1
|
Gopalakrishnan J, Feistel K, Friedrich BM, Grapin‐Botton A, Jurisch‐Yaksi N, Mass E, Mick DU, Müller R, May‐Simera H, Schermer B, Schmidts M, Walentek P, Wachten D. Emerging principles of primary cilia dynamics in controlling tissue organization and function. EMBO J 2023; 42:e113891. [PMID: 37743763 PMCID: PMC10620770 DOI: 10.15252/embj.2023113891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/07/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
Primary cilia project from the surface of most vertebrate cells and are key in sensing extracellular signals and locally transducing this information into a cellular response. Recent findings show that primary cilia are not merely static organelles with a distinct lipid and protein composition. Instead, the function of primary cilia relies on the dynamic composition of molecules within the cilium, the context-dependent sensing and processing of extracellular stimuli, and cycles of assembly and disassembly in a cell- and tissue-specific manner. Thereby, primary cilia dynamically integrate different cellular inputs and control cell fate and function during tissue development. Here, we review the recently emerging concept of primary cilia dynamics in tissue development, organization, remodeling, and function.
Collapse
Affiliation(s)
- Jay Gopalakrishnan
- Institute for Human Genetics, Heinrich‐Heine‐UniversitätUniversitätsklinikum DüsseldorfDüsseldorfGermany
| | - Kerstin Feistel
- Department of Zoology, Institute of BiologyUniversity of HohenheimStuttgartGermany
| | | | - Anne Grapin‐Botton
- Cluster of Excellence Physics of Life, TU DresdenDresdenGermany
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at The University Hospital Carl Gustav Carus and Faculty of Medicine of the TU DresdenDresdenGermany
| | - Nathalie Jurisch‐Yaksi
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
| | - Elvira Mass
- Life and Medical Sciences Institute, Developmental Biology of the Immune SystemUniversity of BonnBonnGermany
| | - David U Mick
- Center for Molecular Signaling (PZMS), Center of Human and Molecular Biology (ZHMB)Saarland School of MedicineHomburgGermany
| | - Roman‐Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD), Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Helen May‐Simera
- Institute of Molecular PhysiologyJohannes Gutenberg‐UniversityMainzGermany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD), Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Miriam Schmidts
- Pediatric Genetics Division, Center for Pediatrics and Adolescent MedicineUniversity Hospital FreiburgFreiburgGermany
- CIBSS‐Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
| | - Peter Walentek
- CIBSS‐Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
- Renal Division, Internal Medicine IV, Medical CenterUniversity of FreiburgFreiburgGermany
| | - Dagmar Wachten
- Institute of Innate Immunity, Biophysical Imaging, Medical FacultyUniversity of BonnBonnGermany
| |
Collapse
|
2
|
Physical Activity, Exercise, and Sports in Individuals with Skeletal Dysplasia: What Is Known about Their Benefits? SUSTAINABILITY 2022. [DOI: 10.3390/su14084487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
There is a lack of knowledge about the practice of physical activity, exercise, and sports in people with skeletal dysplasia (SD). This study aimed to characterize the physical fitness of people with SD; investigate the benefits of physical activity, exercise, or sports programs for people with SD; identify the adapted physical activities that can be prescribed to individuals with SD; and identify the most common and effective structural characteristics and guidelines for the evaluation of individuals with SD and corresponding activity prescriptions. Electronic searches were carried out in the PubMed, Scopus, SPORTDiscus, Psycinfo, and Web of Science databases in October 2021 and March 2022 and included papers published until 3 March 2022. The search strategy terms used were “dwarfism”, “dwarf”, “skeletal dysplasia”, “achondroplasia”, “pseudoachondroplasia”, “hypochondroplasia”, “campomelic dysplasia”, “hair cartilage hypoplasia”, “x-linked hypophosphatemia”, “metaphyseal chondrodysplasia schmid type”, “multiple epiphyseal dysplasia”, “three M syndrome”, “3-M syndrome”, “hypophosphatasia”, “fibrodysplasia ossificans progressive”, “type II collagen disorders”, “type II collagenopathies”, “type II collagenopathy”, “physical activity”, “exercise”, “sport”, “training”, and “physical fitness”, with the Boolean operators “AND” or “OR”. After reading the full texts of the studies, and according to previously defined eligibility criteria, fifteen studies met the inclusion criteria; however, there was not a single intervention study with physical exercise. Several cross-sectional, review, or qualitative studies presented a set of essential aspects that future intervention studies can consider when evaluating, prescribing, and implementing physical exercise programs, as they allowed the physical characterization of the SD population. This study demonstrated an apparent scarcity in the literature of experimental studies with physical exercise implementation in the SD population.
Collapse
|
3
|
Hussain S, Sun M, Guo Y, Mushtaq N, Zhao Y, Yuan Y, Hussain N, Osoro E, Suleiman A, Sadiq M, Zhang F, Han Y, Sun J, Lu S. SFMBT2 positively regulates SOX9 and chondrocyte proliferation. Int J Mol Med 2018; 42:3503-3512. [PMID: 30272322 DOI: 10.3892/ijmm.2018.3894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/19/2018] [Indexed: 11/06/2022] Open
Abstract
SRY‑box 9 (SOX9) is the master regulator of the chondrocyte phenotype, which is essential for differentiating chondrogenic mesenchymal condensations into chondrocytes, and is involved in regulating every stage of chondrocyte differentiation. SOX9 deletion in chondrocytes at the late stages of cartilage development results in decreased chondrocyte proliferation; inhibited expression of cartilage matrix genes, including Indian hedgehog and the downstream parathyroid hormone‑related protein; and premature conversion of proliferating chondrocytes into hypertrophic chondrocytes, which mineralize their matrix prematurely. Therefore, SOX9 is considered vital for the majority of phases of chondrocyte lineage, from early condensations to the differentiation of proliferating chondrocytes, leading to chondrocyte hypertrophy. It has been reported that SOX9 expression is decreased in osteoarthritis (OA) cartilage. Regeneration or repair of cartilage degradation in OA remains a challenge. Previous studies have indicated that overexpression of SOX9 can promote cartilage repair and can be used as a potential therapeutic agent at the early stages of human OA. The present study identified Scm‑like with four malignant brain tumor domains 2 (SFMBT2) as a novel regulator of SOX9 expression in human chondrocytes. Our previous study revealed that SFMBT2 is negatively regulated in OA cartilage, and decreased levels of SFMBT2 contribute to the catabolic phenotype of chondrocytes. The present study detected increased expression levels of SFMBT2 in early cartilage development and during the early phases of chondrogenesis. Overexpression of SFMBT2 in C28/I2 cells upregulated SOX9 expression in a dose‑dependent manner. Furthermore, SFMBT2 positively regulated C28/I2 cell proliferation and restored the decreased levels of SOX9 in chondrocytes following tumor necrosis factor‑α treatment. Additional studies may reveal novel insights into the molecular mechanism involved and the potential role of SFMBT2 in cartilage repair and OA management.
Collapse
Affiliation(s)
- Safdar Hussain
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Mengyao Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Yuanxu Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Nosheen Mushtaq
- Department of Microbiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Yitong Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Ying Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Nazim Hussain
- Centre for Applied Molecular Biology (CAMB), University of The Punjab, Lahore 53700, Pakistan
| | - Ezra Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Abubakar Suleiman
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Muhammad Sadiq
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Jian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
4
|
Kong Y, Guo Y, Zhang J, Zhao B, Wang J. Strontium Promotes Transforming Growth Factors β1 and β2 Expression in Rat Chondrocytes Cultured In Vitro. Biol Trace Elem Res 2018; 184:450-455. [PMID: 29170863 DOI: 10.1007/s12011-017-1208-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 01/18/2023]
Abstract
The transforming growth factors β1 (TGF-β1) and TGF-β2, as two distinct homodimers of TGF-β superfamily, involve in chondrocyte growth and differentiation. Emerging evidence has implied that strontium (Sr) plays an important role in the bone formation and resorption, and has strong effects on stimulating human cartilage matrix formation in vitro. However, the direct effects of Sr on TGF-β1 and TGF-β2 expressions in chondrocytes are not entirely clear. The purpose of this study was to evaluate the influence of different Sr concentrations on the expression of TGF-β1 and TGF-β2 in rat chondrocytes in vitro. Chondrocytes were isolated from Wistar rat articular by enzymatic digestion. Strontium chloride hexahydrate (SrCl2·6H2O) was used as a Sr source in this study. Sr was added to the culture solution at final concentrations of 0, 0.5, 1.0, 2.0, 5.0, 20.0, and 100 μg/mL. After 72 h of continuous culture, TGF-β1 and TGF-β2 mRNA abundance and protein expression levels in the chondrocytes were determined by real-time polymerase chain reaction (real-time PCR) and Western blot, respectively. The results showed that TGF-β1 and TGF-β2 expressions in chondrocytes increased dose-dependently with Sr concentration. The mRNA abundance of TGF-β1 and TGF-β2 were markedly higher than those observed for control (P < 0.01) when the Sr-treated concentration exceeded 1.0 and 5.0 μg/mL, respectively. The TGF-β1 and TGF-β2 protein expression levels were extremely significantly higher than those in the control group (P < 0.01) at above 5.0 μg/mL Sr-treatment. These results indicated that Sr could involve in the chondrocytes metabolism via regulating TGF-β1 and TGF-β2 signalling.
Collapse
Affiliation(s)
- Yezi Kong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yazhou Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jinfeng Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
5
|
Ding M, Jin L, Xie L, Park SH, Tong Y, Wu D, Chhabra AB, Fu Z, Li X. A Murine Model for Human ECO Syndrome Reveals a Critical Role of Intestinal Cell Kinase in Skeletal Development. Calcif Tissue Int 2018; 102:348-357. [PMID: 29098359 PMCID: PMC5820141 DOI: 10.1007/s00223-017-0355-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
Abstract
An autosomal-recessive inactivating mutation R272Q in the human intestinal cell kinase (ICK) gene caused profound multiplex developmental defects in human endocrine-cerebro-osteodysplasia (ECO) syndrome. ECO patients exhibited a wide variety of skeletal abnormalities, yet the underlying mechanisms by which ICK regulates skeletal development remained largely unknown. The goal of this study was to understand the structural and mechanistic basis underlying skeletal anomalies caused by ICK dysfunction. Ick R272Q knock-in transgenic mouse model not only recapitulated major ECO skeletal defects such as short limbs and polydactyly but also revealed a deformed spine with defective intervertebral disk. Loss of ICK function markedly reduced mineralization in the spinal column, ribs, and long bones. Ick mutants showed a significant decrease in the proliferation zone of long bones and the number of type X collagen-expressing hypertrophic chondrocytes in the spinal column and the growth plate of long bones. These results implicate that ICK plays an important role in bone and cartilage development by promoting chondrocyte proliferation and maturation. Our findings provided new mechanistic insights into the skeletal phenotype of human ECO and ECO-like syndromes.
Collapse
Affiliation(s)
- Mengmeng Ding
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Jin
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
| | - Lin Xie
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
- Department of Orthopaedic Surgery, Wuhan Orthopaedic Hospital, Huazhong University of Science & Technology, Hubei, 430030, China
| | - So Hyun Park
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
| | - Yixin Tong
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
- The Gastrointestinal Surgery Center, Tongji Hospital, Huazhong University of Science & Technology, Hubei, 430030, China
| | - Di Wu
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
| | - A Bobby Chhabra
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
| | - Zheng Fu
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA.
| | - Xudong Li
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA.
| |
Collapse
|
6
|
Gouveia CHA, Miranda-Rodrigues M, Martins GM, Neofiti-Papi B. Thyroid Hormone and Skeletal Development. VITAMINS AND HORMONES 2018; 106:383-472. [PMID: 29407443 DOI: 10.1016/bs.vh.2017.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thyroid hormone (TH) is essential for skeletal development from the late fetal life to the onset of puberty. During this large window of actions, TH has key roles in endochondral and intramembranous ossifications and in the longitudinal bone growth. There is evidence that TH acts directly in skeletal cells but also indirectly, specially via the growth hormone/insulin-like growth factor-1 axis, to control the linear skeletal growth and maturation. The presence of receptors, plasma membrane transporters, and activating and inactivating enzymes of TH in skeletal cells suggests that direct actions of TH in these cells are crucial for skeletal development, which has been confirmed by several in vitro and in vivo studies, including mouse genetic studies, and clinical studies in patients with resistance to thyroid hormone due to dominant-negative mutations in TH receptors. This review examines progress made on understanding the mechanisms by which TH regulates the skeletal development.
Collapse
Affiliation(s)
- Cecilia H A Gouveia
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| | | | - Gisele M Martins
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil; Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Bianca Neofiti-Papi
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
7
|
Abstract
Skeletal muscle and bone rely on a number of growth factors to undergo development, modulate growth, and maintain physiological strength. A major player in these actions is insulin-like growth factor I (IGF-I). However, because this growth factor can directly enhance muscle mass and bone density, it alters the state of the musculoskeletal system indirectly through mechanical crosstalk between these two organ systems. Thus, there are clearly synergistic actions of IGF-I that extend beyond the direct activity through its receptor. This review will cover the production and signaling of IGF-I as it pertains to muscle and bone, the chemical and mechanical influences that arise from IGF-I activity, and the potential for therapeutic strategies based on IGF-I. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
|
8
|
Rutkowski TP, Kohn A, Sharma D, Ren Y, Mirando AJ, Hilton MJ. HES factors regulate specific aspects of chondrogenesis and chondrocyte hypertrophy during cartilage development. J Cell Sci 2016; 129:2145-55. [PMID: 27160681 DOI: 10.1242/jcs.181271] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/05/2016] [Indexed: 12/11/2022] Open
Abstract
RBPjκ-dependent Notch signaling regulates multiple processes during cartilage development, including chondrogenesis, chondrocyte hypertrophy and cartilage matrix catabolism. Select members of the HES- and HEY-families of transcription factors are recognized Notch signaling targets that mediate specific aspects of Notch function during development. However, whether particular HES and HEY factors play any role(s) in the processes during cartilage development is unknown. Here, for the first time, we have developed unique in vivo genetic models and in vitro approaches demonstrating that the RBPjκ-dependent Notch targets HES1 and HES5 suppress chondrogenesis and promote the onset of chondrocyte hypertrophy. HES1 and HES5 might have some overlapping function in these processes, although only HES5 directly regulates Sox9 transcription to coordinate cartilage development. HEY1 and HEYL play no discernable role in regulating chondrogenesis or chondrocyte hypertrophy, whereas none of the HES or HEY factors appear to mediate Notch regulation of cartilage matrix catabolism. This work identifies important candidates that might function as downstream mediators of Notch signaling both during normal skeletal development and in Notch-related skeletal disorders.
Collapse
Affiliation(s)
- Timothy P Rutkowski
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Anat Kohn
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deepika Sharma
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yinshi Ren
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anthony J Mirando
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matthew J Hilton
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
9
|
Abstract
Osteoporosis is a global public health problem currently affecting more than 200 million people worldwide. Major research efforts are being made to improve the outcomes for patients with osteoporosis. However, the treatment of fractures associated with osteoporosis remains unsatisfactory. Animal models continue to be an important tool for establishing strategies to treat osteoporotic fractures, and various methods of inducing osteoporosis have been used. Investigators must select a model that best reflects the clinical problem being studied, and the underlying pathophysiology of the osteoporosis in the target patient group. In particular a model for Type I post-menopausal osteoporosis should mimic a fall in oestrogen and rise in osteoclast activity observed with this condition, whereas a model for type II 'senile' osteoporosis should mimic the fall in osteoblast activity. Unfortunately, there is no single all-encompassing model that precisely imitates the underlying osteoporosis or the fracture patterns seen in humans. As such the choice of species and model must be individualised to the scientific question being addressed. This article summarises general considerations when choosing an osteoporotic fracture model and outlines existing models of osteoporosis. The most appropriate model in a range of osteoporotic fracture research scenarios are subsequently considered.
Collapse
Affiliation(s)
- A Hamish Simpson
- Department of Trauma and Orthopaedics, The University of Edinburgh, Edinburgh, UK.
| | - Iain R Murray
- Department of Trauma and Orthopaedics, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
10
|
Bikle DD, Tahimic C, Chang W, Wang Y, Philippou A, Barton ER. Role of IGF-I signaling in muscle bone interactions. Bone 2015; 80:79-88. [PMID: 26453498 PMCID: PMC4600536 DOI: 10.1016/j.bone.2015.04.036] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/11/2015] [Accepted: 04/22/2015] [Indexed: 12/16/2022]
Abstract
Skeletal muscle and bone rely on a number of growth factors to undergo development, modulate growth, and maintain physiological strength. A major player in these actions is insulin-like growth factor I (IGF-I). However, because this growth factor can directly enhance muscle mass and bone density, it alters the state of the musculoskeletal system indirectly through mechanical crosstalk between these two organ systems. Thus, there are clearly synergistic actions of IGF-I that extend beyond the direct activity through its receptor. This review will cover the production and signaling of IGF-I as it pertains to muscle and bone, the chemical and mechanical influences that arise from IGF-I activity, and the potential for therapeutic strategies based on IGF-I. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Daniel D Bikle
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Candice Tahimic
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Wenhan Chang
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Yongmei Wang
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Anastassios Philippou
- National and Kapodistrian University of Athens, Department of Physiology, Medical School, Goudi-Athens, Greece
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
Abstract
Animal models are widely used to investigate the pathogenesis of osteoporosis and for the clinical testing of anti-resorptive drugs. However, osteoporotic fracture models designed to investigate novel ways to treat fractures of osteoporotic bone must fulfil requirements distinct from those of pharmacological testing. Bone strength and toughness, implant fixation and osteointegration and fracture repair are of particular interest. Osteoporotic models should reflect the underlying clinical scenario be that primary type 1 (post-menopausal) osteoporosis, primary type 2 (senile) osteoporosis or secondary osteoporosis. In each scenario, small and large animal models have been developed. While rodent models facilitate the study of fractures in strains specifically established to facilitate understanding of the pathologic basis of disease, concerns remain about the relevance of small animal fracture models to the human situation. There is currently no all-encompassing model, and the choice of species and model must be individualized to the scientific question being addressed.
Collapse
Affiliation(s)
- A Hamish Simpson
- Department of Trauma and Orthopaedics, The University of Edinburgh, Chancellors Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK,
| | | |
Collapse
|
12
|
Alarcón H, Ynsa MD, Dang ZY, Torres-Costa V, Manso-Silván M, Wu JF, Breese MBH, García-Ruiz JP. Conditioned bio-interfaces of silicon/porous silicon micro-patterns lead to the chondrogenesis of hMSCs. RSC Adv 2015. [DOI: 10.1039/c5ra09069e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
hMSCs find attractive both Si and PSi surfaces to develop cell-surface adhesions which are needed in differentiation and the presence of CM-hMSCs bio-interface improves the differentiation process with respect to a control PSi surface.
Collapse
Affiliation(s)
- H. Alarcón
- Molecular Biology Department
- Universidad Autónoma de Madrid
- 28049 Madrid
- Spain
| | - M. D. Ynsa
- Department of Applied Physics and Instituto Nicolás Cabrera
- Universidad Autónoma de Madrid
- 28049 Madrid
- Spain
- Centro de Micro-Análisis de Materiales (CMAM)
| | - Z. Y. Dang
- Centre for Ion Beam Applications (CIBA)
- Department of Physics
- National University of Singapore
- Singapore 117542
| | - V. Torres-Costa
- Department of Applied Physics and Instituto Nicolás Cabrera
- Universidad Autónoma de Madrid
- 28049 Madrid
- Spain
- Centro de Micro-Análisis de Materiales (CMAM)
| | - M. Manso-Silván
- Department of Applied Physics and Instituto Nicolás Cabrera
- Universidad Autónoma de Madrid
- 28049 Madrid
- Spain
| | - J. F. Wu
- Centre for Ion Beam Applications (CIBA)
- Department of Physics
- National University of Singapore
- Singapore 117542
| | - M. B. H. Breese
- Centre for Ion Beam Applications (CIBA)
- Department of Physics
- National University of Singapore
- Singapore 117542
| | - J. P. García-Ruiz
- Molecular Biology Department
- Universidad Autónoma de Madrid
- 28049 Madrid
- Spain
| |
Collapse
|
13
|
Tanasawet S, Withyachumnarnkul B, Changsangfar C, Cummins SF, Sroyraya M, Sangsuwan P, Kitiyanant Y, Asuvapongpatana S, Weerachatyanukul W. Isolation of Organic Matrix Nacreous Proteins fromHaliotis diversicolorand Their Effect OnIn VitroOsteoinduction. MALACOLOGIA 2013. [DOI: 10.4002/040.056.0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
14
|
Wang Y, Bikle DD, Chang W. Autocrine and Paracrine Actions of IGF-I Signaling in Skeletal Development. Bone Res 2013; 1:249-59. [PMID: 26273506 DOI: 10.4248/br201303003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 06/29/2013] [Indexed: 11/10/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) regulates cell growth, survival, and differentiation by acting on the IGF-I receptor, (IGF-IR)-a tyrosine kinase receptor, which elicits diverse intracellular signaling responses. All skeletal cells express IGF-I and IGF-IR. Recent studies using tissue/cell-specific gene knockout mouse models and cell culture techniques have clearly demonstrated that locally produced IGF-I is more critical than the systemic IGF-I in supporting embryonic and postnatal skeletal development and bone remodeling. Local IGF-I/IGF-IR signaling promotes the growth, survival and differentiation of chondrocytes and osteoblasts, directly and indirectly, by altering other autocrine/paracrine signaling pathways in cartilage and bone, and by enhancing interactions among these skeletal cells through hormonal and physical means. Moreover, local IGF-I/IGF-IR signaling is critical for the anabolic bone actions of growth hormone and parathyroid hormone. Herein, we review evidence supporting the actions of local IGF-I/IGF-IR in the above aspects of skeletal development and remodeling.
Collapse
Affiliation(s)
- Yongmei Wang
- Endocrine Unit, University of California, San Francisco, Veterans Affairs Medical Center , San Francisco, CA, USA
| | - Daniel D Bikle
- Endocrine Unit, University of California, San Francisco, Veterans Affairs Medical Center , San Francisco, CA, USA
| | - Wenhan Chang
- Endocrine Unit, University of California, San Francisco, Veterans Affairs Medical Center , San Francisco, CA, USA
| |
Collapse
|
15
|
Abstract
Mutations that exaggerate signalling of the receptor tyrosine kinase fibroblast growth factor receptor 3 (FGFR3) give rise to achondroplasia, the most common form of dwarfism in humans. Here we review the clinical features, genetic aspects and molecular pathogenesis of achondroplasia and examine several therapeutic strategies designed to target the mutant receptor or its signalling pathways, including the use of kinase inhibitors, blocking antibodies, physiologic antagonists, RNAi and chaperone inhibitors. We conclude by discussing the challenges of treating growth plate disorders in children.
Collapse
|
16
|
Yan Q, Feng Q, Beier F. Reduced chondrocyte proliferation, earlier cell cycle exit and increased apoptosis in neuronal nitric oxide synthase-deficient mice. Osteoarthritis Cartilage 2012; 20:144-51. [PMID: 22179029 DOI: 10.1016/j.joca.2011.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 11/21/2011] [Accepted: 11/24/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Nitric oxide (NO) has been implicated in the local regulation of bone metabolism. However, the contribution made by specific nitric oxide synthase (NOS) enzymes to skeletal development is unclear. The objective of this study was to examine the effects of inactivation of neuronal nitric oxide synthase (nNOS) on cartilage development in mice. DESIGN Mice carrying a null mutation in the nNOS gene were used to address our objectives. Histological staining, immunohistochemistry and in situ analyses were employed along with real-time reverse transcriptase - polymerase chain reaction (RT-PCR). RESULTS nNOS-null mice show transient growth retardation and shorter long bones. nNOS-deficient growth plates show a reduction in replicating cells. Reduced chondrocyte numbers may in part be due to slower cell cycle progression and premature cell cycle exit caused by decreased cyclin D1 and increased p57 expression in mutants. In addition, apoptosis was increased as shown by increased cleaved-caspase 3 staining in hypertrophic chondrocytes in mutants. Real-time PCR demonstrated that expression of early chondrocyte markers such as Sox genes was reduced in mutant mice, while expression of prehypertrophic markers such as RORα was increased. Histological sections also demonstrated thinner cortical bone, fewer trabeculae and reduced mineralization in mutant mice. CONCLUSIONS These data identify an important role of nNOS in chondrocyte proliferation and endochondral bone growth and demonstrate that nNOS coordinates cell cycle exit and chondrocyte differentiation in cartilage development.
Collapse
Affiliation(s)
- Q Yan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
17
|
Elefteriou F, Yang X. Genetic mouse models for bone studies--strengths and limitations. Bone 2011; 49:1242-54. [PMID: 21907838 PMCID: PMC3331798 DOI: 10.1016/j.bone.2011.08.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 08/15/2011] [Accepted: 08/18/2011] [Indexed: 11/25/2022]
Abstract
Mice have become a preferred model system for bone research because of their genetic and pathophysiological similarities to humans: a relatively short reproductive period, leading to relatively low cost of maintenance and the availability of the entire mouse genome sequence information. The success in producing the first transgenic mouse line that expressed rabbit β-globin protein in mouse erythrocytes three decades ago marked the beginning of the use of genetically engineered mice as model system to study human diseases. Soon afterward the development of cultured pluripotent embryonic stem cells provided the possibility of gene replacement or gene deletion in mice. These technologies have been critical to identify new genes involved in bone development, growth, remodeling, repair, and diseases, but like many other approaches, they have limitations. This review will introduce the approaches that allow the generation of transgenic mice and global or conditional (tissue-specific and inducible) mutant mice. A list of the various promoters used to achieve bone-specific gene deletion or overexpression is included. The limitations of these approaches are discussed, and general guidelines related to the analysis of genetic mouse models are provided.
Collapse
Affiliation(s)
- Florent Elefteriou
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| | - Xiangli Yang
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| |
Collapse
|
18
|
Akiyama M, Zhou M, Sugimoto R, Hongu T, Furuya M, Funakoshi Y, Kato M, Hasegawa H, Kanaho Y. Tissue- and development-dependent expression of the small GTPase Arf6 in mice. Dev Dyn 2010; 239:3416-35. [DOI: 10.1002/dvdy.22481] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
19
|
Abstract
PURPOSE OF REVIEW Although the genetic defect underlying achondroplasia has been known for over a decade, no effective therapies to stimulate bone growth have emerged. Here we review the recent literature and summarize the molecular mechanisms underlying disease pathology and examine their potential as therapeutic targets. Currently used preclinical models are discussed in the context of recent advances with a special focus on C-type natriuretic peptide. RECENT FINDINGS Research on the mutation in Fibroblast Growth Factor Receptor 3 (FGFR3) that causes achondroplasia suggests that disease results from increased signal transduction from the mutant receptor. Thus, current therapeutic strategies have focused on reducing signals emanating from FGFR3. First-generation therapies directly targeting FGFR3, such as kinase inhibitors and neutralizing antibodies, designed for targeting FGFR3 in cancer, are still in the preclinical phase and have yet to translate into the management of achondroplasia. Counteracting signal transduction pathways downstream of FGFR3 holds promise with the discovery that administration of C-type natriuretic peptide to achondroplastic mice ameliorates their clinical phenotype. However, more research into long-term effectiveness and safety of this strategy is needed. Direct targeting of therapeutic agents to growth plate cartilage may enhance efficacy and minimize side effects of these and future therapies. SUMMARY Current research into the pathogenesis of achondroplasia has expanded our understanding of the mechanisms of FGFR3-induced disease and has increased the number of approaches that we may use to potentially correct it. Further research is needed to validate these approaches in preclinical models of achondroplasia.
Collapse
Affiliation(s)
- Melanie B Laederich
- Research Center, Shriners Hospital for Children, Department of Cell & Developmental Biology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | |
Collapse
|
20
|
Skeletal dysplasias associated with mild myopathy-a clinical and molecular review. J Biomed Biotechnol 2010; 2010:686457. [PMID: 20508815 PMCID: PMC2875749 DOI: 10.1155/2010/686457] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 03/15/2010] [Indexed: 11/17/2022] Open
Abstract
Musculoskeletal system is a complex assembly of tissues which acts as scaffold for the body and enables locomotion. It is often overlooked that different components of this system may biomechanically interact and affect each other. Skeletal dysplasias are diseases predominantly affecting the development of the osseous skeleton. However, in some cases skeletal dysplasia patients are referred to neuromuscular clinics prior to the correct skeletal diagnosis. The muscular complications seen in these cases are usually mild and may stem directly from the muscle defect and/or from the altered interactions between the individual components of the musculoskeletal system. A correct early diagnosis may enable better management of the patients and a better quality of life. This paper attempts to summarise the different components of the musculoskeletal system which are affected in skeletal dysplasias and lists several interesting examples of such diseases in order to enable better understanding of the complexity of human musculoskeletal system.
Collapse
|
21
|
Kake T, Kitamura H, Adachi Y, Yoshioka T, Watanabe T, Matsushita H, Fujii T, Kondo E, Tachibe T, Kawase Y, Jishage KI, Yasoda A, Mukoyama M, Nakao K. Chronically elevated plasma C-type natriuretic peptide level stimulates skeletal growth in transgenic mice. Am J Physiol Endocrinol Metab 2009; 297:E1339-48. [PMID: 19808910 DOI: 10.1152/ajpendo.00272.2009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
C-type natriuretic peptide (CNP) plays a critical role in endochondral ossification through guanylyl cyclase-B (GC-B), a natriuretic peptide receptor subtype. Cartilage-specific overexpression of CNP enhances skeletal growth and rescues the dwarfism in a transgenic achondroplasia model with constitutive active mutation of fibroblast growth factor receptor-3. For future clinical application, the efficacy of CNP administration on skeletal growth must be evaluated. Due to the high clearance of CNP, maintaining a high concentration is technically difficult. However, to model high blood CNP concentration, we established a liver-targeted CNP-overexpressing transgenic mouse (SAP-CNP tgm). SAP-CNP tgm exhibited skeletal overgrowth in proportion to the blood CNP concentration and revealed phenotypes of systemic stimulation of cartilage bones, including limbs, paws, costal bones, spine, and skull. Furthermore, in SAP-CNP tgm, the size of the foramen magnum, the insufficient formation of which results in cervico-medullary compression in achondroplasia, also showed significant increase. CNP primarily activates GC-B, but under high concentrations it cross-reacts with guanylyl cyclase-A (GC-A), a natriuretic peptide receptor subtype of atrial natriuretic peptides (ANP) and brain natriuretic peptides (BNP). Although activation of GC-A could alter cardiovascular homeostasis, leading to hypotension and heart weight reduction, the skeletal overgrowth phenotype in the line of SAP-CNP tgm with mild overexpression of CNP did not accompany decrease of systolic blood pressure or heart weight. These results suggest that CNP administration stimulates skeletal growth without adverse cardiovascular effect, and thus CNP could be a promising remedy targeting achondroplasia.
Collapse
Affiliation(s)
- Takei Kake
- Pharmaceutical Research Department I, Research Division, Chugai Pharmaceutical Company, Shizuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chattah NLT, Sharir A, Weiner S, Shahar R. Determining the elastic modulus of mouse cortical bone using electronic speckle pattern interferometry (ESPI) and micro computed tomography: a new approach for characterizing small-bone material properties. Bone 2009; 45:84-90. [PMID: 19332167 DOI: 10.1016/j.bone.2009.03.664] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 03/09/2009] [Accepted: 03/11/2009] [Indexed: 12/20/2022]
Abstract
Mice phenotypes are invaluable for understanding bone formation and function, as well as bone disease. The elastic modulus is an important property of bones that can provide insights into bone quality. The determination of the elastic modulus of mouse cortical bone is complicated by the small dimensions of the bones. Whole bone bending tests are known to under estimate the elastic modulus compared to nanoindentation tests. The latter however provides information on extremely localized areas that do not necessarily correspond to the bulk elastic modulus under compression. This study presents a novel method for determining the bulk or effective elastic modulus of mouse cortical bone using the femur. We use Electronic Speckle Pattern Interferometry (ESPI), an optical method that enables the measurement of displacements on the bone surface, as it is compressed under water. This data is combined with geometric information obtained from micro-CT to calculate the elastic modulus. Roughly tubular cortical bone segments (2 mm) were cut from the diaphyses of femora of four week old C57BL/6 (B6) female mice and compressed axially using a mechanical tension-compression device. Displacements in the loading direction were mapped on the bone surface after loading the specimen. A linear regression of the displacement vs. axial-position enabled the calculation of the effective strain. Effective stress was calculated using force (N) data from the system's load cell and the mean cross-sectional area of the sample as determined by micro-CT. The effective elastic modulus (E) was calculated from the stress to strain ratio. The method was shown to be accurate and precise using a standard material machined to similar dimensions as those of the mouse femoral segments. Diaphyses of mouse femora were shown to have mean elastic moduli of 10.4+/-0.9 GPa for femora frozen for eight months, 8.6+/-1.4 GPa for femora frozen for two weeks and 8.9+/-1.1 GPa for the fresh femora. These values are much higher than those measured using three-point bending, and lower than values reported in the literature based on nanoindentation tests from mice bones of the same age. We show that this method can be used to accurately and precisely measure the effective elastic modulus of mouse cortical bone.
Collapse
|
23
|
Ulici V, Hoenselaar KD, Gillespie JR, Beier F. The PI3K pathway regulates endochondral bone growth through control of hypertrophic chondrocyte differentiation. BMC DEVELOPMENTAL BIOLOGY 2008; 8:40. [PMID: 18405384 PMCID: PMC2329617 DOI: 10.1186/1471-213x-8-40] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 04/11/2008] [Indexed: 02/07/2023]
Abstract
Background The majority of our bones develop through the process of endochondral ossification that involves chondrocyte proliferation and hypertrophic differentiation in the cartilage growth plate. A large number of growth factors and hormones have been implicated in the regulation of growth plate biology, however, less is known about the intracellular signaling pathways involved. PI3K/Akt has been identified as a major regulator of cellular proliferation, differentiation and death in multiple cell types. Results and Discussion Employing an organ culture system of embryonic mouse tibiae and LY294002, a pharmacological inhibitor of PI3K, we show that inhibition of the pathway results in significant growth reduction, demonstrating that PI3K is required for normal endochondral bone growth in vitro. PI3K inhibition reduces the length of the proliferating and particularly of the hypertrophic zone. Studies with organ cultures and primary chondrocytes in micromass culture show delayed hypertrophic differentiation of chondrocytes and increased apoptosis in the presence of LY294002. Surprisingly, PI3K inhibition had no strong effect on IGF1-induced bone growth, but partially blocked the anabolic effects of C-type natriuretic peptide. Conclusion Our data demonstrate an essential role of PI3K signaling in chondrocyte differentiation and as a consequence of this, in the endochondral bone growth process.
Collapse
Affiliation(s)
- Veronica Ulici
- CIHR Group in Skeletal Development and Remodeling, Department of Physiology & Pharmacology, University of Western Ontario, London, ON, N6A 5C1, Canada
| | | | | | | |
Collapse
|
24
|
Even-Zohar N, Jacob J, Amariglio N, Rechavi G, Potievsky O, Phillip M, Gat-Yablonski G. Nutrition-induced catch-up growth increases hypoxia inducible factor 1alpha RNA levels in the growth plate. Bone 2008; 42:505-15. [PMID: 18201948 DOI: 10.1016/j.bone.2007.10.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 10/01/2007] [Accepted: 10/16/2007] [Indexed: 12/21/2022]
Abstract
Although catch-up growth is a well-known phenomenon, the local pathways at the epiphyseal growth plate that govern this process remain poorly understood. To study the mechanisms governing catch-up growth in the growth plate, we subjected prepubertal rats to 10 days of 40% food restriction, followed by a renewal of the regular food supply to induce catch-up growth. The animals were weighed daily, and their humeral length was measured at sacrifice. The proximal tibial epiphyseal growth plates (EGPs) were studied, and findings were compared with EGPs from animals fed ad libitum and animals under food restriction. The gene expression profile in the growth plates was examined using DNA microarrays, and the expression levels of selected genes were validated by real-time polymerase chain reaction. To localize gene expression in different growth plate zones, microdissection was used. Protein levels and localization were examined using immunohistochemistry. We showed that the expression level of 550 genes decreased during food restriction and increased during catch-up growth, starting already one day after refeeding. HIF-1alpha, as well as several of its downstream targets, was found among these genes. Immunohistochemistry showed a similar pattern for HIF-1alpha protein abundance. Additionally, HIF-1alpha mRNA and protein levels were higher in the proliferating than in the hypertrophic zone, and this distribution was unaffected by nutritional status. These findings indicate that nutrition has a profound effect on gene expression level during growth plate growth, and suggest an important role for HIF-1alpha in the growth plate and its response to nutritional manipulation.
Collapse
Affiliation(s)
- N Even-Zohar
- Institute for Endocrinology and Diabetes, Schneider Children's Medical Center of Israel, Israel.
| | | | | | | | | | | | | |
Collapse
|
25
|
Jones DC, Wein MN, Glimcher LH. Schnurri-3 is an essential regulator of osteoblast function and adult bone mass. Ann Rheum Dis 2007; 66 Suppl 3:iii49-51. [PMID: 17934096 PMCID: PMC2095286 DOI: 10.1136/ard.2007.078352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2007] [Indexed: 11/03/2022]
Abstract
Skeletal remodelling is a cyclical process where under normal physiological conditions, bone formation occurs at sites where bone resorption has previously taken place. Homeostatic remodelling of the skeleton is mediated by osteoclasts, giant multinucleated cells of haematopoietic origin that are responsible for bone resorption and osteoblasts, which originate from mesenchymal stem cells, and synthesise the matrix constituents on bone-forming surfaces.1 Proliferation, differentiation and bone remodelling activities of these cells involve a complex temporal network of growth factors, signalling proteins and transcription factors. Dysregulation of any one component may disrupt the remodelling process and contribute to the pathogenesis of common skeletal disorders, like osteoporosis and Paget's disease. Rare single gene disorders resulting in elevated bone mass due to osteoclast defects are collectively termed osteopetrosis. Rarer still are single gene disorders, collectively termed osteosclerosis, in which elevated bone mass is due to intrinsically elevated osteoblast activity.2 While we have learned much about the molecular control of skeletal formation and remodelling from these mutations, additional genes that regulate bone mass have yet to be characterised.
Collapse
Affiliation(s)
- Dallas C Jones
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 651 Huntington Ave, FXB 205, Boston, MA 02115, USA
| | | | | |
Collapse
|
26
|
Miller LM, Little W, Schirmer A, Sheik F, Busa B, Judex S. Accretion of bone quantity and quality in the developing mouse skeleton. J Bone Miner Res 2007; 22:1037-45. [PMID: 17402847 DOI: 10.1359/jbmr.070402] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED In this work, we found that bone mineral formation proceeded very rapidly in mice by 1 day of age, where the degree of mineralization, the tissue mineral density, and the mineral crystallinity reached 36%, 51%, and 87% of the adult values, respectively. However, even though significant mineralization had occurred, the elastic modulus of 1-day-old bone was only 14% of its adult value, indicating that the intrinsic stiffening of the bone lags considerably behind the initial mineral formation. INTRODUCTION To meet the mechanical challenges during early development, the skeleton requires the rapid accretion of bone quality and bone quantity. Here, we describe early bone development in the mouse skeleton and test the hypothesis that specific compositional properties determine the stiffness of the tissue. MATERIALS AND METHODS Tibias of female BALB mice were harvested at eight time-points (n = 4 each) distributed between 1 and 40 days of age and subjected to morphometric (muCT), chemical (Fourier transform infrared microspectroscopy), and mechanical (nanoindentation) analyses. Tibias of 450-day-old mice served as fully mineralized control specimens. RESULTS Bone growth proceeded very rapidly; at 1 day of age, the degree of mineralization (phosphate/protein ratio), the density of mineralized bone (TMD), and mineral crystallinity had reached 36%, 51%, and 87% of the adult (450 days) values, respectively. Spatially, the variability in mineralization across the mid-diaphysis was very high for the early time-points and declined over time. In contrast to the notable changes in mineralization, carbonate substitution into the mineral lattice (carbonate/phosphate ratio) and collagen cross-linking did not show any significant changes over this time period. Even though significant mineralization had occurred, the elastic modulus of 1-day-old bone was only 14% of the adult value and increased to 89% (of its adult value) after 40 days. Between samples of different time-points, significant positive correlations were observed between the elastic modulus and TMD (r(2) = 0.84), phosphate/protein ratio (r(2) = 0.59), and crystallinity (r(2) = 0.23), whereas collagen cross-linking showed a small but significant negative correlation (r(2) = 0.15). CONCLUSIONS These data indicate that specific chemical and morphometric properties modulate bone's stiffness during early growth. The intrinsic stiffening of the bone, however, lags considerably behind the initial mineral formation, emphasizing the importance of bone mineral quality for optimizing matrix integrity.
Collapse
Affiliation(s)
- Lisa M Miller
- National Synchrotron Light Source, Brookhaven National Laboratory, Upton, New York 11973-5000, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Andrade AC, Nilsson O, Barnes KM, Baron J. Wnt gene expression in the post-natal growth plate: regulation with chondrocyte differentiation. Bone 2007; 40:1361-9. [PMID: 17337262 PMCID: PMC1941840 DOI: 10.1016/j.bone.2007.01.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 12/07/2006] [Accepted: 01/10/2007] [Indexed: 01/10/2023]
Abstract
Longitudinal growth of long bones occurs at the growth plate by endochondral ossification. In the embryonic mouse, this process is regulated by Wnt signaling. Little is known about which members of the Wnt family of secreted signaling proteins might be involved in the regulation of the postnatal growth plate. We used microdissection and real-time PCR to study mRNA expression of Wnt genes in the mouse growth plate. Of the 19 known members of the Wnt family, only six were expressed in postnatal growth plate. Of these, Wnts -2b, -4, and -10b signal through the canonical beta-catenin pathway and Wnts -5a, -5b, and -11 signal through the noncanonical calcium pathway. The spatial expression for these six Wnts was remarkably similar, showing low mRNA expression in the resting zone, increasing expression as the chondrocytes differentiated into the proliferative and prehypertrophic state and then (except Wnt-2b) decreasing expression as the chondrocytes underwent hypertrophic differentiation. This overall pattern is broadly consistent with previous studies of embryonic mouse growth cartilage suggesting that Wnt signaling modulates chondrocyte proliferation and hypertrophic differentiation. We also found that mRNA expression of these Wnt genes persisted at similar levels at 4 weeks, when longitudinal bone growth is waning. In conclusion, we have identified for the first time the specific Wnt genes that are expressed in the postnatal mammalian growth plate. The six identified Wnt genes showed a similar pattern of expression during chondrocyte differentiation, suggesting overlapping or interacting roles in postnatal endochondral bone formation.
Collapse
Affiliation(s)
- Anenisia C Andrade
- Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
28
|
Wang Y, Nishida S, Sakata T, Elalieh HZ, Chang W, Halloran BP, Doty SB, Bikle DD. Insulin-like growth factor-I is essential for embryonic bone development. Endocrinology 2006; 147:4753-61. [PMID: 16857753 PMCID: PMC10645399 DOI: 10.1210/en.2006-0196] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although IGF-I has been identified as an important growth factor for the skeleton, the role of IGF-I on embryonic bone development remains unknown. Here we show that, in IGF-I-deficient (IGF-I(-/-)) mice, skeletal malformations, including short-limbed dwarfism, were evident at days post coitus (dpc) 14.5 to 18.5, accompanied by delays of mineralization in the spinal column, sternum, and fore paws. Reduced chondrocyte proliferation and increased chondrocyte apoptosis were identified in both the spinal ossification center and the growth plate of long bones. Abnormal chondrocyte differentiation and delayed initiation of mineralization was characterized by small size and fewer numbers of type X collagen expressing hypertrophic chondrocytes and lower osteocalcin expression. The Indian hedgehog-PTHrP feedback loop was altered; expression of Indian hedgehog was reduced in IGF-I(-/-) mice in long bones and in the spine, whereas expression of PTHrP was increased. Our results indicate that IGF-I plays an important role in skeletal development by promoting chondrocyte proliferation and maturation while inhibiting apoptosis to form bones of appropriate size and strength.
Collapse
Affiliation(s)
- Yongmei Wang
- Department of Medicine, Endocrine Unit, 111N, Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, California 94121, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Farnum CE, Lenox M, Zipfel W, Horton W, Williams R. In vivo delivery of fluoresceinated dextrans to the murine growth plate: imaging of three vascular routes by multiphoton microscopy. ACTA ACUST UNITED AC 2006; 288:91-103. [PMID: 16342207 PMCID: PMC2573993 DOI: 10.1002/ar.a.20272] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Bone elongation by endochondral ossification occurs through the differentiation cascade of chondrocytes of cartilaginous growth plates. Molecules from the systemic vasculature reach the growth plate from three different directions: epiphyseal, metaphyseal, and a ring vessel and plexus associated with the perichondrium. This study is an analysis of the real-time dynamics of entrance of fluoresceinated tracers of different molecular weights into the growth plate from the systemic vasculature and tests the hypothesis that molecular weight is a key variable in the determination of both the directionality and the extent of tracer movement into the growth plate. Multiphoton microscopy was used for direct in vivo imaging of the murine proximal tibial growth plate in anesthetized 4- to 5-week-old transgenic mice with green fluorescent protein linked to the collagen II promoter. Mice were given an intracardiac injection of either fluorescein (332.3 Da) or fluoresceinated dextrans of 3, 10, 40, 70 kDa, singly or sequentially. For each tracer, directionality and rate of arrival, together with extent of movement within the growth plate, were imaged in real time. For small molecules (up to 10 kDa), vascular access from all three directions was observed and entrance was equally permissive from the metaphyseal and the epiphyseal sides. Within our detection limit (a few percent of vascular concentration), 40 kDa and larger dextrans did not enter. These results have implications both for understanding systemic and paracrine regulation of growth plate chondrocytic differentiation, as well as variables associated with effective drug delivery to growth plate chondrocytes.
Collapse
Affiliation(s)
- Cornelia E Farnum
- Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, USA.
| | | | | | | | | |
Collapse
|
30
|
Cheers MS, Ettensohn CA. P16 is an essential regulator of skeletogenesis in the sea urchin embryo. Dev Biol 2005; 283:384-96. [PMID: 15935341 DOI: 10.1016/j.ydbio.2005.02.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Revised: 02/16/2005] [Accepted: 02/23/2005] [Indexed: 11/27/2022]
Abstract
The primary mesenchyme cells (PMCs) of the sea urchin embryo undergo a dramatic sequence of morphogenetic behaviors that culminates in the formation of the larval endoskeleton. Recent studies have identified components of a gene regulatory network that underlies PMC specification and differentiation. In previous work, we identified novel gene products expressed specifically by PMCs (Illies, M.R., Peeler, M.T., Dechtiaruk, A.M., Ettensohn, C.A., 2002. Identification and developmental expression of new biomineralization proteins in the sea urchin, Strongylocentrotus purpuratus. Dev. Genes Evol. 212, 419-431). Here, we show that one of these gene products, P16, plays an essential role in skeletogenesis. P16 is not required for PMC specification, ingression, migration, or fusion, but is essential for skeletal rod elongation. We have compared the predicted sequences of P16 from two species and show that this small, acidic protein is highly conserved in both structure and function. The predicted amino acid sequence of P16 and the subcellular localization of a GFP-tagged form of the protein suggest that P16 is enriched in the plasma membrane. It may function to receive signals required for skeletogenesis or may play a more direct role in the deposition of biomineral. Finally, we place P16 downstream of Alx1 in the PMC gene network, thereby linking the network to a specific "effector" protein involved in biomineralization.
Collapse
Affiliation(s)
- Melani S Cheers
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
31
|
Valente T, Junyent F, Auladell C. Zac1 is expressed in progenitor/stem cells of the neuroectoderm and mesoderm during embryogenesis: differential phenotype of the Zac1-expressing cells during development. Dev Dyn 2005; 233:667-79. [PMID: 15844099 DOI: 10.1002/dvdy.20373] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Zac1, a new zinc-finger protein that regulates both apoptosis and cell cycle arrest, is abundantly expressed in many neuroepithelia during early brain development. In the present work, we study the expression of Zac1 during early embryogenesis and we determine the cellular phenotype of the Zac1-expressing cells throughout development. Our results show that Zac1 is expressed in the progenitor/stem cells of several tissues (nervous system, skeleton, and skeletal muscle), because they colocalize with several progenitor/stem markers (Nestin, glial fibrillary acidic protein, FORSE-1, proliferating cell nuclear antigen, and bromodeoxyuridine). In postnatal development, Zac1 is expressed in all phases of the life cycle of the chondrocytes (from proliferation to apoptosis), in some limbic gamma-aminobutyric acid-ergic neuronal subpopulations, and during developmental myofibers. Therefore, the intense expression of Zac1 in the progenitor/stem cells of different cellular lineages during the proliferative cycle, before differentiation into postmitotic cells, suggests that Zac1 plays an important role in the control of cell fate during neurogenesis, chondrogenesis, and myogenesis.
Collapse
Affiliation(s)
- Tony Valente
- Departament de Biologia Cellular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
| | | | | |
Collapse
|
32
|
Ward WE, Kim S, Chan D, Fonseca D. Serum equol, bone mineral density and biomechanical bone strength differ among four mouse strains. J Nutr Biochem 2005; 16:743-9. [PMID: 16098733 DOI: 10.1016/j.jnutbio.2005.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 04/03/2005] [Accepted: 04/08/2005] [Indexed: 10/25/2022]
Abstract
The extent of conversion of daidzein to its metabolite, equol, by intestinal microflora may be a critical step that determines if a diet rich in daidzein protects against the deterioration of bone after estrogen withdrawal. The objective was to determine the extent that daidzein is converted to equol. In addition, bone mineral content (BMC), bone mineral density (BMD) and strength of femurs and lumbar vertebrae (LV) in four mouse strains were measured. Mice were ovariectomized and fed control diet (AIN93G) with or without daidzein (200 mg daidzein/kg diet) for 3 weeks, after which serum, femurs and LV were collected. Serum daidzein and equol were elevated in all mice fed daidzein. Among mice fed daidzein, the CD-1 and Swiss-Webster (SW) mice had higher (P<.001) serum equol than C57BL/6 (C57) and C3H mice. Differences due to mouse strain were observed for all bone outcomes. C57 mice had lower femur BMC (P<.001), BMD (P<.001) and peak load at femur midpoint (P<.001) and neck (P<.001) than other mouse strains. C57 mice also had a lower femur midpoint yield load (P<.001) and resilience (P<.001) than C3H mice. C57 mice had a lower LV1-4 BMC (P<.001) and BMD (P<.001) compared with all mouse strains and peak load of LV3 was lower than CD-1 and SW mice. Differences in serum equol, BMD and bone strength properties should be considered when selecting a mouse strain for investigating whether dietary strategies that include isoflavones preserve bone tissue after ovariectomy.
Collapse
Affiliation(s)
- Wendy E Ward
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3E2.
| | | | | | | |
Collapse
|
33
|
Abstract
This review is intended to help the neonatologist who is asked to see a baby or speak to parents who are expecting a baby with signs of a generalised disturbance of bone growth and/or modelling. In this review, we will: define a skeletal dysplasia; discuss the presenting features of a skeletal dysplasia in pregnancy and the newborn period; suggest a clinical approach to find the correct diagnosis; discuss the management of the neonate with a skeletal dysplasia; summarise the clinical features of the most common dysplasias; outline some pitfalls and difficulties in counselling the parents of the baby; and give information on further sources of information about skeletal dysplasias.
Collapse
|
34
|
Ophoff J, Vanderschueren D. The senile osteoporosis mouse model SAMP-6: The ideal animal model for human osteoporosis? ACTA ACUST UNITED AC 2005. [DOI: 10.1138/20050162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Wolf SE, Woodside KJ. Transgenic and gene knock-out techniques and burn research. J Surg Res 2005; 123:328-39. [PMID: 15680397 DOI: 10.1016/j.jss.2004.06.001] [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: 01/12/2004] [Indexed: 02/03/2023]
Abstract
The development of transgenic technology has given researchers a powerful tool to examine biological effects, and the response to injury is no exception. Techniques such as pronuclear injection, targeted homologous recombination, and Cre/loxP gene excision are being used to construct animals with specific genetic designs; these are exploited to learn the role of genes in the response to severe burn. We review the construction of transgenic animals, pitfalls and benefits of this relatively new technique, and how this technique has been used in burn research.
Collapse
Affiliation(s)
- Steven E Wolf
- Department of Surgery, University of Texas Health Science Center--San Antonio, San Antonio, Texas, USA.
| | | |
Collapse
|
36
|
Sugimori K, Matsui K, Motomura H, Tokoro T, Wang J, Higa S, Kimura T, Kitajima I. BMP-2 prevents apoptosis of the N1511 chondrocytic cell line through PI3K/Akt-mediated NF-kappaB activation. J Bone Miner Metab 2005; 23:411-9. [PMID: 16261446 DOI: 10.1007/s00774-005-0622-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Accepted: 05/25/2005] [Indexed: 01/24/2023]
Abstract
The signal transduction pathway by which bone morphogenetic protein-2 (BMP-2) regulates apoptosis in chondrocytes remains largely unknown. We investigated the involvement of phosphatidylinositol 3-kinase (PI3K)/Akt-mediated NF-kappaB activation by BMP-2 stimulation in the modulation of this antiapoptotic process in a chondrocytic cell line, N1511. BMP-2 prevented apoptosis through the inhibition of caspase-3 and -9 and an increase in Bcl-xL expression, and this antiapoptotic effect was inhibited by Noggin. Not only was NF-kappaB p65 activated transiently in the early phase (5-15 min) after treatment with BMP-2 but p65 at serine 536 was phosphorylated from 5 min as well. Akt was rapidly phosphorylated in response to BMP-2 treatment; however, the inhibition of PI3K by Wortmannin markedly reduced the phosphorylation of Akt by BMP-2. Wortmannin also decreased the NF-kappaB transcriptional activity that was up-regulated by BMP-2. Thus, BMP-2-induced NF-kappaB activation is mediated by PI3K/Akt signaling. Wortmannin treatment inhibited the antiapoptotic effect of BMP-2. These data indicate that BMP-2 can utilize a new signal transduction pathway in the NF-kappaB activation system, which plays a crucial role in the survival of the N1511 chondrocytic cell line.
Collapse
Affiliation(s)
- Kazuhito Sugimori
- Department of Orthopaedic Surgery, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Denton CP, Abraham DJ. Transgenic analysis of scleroderma: understanding key pathogenic events in vivo. Autoimmun Rev 2004; 3:285-93. [PMID: 15246024 DOI: 10.1016/j.autrev.2003.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2003] [Accepted: 10/13/2003] [Indexed: 10/26/2022]
Abstract
Modern molecular genetic methods have allowed better understanding of established mouse models of scleroderma and also facilitated the development of new and better defined mouse strains for investigating the pathogenesis of the disease. The best characterized scleroderma animal model is the type 1 tight skin mouse (Tsk1). Backcrossing these animals with other mutant strains has been informative. These experiments implicate the IL-4 ligand-receptor axis in the development of skin fibrosis. Parallel expression analysis of genes using microarrays has provided insight into novel mediators of fibrosis including the C-C chemokine MCP-3. Other experiments suggest that embryonically defined fibroblast-specific regulatory elements may be targets for activation in this model. The same lineage-specific elements have been used to selectively activate TGF beta signaling pathways in fibrosis to generate a novel model for scleroderma and also have been used to develop systems for ligand-dependent fibroblast-specific genetic recombination that will allow further analysis key candidate genes implicated in scleroderma pathogenesis. Better mouse models will improve understanding of this intractable rheumatic disease and can be expected to ultimately lead to improved treatments and outcome.
Collapse
MESH Headings
- Animals
- Chemokine CCL7
- Crosses, Genetic
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Fibrosis
- Forecasting
- Gene Expression
- Genes, Reporter
- Humans
- Mice
- Mice, Inbred Strains
- Mice, Mutant Strains
- Mice, Transgenic
- Models, Biological
- Monocyte Chemoattractant Proteins/genetics
- Monocyte Chemoattractant Proteins/metabolism
- Receptors, Interleukin-4/genetics
- Receptors, Interleukin-4/metabolism
- Recombination, Genetic
- Scleroderma, Systemic/immunology
- Scleroderma, Systemic/metabolism
- Scleroderma, Systemic/pathology
- Signal Transduction
- Transforming Growth Factor beta/metabolism
Collapse
|
38
|
Sorkin AM, Dee KC, Knothe Tate ML. “Culture shock” from the bone cell's perspective: emulating physiological conditions for mechanobiological investigations. Am J Physiol Cell Physiol 2004; 287:C1527-36. [PMID: 15317661 DOI: 10.1152/ajpcell.00059.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bone physiology can be examined on multiple length scales. Results of cell-level studies, typically carried out in vitro, are often extrapolated to attempt to understand tissue and organ physiology. Results of organ- or organism-level studies are often analyzed to deduce the state(s) of the cells within the larger system(s). Although phenomena on all of these scales—cell, tissue, organ, system, organism—are interlinked and contribute to the overall health and function of bone tissue, it is difficult to relate research among these scales. For example, groups of cells in an exogenous, in vitro environment that is well defined by the researcher would not be expected to function similarly to those in a dynamic, endogenous environment, dictated by systemic as well as organismal physiology. This review of the literature on bone cell culture describes potential causes and components of cell “culture shock,” i.e., behavioral variations associated with the transition from in vivo to in vitro environment, focusing on investigations of mechanotransduction and experimental approaches to mimic aspects of bone tissue on a macroscopic scale. The state of the art is reviewed, and new paradigms are suggested to begin bridging the gap between two-dimensional cell cultures in petri dishes and the three-dimensional environment of living bone tissue.
Collapse
Affiliation(s)
- Adam M Sorkin
- Department of Biomedical Engineering, Case Western Reserve Univ., 10900 Euclid Ave., Olin 219, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
39
|
Abstract
OBJECTIVES The goal of this study was to develop and characterize a closed femur fracture model for mice that can be used for the molecular and genetic analysis of fracture healing. STUDY DESIGN Longitudinal time study of species-specific fracture healing. METHODS A protocol was developed for creating reproducible, closed femur fractures in mice. Impending fractures were stabilized by retrograde insertion of a 0.01-inch-diameter, stainless steel wire into the intramedullary canal. The intramedullary wire was held in place with a wedge made from the first 2 mm of a 30-gauge needle. Fractures were produced by 3-point bending. Fracture healing was assessed by radiography, histology, and torsional mechanical testing. RESULTS The mouse femur fracture technique produced good results with minimal loss of animals. Of the 246 mice used in the study, 22 mice were excluded due to poor fracture quality (8), loss of fracture stabilization (6), or to anesthesia death (8). Radiography showed a consistent pattern of fracture healing between mice with peak fracture callus volume evident at 10 (15 mice) to 14 days (18 mice) after fracture. Fracture bridging was apparent in all 3-week postfracture radiographs (35 mice). Histologic examination of 117 specimens at 9 time points showed chondrocyte differentiation within the fracture callus by 7 days after fracture, endochondral ossification occurring by 10 days after fracture, and bone remodeling evident as early as 3 weeks after fracture. Despite radiologic and histologic evidence of fracture bridging after 3 weeks, torsional mechanical testing of 68 mice at 3, 4, 6, and 12 weeks after fracture (group size of 15 to 18 mice at each time point) indicated that significant increases in structural or material strength did not occur until 6 to 12 weeks after fracture. CONCLUSIONS Femur fracture healing in mice follows a typical endochondral ossification pathway with fracture bridging occurring approximately 1 week faster in mice than rats. This fracture model is amenable to the molecular and genetic analysis of fracture healing using different inbred, transgenic, and knockout strains of mice.
Collapse
Affiliation(s)
- Michaele B Manigrasso
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, NJ 07103, USA
| | | |
Collapse
|
40
|
Austin CP, Battey JF, Bradley A, Bucan M, Capecchi M, Collins FS, Dove WF, Duyk G, Dymecki S, Eppig JT, Grieder FB, Heintz N, Hicks G, Insel TR, Joyner A, Koller BH, Lloyd KCK, Magnuson T, Moore MW, Nagy A, Pollock JD, Roses AD, Sands AT, Seed B, Skarnes WC, Snoddy J, Soriano P, Stewart DJ, Stewart F, Stillman B, Varmus H, Varticovski L, Verma IM, Vogt TF, von Melchner H, Witkowski J, Woychik RP, Wurst W, Yancopoulos GD, Young SG, Zambrowicz B. The knockout mouse project. Nat Genet 2004; 36:921-4. [PMID: 15340423 PMCID: PMC2716027 DOI: 10.1038/ng0904-921] [Citation(s) in RCA: 439] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mouse knockout technology provides a powerful means of elucidating gene function in vivo, and a publicly available genome-wide collection of mouse knockouts would be significantly enabling for biomedical discovery. To date, published knockouts exist for only about 10% of mouse genes. Furthermore, many of these are limited in utility because they have not been made or phenotyped in standardized ways, and many are not freely available to researchers. It is time to harness new technologies and efficiencies of production to mount a high-throughput international effort to produce and phenotype knockouts for all mouse genes, and place these resources into the public domain.
Collapse
Affiliation(s)
- Christopher P Austin
- National Human Genome Research Institute, National Institutes of Health, Building 31, Room 4B09, 31 Center Drive, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Boldogköi Z. Gene Network Polymorphism Is the Raw Material of Natural Selection: The Selfish Gene Network Hypothesis. J Mol Evol 2004; 59:340-57. [PMID: 15553089 DOI: 10.1007/s00239-004-2629-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Population genetics, the mathematical theory of modern evolutionary biology, defines evolution as the alteration of the frequency of distinct gene variants (alleles) differing in fitness over the time. The major problem with this view is that in gene and protein sequences we can find little evidence concerning the molecular basis of phenotypic variance, especially those that would confer adaptive benefit to the bearers. Some novel data, however, suggest that a large amount of genetic variation exists in the regulatory region of genes within populations. In addition, comparison of homologous DNA sequences of various species shows that evolution appears to depend more strongly on gene expression than on the genes themselves. Furthermore, it has been demonstrated in several systems that genes form functional networks, whose products exhibit interrelated expression profiles. Finally, it has been found that regulatory circuits of development behave as evolutionary units. These data demonstrate that our view of evolution calls for a new synthesis. In this article I propose a novel concept, termed the selfish gene network hypothesis, which is based on an overall consideration of the above findings. The major statements of this hypothesis are as follows. (1) Instead of individual genes, gene networks (GNs) are responsible for the determination of traits and behaviors. (2) The primary source of microevolution is the intraspecific polymorphism in GNs and not the allelic variation in either the coding or the regulatory sequences of individual genes. (3) GN polymorphism is generated by the variation in the regulatory regions of the component genes and not by the variance in their coding sequences. (4) Evolution proceeds through continuous restructuring of the composition of GNs rather than fixing of specific alleles or GN variants.
Collapse
Affiliation(s)
- Zsolt Boldogköi
- Laboratory of Neuromorphology, Department of Anatomy, Faculty of Medicine, University of Budapest, Budapest, Hungary.
| |
Collapse
|