yong-Hyon Kwon
- Scoliosis Research Institute, Department of Orthopedics, Korea University Guro Hospital, Seoul, Korea
| |
Collapse
|
31
|
David A, Hwa V, Metherell LA, Netchine I, Camacho-Hübner C, Clark AJL, Rosenfeld RG, Savage MO. Evidence for a continuum of genetic, phenotypic, and biochemical abnormalities in children with growth hormone insensitivity. Endocr Rev 2011; 32:472-97. [PMID: 21525302 DOI: 10.1210/er.2010-0023] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
GH insensitivity (GHI) presents in childhood as growth failure and in its severe form is associated with dysmorphic and metabolic abnormalities. GHI may be caused by genetic defects in the GH-IGF-I axis or by acquired states such as chronic illness. This article discusses the former category. The field of GHI due to mutations affecting GH action has evolved considerably since the original description of the extreme phenotype related to homozygous GH receptor (GHR) mutations over 40 yr ago. A continuum of genetic, phenotypic, and biochemical abnormalities can be defined associated with clinically relevant defects in linear growth. The role and mechanisms of the GH-IGF-I axis in normal human growth is discussed, followed by descriptions of mutations in GHR, STAT5B, PTPN11, IGF1, IGFALS, IGF1R, and GH1 defects causing bioinactive GH or anti-GH antibodies. These defects are associated with a range of genetic, clinical, and hormonal characteristics. Genetic abnormalities causing growth failure that is less severe than the extreme phenotype are emphasized, together with an analysis of height and serum IGF-I across the spectrum of different types of GHR defects. An overall view of genotype and phenotype relationships is presented, together with an updated approach to the assessment of the patient with GHI, focusing on investigation of the GH-IGF-I axis and relevant molecular studies contributing to this diagnosis.
Collapse
Affiliation(s)
- Alessia David
- Department of Endocrinology, Barts and the London School of Medicine and Dentistry, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Netchine I, Azzi S, Le Bouc Y, Savage MO. IGF1 molecular anomalies demonstrate its critical role in fetal, postnatal growth and brain development. Best Pract Res Clin Endocrinol Metab 2011; 25:181-90. [PMID: 21396584 DOI: 10.1016/j.beem.2010.08.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The phenotype caused by human genetic insulin-like growth factor-I (IGF-I) defects is characterised by the association of intrauterine and postnatal growth retardation with sensorineural deafness and intellectual deficit. This syndrome is extremely rare and only four cases have been reported. Addition clinical features may include microcephaly and later in life adiposity and insulin resistance. Partial gonadal dysfunction and osteoporosis may also be present. A case of partial IGF-I deficiency has recently been described and was associated with pre- and postnatal growth retardation and microcephaly but the developmental delay was mild and hearing tests were normal. IGF-I deficiency is transmitted as an autosomal recessive trait and is caused by homozygous mutations in the IGF1 gene. Currently these patients can benefit from recombinant IGF-I which is now available for treatment. These observations demonstrate that the integrity of IGF-I signalling is important for normal growth and brain development.
Collapse
Affiliation(s)
- Irène Netchine
- APHP, Hôpital Armand-Trousseau, Explorations Fonctionnelles Endocriniennes, Paris, France.
| | | | | | | |
Collapse
|
33
|
Cartilage engineering from mesenchymal stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2010; 123:163-200. [PMID: 20535603 DOI: 10.1007/10_2010_67] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal progenitor cells known as multipotent mesenchymal stromal cells or mesenchymal stem cells (MSC) have been isolated from various tissues. Since they are able to differentiate along the mesenchymal lineages of cartilage and bone, they are regarded as promising sources for the treatment of skeletal defects. Tissue regeneration in the adult organism and in vitro engineering of tissues is hypothesized to follow the principles of embryogenesis. The embryonic development of the skeleton has been studied extensively with respect to the regulatory mechanisms governing morphogenesis, differentiation, and tissue formation. Various concepts have been designed for engineering tissues in vitro based on these developmental principles, most of them involving regulatory molecules such as growth factors or cytokines known to be the key regulators in developmental processes. Growth factors most commonly used for in vitro cultivation of cartilage tissue belong to the fibroblast growth factor (FGF) family, the transforming growth factor-beta (TGF-β) super-family, and the insulin-like growth factor (IGF) family. In this chapter, in vivo actions of members of these growth factors described in the literature are compared with in vitro concepts of cartilage engineering making use of these growth factors.
Collapse
|
34
|
Ohlsson C, Mohan S, Sjögren K, Tivesten A, Isgaard J, Isaksson O, Jansson JO, Svensson J. The role of liver-derived insulin-like growth factor-I. Endocr Rev 2009; 30:494-535. [PMID: 19589948 PMCID: PMC2759708 DOI: 10.1210/er.2009-0010] [Citation(s) in RCA: 300] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IGF-I is expressed in virtually every tissue of the body, but with much higher expression in the liver than in any other tissue. Studies using mice with liver-specific IGF-I knockout have demonstrated that liver-derived IGF-I, constituting a major part of circulating IGF-I, is an important endocrine factor involved in a variety of physiological and pathological processes. Detailed studies comparing the impact of liver-derived IGF-I and local bone-derived IGF-I demonstrate that both sources of IGF-I can stimulate longitudinal bone growth. We propose here that liver-derived circulating IGF-I and local bone-derived IGF-I to some extent have overlapping growth-promoting effects and might have the capacity to replace each other (= redundancy) in the maintenance of normal longitudinal bone growth. Importantly, and in contrast to the regulation of longitudinal bone growth, locally derived IGF-I cannot replace (= lack of redundancy) liver-derived IGF-I for the regulation of a large number of other parameters including GH secretion, cortical bone mass, kidney size, prostate size, peripheral vascular resistance, spatial memory, sodium retention, insulin sensitivity, liver size, sexually dimorphic liver functions, and progression of some tumors. It is clear that a major role of liver-derived IGF-I is to regulate GH secretion and that some, but not all, of the phenotypes in the liver-specific IGF-I knockout mice are indirect, mediated via the elevated GH levels. All of the described multiple endocrine effects of liver-derived IGF-I should be considered in the development of possible novel treatment strategies aimed at increasing or reducing endocrine IGF-I activity.
Collapse
Affiliation(s)
- Claes Ohlsson
- Division of Endocrinology, Institute of Medicine, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Gevers EF, Hannah MJ, Waters MJ, Robinson ICAF. Regulation of rapid signal transducer and activator of transcription-5 phosphorylation in the resting cells of the growth plate and in the liver by growth hormone and feeding. Endocrinology 2009; 150:3627-36. [PMID: 19359380 DOI: 10.1210/en.2008-0985] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GH has physiological functions in many tissues, but the cellular targets for direct effects of GH remain ill defined in complex tissues such as the growth plate in which the contribution of direct vs. indirect actions of GH remains controversial. The Janus kinase (Jak)-signal transducer and activator of transcription (STAT)-5 pathway is activated by GH, so we developed a method to visualize nuclear Stat5b and phosphorylated Stat5 in single cells in response to a pulse of GH. Hep2 cells did not show a Stat5 phosphorylation (pY-Stat5) response to GH except in cells transfected to express GH receptors. ATDC5 cells express GH receptors and showed GH-induced pY-Stat5 responses, which varied with their state of chondrocyte differentiation. In vivo, Stat5b(+ve) nuclei were seen in the resting and prehypertrophic chondrocytes of the growth plate. After a single ip pulse of human GH or mouse GH, but not prolactin, pY-Stat5 responses were visible in cells in the resting zone and groove of Ranvier, 10-45 min later. Prehypertrophic chondrocytes showed no pY-Stat5 response to GH. GH target cells were also identified in other tissues, and a marked variability in spatiotemporal pY-Stat5 responses was evident. Endogenous hepatic pY-Stat5 was detected in mice with intact GH secretion but only during a GH pulse. Fasting and chronic exposure to GH attenuated the pY-Stat5 response to an acute GH injection. In conclusion, pY-Stat5 responses to GH vary in time and space, are sensitive to nutritional status, and may be inhibited by prior GH exposure. In the growth plate, our data provide direct in vivo support for an early role of GH to regulate the fate of immature chondrocytes.
Collapse
Affiliation(s)
- Evelien F Gevers
- Division of Molecular Neuroendocrinology, Medical Research Council National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom.
| | | | | | | |
Collapse
|
36
|
Fan Y, Menon RK, Cohen P, Hwang D, Clemens T, DiGirolamo DJ, Kopchick JJ, Le Roith D, Trucco M, Sperling MA. Liver-specific deletion of the growth hormone receptor reveals essential role of growth hormone signaling in hepatic lipid metabolism. J Biol Chem 2009; 284:19937-44. [PMID: 19460757 DOI: 10.1074/jbc.m109.014308] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Growth hormone (GH) plays a pivotal role in growth and metabolism, with growth promotion mostly attributed to generation of insulin-like growth factor I (IGF-I) in liver or at local sites of GH action, whereas the metabolic effects of GH are considered to be intrinsic to GH itself. To distinguish the effects of GH from those of IGF-I, we developed a Cre-lox-mediated model of tissue-specific deletion of the growth hormone receptor (GHR). Near total deletion of the GHR in liver (GHRLD) had no effect on total body or bone linear growth despite a >90% suppression of circulating IGF-I; however, total bone density was significantly reduced. Circulating GH was increased 4-fold, and GHRLD displayed insulin resistance, glucose intolerance, and increased circulating free fatty acids. Livers displayed marked steatosis, the result of increased triglyceride synthesis and decreased efflux; reconstitution of hepatic GHR signaling via adenoviral expression of GHR restored triglyceride output to normal, whereas IGF-I infusion did not correct steatosis despite restoration of circulating GH to normal. Thus, with near total absence of circulating IGF-I, GH action at the growth plate, directly and via locally generated IGF-I, can regulate bone growth, but at the expense of diabetogenic, lipolytic, and hepatosteatotic consequences. Our results indicate that IGF-I is essential for bone mineral density, whereas hepatic GH signaling is essential to regulate intrahepatic lipid metabolism. We propose that circulating IGF-I serves to amplify the growth-promoting effects of GH, while simultaneously dampening the catabolic effects of GH.
Collapse
Affiliation(s)
- Yong Fan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Eynon N, Yamin C, Ben-Sira D, Sagiv M. Optimal health and function among the elderly: lessening severity of ADL disability. Eur Rev Aging Phys Act 2009. [DOI: 10.1007/s11556-009-0048-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Abstract
Despite mounting evidence implicating sedentary behavior as a significant risk factor among the elderly, there is a limited amount of information on the type and amount of activity needed to promote optimal health and function in older people. Overall muscle strength and mass decline 30–50% between the ages of 30 and 80. The loss of muscle mass accounts for most of the observed loss of strength. The loss of muscle tissue is due to a decrease in the number of muscle fibers and to atrophy of the type II muscle fibers. The declining strength reduces the capacity to carry out basic activities of daily life and puts people at risk for falls and dependence on others. The objective of the present review is to examine the role of exercise training as a primary tool for increasing cardiopulmonary and muscular fitness in order to lessen the severity of disability in activities of daily living and to attain optimal health and functioning among the elderly.
Collapse
|
38
|
Hindmarsh PC, Brook CG. Auxological and biochemical assessment of short stature. ACTA PAEDIATRICA SCANDINAVICA. SUPPLEMENT 2008; 343:73-6. [PMID: 3057809 DOI: 10.1111/j.1651-2227.1988.tb10804.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Measurements of height and calculation of height velocity are highly accurate and sensitive to changes in a child's health. Definition of normality must be based on height velocity, and it is not acceptable to label a child as short and normal purely on the basis of a biochemical test unless this is substantiated with the demonstration of a normal height velocity. Response to therapy is dependent on the child's pretreatment growth rate and also on the amount of GH administered and the frequency of administration.
Collapse
|
39
|
Isaksson OG, Lindahl A, Nilsson A, Isgaard J. Action of growth hormone: current views. ACTA PAEDIATRICA SCANDINAVICA. SUPPLEMENT 2008; 343:12-8. [PMID: 3057803 DOI: 10.1111/j.1651-2227.1988.tb10794.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- O G Isaksson
- Department of Physiology, University of Gothenburg, Sweden
| | | | | | | |
Collapse
|
40
|
Abstract
Abstract
The growth hormone–insulin-like growth factor I (IGF-I) axis is an important physiological regulator muscle for development. Although there is evidence that aging muscle retains the ability to synthesize IGF-I, there is also evidence that aging may be associated with attenuation of the ability of exercise to induce an isoform of IGF-I that promotes satellite cell proliferation. However, it is clear that overexpression of IGF-I in the muscle can protect against age-related sarcopenia. Strength training appears to be the intervention of choice for the prevention and treatment of sarcopenia. IGF-I has been implicated in the loss of the muscle with age, and IGF-I expression levels change as a consequence of strength training in older adults. However, it seems that advancing age, rather than declining serum levels of IGF-I, appears to be a major determinant of life-time changes in body composition in women and men. We concluded that resistive exercise is a significant determinant of muscle mass and function. Elevated levels of IGF-I have been found in physically active compared to sedentary individuals. Recent work suggests that IGF-I as a mediator plays an important role in muscle hypertrophy and angiogenesis, both of which characterize the anabolic adaptation of muscles to exercise.
Collapse
|
41
|
Hutchison MR, Bassett MH, White PC. Insulin-like growth factor-I and fibroblast growth factor, but not growth hormone, affect growth plate chondrocyte proliferation. Endocrinology 2007; 148:3122-30. [PMID: 17395707 DOI: 10.1210/en.2006-1264] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Of the many factors that regulate linear growth, IGF-I has a central role in epiphyseal chondrocyte development. Whether IGF-I is solely of systemic or also of local origin is uncertain, as is how other growth factors interact with IGF-I at the growth plate. We studied the proliferative effects of IGF-I on juvenile bovine epiphyseal chondrocytes fractionated by density gradient centrifugation. Cell density correlated with size, glycogen content, and gene expression patterns. There was a gradient of response to IGF-I, with the greatest proliferative response in high-density cells corresponding to the reserve zone, as measured by [3H]thymidine uptake. Low-density (hypertrophic zone) cells proliferated only when exposed to IGF-I and basic fibroblast growth factor (FGF). The gradient of IGF-I response correlated with [125I]IGF-I binding as determined by Scatchard analysis: IGF-I receptor number was 10-fold greater in reserve zone cells than in hypertrophic cells. When exposed to basic FGF for 24 hours, IGF-I binding in hypertrophic cells increased 3-fold. In contrast, no specific binding of GH was demonstrated in juvenile bovine chondrocytes. GH produced neither signal transducer and activator of transcription phosphorylation, increased proliferation, nor increased IGF-I mRNA levels in any chondrocyte fraction. IGF-I mRNA levels were extremely low at 800-1100 copies/microg 18S RNA in bovine chondrocytes. These results suggest that the major regulator of chondrocyte proliferation is systemic IGF-I; FGFs may influence the actions of IGF-I at the growth plate by altering its receptor number in chondrocytes.
Collapse
Affiliation(s)
- Michele R Hutchison
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9063, USA.
| | | | | |
Collapse
|
42
|
Barbosa APF, Silva JDP, Fonseca EC, Lopez PM, Fernandes MBC, Balduino A, Duarte MEL. Response of the growth plate of uremic rats to human growth hormone and corticosteroids. Braz J Med Biol Res 2007; 40:1101-9. [PMID: 17665047 DOI: 10.1590/s0100-879x2006005000134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Accepted: 05/08/2007] [Indexed: 11/22/2022] Open
Abstract
Children with chronic renal failure in general present growth retardation that is aggravated by corticosteroids. We describe here the effects of methylprednisolone (MP) and recombinant human growth hormone (rhGH) on the growth plate (GP) of uremic rats. Uremia was induced by subtotal nephrectomy in 30-day-old rats, followed by 20 IU kg-1 day-1 rhGH (N = 7) or 3 mg kg-1 day-1 MP (N = 7) or 20 IU kg-1 day-1 rhGH + 3 mg kg-1 day-1 MP (N = 7) treatment for 10 days. Control rats with intact renal function were sham-operated and treated with 3 mg kg-1 day-1 MP (N = 7) or vehicle (N = 7). Uremic rats (N = 7) were used as untreated control animals. Structural alterations in the GP and the expression of anti-proliferating cell nuclear antigen (PCNA) and anti-insulin-like growth factor I (IGF-I) by epiphyseal chondrocytes were evaluated. Uremic MP rats displayed a reduction in the proliferative zone height (59.08 +/- 4.54 vs 68.07 +/- 7.5 microm, P < 0.05) and modifications in the microarchitecture of the GP. MP and uremia had an additive inhibitory effect on the proliferative activity of GP chondrocytes, lowering the expression of PCNA (19.48 +/- 11.13 vs 68.64 +/- 7.9% in control, P < 0.0005) and IGF-I (58.53 +/- 0.96 vs 84.78 +/- 2.93% in control, P < 0.0001), that was counteracted by rhGH. These findings suggest that in uremic rats rhGH therapy improves longitudinal growth by increasing IGF-I synthesis in the GP and by stimulating chondrocyte proliferation.
Collapse
Affiliation(s)
- A P F Barbosa
- Departamento de Patologia, Universidade de Ciências e Saúde de Alagoas, Maceió, AL, Brazil
| | | | | | | | | | | | | |
Collapse
|
43
|
Giannoni P, Cancedda R. Articular chondrocyte culturing for cell-based cartilage repair: needs and perspectives. Cells Tissues Organs 2007; 184:1-15. [PMID: 17190975 DOI: 10.1159/000096946] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2006] [Indexed: 01/13/2023] Open
Abstract
Articular cartilage displays a limited capacity of self-regeneration after injury. Thus, the biology of this tissue and its cellular components - the chondrocytes - has become the focus of several investigations, driven by tissue engineering and the basic and clinical research fields, aiming to ameliorate the present clinical approaches to cartilage repair. In this work, we present a brief recapitulation of the events that lead to cartilage development during the skeletal embryonal growth. The intrinsic phenotypic plasticity of the mesenchymal precursors and the adult chondrocytes is evaluated, dependent on the cell source, its physiopathological state, and as a function of the donor's age. The phenotypic changes induced by the basic culturing techniques are also taken into account, thus highlighting the phenotypic plasticity of the chondrocyte as the main property which could couple the differentiation process to the repair process. Chondrocyte proliferation and the contemporary maintenance of the chondrogenic differentiation potential are regarded as the two primary goals to be achieved in order to fulfill the quantitative needs of the clinical applications and the qualitative requirements of a properly repaired tissue. In this light, the effects of several growth factors and medium supplements are investigated. Finally, the latest improvements in culturing conditions and their possible clinical applications are presented as well.
Collapse
|
44
|
Huang TS, Yang RS, Tsai TW, Liu SH. Growth hormone cannot enhance the recovery of dexamethasone-induced osteopenia after withdrawal in young female wistar rats. TOHOKU J EXP MED 2005; 204:257-66. [PMID: 15572851 DOI: 10.1620/tjem.204.257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dexamethasone (DEX) suppresses the secretion of and responsiveness to growth hormone (GH). Here we aimed to assess the therapeutic effects of GH on the DEX-induced osteopenia. Female Wistar rats were treated for 2 weeks with DEX (200 microg/day) or saline as a control. DEX significantly decreased body weight gain, bone mineral density (BMD), growth plate thickness, area ratio of trabecular bone, and serum osteocalcin levels. DEX also elongated the tibia primary spongiosa and caused many tiny lipid droplets in the tibia marrow. These results indicated that DEX induced osteopenia in rats. We then assessed the effects of GH on the recovery of osteopenia after withdrawal of DEX. DEX-treated rats were subsequently treated for 1 week with GH (0.1 or 0.3 U/day) or saline, while saline-pretreated rats were treated for 1 week with saline as a control. GH (0.1 or 0.3 U/day)-treated rats showed a catch-up growth in various bone measurements by one week after DEX withdrawal, though most of them remained subnormal. GH treatment did not enhance the recovery of DEX-induced osteopenia. Therefore a short-term exposure to DEX significantly impaired the bone metabolism, which started to recover soon after withdrawal of DEX. Unfortunately, immediate administration of GH after withdrawal of DEX did not enhance the recovery process.
Collapse
Affiliation(s)
- Tien-Shang Huang
- Department of Internal Medicine, National Taiwan University & Hospital, Taipei, Taiwan
| | | | | | | |
Collapse
|
45
|
Caminos JE, Gualillo O, Lago F, Otero M, Blanco M, Gallego R, Garcia-Caballero T, Goldring MB, Casanueva FF, Gomez-Reino JJ, Dieguez C. The endogenous growth hormone secretagogue (ghrelin) is synthesized and secreted by chondrocytes. Endocrinology 2005; 146:1285-92. [PMID: 15576457 DOI: 10.1210/en.2004-1379] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ghrelin, the endogenous ligand for the GH secretagogue receptor (GHS-R), is a recently isolated hormone, prevalently expressed in stomach but also in other tissues such as hypothalamus and placenta. This novel acylated peptide acts at a central level to stimulate GH secretion and, notably, to regulate food intake. However, the existence of further, as yet unknown, effects or presence of ghrelin in peripheral tissues cannot be ruled out. In this report, we provide clear evidence for the expression of ghrelin peptide and mRNA in human, mouse, and rat chondrocytes. Immunoreactive ghrelin was identified by immunohistochemistry in rat cartilage, being localized prevalently in proliferative and maturative zone of the epiphyseal growth plate, and in mouse and human chondrocytic cell lines. Moreover, ghrelin mRNA was detected by RT-PCR and confirmed by Southern analysis in rat cartilage as well as in mouse and human chondrocytes cell lines. Ghrelin mRNA expression has been studied in rat along early life development showing a stable profile of expression throughout. Although ghrelin expression in chondrocytes suggests the presence of an unexpected autocrine/paracrine pathway, we failed to identify the functional GH secretagogue receptor type 1A by RT-PCR. On the other hand, binding analysis with 125I ghrelin suggests the presence of specific receptors different from the 1A isotype. Scatchard analysis revealed the presence of two receptors with respectively high and low affinity. Finally, ghrelin, in vitro, was able to significantly stimulate cAMP production and inhibits chondrocytes metabolic activity both in human and murine chondrocytes. In addition, ghrelin is able to actively decrease both spontaneous or insulin-induced long chain fatty acid uptake in human and mouse chondrocytes. This study is the first to provide evidence for the presence of this novel peptide in chondrocytes and suggests novel potential roles for this newly recognized component of the GH axis in cartilage metabolism.
Collapse
Affiliation(s)
- J E Caminos
- Department of Physiology, University of Santiago de Compostela, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yamanouchi K, Yada E, Hozumi H, Ueno C, Nishihara M. Analyses of hind leg skeletons in human growth hormone transgenic rats. Exp Gerontol 2004; 39:1179-88. [PMID: 15288692 DOI: 10.1016/j.exger.2004.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2004] [Revised: 04/22/2004] [Accepted: 04/30/2004] [Indexed: 11/17/2022]
Abstract
Growth hormone (GH) is essential in the development and growth of the skeleton and for the maintenance of bone mass and density, and its secretion is known to decline with aging. We have previously produced transgenic rats with low circulating GH that represent several age-associated phenotypes such as obesity, insulin-resistance and leptin-resistance. In the present study, the cross-sectional area, bone mineral density, and strength indexes of the hind leg skeletons of the transgenic rats were examined by an X-ray computed tomography scanning. The mean cross-sectional area of the transgenic rats showed no increase after 2 months old up to 8 months old and the strength indexes were significantly lower than their non-transgenic siblings at all ages examined. The trabecular bone mineral density in the transgenic rats drastically decreased at 8 months old, while the cortical bone mineral density was comparable to the non-transgenic rats, suggesting the onset of osteoporosis at this period. The results obtained in this study indicate that the transgenic rats could be useful model to gain insight into the complex mechanism leading to osteoporosis with aging.
Collapse
Affiliation(s)
- Keitaro Yamanouchi
- Department of Veterinary Physiology, Veterinary Medical Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | | | | | | | | |
Collapse
|
47
|
Abe I, Ochiai N, Ichimura H, Tsujino A, Sun J, Hara Y. Internodes can nearly double in length with gradual elongation of the adult rat sciatic nerve. J Orthop Res 2004; 22:571-7. [PMID: 15099637 DOI: 10.1016/j.orthres.2003.08.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2003] [Accepted: 08/25/2003] [Indexed: 02/04/2023]
Abstract
Leg lengthening procedure is used increasingly to treat leg length discrepancy and some forms of dwarfism. We investigated adaptation in rat sciatic nerve to the gradual nerve elongation that occurs with leg lengthening. Indirect nerve elongation was produced by leg lengthening by a total of 15, 30, 45, or 70 mm at a rate of 1 mm/day. One day after leg lengthening completion, transverse semithin sections of sciatic nerve were prepared and examined; a teased-fiber study also was performed. Elongation decreased axon diameter, but not significantly. In teased-fiber preparations, internodal length was increased by 93%, and the longest internode measured 3000 microm after leg lengthening by 70 mm. Slopes of fiber diameter-internodal length regression lines increased with increasing elongation. Paranodal demyelination caused by nerve elongation worsened as elongation increased, stimulating remyelination (i.e., intercalation of a segment). Only 0.8% of axons showed degeneration in the group with 70 mm of elongation. We concluded that adult rat sciatic nerve can adapt itself to leg lengthening procedure with even doubling internodal length.
Collapse
Affiliation(s)
- Ichiro Abe
- Department of Orthopedic Surgery, University of Tsukuba, 1-1-1 Tennodai, Tsukuba-shi, Ibaraki 305-8575, Japan.
| | | | | | | | | | | |
Collapse
|
48
|
Olney RC, Wang J, Sylvester JE, Mougey EB. Growth factor regulation of human growth plate chondrocyte proliferation in vitro. Biochem Biophys Res Commun 2004; 317:1171-82. [PMID: 15094393 DOI: 10.1016/j.bbrc.2004.03.170] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Indexed: 11/25/2022]
Abstract
Linear growth occurs as the result of growth plate chondrocytes undergoing proliferative and hypertrophic phases. Paracrine feedback loops that regulate the entry of chondrocytes into the hypertrophic phase have been shown and similar pathways likely exist for the proliferative phase. Human long-bone growth plate chondrocytes were cultured in vitro. The proliferative effects of a variety of factors were determined by [3H]thymidine uptake and the gene expression profile of these cells was determined by DNA microarray analysis. Serum, insulin-like growth factor (IGF)-I and -II, transforming growth factor-beta (TGF-beta, fibroblast growth factor (FGF)-1, -2, and -18, and platelet-derived growth factor (PDGF)-BB were potent stimulators of proliferation. FGF-10, testosterone, and bone morphogenetic proteins (BMP)-2, -4, and -6 inhibited proliferation. Microarray analysis showed that the genes for multiple members of the IGF-I, TGF-beta, FGF, and BMP pathways were expressed, suggesting the presence of autocrine/paracrine pathways that regulate the proliferative phase of growth plate-mediated growth.
Collapse
Affiliation(s)
- Robert C Olney
- Cellular and Molecular Medicine Laboratory, The Nemours Children's Clinic, 807 Children's Way, Jacksonville, FL 32207, USA.
| | | | | | | |
Collapse
|
49
|
Berlanga J, Infante J, Capó V, de la Fuente J, Castro FO. Characterization of transgenic mice lineages. I. Overexpression of hGH causes the formation of liver intranuclear pseudoinclusion bodies and renal and hepatic injury. ACTA ACUST UNITED AC 2004. [DOI: 10.1002/abio.370130409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
50
|
Abstract
The growth plate is the final target organ for longitudinal growth and results from chondrocyte proliferation and differentiation. During the first year of life, longitudinal growth rates are high, followed by a decade of modest longitudinal growth. The age at onset of puberty and the growth rate during the pubertal growth spurt (which occurs under the influence of estrogens and GH) contribute to sex difference in final height between boys and girls. At the end of puberty, growth plates fuse, thereby ceasing longitudinal growth. It has been recognized that receptors for many hormones such as estrogen, GH, and glucocorticoids are present in or on growth plate chondrocytes, suggesting that these hormones may influence processes in the growth plate directly. Moreover, many growth factors, i.e., IGF-I, Indian hedgehog, PTHrP, fibroblast growth factors, bone morphogenetic proteins, and vascular endothelial growth factor, are now considered as crucial regulators of chondrocyte proliferation and differentiation. In this review, we present an update on the present perception of growth plate function and the regulation of chondrocyte proliferation and differentiation by systemic and local regulators of which most are now related to human growth disorders.
Collapse
Affiliation(s)
- B C J van der Eerden
- Department of Pediatrics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | | |
Collapse
|