1
|
Hughes JM, Guerriere KI, Popp KL, Castellani CM, Pasiakos SM. Exercise for optimizing bone health after hormone-induced increases in bone stiffness. Front Endocrinol (Lausanne) 2023; 14:1219454. [PMID: 37790607 PMCID: PMC10544579 DOI: 10.3389/fendo.2023.1219454] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/17/2023] [Indexed: 10/05/2023] Open
Abstract
Hormones and mechanical loading co-regulate bone throughout the lifespan. In this review, we posit that times of increased hormonal influence on bone provide opportunities for exercise to optimize bone strength and prevent fragility. Examples include endogenous secretion of growth hormones and sex steroids that modulate adolescent growth and exogenous administration of osteoanabolic drugs like teriparatide, which increase bone stiffness, or its resistance to external forces. We review evidence that after bone stiffness is increased due to hormonal stimuli, mechanoadaptive processes follow. Specifically, exercise provides the mechanical stimulus necessary to offset adaptive bone resorption or promote adaptive bone formation. The collective effects of both decreased bone resorption and increased bone formation optimize bone strength during youth and preserve it later in life. These theoretical constructs provide physiologic foundations for promoting exercise throughout life.
Collapse
Affiliation(s)
- Julie M. Hughes
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Katelyn I. Guerriere
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Kristin L. Popp
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA, United States
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Colleen M. Castellani
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA, United States
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Stefan M. Pasiakos
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA, United States
| |
Collapse
|
2
|
|
3
|
Abstract
Bone and skeletal muscle are integrated organs and their coupling has been considered mainly a mechanical one in which bone serves as attachment site to muscle while muscle applies load to bone and regulates bone metabolism. However, skeletal muscle can affect bone homeostasis also in a non-mechanical fashion, i.e., through its endocrine activity. Being recognized as an endocrine organ itself, skeletal muscle secretes a panel of cytokines and proteins named myokines, synthesized and secreted by myocytes in response to muscle contraction. Myokines exert an autocrine function in regulating muscle metabolism as well as a paracrine/endocrine regulatory function on distant organs and tissues, such as bone, adipose tissue, brain and liver. Physical activity is the primary physiological stimulus for bone anabolism (and/or catabolism) through the production and secretion of myokines, such as IL-6, irisin, IGF-1, FGF2, beside the direct effect of loading. Importantly, exercise-induced myokine can exert an anti-inflammatory action that is able to counteract not only acute inflammation due to an infection, but also a condition of chronic low-grade inflammation raised as consequence of physical inactivity, aging or metabolic disorders (i.e., obesity, type 2 diabetes mellitus). In this review article, we will discuss the effects that some of the most studied exercise-induced myokines exert on bone formation and bone resorption, as well as a brief overview of the anti-inflammatory effects of myokines during the onset pathological conditions characterized by the development a systemic low-grade inflammation, such as sarcopenia, obesity and aging.
Collapse
Affiliation(s)
- Marta Gomarasca
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Milan, Italy
| | - Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Lombardi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Milan, Italy; Gdańsk University of Physical Education & Sport, Gdańsk, Pomorskie, Poland.
| |
Collapse
|
4
|
Yarrow JF, Kok HJ, Phillips EG, Conover CF, Lee J, Bassett TE, Buckley KH, Reynolds MC, Wnek RD, Otzel DM, Chen C, Jiron JM, Graham ZA, Cardozo C, Vandenborne K, Bose PK, Aguirre JI, Borst SE, Ye F. Locomotor training with adjuvant testosterone preserves cancellous bone and promotes muscle plasticity in male rats after severe spinal cord injury. J Neurosci Res 2019; 98:843-868. [PMID: 31797423 DOI: 10.1002/jnr.24564] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Abstract
Loading and testosterone may influence musculoskeletal recovery after spinal cord injury (SCI). Our objectives were to determine (a) the acute effects of bodyweight-supported treadmill training (TM) on hindlimb cancellous bone microstructure and muscle mass in adult rats after severe contusion SCI and (b) whether longer-term TM with adjuvant testosterone enanthate (TE) delivers musculoskeletal benefit. In Study 1, TM (40 min/day, 5 days/week, beginning 1 week postsurgery) did not prevent SCI-induced hindlimb cancellous bone loss after 3 weeks. In Study 2, TM did not attenuate SCI-induced plantar flexor muscles atrophy nor improve locomotor recovery after 4 weeks. In our main study, SCI produced extensive distal femur and proximal tibia cancellous bone deficits, a deleterious slow-to-fast fiber-type transition in soleus, lower muscle fiber cross-sectional area (fCSA), impaired muscle force production, and levator ani/bulbocavernosus (LABC) muscle atrophy after 8 weeks. TE alone (7.0 mg/week) suppressed bone resorption, attenuated cancellous bone loss, constrained the soleus fiber-type transition, and prevented LABC atrophy. In comparison, TE+TM concomitantly suppressed bone resorption and stimulated bone formation after SCI, produced near-complete cancellous bone preservation, prevented the soleus fiber-type transition, attenuated soleus fCSA atrophy, maintained soleus force production, and increased LABC mass. 75% of SCI+TE+TM animals recovered voluntary over-ground hindlimb stepping, while no SCI and only 20% of SCI+TE animals regained stepping ability. Positive associations between testosterone and locomotor function suggest that TE influenced locomotor recovery. In conclusion, short-term TM alone did not improve bone, muscle, or locomotor recovery in adult rats after severe SCI, while longer-term TE+TM provided more comprehensive musculoskeletal benefit than TE alone.
Collapse
Affiliation(s)
- Joshua F Yarrow
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.,Brain Rehabilitation Research Center, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.,Division of Endocrinology, Diabetes, and Metabolism, University of Florida College of Medicine, Gainesville, FL, USA
| | - Hui Jean Kok
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.,Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Ean G Phillips
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Christine F Conover
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Jimmy Lee
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Taylor E Bassett
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Kinley H Buckley
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Michael C Reynolds
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Russell D Wnek
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Dana M Otzel
- Brain Rehabilitation Research Center, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Cong Chen
- Divison of Orthopedics and Rehabilitation, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jessica M Jiron
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Zachary A Graham
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA.,Departments of Medicine, Icahn School of Medicine, New York, NY, USA
| | - Christopher Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA.,Departments of Medicine, Icahn School of Medicine, New York, NY, USA.,Rehabilitation Medicine, Icahn School of Medicine, New York, NY, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Prodip K Bose
- Brain Rehabilitation Research Center, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA.,Department of Physiological Sciences, University of Florida, Gainesville, FL, USA.,Division of Neurology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jose Ignacio Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Stephen E Borst
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Fan Ye
- Research Service, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| |
Collapse
|
5
|
Lombardi G, Ziemann E, Banfi G. Physical Activity and Bone Health: What Is the Role of Immune System? A Narrative Review of the Third Way. Front Endocrinol (Lausanne) 2019; 10:60. [PMID: 30792697 PMCID: PMC6374307 DOI: 10.3389/fendo.2019.00060] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/22/2019] [Indexed: 12/12/2022] Open
Abstract
Bone tissue can be seen as a physiological hub of several stimuli of different origin (e.g., dietary, endocrine, nervous, immune, skeletal muscle traction, biomechanical load). Their integration, at the bone level, results in: (i) changes in mineral and protein composition and microarchitecture and, consequently, in shape and strength; (ii) modulation of calcium and phosphorous release into the bloodstream, (iii) expression and release of hormones and mediators able to communicate the current bone status to the rest of the body. Different stimuli are able to act on either one or, as usual, more levels. Physical activity is the key stimulus for bone metabolism acting in two ways: through the biomechanical load which resolves into a direct stimulation of the segment(s) involved and through an indirect load mediated by muscle traction onto the bone, which is the main physiological stimulus for bone formation, and the endocrine stimulation which causes homeostatic adaptation. The third way, in which physical activity is able to modify bone functions, passes through the immune system. It is known that immune function is modulated by physical activity; however, two recent insights have shed new light on this modulation. The first relies on the discovery of inflammasomes, receptors/sensors of the innate immunity that regulate caspase-1 activation and are, hence, the tissue triggers of inflammation in response to infections and/or stressors. The second relies on the ability of certain tissues, and particularly skeletal muscle and adipose tissue, to synthesize and secrete mediators (namely, myokines and adipokines) able to affect, profoundly, the immune function. Physical activity is known to act on both these mechanisms and, hence, its effects on bone are also mediated by the immune system activation. Indeed, that immune system and bone are tightly connected and inflammation is pivotal in determining the bone metabolic status is well-known. The aim of this narrative review is to give a complete view of the exercise-dependent immune system-mediated effects on bone metabolism and function.
Collapse
Affiliation(s)
- Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Physiology and Pharmacology, Faculty of Rehabilitation and Kinesiology, Gdansk University of Physical Education and Sport, Gdansk, Poland
- *Correspondence: Giovanni Lombardi
| | - Ewa Ziemann
- Department of Physiology and Pharmacology, Faculty of Rehabilitation and Kinesiology, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
6
|
A review of the cellular and molecular effects of extracorporeal shockwave therapy. Vet Comp Orthop Traumatol 2017; 29:99-107. [DOI: 10.3415/vcot-15-04-0057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/15/2015] [Indexed: 12/31/2022]
Abstract
SummaryExtracorporeal shockwave therapy (ESWT) is a novel therapeutic modality and its use in promoting connective tissue repair and analgesic effect has been advocated in the literature. It is convenient, cost-effective, and has negligible complications; it therefore bypasses many of the problems associated with surgical interventions. This paper reviews the proposed mechanisms of action in promoting tissue repair and regeneration as well as analysing its efficacy providing an analgesic effect in clinical applications. Further research will be required to not only identify the underlying mechanisms more precisely, but will also be critical for ensuring consistency across the literature so that the most beneficial treatment protocol can be developed. Extracorporeal shockwave therapy stands as a promising alternative modality in promoting tissue repair.
Collapse
|
7
|
Rooney AM, van der Meulen MCH. Mouse models to evaluate the role of estrogen receptor α in skeletal maintenance and adaptation. Ann N Y Acad Sci 2017; 1410:85-92. [PMID: 29148577 DOI: 10.1111/nyas.13523] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 12/15/2022]
Abstract
Estrogen signaling and mechanical loading have individual and combined effects on skeletal maintenance and adaptation. Previous work investigating estrogen signaling both in vitro and in vivo using global estrogen receptor α (ERα) gene knockout mouse models has provided information regarding the role of ERα in regulating bone mass and adaptation to mechanical stimulation. However, these models have inherent limitations that confound interpretation of the data. Therefore, recent studies have focused on mice with targeted deletion of ERα from specific bone cells and their precursors. Cell stage, tissue type, and mouse sex all influence the effects of ERα gene deletion. Lack of ERα in osteoblast progenitor and precursor cells generally affects the periosteum of female and male mice. The absence of ERα in differentiated osteoblasts, osteocytes, and osteoclasts in mice generally resulted in reduced cancellous bone mass, with differing reports of the effect by animal sex and greater deficiencies in bone mass typically occurring in cancellous bone in female mice. Limited data exist for the role of bone cell-specific ERα in skeletal adaptation in vivo. Cell-specific ERα gene knockout mice provide an excellent platform for investigating the function of ERα in regulating skeletal phenotype and response to mechanical loading by sex and age.
Collapse
Affiliation(s)
- Amanda M Rooney
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Marjolein C H van der Meulen
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York.,Research Division, Hospital for Special Surgery, New York, New York
| |
Collapse
|
8
|
Goldring SR. Differential mechanisms of de-regulated bone formation in rheumatoid arthritis and spondyloarthritis. Rheumatology (Oxford) 2017; 55:ii56-ii60. [PMID: 27856661 DOI: 10.1093/rheumatology/kew345] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 08/23/2016] [Indexed: 11/14/2022] Open
Abstract
The inflammatory arthropathies share in common their tendency to produce marked alterations in skeletal remodelling and architecture. This review will focus on RA and the seronegative spondyloarthopathies (SpA), which share common features with respect to their tendency to produce localized bone destruction at sites of articular and peri-articular inflammation. However, there are significant differences in the skeletal pathology in these conditions, which include the unique involvement of the axial skeleton and the presence of inflammation in the extra-articular entheses in SpA. There also are differences in the pattern of bone formation and repair associated with the articular and peri-articular inflammation. This review will highlight the molecular and cellular processes that are involved in the pathogenesis of the skeletal pathology in these two forms of inflammatory arthritis with specific focus on the pathogenic mechanisms underlying the differential patterns of bone formation and repair.
Collapse
Affiliation(s)
- Steven R Goldring
- The Hospital for Special Surgery, Weill College Medical College, New York, NY, USA
| |
Collapse
|
9
|
Ortinau LC, Linden MA, Rector RS, Hinton PS. Exercise improves femoral whole-bone and tissue-level biomechanical properties in hyperphagic OLETF rats. Appl Physiol Nutr Metab 2017; 42:884-892. [PMID: 28460190 DOI: 10.1139/apnm-2017-0077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An often-overlooked comorbidity of type 2 diabetes (T2D) is increased fracture risk. Since traditional T2D therapies focus solely on glucose homeostasis, there is an increased need for effective treatment strategies that improve both metabolic and bone health. The current study evaluated if previously reported protective effects of exercise (EX) on cortical bone geometry and biomechanical properties are due to the prevention of obesity compared with obese Otsuka Long-Evans Tokushima Fatty (OLETF) rats or the unique skeletal effects of exercise. Four-week-old male OLETF rats were randomly assigned to 1 of 3 groups, each fed a standard rodent chow diet until 40 weeks of age: ad libitum-fed sedentary (O-SED), ad libitum-fed EX (O-EX), or a control group body-weight-matched to the O-EX group by caloric restriction (O-CR). Ad libitum-fed sedentary Long-Evans Tokushima Otsuka (L-SED) rats were used as a lean control. EX or CR prevented the excess body mass accumulation and elevated HbA1c observed in O-SED. Total-body bone mineral density was greater in O-EX than O-CR, but similar to L-SED and O-SED. O-EX and O-CR had lower osteocalcin and TRAP5b than O-SED and L-SED. EX or CR prevented the ∼3-fold increase in CTx in O-SED versus L-SED. EX increased femoral cortical mass accumulation and expansion at the mid-diaphysis compared with O-CR. EX or CR significantly increased tissue-level stiffness and strength compared with O-SED and L-SED, but O-EX had greater whole-bone stiffness than all other groups. In summary, EX has distinct advantages over CR for improving bone biomechanical properties in hyperphagic OLETF rats.
Collapse
Affiliation(s)
- Laura C Ortinau
- a Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, 204 Gwynn Hall, Columbia, MO 65211, USA
| | - Melissa A Linden
- a Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, 204 Gwynn Hall, Columbia, MO 65211, USA.,b Research Service-Harry S. Truman Memorial Veterans Medical Center, Columbia, MO, USA
| | - R Scott Rector
- a Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, 204 Gwynn Hall, Columbia, MO 65211, USA.,b Research Service-Harry S. Truman Memorial Veterans Medical Center, Columbia, MO, USA.,c Department of Medicine, Gastroenterology and Hepatology, University of Missouri, Columbia, MO, USA
| | - Pamela S Hinton
- a Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, 204 Gwynn Hall, Columbia, MO 65211, USA
| |
Collapse
|
10
|
Hinton PS, Nigh P, Thyfault J. Serum sclerostin decreases following 12months of resistance- or jump-training in men with low bone mass. Bone 2017; 96:85-90. [PMID: 27744012 PMCID: PMC5328803 DOI: 10.1016/j.bone.2016.10.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/07/2016] [Accepted: 10/10/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE We previously reported that 12months of resistance training (RT, 2×/wk, N=19) or jump training (JUMP, 3×/wk, N=19) increased whole body and lumbar spine BMD and increased serum bone formation markers relative to resorption in physically active (≥4h/wk) men (mean age: 44±2y; median: 44y) with osteopenia of the hip or spine. The purpose of this secondary analysis was to examine the effects of the RT or JUMP intervention on potential endocrine mediators of the exercise effects on bone, specifically IGF-I, PTH and sclerostin. METHODS Fasting blood samples were collected after a 24-h period of no exercise at baseline and after 12months of RT or JUMP. IGF-I, PTH and sclerostin were measured in serum by ELISA. The effects of RT or JUMP on IGF-I, PTH and sclerostin were evaluated using 2×2 repeated measures ANOVA (time, group). This study was conducted in accordance with the Declaration of Helsinki and was approved by the University of Missouri IRB. RESULTS Sclerostin concentrations in serum significantly decreased and IGF-I significantly increased after 12months of RT or JUMP; while PTH remained unchanged. CONCLUSION The beneficial effects of long-term, progressive-intensity RT or JUMP on BMD in moderately active men with low bone mass are associated with decreased sclerostin and increased IGF-I.
Collapse
Affiliation(s)
- Pamela S Hinton
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, United States.
| | - Peggy Nigh
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, United States
| | - John Thyfault
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, United States; Internal Medicine - Division of GI and Hepatology, University of Missouri, Columbia, MO 65211, United States
| |
Collapse
|
11
|
Iwaniec UT, Turner RT. Influence of body weight on bone mass, architecture and turnover. J Endocrinol 2016; 230:R115-30. [PMID: 27352896 PMCID: PMC4980254 DOI: 10.1530/joe-16-0089] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
Weight-dependent loading of the skeleton plays an important role in establishing and maintaining bone mass and strength. This review focuses on mechanical signaling induced by body weight as an essential mechanism for maintaining bone health. In addition, the skeletal effects of deviation from normal weight are discussed. The magnitude of mechanical strain experienced by bone during normal activities is remarkably similar among vertebrates, regardless of size, supporting the existence of a conserved regulatory mechanism, or mechanostat, that senses mechanical strain. The mechanostat functions as an adaptive mechanism to optimize bone mass and architecture based on prevailing mechanical strain. Changes in weight, due to altered mass, weightlessness (spaceflight), and hypergravity (modeled by centrifugation), induce an adaptive skeletal response. However, the precise mechanisms governing the skeletal response are incompletely understood. Furthermore, establishing whether the adaptive response maintains the mechanical competence of the skeleton has proven difficult, necessitating the development of surrogate measures of bone quality. The mechanostat is influenced by regulatory inputs to facilitate non-mechanical functions of the skeleton, such as mineral homeostasis, as well as hormones and energy/nutrient availability that support bone metabolism. Although the skeleton is very capable of adapting to changes in weight, the mechanostat has limits. At the limits, extreme deviations from normal weight and body composition are associated with impaired optimization of bone strength to prevailing body size.
Collapse
Affiliation(s)
- Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| |
Collapse
|
12
|
Gombos GC, Bajsz V, Pék E, Schmidt B, Sió E, Molics B, Betlehem J. Direct effects of physical training on markers of bone metabolism and serum sclerostin concentrations in older adults with low bone mass. BMC Musculoskelet Disord 2016; 17:254. [PMID: 27278385 PMCID: PMC4899888 DOI: 10.1186/s12891-016-1109-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
Abstract
Background Both gravitational loading and the forces generated by muscle contraction have direct effects on serum markers of bone metabolism. The object of this study was to examine the direct effects of a single session of resistance exercise or walking on biochemical markers of bone metabolism in participants with low bone mass. Methods A total of 150 otherwise healthy female subjects (mean age = 59.1 ± 7.1 years) diagnosed with osteoporosis or osteopenia were randomly allocated to either a resistance exercise group (RG; n = 50), walking group (WG; n = 50), or control group (CG; n = 50). Changes in bone-specific alkaline phosphatase (BALP), carboxy-terminal cross-linked telopeptide of type I collagen (CTX), and serum sclerostin concentrations were measured before and immediately after a single exercise intervention. Results There was no significant change in BALP values in any of the groups. Sclerostin levels increased in the RG and WG, and there was significant difference between the WG and CG after the exercise intervention (P < 0.01). In contrast, the changes in CTX concentrations from baseline were significant in the RG (P < 0.01) but not in the WG (P = 0.11), and there was a significant difference between resistance exercise and walking (P < 0.01). Conclusions In participants with low bone mass, resistance exercise influenced the serum concentrations of CTX, a marker of bone resorption, but walking did not. Trial registration Current Controlled Trials ISRCTN16329455; retrospectively registered on 05/05/2016.
Collapse
Affiliation(s)
| | - Viktória Bajsz
- University of Pécs, Faculty of Health Sciences, 33 Landorhegyi Road, Zalaegerszeg, 8900, Hungary
| | - Emese Pék
- University of Pécs, Faculty of Health Sciences, 33 Landorhegyi Road, Zalaegerszeg, 8900, Hungary
| | - Béla Schmidt
- University of Pécs, Faculty of Health Sciences, 33 Landorhegyi Road, Zalaegerszeg, 8900, Hungary
| | - Eszter Sió
- University of Pécs, Faculty of Health Sciences, 33 Landorhegyi Road, Zalaegerszeg, 8900, Hungary
| | - Bálint Molics
- University of Pécs, Faculty of Health Sciences, 33 Landorhegyi Road, Zalaegerszeg, 8900, Hungary
| | - József Betlehem
- University of Pécs, Faculty of Health Sciences, 33 Landorhegyi Road, Zalaegerszeg, 8900, Hungary
| |
Collapse
|
13
|
Lee SH, Oh KN, Han Y, Choi YH, Lee KY. Estrogen Receptor α Regulates Dlx3-Mediated Osteoblast Differentiation. Mol Cells 2016; 39:156-62. [PMID: 26674964 PMCID: PMC4757804 DOI: 10.14348/molcells.2016.2291] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 10/31/2015] [Accepted: 11/03/2015] [Indexed: 01/01/2023] Open
Abstract
Estrogen receptor α (ER-α), which is involved in bone metabolism and breast cancer, has been shown to have transcriptional targets. Dlx3 is essential for the skeletal development and plays an important role in osteoblast differentiation. Various osteogenic stimulators and transcription factors can induce the protein expression of Dlx3. However, the regulatory function of ER-α in the Dlx3 mediated osteogenic process remains unknown. Therefore, we investigated the regulation of Dlx3 and found that ER-α is a positive regulator of Dlx3 transcription in BMP2-induced osteoblast differentiation. We also found that ER-α interacts with Dlx3 and increases its transcriptional activity and DNA binding affinity. Furthermore, we demonstrated that the regulation of Dlx3 activity by ER-α is independent of the ligand (estradiol) binding domain. These results indicate that Dlx3 is a novel target of ER-α, and that ER-α regulates the osteoblast differentiation through modulation of Dlx3 expression and/or interaction with Dlx3.
Collapse
Affiliation(s)
- Sung Ho Lee
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757,
Korea
| | - Kyo-Nyeo Oh
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757,
Korea
| | - Younho Han
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757,
Korea
| | - You Hee Choi
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757,
Korea
| | - Kwang-Youl Lee
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757,
Korea
| |
Collapse
|
14
|
Zhang D, Park BM, Kang M, Nam H, Kim EJ, Bae C, Lim SK. The systemic effects of sclerostin overexpression using ΦC31 integrase in mice. Biochem Biophys Res Commun 2016; 472:471-6. [PMID: 26845353 DOI: 10.1016/j.bbrc.2016.01.178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/29/2016] [Indexed: 11/29/2022]
Abstract
Sclerostin, encoded by the Sost gene, is mainly produced by osteocytes in bone and antagonizes the Wnt/β-catenin signaling pathway, which is a requisite for bone formation. Currently, human anti-sclerostin antibodies are being tested in phase III clinical trials. In addition, serum sclerostin levels are reported to be associated with bone mineral density and fracture risk in normal individuals; however, the correlation between serum sclerostin and bone mass remains controversial. To study the effects of the continuous exposure of exogenous sclerostin on bone, a ΦC31 integrase system, which has the characteristics of site-specificity and efficiency, was applied for the delivery of the Sost gene in this study. We injected Sost-attB plasmid with or without ΦC31 integrase plasmid into the mouse tail vein using a hydrodynamic-based method. The site-specific integration of the Sost gene into the mouse genome was confirmed by examining a pseudo-attP site on the hepatic genomic DNA. Sclerostin was expressed in the hepatocytes, secreted into the blood flow, and maintained at high concentrations in the mice with both Sost-attB plasmid and ΦC31 integrase plasmid injections, which was observed by serial measurement. Moreover, the mice with long-term high levels of serum sclerostin showed trabecular bone loss on micro-CT analysis. Peripheral B cell populations were not affected. Our results suggested that sclerostin could be expressed in the liver and sustained successfully at high levels in the blood by using the ΦC31 integrase system, leading to trabecular bone loss. These findings may help to further ascertain the effects of sclerostin introduced exogenously on the skeleton.
Collapse
Affiliation(s)
- Dongdong Zhang
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea; Division of Endocrinology & Metabolism, Department of Internal Medicine, Affiliated Yantai Hospital of Binzhou Medical University, Yantai, PR China
| | - Bo Mi Park
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Myengmo Kang
- Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - HeeJin Nam
- Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jin Kim
- Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - ChuHyun Bae
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Sung Kil Lim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea; Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Meakin LB, Udeh C, Galea GL, Lanyon LE, Price JS. Exercise does not enhance aged bone's impaired response to artificial loading in C57Bl/6 mice. Bone 2015; 81:47-52. [PMID: 26142929 PMCID: PMC4652609 DOI: 10.1016/j.bone.2015.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/24/2015] [Accepted: 06/30/2015] [Indexed: 01/06/2023]
Abstract
Bones adapt their structure to their loading environment and so ensure that they become, and are maintained, sufficiently strong to withstand the loads to which they are habituated. The effectiveness of this process declines with age and bones become fragile fracturing with less force. This effect in humans also occurs in mice which experience age-related bone loss and reduced adaptation to loading. Exercise engenders many systemic and local muscular physiological responses as well as engendering local bone strain. To investigate whether these physiological responses influence bones' adaptive responses to mechanical strain we examined whether a period of treadmill exercise influenced the adaptive response to an associated period of artificial loading in young adult (17-week) and old (19-month) mice. After treadmill acclimatization, mice were exercised for 30 min three times per week for two weeks. Three hours after each exercise period, right tibiae were subjected to 40 cycles of non-invasive axial loading engendering peak strain of 2250 με. In both young and aged mice exercise increased cross-sectional muscle area and serum sclerostin concentration. In young mice it also increased serum IGF1. Exercise did not affect bone's adaptation to loading in any measured parameter in young or aged bone. These data demonstrate that a level of exercise sufficient to cause systemic changes in serum, and adaptive changes in local musculature, has no effect on bone's response to loading 3h later. This study provides no support for the beneficial effects of exercise on bone in the elderly being mediated by systemic or local muscle-derived effects rather than local adaptation to altered mechanical strain.
Collapse
Affiliation(s)
- Lee B Meakin
- School of Veterinary Sciences, University of Bristol, Bristol BS40 5DU, UK.
| | - Chinedu Udeh
- School of Clinical Sciences, University of Bristol, Bristol BS2 8DZ, UK
| | - Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Bristol BS40 5DU, UK
| | - Lance E Lanyon
- School of Veterinary Sciences, University of Bristol, Bristol BS40 5DU, UK
| | - Joanna S Price
- School of Veterinary Sciences, University of Bristol, Bristol BS40 5DU, UK
| |
Collapse
|
16
|
Sontam DM, Firth EC, Tsai P, Vickers MH, O'Sullivan JM. Different exercise modalities have distinct effects on the integrin-linked kinase (ILK) and Ca2+ signaling pathways in the male rat bone. Physiol Rep 2015; 3:3/10/e12568. [PMID: 26471755 PMCID: PMC4632950 DOI: 10.14814/phy2.12568] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mechanical loading is essential to maintain optimal skeletal health. Despite the fact that early-life exercise has positive, long-lasting effects on the musculo-skeletal system, the response of the musculo-skeletal system to spontaneous low-impact exercise has been poorly studied. Previously, we identified subtle morphological changes in the femoral diaphysis of exercised animals compared to nonexercised controls. We hypothesized that significant changes in gene expression of cells should precede significant measurable phenotypic changes in the tissues of which they are part. Here, we employed RNA-Seq to analyse the transcriptome of the cortical bone from the femoral mid-diaphysis of prepubertal male Sprague-Dawley rats that were assigned to control (CON); bipedal stance (BPS); or wheel exercise (WEX) groups for 15 days. We identified 808 and 324 differentially expressed transcripts in the BPS and WEX animals respectively. While a number of transcripts change their levels in an exercise-specific manner, we identified 191 transcripts that were differentially expressed in both BPS and WEX. Importantly, we observed that the exercise mode had diametrically opposite effects on transcripts for multiple genes within the integrin-linked kinase (ILK) and Ca(2+) signaling pathways such that they were up-regulated in BPS and down-regulated in WEX. The findings are important for our understanding of possible ways in which different exercise regimens might affect bone when normal activities apply mechanical stimuli during postnatal growth and development.
Collapse
Affiliation(s)
- Dharani M Sontam
- The Liggins Institute, University of Auckland, Auckland, New Zealand Gravida: National Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | - Elwyn C Firth
- The Liggins Institute, University of Auckland, Auckland, New Zealand Gravida: National Centre for Growth and Development, University of Auckland, Auckland, New Zealand Department of Sport and Exercise Science, University of Auckland, Auckland, New Zealand
| | - Peter Tsai
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- The Liggins Institute, University of Auckland, Auckland, New Zealand Gravida: National Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | - Justin M O'Sullivan
- The Liggins Institute, University of Auckland, Auckland, New Zealand Gravida: National Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| |
Collapse
|
17
|
Melville KM, Kelly NH, Surita G, Buchalter DB, Schimenti JC, Main RP, Ross FP, van der Meulen MCH. Effects of Deletion of ERα in Osteoblast-Lineage Cells on Bone Mass and Adaptation to Mechanical Loading Differ in Female and Male Mice. J Bone Miner Res 2015; 30:1468-80. [PMID: 25707500 PMCID: PMC4506717 DOI: 10.1002/jbmr.2488] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 12/30/2022]
Abstract
Estrogen receptor alpha (ERα) has been implicated in bone's response to mechanical loading in both males and females. ERα in osteoblast lineage cells is important for determining bone mass, but results depend on animal sex and the cellular stage at which ERα is deleted. We demonstrated previously that when ERα is deleted from mature osteoblasts and osteocytes in mixed-background female mice, bone mass and strength are decreased. However, few studies exist examining the skeletal response to loading in bone cell-specific ERαKO mice. Therefore, we crossed ERα floxed (ERα(fl/fl)) and osteocalcin-Cre (OC-Cre) mice to generate animals lacking ERα in mature osteoblasts and osteocytes (pOC-ERαKO) and littermate controls (LC). At 10 weeks of age, the left tibia was loaded in vivo for 2 weeks. We analyzed bone mass through micro-CT, bone formation rate by dynamic histomorphometry, bone strength from mechanical testing, and osteoblast and osteoclast activity by serum chemistry and immunohistochemistry. ERα in mature osteoblasts differentially regulated bone mass in males and females. Compared with LC, female pOC-ERαKO mice had decreased cortical and cancellous bone mass, whereas male pOC-ERαKO mice had equal or greater bone mass than LC. Bone mass results correlated with decreased compressive strength in pOC-ERαKO female L(5) vertebrae and with increased maximum moment in pOC-ERαKO male femora. Female pOC-ERαKO mice responded more to mechanical loading, whereas the response of pOC-ERαKO male animals was similar to their littermate controls.
Collapse
Affiliation(s)
- Katherine M. Melville
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
- Department of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Natalie H. Kelly
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
- Department of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Gina Surita
- Department of Biological Sciences, Cornell University, Ithaca, NY
| | | | | | - Russell P. Main
- College of Veterinary Medicine, Purdue University, West Lafayette, IN
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - F. Patrick Ross
- Research Division, Hospital for Special Surgery, New York, NY
| | - Marjolein C. H. van der Meulen
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
- Department of Biomedical Engineering, Cornell University, Ithaca, NY
- Research Division, Hospital for Special Surgery, New York, NY
| |
Collapse
|
18
|
Hinton PS, Shankar K, Eaton LM, Rector RS. Obesity-related changes in bone structural and material properties in hyperphagic OLETF rats and protection by voluntary wheel running. Metabolism 2015; 64:905-16. [PMID: 25963848 DOI: 10.1016/j.metabol.2015.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/25/2015] [Accepted: 04/24/2015] [Indexed: 12/11/2022]
Abstract
PURPOSE To examine how the development of obesity and the associated insulin resistance affect bone structural and material properties, and bone formation and resorption markers in the Otsuka Long-Evans Tokushima Fatty (OLETF) rat model. METHODS This was a 36-week study of sedentary, hyperphagic, male OLETF rats (OLETF-SED), exercise-treated OLETF rats (OLETF-EX) and sedentary non-hyperphagic controls (LETO-SED) with data collection at 13, 20, and 40 weeks of age (n = 5-8 animals per group per timepoint). RESULTS Body mass and fat (%) were significantly greater in OLETF-SED versus controls. OLETF-SED were insulin resistant at 13 and 20 weeks, with overt diabetes by 40 weeks. At 13weeks, OLETF-SED had lower total body BMC and BMD and serum P1NP compared with LETO-SED. Differences in total body BMC and BMD between OLETF-SED and LETO-SED persisted at 20 weeks, with reductions in total and cortical BMD of the tibia. OLETF-SED also had lesser femur diameter, cross-sectional area, polar moment of area, and torque at fracture than LETO-SED. By 40 weeks, OLETF-SED had elevated bone resorption and reduced intrinsic bone strength. OLETF-EX did not show the excessive weight gain, obesity, insulin resistance or diabetes observed in OLETF-SED. OLETF-EX had greater BMD than OLETF-SED, and structural and material properties of the femur were significantly increased in OLETF-EX relative to OLETF-SED and LETO-SED. CONCLUSIONS The negative skeletal effects of excessive adiposity and insulin resistance were evident early in the progressive obesity with lasting negative impacts on intrinsic and extrinsic bone strength. Exercise protected against obesity-associated skeletal changes with marked benefits on the biomechanical properties of bone.
Collapse
Affiliation(s)
- Pamela S Hinton
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, MO, USA.
| | - Kartik Shankar
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA, Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Lynn M Eaton
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - R Scott Rector
- Research Service-Harry S Truman Memorial Veterans Medical Center, and Departments of Medicine-Division of Gastroenterology and Hepatology and Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, MO, USA
| |
Collapse
|
19
|
Galea GL, Meakin LB, Savery D, Taipaleenmaki H, Delisser P, Stein GS, Copp AJ, van Wijnen AJ, Lanyon LE, Price JS. Planar cell polarity aligns osteoblast division in response to substrate strain. J Bone Miner Res 2015; 30:423-35. [PMID: 25264362 PMCID: PMC4333081 DOI: 10.1002/jbmr.2377] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/21/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023]
Abstract
Exposure of bone to dynamic strain increases the rate of division of osteoblasts and also influences the directional organization of the cellular and molecular structure of the bone tissue that they produce. Here, we report that brief exposure to dynamic substrate strain (sufficient to rapidly stimulate cell division) influences the orientation of osteoblastic cell division. The initial proliferative response to strain involves canonical Wnt signaling and can be blocked by sclerostin. However, the strain-related orientation of cell division is independently influenced through the noncanonical Wnt/planar cell polarity (PCP) pathway. Blockade of Rho-associated coiled kinase (ROCK), a component of the PCP pathway, prevents strain-related orientation of division in osteoblast-like Saos-2 cells. Heterozygous loop-tail mutation of the core PCP component van Gogh-like 2 (Vangl2) in mouse osteoblasts impairs the orientation of division in response to strain. Examination of bones from Vangl2 loop-tail heterozygous mice by µCT and scanning electron microscopy reveals altered bone architecture and disorganized bone-forming surfaces. Hence, in addition to the well-accepted role of PCP involvement in response to developmental cues during skeletal morphogenesis, our data reveal that this pathway also acts postnatally, in parallel with canonical Wnt signaling, to transduce biomechanical cues into skeletal adaptive responses. The simultaneous and independent actions of these two pathways appear to influence both the rate and orientation of osteoblast division, thus fine-tuning bone architecture to meet the structural demands of functional loading.
Collapse
Affiliation(s)
- Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Exercise and Regulation of Bone and Collagen Tissue Biology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 135:259-91. [DOI: 10.1016/bs.pmbts.2015.07.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Galea GL, Price JS. Four-point bending protocols to study the effects of dynamic strain in osteoblastic cells in vitro. Methods Mol Biol 2015; 1226:117-130. [PMID: 25331047 DOI: 10.1007/978-1-4939-1619-1_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Strain engendered within bone tissue by mechanical loading of the skeleton is a major influence on the processes of bone modeling and remodeling and so a critical determinant of bone mass and architecture. The cells best placed to respond to strain in bone tissue are the resident osteocytes and osteoblasts. To address the mechanisms of strain-related responses in osteoblast-like cells, our group uses both in vivo and in vitro approaches, including a system of four-point bending of the substrate on which cells are cultured. A range of cell lines can be studied using this system but we routinely compare their responses to those in primary cultures of osteoblast-like cells derived from explants of mouse long bones. These cells show a range of well-characterized responses to physiological levels of strain, including increased proliferation, which in vivo is a feature of the osteogenic response.
Collapse
Affiliation(s)
- Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Southwell Str, Bristol, BS2 8EJ, UK
| | | |
Collapse
|
22
|
Thaler R, Sturmlechner I, Spitzer S, Riester SM, Rumpler M, Zwerina J, Klaushofer K, van Wijnen AJ, Varga F. Acute-phase protein serum amyloid A3 is a novel paracrine coupling factor that controls bone homeostasis. FASEB J 2014; 29:1344-59. [PMID: 25491310 DOI: 10.1096/fj.14-265512] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/17/2014] [Indexed: 11/11/2022]
Abstract
Serum amyloid A (A-SAA/Saa3) was shown before to affect osteoblastic metabolism. Here, using RT-quantitative PCR and/or immunoblotting, we show that expression of mouse Saa3 and human SAA1 and SAA2 positively correlates with increased cellular maturation toward the osteocyte phenotype. Expression is not detected in C3H10T1/2 embryonic fibroblasts but is successively higher in preosteoblastic MC3T3-E1 cells, late osteoblastic MLO-A5 cells, and MLO-Y4 osteocytes, consistent with findings using primary bone cells from newborn mouse calvaria. Recombinant Saa3 protein functionally inhibits osteoblast differentiation as reflected by reductions in the expression of osteoblast markers and decreased mineralization in newborn mouse calvaria. Yet, Saa3 protein enhances osteoclastogenesis in mouse macrophages/monocytes based on the number of multinucleated and tartrate-resistant alkaline phosphatase-positive cells and Calcr mRNA expression. Depletion of Saa3 in MLO osteocytes results in the loss of the mature osteocyte phenotype. Recombinant osteocalcin, which is reciprocally regulated with Saa3 at the osteoblast/osteocyte transition, attenuates Saa3 expression in MLO-Y4 osteocytes. Mechanistically, Saa3 produced by MLO-Y4 osteocytes is integrated into the extracellular matrix of MC3T3-E1 osteoblasts, where it associates with the P2 purinergic receptor P2rx7 to stimulate Mmp13 expression via the P2rx7/MAPK/ERK/activator protein 1 axis. Our data suggest that Saa3 may function as an important coupling factor in bone development and homeostasis.
Collapse
Affiliation(s)
- Roman Thaler
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ines Sturmlechner
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Silvia Spitzer
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott M Riester
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Monika Rumpler
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jochen Zwerina
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Klaus Klaushofer
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andre J van Wijnen
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Franz Varga
- *Ludwig Boltzmann Institute of Osteology, Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt, Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; and Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Morse A, McDonald MM, Kelly NH, Melville KM, Schindeler A, Kramer I, Kneissel M, van der Meulen MCH, Little DG. Mechanical load increases in bone formation via a sclerostin-independent pathway. J Bone Miner Res 2014; 29:2456-67. [PMID: 24821585 PMCID: PMC4501925 DOI: 10.1002/jbmr.2278] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/22/2014] [Accepted: 05/06/2014] [Indexed: 01/24/2023]
Abstract
Sclerostin, encoded by the Sost gene, is an important negative regulator of bone formation that has been proposed to have a key role in regulating the response to mechanical loading. To investigate the effect of long-term Sclerostin deficiency on mechanotransduction in bone, we performed experiments on unloaded or loaded tibiae of 10 week old female Sost-/- and wild type mice. Unloading was induced via 0.5U botulinum toxin (BTX) injections into the right quadriceps and calf muscles, causing muscle paralysis and limb disuse. On a separate group of mice, increased loading was performed on the left tibiae through unilateral cyclic axial compression of equivalent strains (+1200 µe) at 1200 cycles/day, 5 days/week. Another cohort of mice receiving equivalent loads (-9.0 N) also were assessed. Contralateral tibiae served as normal load controls. Loaded/unloaded and normal load tibiae were assessed at day 14 for bone volume (BV) and formation changes. Loss of BV was seen in the unloaded tibiae of wild type mice, but BV was not different between normal load and unloaded Sost-/- tibiae. An increase in BV was seen in the loaded tibiae of wild type and Sost-/- mice over their normal load controls. The increased BV was associated with significantly increased mid-shaft periosteal mineralizing surface/bone surface (MS/BS), mineral apposition rate (MAR), and bone formation rate/bone surface (BFR/BS), and endosteal MAR and BFR/BS. Notably, loading induced a greater increase in periosteal MAR and BFR/BS in Sost-/- mice than in wild type controls. Thus, long-term Sclerostin deficiency inhibits the bone loss normally induced with decreased mechanical load, but it can augment the increase in bone formation with increased load.
Collapse
Affiliation(s)
- A Morse
- Orthopaedic Research and Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Campoli G, Baka N, Kaptein BL, Valstar ER, Zachow S, Weinans H, Zadpoor AA. Relationship between the shape and density distribution of the femur and its natural frequencies of vibration. J Biomech 2014; 47:3334-43. [PMID: 25171786 DOI: 10.1016/j.jbiomech.2014.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/04/2014] [Accepted: 08/08/2014] [Indexed: 11/17/2022]
Abstract
It has been recently suggested that mechanical loads applied at frequencies close to the natural frequencies of bone could enhance bone apposition due to the resonance phenomenon. Other applications of bone modal analysis are also suggested. For the above-mentioned applications, it is important to understand how patient-specific bone shape and density distribution influence the natural frequencies of bones. We used finite element models to study the effects of bone shape and density distribution on the natural frequencies of the femur in free boundary conditions. A statistical shape and appearance model that describes shape and density distribution independently was created, based on a training set of 27 femora. The natural frequencies were then calculated for different shape modes varied around the mean shape while keeping the mean density distribution, for different appearance modes around the mean density distribution while keeping the mean bone shape, and for the 27 training femora. Single shape or appearance modes could cause up to 15% variations in the natural frequencies with certain modes having the greatest impact. For the actual femora, shape and density distribution changed the natural frequencies by up to 38%. First appearance mode that describes the general cortical bone thickness and trabecular bone density had one of the strongest impacts. The first appearance mode could therefore provide a sensitive measure of general bone health and disease progression. Since shape and density could cause large variations in the calculated natural frequencies, patient-specific FE models are needed for accurate estimation of bone natural frequencies.
Collapse
Affiliation(s)
- G Campoli
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - N Baka
- Biomechanics and Imaging Group, Department of Orthopaedics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - B L Kaptein
- Biomechanics and Imaging Group, Department of Orthopaedics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - E R Valstar
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands; Biomechanics and Imaging Group, Department of Orthopaedics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - S Zachow
- Visualization & Data Analysis Medical Planning & Computational Medicine Groups, Zuse Institute Berlin (ZIB), Berlin, Germany
| | - H Weinans
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands; Departments of Orthopedics and Rheumatology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - A A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands.
| |
Collapse
|
25
|
Mosti MP, Carlsen T, Aas E, Hoff J, Stunes AK, Syversen U. Maximal Strength Training Improves Bone Mineral Density and Neuromuscular Performance in Young Adult Women. J Strength Cond Res 2014; 28:2935-45. [DOI: 10.1519/jsc.0000000000000493] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
26
|
Gombos Császár G, Bajsz V, Sió E, Steinhausz Tóth V, Schmidt B, Szekeres L, Kránicz J. The direct effect of specific training and walking on bone metabolic markers in young adults with peak bone mass. ACTA ACUST UNITED AC 2014; 101:205-15. [PMID: 24603147 DOI: 10.1556/aphysiol.101.2014.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As a prevention, a physically active lifestyle including the performance of weight-bearing exercises is important to enhance and maintain bone mineral content. Fifty young women were selected for the study. Twenty-five women carried out a specific training directed by a physiotherapist in the training group (TG), while 25 women were walking for 60 minutes in the control group (CG). Total and bone-specific alkaline phosphatase (ALP and BALP) and C-terminal cross-linked telopeptide (CTX) levels were measured at the beginning and at the end of exercise. The most remarkable change was seen in CTX levels (TG -28.89%, p < 0.001; CG -52.54%, p < 0.001), and there was also a significant difference in the values of CTX between TG and CG (p = 0.012). Therefore, walking more significantly reduced the level of CTX than special exercise. The decrease of BALP in TG was considerable but not significant (TG -4.63%, p = 0.091), while BALP levels dropped significantly in CG (-7.65%, p = 0.011), and there was a non-significant difference between the two groups (p = 0.22). Regarding the ALP level, a significant reduction was detected in TG and CG (-6.84%, p < 0.001 vs. -4.57%, p < 0.001). This study reveals that the 60-minute, middle-intensity training and the brisk walking have an immediate effect on bone metabolic markers.
Collapse
Affiliation(s)
- G Gombos Császár
- University of Pécs Faculty of Health Sciences Zalaegerszeg Hungary Landorhegyi utca 33 H-8900 Zalaegerszeg Hungary
| | - V Bajsz
- University of Pécs Faculty of Health Sciences Zalaegerszeg Hungary
| | - E Sió
- University of Pécs Faculty of Health Sciences Zalaegerszeg Hungary
| | | | - B Schmidt
- University of Pécs Faculty of Health Sciences Zalaegerszeg Hungary
| | - L Szekeres
- Hévíz Spa and St. Andrew Rheumatology Hospital Hévíz Hungary
| | - J Kránicz
- University of Pécs Faculty of Health Sciences Pécs Hungary
| |
Collapse
|
27
|
Amrein K, Dobnig H, Wagner D, Piswanger-Sölkner C, Pieber TR, Pilz S, Tomaschitz A, Dimai HP, Fahrleitner-Pammer A. Sclerostin in Institutionalized Elderly Women: Associations with Quantitative Bone Ultrasound, Bone Turnover, Fractures, and Mortality. J Am Geriatr Soc 2014; 62:1023-9. [DOI: 10.1111/jgs.12791] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Karin Amrein
- Division of Endocrinology and Metabolism; Medical University of Graz; Graz Austria
| | - Harald Dobnig
- Division of Endocrinology and Metabolism; Medical University of Graz; Graz Austria
| | - Doris Wagner
- Division of Transplantation Surgery; Department of Surgery; Medical University of Graz; Graz Austria
| | | | - Thomas R. Pieber
- Division of Endocrinology and Metabolism; Medical University of Graz; Graz Austria
| | - Stefan Pilz
- Division of Endocrinology and Metabolism; Medical University of Graz; Graz Austria
| | - Andreas Tomaschitz
- Division of Cardiology; Department of Internal Medicine; Medical University of Graz; Graz Austria
| | - Hans Peter Dimai
- Division of Endocrinology and Metabolism; Medical University of Graz; Graz Austria
| | | |
Collapse
|
28
|
Delgado-Calle J, Riancho JA, Klein-Nulend J. Nitric oxide is involved in the down-regulation of SOST expression induced by mechanical loading. Calcif Tissue Int 2014; 94:414-22. [PMID: 24322886 DOI: 10.1007/s00223-013-9821-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/23/2013] [Indexed: 01/08/2023]
Abstract
UNLABELLED Mechanical stimulation reduces sclerostin expression in rodents. However, few data are available about the effect of physical stimuli in human systems. Recently we observed that the demethylating agent AzadC induces SOST expression in bone cells. This allowed us in this study to explore the effect of mechanical loading on SOST expression by subjecting AzadC-treated human bone cells to pulsating fluid flow (PFF). PFF significantly decreased the AzadC-induced expression of SOST. This effect persisted for at least 24 h, and in fact SOST expression was lower at 24 h after PFF treatment than at 1 h after PFF treatment (PFF/static ratio 0.47 ± 0.04 vs. 0.63 ± 0.03 respectively, p = 0.03). The PFF-induced decrease in SOST expression was not due to a change in the methylation profile of the SOST promoter. However, PFF stimulated nitric oxide (NO) synthesis, which appeared essential for the PFF effect on SOST expression. In fact, the NO synthase inhibitor 1400 W prevented the effect of PFF on SOST expression. Moreover, the NO-donor SNAP decreased SOST mRNA in bone organ cultures. The conditioned medium (CM) of cells subjected to PFF induced a 38 ± 4 % decrease in SOST expression (p = 0.03) in static cultures and diminished the transcriptional activity of reporter vectors with the cloned SOST promoter (Static-CM: 1.47 ± 0.10 vs. PFF-CM 0.78 ± 0.09, p = 0.02). This is consistent with a PFF-induced secretion of factors that modulate SOST. Our results suggest that NO and other soluble factors are involved in the inhibition of SOST expression by PFF.
Collapse
Affiliation(s)
- Jesús Delgado-Calle
- Department of Internal Medicine, Hospital U. M. Valdecilla-IFIMAV, University of Cantabria, RETICEF, Av., Marqués de Valdecilla, s/n, 39008, Santander, Spain
| | | | | |
Collapse
|
29
|
Knapik DM, Perera P, Nam J, Blazek AD, Rath B, Leblebicioglu B, Das H, Wu LC, Hewett TE, Agarwal SK, Robling AG, Flanigan DC, Lee BS, Agarwal S. Mechanosignaling in bone health, trauma and inflammation. Antioxid Redox Signal 2014; 20:970-85. [PMID: 23815527 PMCID: PMC3924811 DOI: 10.1089/ars.2013.5467] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE Mechanosignaling is vital for maintaining the structural integrity of bone under physiologic conditions. These signals activate and suppress multiple signaling cascades regulating bone formation and resorption. Understanding these pathways is of prime importance to exploit their therapeutic potential in disorders associated with bone loss due to disuse, trauma, or disruption of homeostatic mechanisms. RECENT ADVANCES In the case of cells of the bone, an impressive amount of data has been generated that provides evidence of a complex mechanism by which mechanical signals can maintain or disrupt cellular homeostasis by driving transcriptional regulation of growth factors, matrix proteins and inflammatory mediators in health and inflammation. Mechanical signals act on cells in a magnitude dependent manner to induce bone deposition or resorption. During health, physiological levels of these signals are essential for maintaining bone strength and architecture, whereas during inflammation, similar signals can curb inflammation by suppressing the nuclear factor kappa B (NF-κB) signaling cascade, while upregulating matrix synthesis via mothers against decapentaplegic homolog and/or Wnt signaling cascades. Contrarily, excessive mechanical forces can induce inflammation via activation of the NF-κB signaling cascade. CRITICAL ISSUES Given the osteogenic potential of mechanical signals, it is imperative to exploit their therapeutic efficacy for the treatment of bone disorders. Here we review select signaling pathways and mediators stimulated by mechanical signals to modulate the strength and integrity of the bone. FUTURE DIRECTIONS Understanding the mechanisms of mechanotransduction and its effects on bone lay the groundwork for development of nonpharmacologic mechanostimulatory approaches for osteodegenerative diseases and optimal bone health.
Collapse
Affiliation(s)
- Derrick M Knapik
- 1 Department of Orthopaedic Surgery, The Ohio State University College of Medicine , Columbus, Ohio
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Liedert A, Schinke T, Ignatius A, Amling M. The role of midkine in skeletal remodelling. Br J Pharmacol 2014; 171:870-8. [PMID: 24102259 PMCID: PMC3925025 DOI: 10.1111/bph.12412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/02/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Bone tissue is subjected to continuous remodelling, replacing old or damaged bone throughout life. In bone remodelling, the coordinated activities of bone-forming osteoblasts and bone-resorbing osteoclasts ensure the maintenance of bone mass and strength. In early life, the balance of these cellular activities is tightly regulated by various factors, including systemic hormones, the mechanical environment and locally released growth factors. Age-related changes in the activity of these factors in bone remodelling can result in diseases with low bone mass, such as osteoporosis. Osteoporosis is a systemic and age-related skeletal disease characterized by low bone mass and structural degeneration of bone tissue, predisposing the patient to an increased fracture risk. The growth factor midkine (Mdk) plays a key role in bone remodelling and it is expressed during bone formation and fracture repair. Using a mouse deficient in Mdk, our group have identified this protein as a negative regulator of bone formation and mechanically induced bone remodelling. Thus, specific Mdk antagonists might represent a therapeutic option for diseases characterized by low bone mass, such as osteoporosis. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- A Liedert
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
31
|
Melville KM, Kelly NH, Khan SA, Schimenti JC, Ross FP, Main RP, van der Meulen MCH. Female mice lacking estrogen receptor-alpha in osteoblasts have compromised bone mass and strength. J Bone Miner Res 2014; 29:370-9. [PMID: 24038209 DOI: 10.1002/jbmr.2082] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/02/2013] [Accepted: 08/17/2013] [Indexed: 12/24/2022]
Abstract
Reduced bioavailability of estrogen increases skeletal fracture risk in postmenopausal women, but the mechanisms by which estrogen regulates bone mass are incompletely understood. Because estrogen signaling in bone acts, in part, through estrogen receptor alpha (ERα), mice with global deletion of ERα (ERαKO) have been used to determine the role of estrogen signaling in bone biology. These animals, however, have confounding systemic effects arising from other organs, such as increased estrogen and decreased insulin-like growth factor 1 (IGF-1) serum levels, which may independently affect bone. Mice with tissue-specific ERα deletion in chondrocytes, osteoblasts, osteocytes, or osteoclasts lack the systemic effects seen in the global knockout, but show that presence of the receptor is important for the function of each cell type. Although bone mass is reduced when ERα is deleted from osteoblasts, no study has determined if this approach reduces whole bone strength. To address this issue, we generated female osteoblast-specific ERαKO mice (pOC-ERαKO) by crossing mice expressing a floxed ERα gene (ERα(fl/fl)) with mice transgenic for the osteocalcin-Cre promoter (OC-Cre). Having confirmed that serum levels of estrogen and IGF-1 were unaltered, we focused on relating bone mechanics to skeletal phenotype using whole bone mechanical testing, microcomputed tomography, histology, and dynamic histomorphometry. At 12 and 18 weeks of age, pOC-ERαKO mice had decreased cancellous bone mass in the proximal tibia, vertebra, and distal femur, and decreased cortical bone mass in the tibial midshaft, distal femoral cortex, and L5 vertebral cortex. Osteoblast activity was reduced in cancellous bone of the proximal tibia, but osteoclast number was unaffected. Both femora and vertebrae had decreased whole bone strength in mechanical tests to failure, indicating that ERα in osteoblasts is required for appropriate bone mass and strength accrual in female mice. This pOC-ERαKO mouse is an important animal model that could enhance our understanding of estrogen signaling in bone cells in vivo.
Collapse
Affiliation(s)
- Katherine M Melville
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA; Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Meakin LB, Price JS, Lanyon LE. The Contribution of Experimental in vivo Models to Understanding the Mechanisms of Adaptation to Mechanical Loading in Bone. Front Endocrinol (Lausanne) 2014; 5:154. [PMID: 25324829 PMCID: PMC4181237 DOI: 10.3389/fendo.2014.00154] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/12/2014] [Indexed: 12/12/2022] Open
Abstract
Changing loading regimens by natural means such as exercise, with or without interference such as osteotomy, has provided useful information on the structure:function relationship in bone tissue. However, the greatest precision in defining those aspects of the overall strain environment that influence modeling and remodeling behavior has been achieved by relating quantified changes in bone architecture to quantified changes in bones' strain environment produced by direct, controlled artificial bone loading. Jiri Hert introduced the technique of artificial loading of bones in vivo with external devices in the 1960s using an electromechanical device to load rabbit tibiae through transfixing stainless steel pins. Quantifying natural bone strains during locomotion by attaching electrical resistance strain gages to bone surfaces was introduced by Lanyon, also in the 1960s. These studies in a variety of bones in a number of species demonstrated remarkable uniformity in the peak strains and maximum strain rates experienced. Experiments combining strain gage instrumentation with artificial loading in sheep, pigs, roosters, turkeys, rats, and mice has yielded significant insight into the control of strain-related adaptive (re)modeling. This diversity of approach has been largely superseded by non-invasive transcutaneous loading in rats and mice, which is now the model of choice for many studies. Together such studies have demonstrated that over the physiological strain range, bone's mechanically adaptive processes are responsive to dynamic but not static strains; the size and nature of the adaptive response controlling bone mass is linearly related to the peak loads encountered; the strain-related response is preferentially sensitive to high strain rates and unresponsive to static ones; is most responsive to unusual strain distributions; is maximized by remarkably few strain cycles, and that these are most effective when interrupted by short periods of rest between them.
Collapse
Affiliation(s)
- Lee B. Meakin
- School of Veterinary Sciences, University of Bristol, Bristol, UK
- *Correspondence: Lee B. Meakin, School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU, UK e-mail:
| | - Joanna S. Price
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Lance E. Lanyon
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
33
|
Abstract
The seronegative spondyloarthopathies (SpA) share certain common articular and peri-articular features that differ from rheumatoid arthritis (RA) and other forms of inflammatory arthritis. These include the tendency of the SpAs to involve the axial skeleton in addition to the diarthrodial joints, and the prominent involvement of the extra-articular entheses (sites of ligamentous and tendon insertion), which are not common sites of primary pathology in RA and other inflammatory arthropathies. The differential anatomic sites of bone pathology in the SpAs in comparison to the other forms of arthritis suggest that the underlying pathogenic processes and cellular and molecular mechanisms that account for the peri-articular bone pathology involve different underlying disease mechanisms. This review will highlight the molecular and cellular processes that are involved in the pathogenesis of the skeletal pathology in the SpAs, and provide evidence that many of the factors involved in regulation of bone cell function exhibit potent immune-regulatory activity, providing support for the general concept of osteoimmunology.
Collapse
|
34
|
Meakin LB, Sugiyama T, Galea GL, Browne WJ, Lanyon LE, Price JS. Male mice housed in groups engage in frequent fighting and show a lower response to additional bone loading than females or individually housed males that do not fight. Bone 2013; 54:113-7. [PMID: 23356987 PMCID: PMC3607215 DOI: 10.1016/j.bone.2013.01.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 12/21/2012] [Accepted: 01/16/2013] [Indexed: 12/11/2022]
Abstract
Experiments to investigate bone's physiological adaptation to mechanical loading frequently employ models that apply dynamic loads to bones in vivo and assess the changes in mass and architecture that result. It is axiomatic that bones will only show an adaptive response if the applied artificial loading environment differs in a significant way from that to which the bones have been habituated by normal functional loading. It is generally assumed that this normal loading is similar between experimental groups. In the study reported here we found that this was not always the case. Male and female 17-week-old C57BL/6 mice were housed in groups of six, and a single episode (40 cycles) of non-invasive axial loading, engendering 2,200 με on the medial surface of the proximal tibiae in sample mice, was applied to right tibiae on alternate days for two weeks. This engendered an adaptive increase in bone mass in females, but not males. Observation revealed the main difference in behaviour between males and females was that males were involved in fights 1.3 times per hour, whereas the females never fought. We therefore housed all mice individually. In females, there was a similar significant osteogenic response to loading in cortical and trabecular bone of both grouped and individual mice. In contrast, in males, adaptive increases in the loaded compared with non-loaded control bones was only apparent in animals housed individually. Our interpretation of these findings is that the frequent vigorous fighting that occurs between young adult males housed in groups could be sufficient to engender peak strains and strain rates that equal or exceed the stimulus derived from artificial loading. This indicates the importance of ensuring that physical activity is consistent between groups. Reducing the background level of the naturally engendered strain environment allows adaptive responses to artificial loading to be demonstrated at lower loads.
Collapse
Affiliation(s)
- Lee B Meakin
- School of Veterinary Sciences, University of Bristol, Bristol BS40 5DU, UK.
| | | | | | | | | | | |
Collapse
|
35
|
A rehabilitation exercise program induces severe bone mineral deficits in estrogen-deficient rats after extended disuse. Menopause 2013; 19:1267-76. [PMID: 22713863 DOI: 10.1097/gme.0b013e318255657f] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Both estrogen and mechanical loading regulate bone maintenance. However, mechanical overload seems less effective in enhancing bone mineral density (BMD) in estrogen-deficient women. The aim of this study was to determine whether estradiol (E2) influences early-phase bone adaptations to reambulation (REAMB) and/or rehabilitation exercises after hindlimb unloading (HLU) of ovariectomized rats. METHODS Eighty-one 5-month-old female Sprague-Dawley rats were randomized into the following groups: (1) intact controls, (2) ovariectomy (OVX), (3) OVX + E2, (4) OVX + 4 weeks of HLU, (5) OVX + E2 + HLU, (6) OVX + HLU + 2 weeks of quadrupedal REAMB, (7) OVX + E2 + HLU + REAMB, (8) OVX + HLU + REAMB + supplemental climbing, jumping, and balance exercises (EX), or (9) OVX + E2 + HLU + REAMB + EX. Serial dual-energy x-ray absorptiometry scans were performed to track total body bone characteristics throughout the study, and peripheral quantitative computerized tomography was used to determine distal femoral metaphyseal bone mineral characteristics. RESULTS Total body BMD increased by 4% to 8% in all animals receiving supplemental E2, whereas BMD did not change in animals without E2. OVX reduced trabecular BMD at the femoral metaphysis, and HLU exacerbated this loss while also reducing cortical BMD. E2 protected against OVX + HLU-induced bone loss at the femoral metaphysis. Conversely, REAMB did not alter BMD, regardless of estrogen status. In the absence of E2, REAMB + EX resulted in severe bone loss after OVX + HLU, with trabecular BMD and cortical BMD measurements that were 91% and 7% below those of controls, respectively (P ≤ 0.001). However, in the presence of E2, REAMB + EX did not negatively influence bone mineral characteristics. CONCLUSIONS E2 protects against bone loss resulting from combined OVX + HLU of rodents. In the absence of estrogen, exercise induces disadvantageous early-phase bone adaptations after extended disuse.
Collapse
|
36
|
Jastrzebski S, Kalinowski J, Stolina M, Mirza F, Torreggiani E, Kalajzic I, Won HY, Lee SK, Lorenzo J. Changes in bone sclerostin levels in mice after ovariectomy vary independently of changes in serum sclerostin levels. J Bone Miner Res 2013; 28:618-26. [PMID: 23044658 PMCID: PMC3554870 DOI: 10.1002/jbmr.1773] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 09/10/2012] [Accepted: 09/14/2012] [Indexed: 01/09/2023]
Abstract
We examined the effects that ovariectomy had on sclerostin mRNA and protein levels in the bones of 8-week-old mice that were either sham-operated (SHAM) or ovariectomized (OVX) and then euthanized 3 or 6 weeks later. In this model, bone loss occurred between 3 and 5 weeks postsurgery. In calvaria, ovariectomy significantly decreased sclerostin mRNA levels at 6 weeks postsurgery (by 52%) but had no significant effect at 3 weeks. In contrast, sclerostin mRNA levels were significantly lower in OVX femurs at 3 weeks postsurgery (by 53%) but equal to that of SHAM at 6 weeks. The effects of ovariectomy on sclerostin were not a global response of osteocytes because they were not mimicked by changes in the mRNA levels for two other relatively osteocyte-specific genes: DMP-1 and FGF-23. Sclerostin protein decreased by 83% and 60%, at 3 and 6 weeks postsurgery in calvaria, respectively, and by 38% in lumbar vertebrae at 6 weeks. We also detected decreases in sclerostin by immunohistochemistry in cortical osteocytes of the humerus at 3 weeks postsurgery. However, there were no significant effects of ovariectomy on sclerostin protein in femurs or on serum sclerostin at 3 and 6 weeks postsurgery. These results demonstrate that ovariectomy has variable effects on sclerostin mRNA and protein in mice, which are dependent on the bones examined and the time after surgery. Given the discrepancy between the effects of ovariectomy on serum sclerostin levels and sclerostin mRNA and protein levels in various bones, these results argue that, at least in mice, serum sclerostin levels may not accurately reflect changes in the local production of sclerostin in bones. Additional studies are needed to evaluate whether this is also the case in humans.
Collapse
Affiliation(s)
- Sandra Jastrzebski
- The Department of Medicine, The University of Connecticut Health Center, Farmington, CT, USA
| | - Judith Kalinowski
- The Department of Medicine, The University of Connecticut Health Center, Farmington, CT, USA
| | | | - Faryal Mirza
- The Department of Medicine, The University of Connecticut Health Center, Farmington, CT, USA
| | - Elena Torreggiani
- The Department of Reconstructive Sciences, The University of Connecticut Health Center, Farmington, CT, USA
| | - Ivo Kalajzic
- The Department of Reconstructive Sciences, The University of Connecticut Health Center, Farmington, CT, USA
| | - Hee Yeon Won
- The Center on Aging, The University of Connecticut Health Center, Farmington, CT, USA
| | - Sun-Kyeong Lee
- The Center on Aging, The University of Connecticut Health Center, Farmington, CT, USA
| | - Joseph Lorenzo
- The Department of Medicine, The University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
37
|
Kuriwaka-Kido R, Kido S, Miyatani Y, Ito Y, Kondo T, Omatsu T, Dong B, Endo I, Miyamoto KI, Matsumoto T. Parathyroid hormone (1-34) counteracts the suppression of interleukin-11 expression by glucocorticoid in murine osteoblasts: a possible mechanism for stimulating osteoblast differentiation against glucocorticoid excess. Endocrinology 2013; 154:1156-67. [PMID: 23397032 DOI: 10.1210/en.2013-1915] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glucocorticoid (GC) excess causes a rapid loss of bone with a reduction in bone formation. Intermittent PTH (1-34) administration stimulates bone formation and counteracts the inhibition of bone formation by GC excess. We have previously demonstrated that mechanical strain enhances interleukin (IL)-11 gene transcription by a rapid induction of ΔFosB expression and protein kinase C (PKC)-δ-mediated phosphorylation of phosphorylated mothers against decapentaplegic (Smad)-1. Because IL-11 suppresses the expression of dickkopf-1 and -2 and stimulates Wnt signaling, IL-11 appears to mediate at least a part of the effect of mechanical strain on osteoblast differentiation and bone formation. The present study was undertaken to examine the effect of PTH(1-34) and GCs on IL-11 expression in murine primary osteoblasts (mPOBs). PTH(1-34) treatment of mPOBs enhanced IL-11 expression in a time- and dose-dependent manner. PTH(1-34) also stimulated ΔFosB expression and Smad1 phosphorylation, which cooperatively stimulated IL-11 gene transcription. PTH(1-34)-induced Smad1 phosphorylation was mediated via PKCδ and was abrogated in mPOBs from PKCδ knockout mice. Dexamethasone suppressed IL-11 gene transcription enhanced by PTH(1-34) without affecting ΔFosB expression or Smad1 phosphorylation, and dexamethasone-GC receptor complex was bound to JunD, which forms heterodimers with ΔFosB. High doses of PTH(1-34) counteracted the effect of dexamethasone on apoptosis of mPOBs, which was blunted by neutralizing anti-IL-11 antibody or IL-11 small interfering RNA. These results demonstrate that PTH(1-34) and GCs interact to regulate IL-11 expression in parallel with osteoblast differentiation and apoptosis and suggest that PTH(1-34) and dexamethasone may regulate osteoblast differentiation and apoptosis via their effect on IL-11 expression.
Collapse
Affiliation(s)
- Rika Kuriwaka-Kido
- MD, Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medicine, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cidem M, Usta TA, Karacan I, Kucuk SH, Uludag M, Gun K. Effects of sex steroids on serum sclerostin levels during the menstrual cycle. Gynecol Obstet Invest 2013; 75:179-84. [PMID: 23429230 DOI: 10.1159/000347013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 01/02/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Previous studies on the effects of estrogen on sclerostin regulation were conducted in postmenopausal women in humans or animals following estrogen treatment or induced estrogen deficiency. The aim of this study was to evaluate the effects of sex hormones on serum sclerostin levels in premenopausal women with normal menstrual cycles. METHODS A prospective observational clinical study. 80 voluntary premenopausal women were recruited for the study. Data from 31 patients were available for the statistical analysis. Serum sclerostin, free estradiol, free testosterone, and progesterone levels were measured during the menstruation, late follicular and mid-luteal phases. The unique protocol IDs were BEAH FTR-4 and NCT01418924 at ClinicalTrials.gov ID. RESULTS Serum sclerostin values were 1.03 ± 0.58 ng/ml during the menstruation phase, 1.0 ± 0.36 ng/ml during the late follicular phase, and 1.18 ± 0.67 ng/ml during the mid-luteal phase (p = 0.543). There was no significant relationship between serum levels of sex steroids and sclerostin. CONCLUSIONS Previous studies have not investigated the impact of sex hormone fluctuations on serum sclerostin levels during the menstrual cycle. The present study shows that serum sclerostin levels were not affected by sex steroids in premenopausal women with normal menstrual cycles.
Collapse
Affiliation(s)
- Muharrem Cidem
- Department of Physical Medicine and Rehabilitation, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
39
|
Komori T. Functions of the osteocyte network in the regulation of bone mass. Cell Tissue Res 2013; 352:191-8. [PMID: 23329124 PMCID: PMC3637644 DOI: 10.1007/s00441-012-1546-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 12/05/2012] [Indexed: 12/11/2022]
Abstract
Osteocytes establish an extensive intracellular and extracellular communication system via gap-junction-coupled cell processes and canaliculi throughout bone and the communication system is extended to osteoblasts on the bone surface. The osteocyte network is an ideal mechanosensory system and suitable for mechanotransduction. However, the overall function of the osteocyte network remains to be clarified, since bone resorption is enhanced by osteocyte apoptosis, which is followed by a process of secondary necrosis attributable to the lack of scavengers. The enhanced bone resorption is caused by the release of intracellular content, including immunostimulatory molecules that activate osteoclastogenesis through the canaliculi. Therefore, a mouse model is required in which the osteocyte network is disrupted but in which no bone resorption is induced, in order to evaluate the overall functions of the osteocyte network. One such model is the BCL2 transgenic mouse, in which the osteocyte network, including both intracellular and extracellular networks, is disrupted. Another model is the osteocyte-specific Gja1 knockout mouse, in which intercellular communication through gap junctions is impaired but the canalicular system is intact. Combining the findings from these mouse models with previous histological observations showing the inverse linkage between osteocyte density and bone formation, we conclude that the osteocyte network enhances bone resorption and inhibits bone formation under physiological conditions. Further, studies with BCL2 transgenic mice show that these osteocyte functions are augmented in the unloaded condition. In this condition, Rankl upregulation in osteoblasts and Sost upregulation in osteocytes are, at least in part, responsible for enhanced bone resorption and suppressed bone formation, respectively.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Cell Biology, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| |
Collapse
|
40
|
Nguyen J, Tang SY, Nguyen D, Alliston T. Load regulates bone formation and Sclerostin expression through a TGFβ-dependent mechanism. PLoS One 2013; 8:e53813. [PMID: 23308287 PMCID: PMC3538690 DOI: 10.1371/journal.pone.0053813] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 12/04/2012] [Indexed: 11/19/2022] Open
Abstract
Bone continually adapts to meet changing physical and biological demands. Osteoblasts, osteoclasts, and osteocytes cooperate to integrate these physical and biochemical cues to maintain bone homeostasis. Although TGFβ acts on all three of these cell types to maintain bone homeostasis, the extent to which it participates in the adaptation of bone to mechanical load is unknown. Here, we investigated the role of the TGFβ pathway in load-induced bone formation and the regulation of Sclerostin, a mechanosensitive antagonist of bone anabolism. We found that mechanical load rapidly represses the net activity of the TGFβ pathway in osteocytes, resulting in reduced phosphorylation and activity of key downstream effectors, Smad2 and Smad3. Loss of TGFβ sensitivity compromises the anabolic response of bone to mechanical load, demonstrating that the mechanosensitive regulation of TGFβ signaling is essential for load-induced bone formation. Furthermore, sensitivity to TGFβ is required for the mechanosensitive regulation of Sclerostin, which is induced by TGFβ in a Smad3-dependent manner. Together, our results show that physical cues maintain bone homeostasis through the TGFβ pathway to regulate Sclerostin expression and the deposition of new bone.
Collapse
Affiliation(s)
- Jacqueline Nguyen
- Graduate Program in Oral and Craniofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Simon Y. Tang
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Daniel Nguyen
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Tamara Alliston
- Graduate Program in Oral and Craniofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Department of Otolaryngology Head and Neck Surgery, University of California San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Sugiyama T, Meakin LB, Galea GL, Lanyon LE, Price JS. The cyclooxygenase-2 selective inhibitor NS-398 does not influence trabecular or cortical bone gain resulting from repeated mechanical loading in female mice. Osteoporos Int 2013; 24:383-8. [PMID: 22349912 PMCID: PMC3536947 DOI: 10.1007/s00198-012-1922-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/17/2012] [Indexed: 11/30/2022]
Abstract
UNLABELLED A single injection of the cyclooxygenase-2 (COX-2) selective inhibitor NS-398 reduces bone's osteogenic response to a single period of mechanical loading in female rats, while women taking COX-2 selective inhibitors do not have lower bone mass. We show that daily NS-398 injection does not influence bone gain from repeated loading in female mice. INTRODUCTION Prostaglandins are mediators of bone cells' early response to mechanical stimulation. COX-2 expression is up-regulated by exposure of these cells to mechanical strain or fluid flow, and the osteogenic response to a single loading period is reduced by COX-2 inhibition. This study determined, in female mice in vivo, the effect of longer term COX-2 inhibition on adaptive (re)modelling of cortical and trabecular bone in response to repeated loading. METHODS Nineteen-week-old female C57BL/6 mice were injected with vehicle or NS-398 (5 mg/kg/day) 5 days a week for 2 weeks. On three alternate days each week, the right tibiae/fibulae were axially loaded [40 cycles (7 min)/day] three hours after injection. Left limbs acted as internal controls. Changes in three-dimensional bone architecture were analysed by high-resolution micro-computed tomography. RESULTS In control limbs NS-398 was associated with reduced trabecular number but had no influence on cortical bone. In loaded limbs trabecular thickness and cortical periosteally enclosed volume increased. NS-398 showed no effect on this response. CONCLUSION Pharmacological inhibition of COX-2 by NS-398 does not affect trabecular or cortical bone's response to repeated mechanical loading in female mice and thus would not be expected to impair the functional adaptation of bone to physical activity in women.
Collapse
Affiliation(s)
- T Sugiyama
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK.
| | | | | | | | | |
Collapse
|
42
|
Ke HZ, Richards WG, Li X, Ominsky MS. Sclerostin and Dickkopf-1 as therapeutic targets in bone diseases. Endocr Rev 2012; 33:747-83. [PMID: 22723594 DOI: 10.1210/er.2011-1060] [Citation(s) in RCA: 304] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The processes of bone growth, modeling, and remodeling determine the structure, mass, and biomechanical properties of the skeleton. Dysregulated bone resorption or bone formation may lead to metabolic bone diseases. The Wnt pathway plays an important role in bone formation and regeneration, and expression of two Wnt pathway inhibitors, sclerostin and Dickkopf-1 (DKK1), appears to be associated with changes in bone mass. Inactivation of sclerostin leads to substantially increased bone mass in humans and in genetically manipulated animals. Studies in various animal models of bone disease have shown that inhibition of sclerostin using a monoclonal antibody (Scl-Ab) increases bone formation, density, and strength. Additional studies show that Scl-Ab improves bone healing in models of bone repair. Inhibition of DKK1 by monoclonal antibody (DKK1-Ab) stimulates bone formation in younger animals and to a lesser extent in adult animals and enhances fracture healing. Thus, sclerostin and DKK1 are emerging as the leading new targets for anabolic therapies to treat bone diseases such as osteoporosis and for bone repair. Clinical trials are ongoing to evaluate the effects of Scl-Ab and DKK1-Ab in humans for the treatment of bone loss and for bone repair.
Collapse
Affiliation(s)
- Hua Zhu Ke
- Metabolic Disorders Research, Amgen Inc., One Amgen Center Drive, MS 29-M-B, Thousand Oaks, California 91320, USA.
| | | | | | | |
Collapse
|
43
|
Miller PD. A review of the efficacy and safety of denosumab in postmenopausal women with osteoporosis. Ther Adv Musculoskelet Dis 2012; 3:271-82. [PMID: 22870485 DOI: 10.1177/1759720x11424220] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Denosumab, a fully human monoclonal antibody to RANK ligand [RANK-L]) was approved for the treatment of postmenopausal osteoporosis in June 2010, and is highly effective in reducing the risk of vertebral, nonvertebral, and hip fracture risk. The registration of denosumab was the culmination of the discovery and clarification of the internal bone microenvironment regulation of bone remodeling: the osteoblast-produced competitors: RANK-L and osteoprotogerin; and the osteoclast receptor: RANK. When RANK-L is upregulated in the estrogen-deficient state and exceeds the amount of osteoprotogerin, there is an increase in osteoclastogenesis and bone resorption, and this is the major mechanism for bone mass loss and osteoporotic fractures in the postmenopausal state. The subsequent development of the human monoclonal antibody to RANK-L (denosumab) was the first product developed to reduce bone resorption by inhibiting RANK-L binding to RANK. Denosumab does not accumulate in bone, and has a unique pharmacokinetics so that its biological effect at the registered dose of 60 mg by subcutaneous injection every 6 months is no longer effective, at least as measured by an increase in the bone resorption marker collagen-cross-link C-telopeptide and a decline in bone mineral density as measured by dual energy X-ray absorptiometry. This unique pharmacokinetic profile thus suggests that in order to maintain the effectiveness of denosumab, continuous administration might be necessary. Extension of the registration trial ('FREEDOM') 5-year data indicates continued safety and efficacy, and will be extended to 10 years so that even longer-term data will be forthcoming. The profound but reversible suppression of bone turnover that is seen with denosumab partly explains the continuous increase in bone mineral density seen through 8 years of the phase II clinical trials. Denosumab offers a highly effective and safe parenteral therapy for osteoporosis and is being studied long term with the extension of the FREEDOM trial, and in other osteoporotic states - in men and glucocorticoid-induced osteoporosis.
Collapse
|
44
|
Thompson WR, Rubin CT, Rubin J. Mechanical regulation of signaling pathways in bone. Gene 2012; 503:179-93. [PMID: 22575727 DOI: 10.1016/j.gene.2012.04.076] [Citation(s) in RCA: 279] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/20/2012] [Accepted: 04/22/2012] [Indexed: 12/21/2022]
Abstract
A wide range of cell types depend on mechanically induced signals to enable appropriate physiological responses. The skeleton is particularly dependent on mechanical information to guide the resident cell population towards adaptation, maintenance and repair. Research at the organ, tissue, cell and molecular levels has improved our understanding of how the skeleton can recognize the functional environment, and how these challenges are translated into cellular information that can site-specifically alter phenotype. This review first considers those cells within the skeleton that are responsive to mechanical signals, including osteoblasts, osteoclasts, osteocytes and osteoprogenitors. This is discussed in light of a range of experimental approaches that can vary parameters such as strain, fluid shear stress, and pressure. The identity of mechanoreceptor candidates is approached, with consideration of integrins, pericellular tethers, focal adhesions, ion channels, cadherins, connexins, and the plasma membrane including caveolar and non-caveolar lipid rafts and their influence on integral signaling protein interactions. Several mechanically regulated intracellular signaling cascades are detailed including activation of kinases (Akt, MAPK, FAK), β-catenin, GTPases, and calcium signaling events. While the interaction of bone cells with their mechanical environment is complex, an understanding of mechanical regulation of bone signaling is crucial to understanding bone physiology, the etiology of diseases such as osteoporosis, and to the development of interventions to improve bone strength.
Collapse
Affiliation(s)
- William R Thompson
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
45
|
Johannesdottir F, Aspelund T, Siggeirsdottir K, Jonsson BY, Mogensen B, Sigurdsson S, Harris TB, Gudnason VG, Lang TF, Sigurdsson G. Mid-thigh cortical bone structural parameters, muscle mass and strength, and association with lower limb fractures in older men and women (AGES-Reykjavik Study). Calcif Tissue Int 2012; 90:354-64. [PMID: 22451219 PMCID: PMC5111551 DOI: 10.1007/s00223-012-9585-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 03/04/2012] [Indexed: 10/28/2022]
Abstract
In a cross-sectional study we investigated the relationship between muscle and bone parameters in the mid-thigh in older people using data from a single axial computed tomographic section through the mid-thigh. Additionally, we studied the association of these variables with incident low-trauma lower limb fractures. A total of 3,762 older individuals (1,838 men and 1,924 women), aged 66-96 years, participants in the AGES-Reykjavik study, were studied. The total cross-sectional muscular area and knee extensor strength declined with age similarly in both sexes. Muscle parameters correlated most strongly with cortical area and total shaft area (adjusted for age, height, and weight) but explained <10 % of variability in those bone parameters. The increment in medullary area (MA) and buckling ratio (BR) with age was almost fourfold greater in women than men. The association between MA and muscle parameters was nonsignificant. During a median follow-up of 5.3 years, 113 women and 66 men sustained incident lower limb fractures. Small muscular area, low knee extensor strength, large MA, low cortical thickness, and high BR were significantly associated with fractures in both sexes. Our results show that bone and muscle loss proceed at different rates and with different gender patterns.
Collapse
Affiliation(s)
| | - Thor Aspelund
- University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | | | | | - Brynjolfur Mogensen
- University of Iceland, Reykjavik, Iceland
- Landspitali-Univerisity Hospital, Reykjavik, Iceland
| | | | | | - Vilmundur G. Gudnason
- University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | | | - Gunnar Sigurdsson
- University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
- Landspitali-Univerisity Hospital, Reykjavik, Iceland
| |
Collapse
|
46
|
Moustafa A, Sugiyama T, Prasad J, Zaman G, Gross TS, Lanyon LE, Price JS. Mechanical loading-related changes in osteocyte sclerostin expression in mice are more closely associated with the subsequent osteogenic response than the peak strains engendered. Osteoporos Int 2012; 23:1225-34. [PMID: 21573880 PMCID: PMC3304063 DOI: 10.1007/s00198-011-1656-4] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 04/20/2011] [Indexed: 01/10/2023]
Abstract
UNLABELLED Osteocyte sclerostin is regulated by loading and disuse in mouse tibiae but is more closely related to subsequent local osteogenesis than the peak strains engendered. INTRODUCTION The purpose of this study was to assess the relationship between loading-related change in osteocyte sclerostin expression, local strain magnitude, and local bone modeling/remodeling. METHODS The right tibiae of 19-week-old female C57BL/6 mice were subjected to non-invasive, dynamic axial loading and/or to sciatic neurectomy-induced disuse. The sclerostin status of osteocytes was evaluated immunohistochemically, changes in bone mass by micro-computed tomography, new bone formation by histomorphometry, and loading-induced strain by strain gauges and finite element analysis. RESULTS In cortical bone of the tibial shaft, loading engendered strains of similar peak magnitude proximally and distally. Proximally, sclerostin-positive osteocytes decreased and new bone formation increased. Distally, there was neither decrease in sclerostin-positive osteocytes nor increased osteogenesis. In trabecular bone of the proximal secondary spongiosa, loading decreased sclerostin-positive osteocytes and increased bone volume. Neither occurred in the primary spongiosa. Disuse increased sclerostin-positive osteocytes and decreased bone volume at all four sites. Loading reversed this sclerostin upregulation to a level below baseline in the proximal cortex and secondary spongiosa. CONCLUSION Loading-related sclerostin downregulation in osteocytes of the mouse tibia is associated preferentially with regions where new bone formation is stimulated rather than where high peak strains are engendered. The mechanisms involved remain unclear, but could relate to peak surface strains not accurately reflecting the strain-related osteogenic stimulus or that sclerostin regulation occurs after sufficient signal processing to distinguish between local osteogenic and non-osteogenic responses.
Collapse
Affiliation(s)
- A. Moustafa
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK
- Department of Surgery, Faculty of Veterinary Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - T. Sugiyama
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU UK
| | - J. Prasad
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA USA
| | - G. Zaman
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK
| | - T. S. Gross
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA USA
| | - L. E. Lanyon
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU UK
| | - J. S. Price
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU UK
| |
Collapse
|
47
|
Bergström I, Parini P, Gustafsson SA, Andersson G, Brinck J. Physical training increases osteoprotegerin in postmenopausal women. J Bone Miner Metab 2012; 30:202-7. [PMID: 21823052 DOI: 10.1007/s00774-011-0304-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 07/04/2011] [Indexed: 11/28/2022]
Abstract
The purpose of this study was to explore whether mechanical loading by exercise over a 1-year period in postmenopausal women had an effect on the receptor activator for nuclear factor kappa B ligand/osteoprotegerin (RANKL/OPG) system or the levels of the Wnt-signaling antagonist sclerostin. A total of 112 postmenopausal were randomized to either sedentary life (controls) or physical activity (training group). Ninety-two women fulfilled the study protocol. The training program consisted of three fast 30-min walks and one or two 1-h aerobic training sessions per week. The effect on the bone mineral density of the hip assessed with dual X-ray absorptiometry was positive as reported earlier. Blood samples were taken from participants at baseline and after 1 year and serum levels of OPG, RANKL and sclerostin were quantified together with the bone metabolism markers C-terminal telopeptide of collagen type I (CTX) and bone-specific alkaline phosphatase (BALP). The results were analyzed using an analysis of covariance model using baseline values as the covariate. The training group displayed a clear mean increase of OPG +7.55 pg/ml compared to controls (p = 0.007). The mean changes for RANKL +0.19 pg/ml (square-root transformed data) and sclerostin +0.62 pmol/l were non-significant (p = 0.13 and p = 0.34). The changes in bone turnover markers CTX and BALP showed a tendency to decrease in the training group versus controls but the changes were small and non-significant. Although our study is limited in number of participating women, we have been able to show an OPG-associated, and RANKL- and sclerostin-independent, training-induced inhibition of postmenopausal bone loss.
Collapse
Affiliation(s)
- Ingrid Bergström
- Division of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
48
|
McGonnell IM, Grigoriadis AE, Lam EWF, Price JS, Sunters A. A specific role for phosphoinositide 3-kinase and AKT in osteoblasts? Front Endocrinol (Lausanne) 2012; 3:88. [PMID: 22833734 PMCID: PMC3400941 DOI: 10.3389/fendo.2012.00088] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 06/29/2012] [Indexed: 12/25/2022] Open
Abstract
The phosphoinositide 3-kinase and AKT (protein kinase B) signaling pathway (PI3K/AKT) plays a central role in the control of cell survival, growth, and proliferation throughout the body. With regard to bone, and particularly in osteoblasts, there is an increasing amount of evidence that the many signaling molecules exert some of their bone-specific effects in part via selectively activating some of the generic effects of the PI3K/AKT pathway in osteoblasts. There is further data demonstrating that PI3K/AKT has the capacity to specifically cross-talk with other signaling pathways and transcriptional networks controlling bone cells' development in order to fine-tune the osteoblast phenotype. There is also evidence that perturbations in the PI3K/AKT pathway may well be responsible for certain bone pathologies. In this review, we discuss some of these findings and suggest that the PI3K/AKT pathway is a central nexus in the extensive network of extracellular signaling pathways that control the osteoblast.
Collapse
Affiliation(s)
- Imelda M. McGonnell
- Department of Veterinary Basic Sciences, The Royal Veterinary College,London, UK
| | - Agamemnon E. Grigoriadis
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, Guy’s Hospital,London, UK
| | - Eric W.-F. Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital,London, UK
| | - Joanna S. Price
- School of Veterinary Sciences, University of Bristol,Bristol, UK
| | - Andrew Sunters
- Department of Veterinary Basic Sciences, The Royal Veterinary College,London, UK
- *Correspondence: Andrew Sunters, Department of Veterinary Basic Sciences, The Royal Veterinary College, Royal College Street, Camden, London NW1 0TU, UK. e-mail:
| |
Collapse
|
49
|
Abstract
With an increase in the average life span especially in the Western hemisphere, there is renewed interest in treating maladies of old age including osteoporosis. Age-related bone loss and resultant osteoporosis substantially increase risk of fractures and morbidity in the geriatric population leading to both a decline in the quality of life for the elderly as well as a substantial burden on the health care system. Herein, we review recent research in murine and rodent models looking at how both extrinsic and intrinsic factors such as hormones, biochemicals, neuromodulators, inflammatory cytokines, oxidative stress, nutrition, and exercise influence the skeleton with age. Recent studies on the relationship between bone and fat in the marrow, and the fate of the marrow mesenchymal stromal cell population, which can give rise to either bone-forming osteoblasts or fat-forming adipocytic cells as a function of age, have also been highlighted. An appreciable range of studies using aging murine as well as cellular models are discussed, as these studies have broadened our understanding of the pathways and players in the aging bone. Impactful information regarding aging and the bone may then allow the application of better pharmacologic as well as nonpharmacologic regimens to alleviate bone loss due to aging.
Collapse
Affiliation(s)
- Farhan A Syed
- Abbott Bioresearch Center, Worcester, MA 01545, USA.
| | | |
Collapse
|
50
|
Shah K, Armamento-Villareal R, Parimi N, Chode S, Sinacore DR, Hilton TN, Napoli N, Qualls C, Villareal DT. Exercise training in obese older adults prevents increase in bone turnover and attenuates decrease in hip bone mineral density induced by weight loss despite decline in bone-active hormones. J Bone Miner Res 2011; 26:2851-9. [PMID: 21786319 PMCID: PMC3206995 DOI: 10.1002/jbmr.475] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Weight loss therapy to improve health in obese older adults is controversial because it causes further bone loss. Therefore, it is recommended that weight loss therapy should include an intervention such as exercise training (ET) to minimize bone loss. The purpose of this study was to determine the independent and combined effects of weight loss and ET on bone metabolism in relation to bone mineral density (BMD) in obese older adults. One-hundred-seven older (age >65 years) obese (body mass index [BMI] ≥ 30 kg/m(2) ) adults were randomly assigned to a control group, diet group, exercise group, and diet-exercise group for 1 year. Body weight decreased in the diet (-9.6%) and diet-exercise (-9.4%) groups, not in the exercise (-1%) and control (-0.2%) groups (between-group p < 0.001). However, despite comparable weight loss, bone loss at the total hip was relatively less in the diet-exercise group (-1.1%) than in the diet group (-2.6%), whereas BMD increased in the exercise group (1.5%) (between-group p < 0.001). Serum C-terminal telopeptide (CTX) and osteocalcin concentrations increased in the diet group (31% and 24%, respectively), whereas they decreased in the exercise group (-13% and -15%, respectively) (between-group p < 0.001). In contrast, similar to the control group, serum CTX and osteocalcin concentrations did not change in the diet-exercise group. Serum procollagen propeptide concentrations decreased in the exercise group (-15%) compared with the diet group (9%) (p = 0.04). Serum leptin and estradiol concentrations decreased in the diet (-25% and -15%, respectively) and diet-exercise (-38% and -13%, respectively) groups, not in the exercise and control groups (between-group p = 0.001). Multivariate analyses revealed that changes in lean body mass (β = 0.33), serum osteocalcin (β = -0.24), and one-repetition maximum (1-RM) strength (β = 0.23) were independent predictors of changes in hip BMD (all p < 0.05). In conclusion, the addition of ET to weight loss therapy among obese older adults prevents weight loss-induced increase in bone turnover and attenuates weight loss-induced reduction in hip BMD despite weight loss-induced decrease in bone-active hormones.
Collapse
Affiliation(s)
- Krupa Shah
- Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|