51
|
Cannarella R, Barbagallo F, Condorelli RA, Aversa A, La Vignera S, Calogero AE. Osteoporosis from an Endocrine Perspective: The Role of Hormonal Changes in the Elderly. J Clin Med 2019; 8:jcm8101564. [PMID: 31581477 PMCID: PMC6832998 DOI: 10.3390/jcm8101564] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/09/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Introduction: Osteoporosis is increasingly prevalent in the elderly, with fractures mostly occurring in women and men who are older than 55 and 65 years of age, respectively. The aim of this review was to examine the evidence regarding the influence of hormones on bone metabolism, followed by clinical data of hormonal changes in the elderly, in the attempt to provide possible poorly explored diagnostic and therapeutic candidate targets for the management of primary osteoporosis in the aging population. Material and methods: An extensive Medline search using PubMed, Embase, and Cochrane Library was performed. Results: While the rise in Thyroid-stimulating hormone (TSH) levels has a protective role on bone mass, the decline of estrogen, testosterone, Insulin-like growth factor 1 (IGF1), and vitamin D and the rise of cortisol, parathyroid hormone, and follicle-stimulating hormone (FSH) favor bone loss in the elderly. Particularly, the AA rs6166 FSH receptor (FSHR) genotype, encoding for a more sensitive FSHR than that encoded by the GG one, is associated with low total body mass density (BMD), independently of circulating estrogen. A polyclonal antibody with a FSHR-binding sequence against the β-subunit of murine FSH seems to be effective in ameliorating bone loss in ovariectomized mice. Conclusions: A complete hormonal assessment should be completed for both women and men during bone loss evaluation. Novel possible diagnostic and therapeutic tools might be developed for the management of male and female osteoporosis.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Federica Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
52
|
Farr JN, Rowsey JL, Eckhardt BA, Thicke BS, Fraser DG, Tchkonia T, Kirkland JL, Monroe DG, Khosla S. Independent Roles of Estrogen Deficiency and Cellular Senescence in the Pathogenesis of Osteoporosis: Evidence in Young Adult Mice and Older Humans. J Bone Miner Res 2019; 34:1407-1418. [PMID: 30913313 PMCID: PMC6697189 DOI: 10.1002/jbmr.3729] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/27/2019] [Accepted: 03/15/2019] [Indexed: 11/10/2022]
Abstract
Estrogen deficiency is a seminal mechanism in the pathogenesis of osteoporosis. Mounting evidence, however, establishes that cellular senescence, a fundamental mechanism that drives multiple age-related diseases, also causes osteoporosis. Recently, we systematically identified an accumulation of senescent cells, characterized by increased p16Ink4a and p21Cip1 levels and development of a senescence-associated secretory phenotype (SASP), in mouse bone/marrow and human bone with aging. We then demonstrated that elimination of senescent cells prevented age-related bone loss using multiple approaches, eg, treating old mice expressing a "suicide" transgene, INK-ATTAC, with AP20187 to induce apoptosis of p16Ink4a -senescent cells or periodically treating old wild-type mice with "senolytics," ie, drugs that eliminate senescent cells. Here, we investigate a possible role for estrogen in the regulation of cellular senescence using multiple approaches. First, sex steroid deficiency 2 months after ovariectomy (OVX, n = 15) or orchidectomy (ORCH, n = 15) versus sham surgery (SHAM, n = 15/sex) in young adult (4-month-old) wild-type mice did not alter senescence biomarkers or induce a SASP in bone. Next, in elderly postmenopausal women, 3 weeks of estrogen therapy (n = 10; 74 ± 5 years) compared with no treatment (n = 10; 78 ± 5 years) did not alter senescence biomarkers or the SASP in human bone biopsies. Finally, young adult (4-month-old) female INK-ATTAC mice were randomized (n = 17/group) to SHAM+Vehicle, OVX+Vehicle, or OVX+AP20187 for 2 months. As anticipated, OVX+Vehicle caused significant trabecular/cortical bone loss compared with SHAM+Vehicle. However, treatment with AP20187, which eliminates senescent cells in INK-ATTAC mice, did not rescue the OVX-induced bone loss or alter senescence biomarkers. Collectively, our data establish independent roles of estrogen deficiency and cellular senescence in the pathogenesis of osteoporosis, which has important implications for testing novel senolytics for skeletal efficacy, as these drugs will need to be evaluated in preclinical models of aging as opposed to the current FDA model of prevention of OVX-induced bone loss. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Joshua N Farr
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jennifer L Rowsey
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brittany A Eckhardt
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brianne S Thicke
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Daniel G Fraser
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - David G Monroe
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
53
|
Shih YRV, Liu M, Kwon SK, Iida M, Gong Y, Sangaj N, Varghese S. Dysregulation of ectonucleotidase-mediated extracellular adenosine during postmenopausal bone loss. SCIENCE ADVANCES 2019; 5:eaax1387. [PMID: 31457100 PMCID: PMC6703860 DOI: 10.1126/sciadv.aax1387] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/12/2019] [Indexed: 05/25/2023]
Abstract
Adenosine and its receptors play a key role in bone homeostasis and regeneration. Extracellular adenosine is generated from CD39 and CD73 activity in the cell membrane, through conversion of adenosine triphosphate to adenosine monophosphate (AMP) and AMP to adenosine, respectively. Despite the relevance of CD39/CD73 to bone health, the roles of these enzymes in bona fide skeletal disorders remain unknown. We demonstrate that CD39/CD73 expression and extracellular adenosine levels in the bone marrow are substantially decreased in animals with osteoporotic bone loss. Knockdown of estrogen receptors ESR1 and ESR2 in primary osteoprogenitors and osteoclasts undergoing differentiation showed decreased coexpression of membrane-bound CD39 and CD73 and lower extracellular adenosine. Targeting the adenosine A2B receptor using an agonist attenuated bone loss in ovariectomized mice. Together, these findings suggest a pathological association of purine metabolism with estrogen deficiency and highlight the potential of A2B receptor as a target to treat osteoporosis.
Collapse
Affiliation(s)
- Yu-Ru V. Shih
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Mengqian Liu
- Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Seong Keun Kwon
- Department of Otorhinolaryngology–Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | | | - Ya Gong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nivedita Sangaj
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
- Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
54
|
Shahoei SH, Nelson ER. Nuclear receptors, cholesterol homeostasis and the immune system. J Steroid Biochem Mol Biol 2019; 191:105364. [PMID: 31002862 PMCID: PMC6589364 DOI: 10.1016/j.jsbmb.2019.04.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/30/2022]
Abstract
Cholesterol is essential for maintaining membrane fluidity in eukaryotes. Additionally, the synthetic cascade of cholesterol results in precursor molecules important for cellular function such as lipid raft formation and protein prenylation. As such, cholesterol homeostasis is tightly regulated. Interestingly, it is now known that some cholesterol precursors and many metabolites serve as active signaling molecules, binding to different classes of receptors including the nuclear receptors. Furthermore, many cholesterol metabolites or their nuclear receptors have been implicated in the regulation of the immune system in normal physiology and disease. Therefore, in this focused review, cholesterol homeostasis and nuclear receptors involved in this regulation will be discussed, with particular emphasis on how these cascades influence the immune system.
Collapse
Affiliation(s)
- Sayyed Hamed Shahoei
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, United States; Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL, United States.
| |
Collapse
|
55
|
The Role of Macrophage in the Pathogenesis of Osteoporosis. Int J Mol Sci 2019; 20:ijms20092093. [PMID: 31035384 PMCID: PMC6539137 DOI: 10.3390/ijms20092093] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/20/2019] [Accepted: 04/26/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is a systemic disease with progressive bone loss. The bone loss is associated with an imbalance between bone resorption via osteoclasts and bone formation via osteoblasts. Other cells including T cells, B cells, macrophages, and osteocytes are also involved in the pathogenesis of osteoporosis. Different cytokines from activated macrophages can regulate or stimulate the development of osteoclastogenesis-associated bone loss. The fusion of macrophages can form multinucleated osteoclasts and, thus, cause bone resorption via the expression of IL-4 and IL-13. Different cytokines, endocrines, and chemokines are also expressed that may affect the presentation of macrophages in osteoporosis. Macrophages have an effect on bone formation during fracture-associated bone repair. However, activated macrophages may secrete proinflammatory cytokines that induce bone loss by osteoclastogenesis, and are associated with the activation of bone resorption. Targeting activated macrophages at an appropriate stage may help inhibit or slow the progression of bone loss in patients with osteoporosis.
Collapse
|
56
|
Henstridge DC, Abildgaard J, Lindegaard B, Febbraio MA. Metabolic control and sex: A focus on inflammatory-linked mediators. Br J Pharmacol 2019; 176:4193-4207. [PMID: 30820935 DOI: 10.1111/bph.14642] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/05/2018] [Accepted: 01/21/2019] [Indexed: 12/15/2022] Open
Abstract
Men and women have many differing biological and physiological characteristics. Thus, it is no surprise that the control of metabolic processes and the mechanisms underlying metabolic-related diseases have sex-specific components. There is a clear metabolic sexual dimorphism in that up until midlife, men have a far greater likelihood of acquiring cardio-metabolic disease than women. Following menopause, however, this difference is reduced, suggestive of a protective role of the female sex hormones. Inflammatory processes have been implicated in the pathogenesis of cardio-metabolic disease with human studies correlating metabolic disease acquisition or risk with levels of various inflammatory markers. Rodent studies employing genetic modifications or novel pharmacological approaches have provided mechanistic insight into the role of these inflammatory mediators. Sex differences impact inflammatory processes and the subsequent biological response. As a consequence, this may affect how inflammation alters metabolic processes between the sexes. Recently, some of our work in the field of inflammatory genes and metabolic control identified a sexual dimorphism in a preclinical model and caused us to question the frequency and scale of such findings in the literature. This review concentrates on inflammatory-related signalling in relation to obesity, insulin resistance, and type 2 diabetes and highlights the differences observed between males and females. Differences in the activation and signalling of various inflammatory genes and proteins present another reason why studying both male and female patients or animals is important in the context of understanding and finding therapeutics for metabolic-related disease. LINKED ARTICLES: This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Darren C Henstridge
- Molecular Metabolism & Aging Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Julie Abildgaard
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Lindegaard
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Pulmonary and Infectious Diseases, Nordsjaellands Hospital, Hillerød, Denmark
| | - Mark A Febbraio
- Division of Diabetes & Metabolism, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,Drug Discover Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| |
Collapse
|
57
|
Yuksel S, Guleç MA, Gultekin Z, Caglar A, Beytemur O, Alagoz E, Eker AA, Subaşı C, Karaoz E. Histopathological, immunohistochemical, and biomechanical effects of splenectomy on Achilles tendon healing in rats. Connect Tissue Res 2019; 60:200-208. [PMID: 29860899 DOI: 10.1080/03008207.2018.1483361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE This study aimed to assess Achilles tendon repair in rats following splenectomy to simulate patients with musculoskeletal system injury who had splenectomy after spleen injury, a situation often seen in orthopedics and traumatology practice. MATERIALS AND METHODS The study included 32 male Sprague-Dawley rats (10 months old; average weight, 394.5 ± 28.3 g). The rats were fed with standard rodent food ad libitum at 22°C in a dark environment for 12 h. They were divided into two groups, namely the splenectomy (total splenectomy and Achilles tendon repair) and control groups (only Achilles tendon repair; n = 16). Four weeks after the surgery, the rats were euthanized, and their Achilles tendons were examined histopathologically, immunohistochemically, and biomechanically. RESULTS In the splenectomy group, proinflammatory cytokines, such as interleukin-1β, tumor necrosis factor-α, and interferon-γ, showed significantly lower values than those in the control group (p ˂0.01); moreover, the levels of anti-inflammatory cytokines like vascular endothelial growth factor, transforming growth factor-β1, interleukin-2, interleukin-10, and hepatocyte growth factor were significantly higher than in the control group (p ˂ 0.001). The average ultimate tensile strengths were 2.58 ± 0.5 in the splenectomy and 2.78 ± 0.3 in the control group (p = 0.043). The average εUTS values were 0.33 ± 0.5 in the splenectomy and 0.44 ± 0.1 in the control group (p = 0.021). CONCLUSION Splenectomy may positively influence Achilles tendon healing through modification of the proinflammatory/anti-inflammatory ratio in favor of anti-inflammatory cytokines by causing a decrease in spleen-originated inflammatory cells.
Collapse
Affiliation(s)
- Serdar Yuksel
- a Bağcılar Training and Research Hospital , Department of Orthopedics and Traumatology , Istanbul , Turkey
| | - M Akif Guleç
- a Bağcılar Training and Research Hospital , Department of Orthopedics and Traumatology , Istanbul , Turkey
| | - Zeki Gultekin
- a Bağcılar Training and Research Hospital , Department of Orthopedics and Traumatology , Istanbul , Turkey
| | - Aysel Caglar
- b Bağcılar Training and Research Hospital , Department of Pathology , Istanbul , Turkey
| | - Ozan Beytemur
- a Bağcılar Training and Research Hospital , Department of Orthopedics and Traumatology , Istanbul , Turkey
| | - Ender Alagoz
- a Bağcılar Training and Research Hospital , Department of Orthopedics and Traumatology , Istanbul , Turkey
| | - A Akdogan Eker
- c Yıldız Technical University , Department of Mechanical Engineering , Istanbul , Turkey
| | - Cansu Subaşı
- d Center for Regenerative Medicine and Stem Cell Research & Manufacturing (LivMedCell) , Istanbul , Turkey
| | - Erdal Karaoz
- d Center for Regenerative Medicine and Stem Cell Research & Manufacturing (LivMedCell) , Istanbul , Turkey.,e İstinye University, Center for Stem Cell and Tissue Engineering Research & Practice , İstanbul , Turkey
| |
Collapse
|
58
|
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
|
59
|
Gender-independent efficacy of mesenchymal stem cell therapy in sex hormone-deficient bone loss via immunosuppression and resident stem cell recovery. Exp Mol Med 2018; 50:1-14. [PMID: 30559383 PMCID: PMC6297134 DOI: 10.1038/s12276-018-0192-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis develops with high prevalence in both postmenopausal women and hypogonadal men. Osteoporosis results in significant morbidity, but no cure has been established. Mesenchymal stem cells (MSCs) critically contribute to bone homeostasis and possess potent immunomodulatory/anti-inflammatory capability. Here, we investigated the therapeutic efficacy of using an infusion of MSCs to treat sex hormone-deficient bone loss and its underlying mechanisms. In particular, we compared the impacts of MSC cytotherapy in the two genders with the aim of examining potential gender differences. Using the gonadectomy (GNX) model, we confirmed that the osteoporotic phenotypes were substantially consistent between female and male mice. Importantly, systemic MSC transplantation (MSCT) not only rescued trabecular bone loss in GNX mice but also restored cortical bone mass and bone quality. Unexpectedly, no differences were detected between the genders. Furthermore, MSCT demonstrated an equal efficiency in rectifying the bone remodeling balance in both genders of GNX animals, as proven by the comparable recovery of bone formation and parallel normalization of bone resorption. Mechanistically, using green fluorescent protein (GFP)-based cell-tracing, we demonstrated rapid engraftment but poor inhabitation of donor MSCs in the GNX recipient bone marrow of each gender. Alternatively, MSCT uniformly reduced the CD3+T-cell population and suppressed the serum levels of inflammatory cytokines in reversing female and male GNX osteoporosis, which was attributed to the ability of the MSC to induce T-cell apoptosis. Immunosuppression in the microenvironment eventually led to functional recovery of endogenous MSCs, which resulted in restored osteogenesis and normalized behavior to modulate osteoclastogenesis. Collectively, these data revealed recipient sexually monomorphic responses to MSC therapy in gonadal steroid deficiency-induced osteoporosis via immunosuppression/anti-inflammation and resident stem cell recovery.
Collapse
|
60
|
Hormone Replacement Therapy: Would it be Possible to Replicate a Functional Ovary? Int J Mol Sci 2018; 19:ijms19103160. [PMID: 30322209 PMCID: PMC6214095 DOI: 10.3390/ijms19103160] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 01/05/2023] Open
Abstract
Background: Throughout history, menopause has been regarded as a transition in a woman’s life. With the increase in life expectancy, women now spend more than a third of their lives in menopause. During these years, women may experience intolerable symptoms both physically and mentally, leading them to seek clinical advice. It is imperative for healthcare providers to improve the quality of life by reducing bothersome menopausal symptoms and preventing disorders such as osteoporosis and atherosclerosis. The current treatment in the form of hormone replacement therapy (HRT) is sometimes inadequate with several limitations and adverse effects. Objective and rationale: The current review aims to discuss the need, efficacy, and limitations of current HRT; the role of other ovarian hormones, and where we stand in comparison with ovary-in situ; and finally, explore towards the preparation of an HRT model by regeneration of ovaries tissues through stem cells which can replicate a functional ovary. Search methods: Four electronic databases (MEDLINE, Embase, Web of Science and CINAHL) were searched from database inception until 26 April 2018, using a combination of relevant controlled vocabulary terms and free-text terms related to ‘menopause’, ‘hormone replacement therapy’, ‘ovary regeneration’, ‘stem cells’ and ‘ovarian transplantation’. Outcomes: We present a synthesis of the existing data on the efficacy and limitations of HRT. HRT is far from adequate in postmenopausal women with symptoms of hormone deprivation as it fails to deliver all hormones secreted by naïve ovarian tissue. Moreover, the pharmacokinetics of synthetic hormones makes them substantially different from natural ones. Not only does the number and type of hormones given in HRT matter, but the route of delivering and their release in circulation are also imperative. The hormones are delivered either orally or topically in a non-physiological uniform manner, which brings along with it several side effects. These identify the need for a hormone delivery system which replicates, integrates and reacts as per the requirement of the female body. Wider implications: The review outlines the strengths and weaknesses of HRT and highlights the potential areas for future research. There is a tremendous potential for research in this field to understand the collective roles of the various ovarian hormones and to devise an auto-regulated hormone delivery system which replicates the normal physiology. Its clinical applications can prove to be transformative for postmenopausal women helping them to lead a healthy and productive life.
Collapse
|
61
|
Jones RM, Mulle JG, Pacifici R. Osteomicrobiology: The influence of gut microbiota on bone in health and disease. Bone 2018; 115:59-67. [PMID: 28433758 DOI: 10.1016/j.bone.2017.04.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/18/2017] [Accepted: 04/18/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Rheinallt M Jones
- Department of Pediatrics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Jennifer G Mulle
- Department of Human Genetics, Emory University School of Medicine, United States
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, United States; Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, United States.
| |
Collapse
|
62
|
Tang M, Tian L, Luo G, Yu X. Interferon-Gamma-Mediated Osteoimmunology. Front Immunol 2018; 9:1508. [PMID: 30008722 PMCID: PMC6033972 DOI: 10.3389/fimmu.2018.01508] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/18/2018] [Indexed: 02/05/2023] Open
Abstract
Osteoimmunology is the interdiscipline that focuses on the relationship between the skeletal and immune systems. They are interconnected by shared signal pathways and cytokines. Interferon-gamma (IFN-γ) plays important roles in immune responses and bone metabolism. IFN-γ enhances macrophage activation and antigen presentation. It regulates antiviral and antibacterial immunity as well as signal transduction. IFN-γ can promote osteoblast differentiation and inhibit bone marrow adipocyte formation. IFN-γ plays dual role in osteoclasts depending on its stage. Furthermore, IFN-γ is an important pathogenetic factor in some immune-mediated bone diseases including rheumatoid arthritis, postmenopausal osteoporosis, and acquired immunodeficiency syndrome. This review will discuss the contradictory findings of IFN-γ in osteoimmunology and its clinical application potential.
Collapse
Affiliation(s)
| | | | | | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
63
|
Shao B, Fu X, Yu Y, Yang D. Regulatory effects of miRNA‑181a on FasL expression in bone marrow mesenchymal stem cells and its effect on CD4+T lymphocyte apoptosis in estrogen deficiency‑induced osteoporosis. Mol Med Rep 2018; 18:920-930. [PMID: 29845202 PMCID: PMC6059724 DOI: 10.3892/mmr.2018.9026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 04/19/2018] [Indexed: 12/29/2022] Open
Abstract
Post-menopausal osteoporosis is a bone formation disorder induced by estrogen deficiency. Estrogen deficiency facilitates the differentiation and maturation of osteoclasts by activating T lymphocytes. In our previous study, it was demonstrated that estrogen promotes bone marrow mesenchymal stem cell (BMMSC)‑induced osteoclast apoptosis through downregulation of microRNA (miR)‑181a and subsequent Fas ligand (FasL) protein accumulation. In the present study, the regulatory effects of miR‑181a on FasL expression in BMMSCs and the apoptotic effects of BMMSCs on cluster of differentiation (CD)4+T lymphocytes were investigated. An ovariectomized mouse model of osteoporosis (OVX) was established and CD4+T lymphocytes were isolated from the bones of these mice. The results demonstrated that the number of CD4+T lymphocytes was increased in the OVX group compared within the control group, thus suggesting that estrogen deficiency may increase CD4+T lymphocyte number. CD4+T lymphocytes were subsequently co‑cultured with estrogen‑treated BMMSCs, after which it was demonstrated that estrogen significantly promoted the apoptosis of CD4+T lymphocytes. Western blot analysis indicated that estrogen promoted the apoptosis of CD4+T lymphocytes through regulation of FasL expression in BMMSCs in a concentration‑dependent manner. Finally, miR‑181a was transfected into BMMSCs, which were co‑cultured with CD4+T lymphocytes in vitro and in vivo. The results revealed that miR‑181a exerted a negative regulatory effect on BMMSC‑induced CD4+T lymphocyte apoptosis by regulating FasL protein expression in BMMSCs; this maybe a key mechanism underlying the development of estrogen deficiency‑induced osteoporosis.
Collapse
Affiliation(s)
- Bingyi Shao
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Xiaohui Fu
- Department of Orthodontics, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yang Yu
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Deqin Yang
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing 401147, P.R. China
| |
Collapse
|
64
|
Abstract
Exposed surfaces of mammals are colonized with 100 trillion indigenous bacteria, fungi, and viruses, creating a diverse ecosystem known as the microbiome. The gastrointestinal tract harbors the greatest numbers of these microorganisms, which regulate human nutrition, metabolism, and immune system function. Moreover, the intestinal microbiota contains pro- and anti-inflammatory products that modulate immune responses and may play a role in maintaining gut barrier function. Therefore, the community composition of the microbiota has profound effects on the immune status of the host and impacts the development and/or progression of inflammatory diseases. Accordingly, numerous studies have shown differences in the microbiota of patients with and without a given inflammatory condition. There is now strong evidence that the gut microbiome regulates bone homeostasis in health and disease, and that prebiotic and probiotics protect against bone loss. Herein, the evidence supporting the role of the microbiota and the effects of prebiotic and probiotics will be reviewed.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
65
|
Graef JL, Ouyang P, Wang Y, Rendina-Ruedy E, Lerner MR, Marlow D, Lucas EA, Smith BJ. Dried Plum Polyphenolic Extract Combined with Vitamin K and Potassium Restores Trabecular and Cortical Bone in Osteopenic Model of Postmenopausal Bone Loss. J Funct Foods 2018; 42:262-270. [PMID: 30319713 DOI: 10.1016/j.jff.2017.12.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Dried plum has unique anabolic effects on bone, but the responsible bioactive components have remained unclear. This study investigated components of dried plum with potential osteoprotective activity utilizing aged, osteopenic Sprague Dawley rats fed diets supplemented with a crude polyphenol extract, potassium, vitamin K or their combination. Whole body and femoral bone mineral density were restored with the polyphenol and combination treatments to a similar extent as the dried fruit. The combination treatment reversed trabecular bone loss in the spine and cortical bone in the femur mid-diaphysis in a similar manner. Biomarkers of bone resorption were reduced by the polyphenol and combination treatments. The polyphenol extract accounted for most of the anabolic effect of dried plum on bone. This study is the first to show the bioactive components in dried plum responsible for restoring bone in vivo.
Collapse
Affiliation(s)
- Jennifer L Graef
- Department of Nutritional Sciences, Oklahoma State University, 301 Human Sciences, Stillwater, OK 74078
| | - Ping Ouyang
- Department of Nutritional Sciences, Oklahoma State University, 301 Human Sciences, Stillwater, OK 74078
| | - Yan Wang
- Department of Nutritional Sciences, Oklahoma State University, 301 Human Sciences, Stillwater, OK 74078
| | - Elizabeth Rendina-Ruedy
- Department of Nutritional Sciences, Oklahoma State University, 301 Human Sciences, Stillwater, OK 74078
| | - Megan R Lerner
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Denver Marlow
- Comparative Medicine Group, Kansas State University, Manhattan, KS 66506
| | - Edralin A Lucas
- Department of Nutritional Sciences, Oklahoma State University, 301 Human Sciences, Stillwater, OK 74078
| | - Brenda J Smith
- Department of Nutritional Sciences, Oklahoma State University, 301 Human Sciences, Stillwater, OK 74078
| |
Collapse
|
66
|
Xu X, Jia X, Mo L, Liu C, Zheng L, Yuan Q, Zhou X. Intestinal microbiota: a potential target for the treatment of postmenopausal osteoporosis. Bone Res 2017; 5:17046. [PMID: 28983411 PMCID: PMC5627629 DOI: 10.1038/boneres.2017.46] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Postmenopausal osteoporosis (PMO) is a prevalent metabolic bone disease characterized by bone loss and structural destruction, which increases the risk of fracture in postmenopausal women. Owing to the high morbidity and serious complications of PMO, many efforts have been devoted to its prophylaxis and treatment. The intestinal microbiota is the complex community of microorganisms colonizing the gastrointestinal tract. Probiotics, which are dietary or medical supplements consisting of beneficial intestinal bacteria, work in concert with endogenous intestinal microorganisms to maintain host health. Recent studies have revealed that bone loss in PMO is closely related to host immunity, which is influenced by the intestinal microbiota. The curative effects of probiotics on metabolic bone diseases have also been demonstrated. The effects of the intestinal microbiota on bone metabolism suggest a promising target for PMO management. This review seeks to summarize the critical effects of the intestinal microbiota and probiotics on PMO, with a focus on the molecular mechanisms underlying the pathogenic relationship between bacteria and host, and to define the possible treatment options.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyue Jia
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Longyi Mo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Dental Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
67
|
Graef JL, Rendina-Ruedy E, Crockett EK, Ouyang P, Wu L, King JB, Cichewicz RH, Lin D, Lucas EA, Smith BJ. Osteoclast Differentiation is Downregulated by Select Polyphenolic Fractions from Dried Plum via Suppression of MAPKs and Nfatc1 in Mouse C57BL/6 Primary Bone Marrow Cells. Curr Dev Nutr 2017; 1:e000406. [PMID: 29955675 PMCID: PMC5998775 DOI: 10.3945/cdn.117.000406] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/08/2017] [Accepted: 09/06/2017] [Indexed: 02/04/2023] Open
Abstract
Background: Clinical and preclinical studies have shown that dietary supplementation with dried plum improves bone health. These osteoprotective effects are a result, in part, of the antiresorptive properties of the fruit, which appear to be mediated by its polyphenolic compounds. Objective: This study was designed to determine if certain fractions of the polyphenolic compounds in dried plums are responsible for the antiresorptive effects and whether they alter mitogen-activated protein kinase (MAPK) and calcium signaling, which are essential to osteoclast differentiation and activity, under normal and inflammatory conditions. Methods: Six polyphenolic fractions were derived from the total polyphenolic extract of dried plum based on solubility. Initial screening, with the use of the Raw 264.7 monocyte and macrophage cell line, showed that 3 fractions had the most marked capacity to downregulate osteoclast differentiation. This response was confirmed in 2 of the fractions by using primary bone marrow-derived cultures and in all subsequent experiments to determine how osteoclast differentiation and function were altered with a focus on these 2 fractions in primary cultures. Data were analyzed by using ANOVA followed by post hoc analyses. Results: Both of the polyphenol fractions decreased osteoclast differentiation and activity coincident with downregulating nuclear factor of activated T cells, cytoplasmic, calcineurin-dependent 1 (Nfatc1), which is required for osteoclast differentiation. Calcium signaling, essential for the auto-amplification of Nfatc1, was suppressed by the polyphenolic fractions under normal conditions as indicated by suppressed mRNA expression of costimulatory receptors osteoclast-associated receptor (Oscar), signaling regulatory protein β1 (Sirpb1), and triggering receptor expressed on myeloid cells 2 (Trem2). In contrast, in the presence of tumor necrosis factor α (TNF-α), only Sirpb1 was downregulated. In addition to calcium signaling, phosphorylation of extracellular signal-regulated kinase (Erk) and p38 MAPK, involved in the expression and activation of Nfatc1, was also suppressed by the polyphenolic fractions. Conclusion: These results show that certain types of polyphenolic compounds from dried plum downregulate calcium and MAPK signaling, resulting in suppression of Nfatc1 expression, which ultimately decreases osteoclast formation and activity.
Collapse
Affiliation(s)
- Jennifer L Graef
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| | | | - Erica K Crockett
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| | - Ping Ouyang
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| | - Lei Wu
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| | - Jarrod B King
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK
| | - Robert H Cichewicz
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK
| | - Dingbo Lin
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| | - Edralin A Lucas
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| | - Brenda J Smith
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
| |
Collapse
|
68
|
Abstract
Osteoporosis increases fracture risk, a cause of crippling morbidity and mortality. The immunoskeletal interface (ISI) is a centralization of cell and cytokine effectors shared between skeletal and immune systems. Consequently, the immune system mediates powerful effects on bone turnover. Physiologically, B cells secrete osteoprotegerin (OPG), a potent anti-osteoclastogenic factor that preserves bone mass. However, activated T cells and B cells secrete pro-osteoclastogenic factors including receptor activator of Nuclear factor-kappaB (NF-kB) ligand (RANKL), Interleukin (IL)-17A, and tumor necrosis factor (TNF)-α promoting bone loss in inflammatory states such as rheumatoid arthritis. Recently, ISI disruption has been linked to osteoporosis in human immunodeficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS), where elevated B cell RANKL and diminished OPG drive bone resorption. HIV-antiretroviral therapy paradoxically intensifies bone loss during disease reversal, as immune reconstitution produces osteoclastogenic cytokines. Interestingly, in estrogen deficiency, activated T cells secrete RANKL, TNF, and IL-17A that amplify bone resorption and contribute to postmenopausal osteoporosis. T cell-produced TNF and IL-17A further contribute to bone loss in hyperparathyroidism, while T cell production of the anabolic Wingless integration site (Wnt) ligand, Wnt10b, promotes bone formation in response to anabolic parathyroid hormone and the immunomodulatory costimulation inhibitor cytotoxic T lymphocyte-associated protein-4-IgG (abatacept). These findings provide a window into the workings of the ISI and suggest novel targets for future therapeutic interventions to reduce fracture risk.
Collapse
Affiliation(s)
- M Neale Weitzmann
- 1 Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, Georgia, USA
- 2 Department of Medicine, Division of Endocrinology and Metabolism and Lipids, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
69
|
Okamoto K, Nakashima T, Shinohara M, Negishi-Koga T, Komatsu N, Terashima A, Sawa S, Nitta T, Takayanagi H. Osteoimmunology: The Conceptual Framework Unifying the Immune and Skeletal Systems. Physiol Rev 2017; 97:1295-1349. [DOI: 10.1152/physrev.00036.2016] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.
Collapse
Affiliation(s)
- Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Tomoki Nakashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masahiro Shinohara
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takako Negishi-Koga
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Noriko Komatsu
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Asuka Terashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Shinichiro Sawa
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takeshi Nitta
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
70
|
Collins FL, Rios-Arce ND, McCabe LR, Parameswaran N. Cytokine and hormonal regulation of bone marrow immune cell Wnt10b expression. PLoS One 2017; 12:e0181979. [PMID: 28800644 PMCID: PMC5553813 DOI: 10.1371/journal.pone.0181979] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/10/2017] [Indexed: 11/28/2022] Open
Abstract
Background & aims Wnt10b is a crucial regulator of bone density through its ability to promote osteoblastogenesis. Parathyroid hormone has been shown to regulate Wnt10b expression in CD8+ T cells. However, the relative expression and other source(s) of Wnt10b in the bone marrow immune cells (BMICs) is unknown. Sex hormones and cytokines such as, estrogen and TNFα are critical regulators of bone physiology but whether they regulate BMIC Wnt10b expression is unclear. To determine the potential regulation of Wnt10b by estrogen and TNFα, we assessed Wnt10b expression by flow cytometry under estrogen- and TNFα-deficient conditions. Methods Effects of TNFα was determined in male and female C57BL/6 wildtype and TNFα knockout mice. Effect of estrogen was investigated 4, 6 and 8 weeks post-surgery in ovariectomized Balb/c mice. Intracellular Wnt10b was detected using goat anti-mouse Wnt10b and a conjugated secondary antibody and analyzed by flow cytometry. Results Wnt10b expression was sex- and lineage-specific. Females had 1.8-fold higher Wnt10b signal compared to males. Percent of Wnt10b+ myeloid cells was higher in females than males (8.9% Vs 5.4%) but Wnt10b+ lymphoid cells was higher in males than females (6.3% Vs 2.5%). TNFα ablation in males increased total BM Wnt10b expression 1.5-fold but significantly reduced the percentage of BM Wnt10b+ CD4+ T cells (65%), CD8+ T cells (59%), dendritic cells (59%), macrophages (56%) and granulocytes (52%). These effects of TNFα on Wnt10b were observed only in males. In contrast to TNFα, estrogen-deficiency had indirect effects on BMIC Wnt10b levels; reducing the average percentage of BM Wnt10b+ CD8+ T cells (25%) and granulocytes (26%) across an 8-week time course. Conclusion Our results demonstrate unique cell type- and sex-dependent effects on BMIC Wnt10b expression. Together, our results reveal myeloid cells in the bone marrow as an important source of Wnt10b under complex hormonal and cytokine regulation.
Collapse
Affiliation(s)
- Fraser L. Collins
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
| | - Laura R. McCabe
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
- Department of Radiology, Michigan State University, East Lansing, Michigan, United States of America
- Biomedical Imaging Research Centre, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail: (NP); (LRM)
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail: (NP); (LRM)
| |
Collapse
|
71
|
Abstract
PURPOSE OF REVIEW Mounting evidence supporting the critical contribution of macrophages, in particular osteal macrophages, to bone regeneration is reviewed. We specifically examine the potential role of macrophages in the basic multicellular units coordinating lifelong bone regeneration via remodelling and bone regeneration in response to injury. We review and discuss the distinctions between macrophage and osteoclast contributions to bone homeostasis, particularly the dichotomous role of the colony-stimulating factor 1-colony-stimulating factor 1 receptor axis. RECENT FINDINGS The impact of inflammation associated with aging and other hallmarks of aging, including senescence, on macrophage function is addressed in the context of osteoporosis and delayed fracture repair. Resident macrophages versus recruited macrophage contributions to fracture healing are also discussed. We identify some of the remaining knowledge gaps that will need to be closed in order to maximise benefits from therapeutically modulating or mimicking the function of macrophages to improve bone health and regeneration over a lifetime.
Collapse
Affiliation(s)
- Lena Batoon
- Bones and Immunology Laboratory, Cancer Biology and Care Program, Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Susan Marie Millard
- Bones and Immunology Laboratory, Cancer Biology and Care Program, Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Liza Jane Raggatt
- Bones and Immunology Laboratory, Cancer Biology and Care Program, Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4092, Australia
| | - Allison Robyn Pettit
- Bones and Immunology Laboratory, Cancer Biology and Care Program, Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4092, Australia.
| |
Collapse
|
72
|
Li J, Wang Q, Yang R, Zhang J, Li X, Zhou X, Miao D. BMI-1 Mediates Estrogen-Deficiency-Induced Bone Loss by Inhibiting Reactive Oxygen Species Accumulation and T Cell Activation. J Bone Miner Res 2017; 32:962-973. [PMID: 27943387 DOI: 10.1002/jbmr.3059] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/03/2016] [Accepted: 12/08/2016] [Indexed: 11/07/2022]
Abstract
Previous studies have shown that estrogen regulates bone homeostasis through regulatory effects on oxidative stress. However, it is unclear how estrogen deficiency triggers reactive oxygen species (ROS) accumulation. Recent studies provide evidence that the B lymphoma Mo-MLV insertion region 1 (BMI-1) plays a critical role in protection against oxidative stress and that this gene is directly regulated by estrogen via estrogen receptor (ER) at the transcriptional level. In this study, ovariectomized mice were given drinking water with/without antioxidant N-acetyl-cysteine (NAC, 1 mg/mL) supplementation, and compared with each other and with sham mice. Results showed that ovariectomy resulted in bone loss with increased osteoclast surface, increased ROS levels, T cell activation, and increased TNF and RANKL levels in serum and in CD4 T cells; NAC supplementation largely prevented these alterations. BMI-1 expression levels were dramatically downregulated in CD4 T cells from ovariectomized mice. We supplemented drinking water to BMI-1-deficient mice with/without NAC and compared them with each other and with wild-type (WT) mice. We found that BMI-1 deficiency mimicked alterations observed in ovariectomy whereas NAC supplementation reversed all alterations induced by BMI-1 deficiency. Because T cells are critical in mediating ovariectomy-induced bone loss, we further assessed whether BMI-1 overexpression in lymphocytes can protect against estrogen deficiency-induced osteoclastogenesis and bone loss by inhibiting oxidative stress, T cell activation, and RANKL production. When WT and Eμ-BMI-1 transgenic mice with BMI-1 specifically overexpressed in lymphocytes were ovariectomized and compared with each other and with WT sham mice, we found that BMI-1 overexpression in lymphocytes clearly reversed all alterations induced by ovariectomy. Results from this study indicate that estrogen deficiency downregulates BMI-1 and subsequently increases ROS, T cell activation, and RANKL production in T cells, thus enhancing osteoclastogenesis and accelerating bone loss. This study clarifies a novel mechanism regulating estrogen deficiency-induced bone loss. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jinbo Li
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Wang
- Department of Human Anatomy, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Renlei Yang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaqi Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xing Li
- Department of Immuno-oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xichao Zhou
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
73
|
|
74
|
Golub LM, Payne JB, Reinhardt RA, Nieman G. Can Systemic Diseases Co-induce (Not Just Exacerbate) Periodontitis? A Hypothetical “Two-hit” Model. J Dent Res 2016; 85:102-5. [PMID: 16434727 DOI: 10.1177/154405910608500201] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- L M Golub
- Department of Oral Biology & Pathology, School of Dental Medicine, SUNY at Stony Brook, Stony Brook, NY, USA.
| | | | | | | |
Collapse
|
75
|
Ohta K, Naruse T, Ishida Y, Shigeishi H, Nakagawa T, Fukui A, Nishi H, Sasaki K, Ogawa I, Takechi M. TNF-α-induced IL-6 and MMP-9 expression in immortalized ameloblastoma cell line established by hTERT. Oral Dis 2016; 23:199-209. [PMID: 27868311 DOI: 10.1111/odi.12594] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Ameloblastoma (AM) shows locally invasive behaviour. However, biological investigations regarding regulation of gene expression associated with AM pathological features are difficult to perform, because AM cells can be passaged for a few generations due to senescence. We report a newly established immortalized AM cell line, AMB cells, by transfection with human telomerase reverse transcriptase (hTERT). Furthermore, we examined whether TNF-α modulates bone resorption-related genes, IL-6 and MMP-9 in cooperation with TGF-β or IFN-γ. MATERIALS AND METHODS Following transfection of an hTERT expression vector into AM cells using a non-viral method, the effects of cytokines on the expressions of IL-6 and MMP-9 mRNA were examined using real-time PCR. TNF-α-induced NF-κB activity was examined by western blotting and transcription factor assays. RESULTS AMB cells continued to grow for more than 100 population doublings. Stimulation with TNF-α increased IL-6 and MMP-9 mRNA expressions, as well as NF-κB activation. Furthermore, TGF-β and IFN-γ dramatically increased TNF-α-mediated expressions of MMP-9 and IL-6 mRNA, respectively, while those responses were suppressed by NF-κB inhibitor. CONCLUSION We established an immortalized AM cell line by hTERT transfection. TNF-α-mediated regulation of MMP-9 and IL-6 via NF-κB may play an important role in the pathological behaviour of AMs, such as bone resorption.
Collapse
Affiliation(s)
- K Ohta
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - T Naruse
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Y Ishida
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - H Shigeishi
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - T Nakagawa
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - A Fukui
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - H Nishi
- Department General Dentistry, Hiroshima University Hospital, Hiroshima, Japan
| | - K Sasaki
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - I Ogawa
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima, Japan
| | - M Takechi
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostmatology, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
76
|
Abstract
Bone mass in the skeleton is dependent on the coordinated activities of bone-forming osteoblasts and bone-resorbing osteoclasts in discrete bone multi-cellular units. Remodeling of bone in these units is important not only for maintaining bone mass, but also to repair microdamage, to prevent accumulation of too much old bone, and for mineral homeostasis. The activities of osteoblasts and osteoclasts are controlled by a variety of hormones and cytokines, as well as by mechanical loading. Most importantly, sex hormones are very crucial for keeping bone mass in balance, and the lack of either estrogen or testosterone leads to decreased bone mass and increased risk for osteoporosis. The prevalence of osteoporotic fractures is increasing dramatically in the Western part of the world and is a major health problem in many countries. In the present review, the cellular and molecular mechanisms controlling bone remodeling and the influence of sex hormones on these processes are summarized. In a separate paper in this issue, the pathogenesis of post-menopausal osteoporosis will be compared with that of inflammation-induced bone remodeling, including the evidence for and against the hypothesis that concomitant post-menopausal osteoporotic disease influences the progression of periodontal disease.
Collapse
Affiliation(s)
- U H Lerner
- Department of Oral Cell Biology, Umeå University, Umeå SE-901 87, Sweden.
| |
Collapse
|
77
|
Lv YG, Kang L, Wu G. Fluorosis increases the risk of postmenopausal osteoporosis by stimulating interferon γ. Biochem Biophys Res Commun 2016; 479:372-379. [PMID: 27644876 DOI: 10.1016/j.bbrc.2016.09.083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 09/16/2016] [Indexed: 10/21/2022]
Abstract
Estrogen deficiency in postmenopausal women frequently activates osteoclasts (OC), accelerates bone resorption, and leads to osteoporosis (OP). Previous studies have demonstrated that interferon γ (IFNγ) could increase bone resorption and may be involved in postmenopausal OP. Fluorosis also increased the risk of fractures and dental fluorosis, and fluoride may enhance osteoclast formation and induce osteoclastic bone destruction in postmenopausal women, but the underlying mechanisms are as yet unknown. Here, we show that serum fluoride and IFNγ levels are negatively correlated with bone mineral density (BMD) in postmenopausal women residing in a fluorotic area. Estrogen suppresses IFNγ, which is elevated by fluoride, playing a pivotal role in triggering bone loss in estrogen-deficient conditions. In vitro, IFNγ is inhibited by estrogen treatment and increased by fluoride in Raw264.7 cell, an osteoclast progenitor cell line. In ovariectomized (Ovx) mice, estrogen loss and IFNγ promote OC activation and subsequent bone loss in vivo. However, IFNγ deficiency prevents bone loss in Ovx mice even in fluoride conditions. Interestingly, fluoride fails to increase IFNγ expression in estrogen receptor α (ERα)-deficient conditions, but not in ERβ-deficient conditions. These findings demonstrate that fluorosis increases the bone loss in postmenopausal OP through an IFNγ-dependent mechanism. IFNγ signaling activates OC and aggravates estrogen deficiency inducing OP. Thus, stimulation of IFNγ production is a pivotal ''upstream'' mechanism by which fluoride promotes bone loss. Suppression of IFNγ levels may constitute a therapeutic approach for preventing bone loss.
Collapse
Affiliation(s)
- Yun-Gang Lv
- Department of Magnetic Resonance Imaging, Zhongnan Hospital, Wuhan University, No. 185 Donghu Road, Wuhan 430071, PR China.
| | - Li Kang
- Department of Interventional Radiology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia 010017, PR China.
| | - Guangyao Wu
- Department of Magnetic Resonance Imaging, Zhongnan Hospital, Wuhan University, No. 185 Donghu Road, Wuhan 430071, PR China.
| |
Collapse
|
78
|
Abstract
Osteoporosis develops when the rate of osteoclastic bone breakdown (resorption) exceeds that of osteoblastic bone formation, which leads to loss of BMD and deterioration of bone structure and strength. Osteoporosis increases the risk of fragility fractures, a cause of substantial morbidity and mortality, especially in elderly patients. This imbalance between bone formation and bone resorption is brought about by natural ageing processes, but is frequently exacerbated by a number of pathological conditions. Of importance to the aetiology of osteoporosis are findings over the past two decades attesting to a deep integration of the skeletal system with the immune system (the immuno-skeletal interface (ISI)). Although protective of the skeleton under physiological conditions, the ISI might contribute to bone destruction in a growing number of pathophysiological states. Although numerous research groups have investigated how the immune system affects basal and pathological osteoclastic bone resorption, recent findings suggest that the reach of the adaptive immune response extends to the regulation of osteoblastic bone formation. This Review examines the evolution of the field of osteoimmunology and how advances in our understanding of the ISI might lead to novel approaches to prevent and treat bone loss, and avert fractures.
Collapse
Affiliation(s)
- M Neale Weitzmann
- The Atlanta Department of Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, Georgia, 30033, USA
- Department of Medicine, Division of Endocrinology and Metabolism and Lipids, Emory University School of Medicine, 101 Woodruff Circle, 1305 WMB, Atlanta, Georgia 30322, USA
| | - Ighovwerha Ofotokun
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, 49 Jesse Hill Jr Drive, Atlanta, Georgia 30303, USA
- Grady Healthcare System, 80 Jesse Hill Jr Drive SE, Atlanta, Georgia, 30303, USA
| |
Collapse
|
79
|
FSH aggravates bone loss in ovariectomised rats with experimental periapical periodontitis. Mol Med Rep 2016; 14:2997-3006. [PMID: 27510616 PMCID: PMC5042797 DOI: 10.3892/mmr.2016.5613] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 07/22/2016] [Indexed: 11/12/2022] Open
Abstract
Periapical bone loss is one of the prominent pathological and clinical features of periapical periodontitis. Previous studies have demonstrated that follicle-stimulating hormone (FSH) could directly affect skeletal remodelling by stimulating the formation and the function of osteoclasts in vitro and in vivo. However, the effect of FSH on periapical bone loss remained to be fully elucidated. In the current study, a rat model was established in order to verify the effect of FSH in experimental periapical lesions. It was identified that FSH aggravated the bone loss of periapical lesions. In addition, RANKL-, TRAP-, TNF-α- and IL-1β-positive cells were increased significantly in FSH-treated groups, which indicated that the function of FSH in bone loss may be mediated through the increasing activity of osteoclasts and the increased secretion of inflammatory cytokines. The results of the current study suggested that FSH, independent of oestrogen, may aggravate periapical bone loss by FSH receptors, which may serve an important role in the immune and inflammatory response of the host to root canal and periradicular infection during menopause.
Collapse
|
80
|
Tseng HC, Kanayama K, Kaur K, Park SH, Park S, Kozlowska A, Sun S, McKenna CE, Nishimura I, Jewett A. Bisphosphonate-induced differential modulation of immune cell function in gingiva and bone marrow in vivo: role in osteoclast-mediated NK cell activation. Oncotarget 2016; 6:20002-25. [PMID: 26343372 PMCID: PMC4652983 DOI: 10.18632/oncotarget.4755] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/16/2015] [Indexed: 11/25/2022] Open
Abstract
The aim of this study is to establish osteoclasts as key immune effectors capable of activating the function of Natural Killer (NK) cells, and expanding their numbers, and to determine in vivo and in vitro effect of bisphosphonates (BPs) during NK cell interaction with osteoclasts and on systemic and local immune function. The profiles of 27 cytokines, chemokines and growth factors released from osteoclasts were found to be different from dendritic cells and M1 macrophages but resembling to untreated monocytes and M2 macrophages. Nitrogen-containing BPs Zoledronate (ZOL) and Alendronate (ALN), but not non-nitrogen-containing BPs Etidronate (ETI), triggered increased release of pro-inflammatory mediators from osteoclasts while all three BPs decreased pit formation by osteoclasts. ZOL and ALN mediated significant release of IL-6, TNF-` and IL-1β, whereas they inhibited IL-10 secretion by osteoclasts. Treatment of osteoclasts with ZOL inhibited NK cell mediated cytotoxicity whereas it induced significant secretion of cytokines and chemokines. NK cells lysed osteoclasts much more than their precursor cells monocytes, and this correlated with the decreased expression of MHC class I expression on osteoclasts. Intravenous injection of ZOL in mice induced pro-inflammatory microenvironment in bone marrow and demonstrated significant immune activation. By contrast, tooth extraction wound of gingival tissues exhibited profound immune suppressive microenvironment associated with dysregulated wound healing to the effect of ZOL which could potentially be responsible for the pathogenesis of Osteonecrosis of the Jaw (ONJ). Finally, based on the data obtained in this paper we demonstrate that osteoclasts can be used as targets for the expansion of NK cells with superior function for immunotherapy of cancer.
Collapse
Affiliation(s)
- Han-Ching Tseng
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Keiichi Kanayama
- Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Department of Periodontology, Asahi University School of Dentistry, Gifu, Japan
| | - Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - So-Hyun Park
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Sil Park
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Anna Kozlowska
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Department of Tumor Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Shuting Sun
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Charles E McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Ichiro Nishimura
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
81
|
Novack DV, Mbalaviele G. Osteoclasts-Key Players in Skeletal Health and Disease. Microbiol Spectr 2016; 4:10.1128/microbiolspec.MCHD-0011-2015. [PMID: 27337470 PMCID: PMC4920143 DOI: 10.1128/microbiolspec.mchd-0011-2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Indexed: 12/12/2022] Open
Abstract
The differentiation of osteoclasts (OCs) from early myeloid progenitors is a tightly regulated process that is modulated by a variety of mediators present in the bone microenvironment. Once generated, the function of mature OCs depends on cytoskeletal features controlled by an αvβ3-containing complex at the bone-apposed membrane and the secretion of protons and acid-protease cathepsin K. OCs also have important interactions with other cells in the bone microenvironment, including osteoblasts and immune cells. Dysregulation of OC differentiation and/or function can cause bone pathology. In fact, many components of OC differentiation and activation have been targeted therapeutically with great success. However, questions remain about the identity and plasticity of OC precursors and the interplay between essential networks that control OC fate. In this review, we summarize the key principles of OC biology and highlight recently uncovered mechanisms regulating OC development and function in homeostatic and disease states.
Collapse
Affiliation(s)
- Deborah Veis Novack
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Department of Medicine
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gabriel Mbalaviele
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Department of Medicine
| |
Collapse
|
82
|
D'Amelio P, Sassi F. Osteoimmunology: from mice to humans. BONEKEY REPORTS 2016; 5:802. [PMID: 27195109 DOI: 10.1038/bonekey.2016.29] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/02/2016] [Indexed: 12/15/2022]
Abstract
The immune system has been recognized as one of the most important regulators of bone turnover and its deregulation is implicated in several bone diseases such as postmenopausal osteoporosis and inflammatory bone loss; recently it has been suggested that the gut microbiota may influence bone turnover by modulation of the immune system. The study of the relationship between the immune system and bone metabolism is generally indicated under the term 'osteoimmunology'. The vast majority of these studies have been performed in animal models; however, several data have been confirmed in humans as well: this review summarizes recent data on the relationship between the immune system and bone with particular regard to the data confirmed in humans.
Collapse
Affiliation(s)
- Patrizia D'Amelio
- Department of Medical Science-Section of Gerontology-University of Torino , Torino, Italy
| | - Francesca Sassi
- Department of Medical Science-Section of Gerontology-University of Torino , Torino, Italy
| |
Collapse
|
83
|
Shahnazari M, Turner RT, Iwaniec UT, Wronski TJ, Li M, Ferruzzi MG, Nissenson RA, Halloran BP. Dietary dried plum increases bone mass, suppresses proinflammatory cytokines and promotes attainment of peak bone mass in male mice. J Nutr Biochem 2016; 34:73-82. [PMID: 27239754 DOI: 10.1016/j.jnutbio.2016.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/22/2016] [Accepted: 04/28/2016] [Indexed: 12/09/2022]
Abstract
Nutrition is an important determinant of bone health and attainment of peak bone mass. Diets containing dried plum (DP) have been shown to increase bone volume and strength. These effects may be linked to the immune system and DP-specific polyphenols. To better understand these relationships, we studied DP in skeletally mature (6-month-old) and growing (1- and 2-month-old) C57Bl/6 male mice. In adult mice, DP rapidly (<2 weeks) increased bone volume (+32%) and trabecular thickness (+24%). These changes were associated with decreased osteoclast surface (Oc.S/BS) and decreased serum CTX, a marker of bone resorption. The reduction in Oc.S/BS was associated with a reduction in the osteoclast precursor pool. Osteoblast surface (Ob.S/BS) and bone formation rate were also decreased suggesting that the gain in bone in adult mice is a consequence of diminished bone resorption and formation, but resorption is reduced more than formation. The effects of DP on bone were accompanied by a decline in interleukins, TNF and MCP-1, suggesting that DP is acting in part through the immune system to suppress inflammatory activity and reduce the size of the osteoclast precursor pool. Feeding DP was accompanied by an increase in plasma phenolics, some of which have been shown to stimulate bone accrual. In growing and young adult mice DP at levels as low as 5% of diet (w/w) increased bone volume. At higher levels (DP 25%), bone volume was increased by as much as 94%. These data demonstrate that DP feeding dramatically increases peak bone mass during growth.
Collapse
Affiliation(s)
- Mohammad Shahnazari
- Veterans Affairs Medical Center, and Department of Medicine, University of California, San Francisco, CA.
| | - Russell T Turner
- Skeletal Biology Laboratory, College of Public Health and Human Science, Oregon State University, Corvallis, OR
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, College of Public Health and Human Science, Oregon State University, Corvallis, OR
| | - Thomas J Wronski
- Department of Physiological Sciences, University of Florida, Gainesville, FL
| | - Min Li
- Departments of Food Science and Nutrition Science, Purdue University, West Lafayette, IN
| | - Mario G Ferruzzi
- Departments of Food Science and Nutrition Science, Purdue University, West Lafayette, IN
| | - Robert A Nissenson
- Veterans Affairs Medical Center, and Department of Medicine, University of California, San Francisco, CA
| | - Bernard P Halloran
- Veterans Affairs Medical Center, and Department of Medicine, University of California, San Francisco, CA
| |
Collapse
|
84
|
Li JY, Chassaing B, Tyagi AM, Vaccaro C, Luo T, Adams J, Darby TM, Weitzmann MN, Mulle JG, Gewirtz AT, Jones RM, Pacifici R. Sex steroid deficiency-associated bone loss is microbiota dependent and prevented by probiotics. J Clin Invest 2016; 126:2049-63. [PMID: 27111232 DOI: 10.1172/jci86062] [Citation(s) in RCA: 439] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/01/2016] [Indexed: 12/22/2022] Open
Abstract
A eubiotic microbiota influences many physiological processes in the metazoan host, including development and intestinal homeostasis. Here, we have shown that the intestinal microbiota modulates inflammatory responses caused by sex steroid deficiency, leading to trabecular bone loss. In murine models, sex steroid deficiency increased gut permeability, expanded Th17 cells, and upregulated the osteoclastogenic cytokines TNFα (TNF), RANKL, and IL-17 in the small intestine and the BM. In germ-free (GF) mice, sex steroid deficiency failed to increase osteoclastogenic cytokine production, stimulate bone resorption, and cause trabecular bone loss, demonstrating that the gut microbiota is central in sex steroid deficiency-induced trabecular bone loss. Furthermore, we demonstrated that twice-weekly treatment of sex steroid-deficient mice with the probiotics Lactobacillus rhamnosus GG (LGG) or the commercially available probiotic supplement VSL#3 reduces gut permeability, dampens intestinal and BM inflammation, and completely protects against bone loss. In contrast, supplementation with a nonprobiotic strain of E. coli or a mutant LGG was not protective. Together, these data highlight the role that the gut luminal microbiota and increased gut permeability play in triggering inflammatory pathways that are critical for inducing bone loss in sex steroid-deficient mice. Our data further suggest that probiotics that decrease gut permeability have potential as a therapeutic strategy for postmenopausal osteoporosis.
Collapse
|
85
|
Changes in bone mass during the perimenopausal transition in naturally menopausal cynomolgus monkeys. Menopause 2016; 23:87-99. [DOI: 10.1097/gme.0000000000000556] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
86
|
Liu H, Li W, Liu Y, Zhang X, Zhou Y. Co-administration of aspirin and allogeneic adipose-derived stromal cells attenuates bone loss in ovariectomized rats through the anti-inflammatory and chemotactic abilities of aspirin. Stem Cell Res Ther 2015; 6:200. [PMID: 26474767 PMCID: PMC4609080 DOI: 10.1186/s13287-015-0195-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/20/2015] [Accepted: 10/01/2015] [Indexed: 01/21/2023] Open
Abstract
Introduction Osteoporosis is a syndrome of excessive skeletal fragility characterized by the loss of mass and deterioration of microarchitecture in bone. Single use of aspirin or adipose-derived stromal cells (ASCs) has been recognized recently to be effective against osteoporosis. The goal of the study was to evaluate the osteogenic effects of the co-administration of aspirin and allogeneic rat adipose-derived stromal cells (rASCs) on ovariectomized (OVX)-induced bone loss in rats. The underlying mechanisms were investigated in vitro and in vivo. Methods Firstly, allogeneic rASCs were isolated and cultured, and the conditioned medium (CM) from the maintenance of rASCs was collected. Secondly, the OVX rats were administrated CM, rASCs, aspirin (ASP) or rASCs + ASP, respectively. Twelve weeks later, the anti-inflammatory and osteogenic effects were assessed by micro-CT, undecalcified histological sections, dynamic histomorphometric analyses and serologic assays for biochemical markers. Finally, a Transwell migration assay in vitro and cell-trafficking analyses in vivo were used to explore the effects of aspirin on rASC migration. Results Systemic administration of aspirin and rASCs attenuated OVX-induced bone loss better than single use of aspirin or ASCs (p < 0.05, respectively). Next, we analyzed the underlying mechanisms of the anti-inflammatory and chemotactic abilities of aspirin. Aspirin suppressed serum levels of the pro-inflammatory cytokines on tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and the anti-inflammatory ability was positively associated with bone morphometry. Also, aspirin exhibited excellent chemotactic effects in vitro and accelerated the homing of allogeneic rASCs into bone marrow during early in vivo stages. Conclusions Co-administered aspirin and allogeneic ASCs can partially reverse OVX-induced bone loss in rats. This effect appears to be mediated by the anti-inflammatory and chemotactic abilities of aspirin.
Collapse
Affiliation(s)
- Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Wei Li
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China. .,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
87
|
Abstract
Breast cancer is one of the most common malignancies of women. The majority of breast cancers express estrogen and/or progesterone receptors, permitting anticancer targeting strategies to reduce estrogen signaling in the cancer cells and thereby lowering the risk of breast cancer recurrence. The development of the selective estrogen receptor modulator (SERM) tamoxifen marked a significant milestone in breast cancer care that transcended older estrogen ablative strategies such as oophorectomy and ovarian irradiation. An unintended benefit of tamoxifen in postmenopausal women was bone density preservation. The third generation of aromatase inhibitors (AIs) have demonstrated superior efficacy to tamoxifen in improving disease-free survival in postmenopausal women. However, the AIs significantly increase bone resorption, reduce bone mineral density, and increase the risk of fracture above that of tamoxifen. As a consequence of this, clinical oncologists have assumed a larger role in the screening and treatment of the skeletal complications of breast cancer therapies. The key features of managing bone health in women with early stage breast cancer receiving adjuvant endocrine therapy are reviewed here.
Collapse
Affiliation(s)
- Gregory A Clines
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA,
| | | | | |
Collapse
|
88
|
Sun D, Zheng X, Chen Y, Jia C, Xu S, Lin C, Zhang P, Zhang Z, Cai D, Jin D, Zhang B, Bai X. Enhancement of osteogenesis post-splenectomy does not attenuate bone loss in ovariectomized rats. J Orthop Res 2015; 33:1356-63. [PMID: 25640957 DOI: 10.1002/jor.22825] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 12/30/2014] [Indexed: 02/04/2023]
Abstract
The roles of different immune cell populations and cytokines in bone metabolism have been extensively investigated. However, the influence of whole immune organ removal on osteopathology remains unknown. In the current study, we investigated the effects of splenectomy on bone metabolism and microarchitecture in rats with or without concurrent ovariectomy. Ovariectomized (OVX) rats were used as osteoporosis model. Sixty 12-week-old female rats were randomized into 4 groups (n = 15): sham, splenectomized (SP), ovariectomized, as well as ovariectomized and splenectomized (OVX + SP). Bone microarchitecture was assessed by micro CT analysis at 4 week and 12 week post-operation, respectively. Bone pathology and metabolism were evaluated via immunohistochemical staining. The serum levels of alkaline phosphatase (ALP), tumor necrosis factor-alpha (TNF-α), tartrate-resistant acid phosphatase 5b (Tracp5b), and C-terminal telopeptide (CTx) were analyzed at 4 and 12 weeks post-operation. Removal of the spleen led to alterations in the homeostasis of bone metabolism and increased bone formation in rats. In this study, our findings indicate that the spleen is involved in skeletal metabolism.
Collapse
Affiliation(s)
- Dawei Sun
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China.,Department of Orthopedics & Microsurgery, Guangdong No. 2 Provincial People's Hospital, Guangzhou, 510317, China
| | - Xiaochen Zheng
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China.,Department of Orthopedics, the Second Affiliated Hospital of Luohe Medical College, Luohe, 462300, China
| | - Yuhui Chen
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China
| | - Chunhong Jia
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Song Xu
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China.,Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| | - Chuangxin Lin
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China
| | - Pei Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhongmin Zhang
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China
| | - Daozhang Cai
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China
| | - Dadi Jin
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China
| | - Baiyu Zhang
- Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaochun Bai
- Department of Orthopedics, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, China.,Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
89
|
Buchwald ZS, Yang C, Nellore S, Shashkova EV, Davis JL, Cline A, Ko J, Novack DV, DiPaolo R, Aurora R. A Bone Anabolic Effect of RANKL in a Murine Model of Osteoporosis Mediated Through FoxP3+ CD8 T Cells. J Bone Miner Res 2015; 30:1508-22. [PMID: 25656537 PMCID: PMC4506715 DOI: 10.1002/jbmr.2472] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/15/2015] [Accepted: 01/30/2015] [Indexed: 12/30/2022]
Abstract
TNF-α and IL-17 secreted by proinflammatory T cells (T(EFF)) promote bone erosion by activating osteoclasts. We previously demonstrated that in addition to bone resorption, osteoclasts act as antigen-presenting cells to induce FoxP3 in CD8 T cells (Tc(REG)). The osteoclast-induced regulatory CD8 T cells limit bone resorption in ovariectomized mice (a murine model of postmenopausal osteoporosis). Here we show that although low-dose receptor activator of NF-κB ligand (RANKL) maximally induces Tc(REG) via Notch signaling pathway to limit bone resorption, high-dose RANKL promotes bone resorption. In vitro, both TNF-α and IL-17, cytokines that are abundant in ovariectomized animals, suppress Tc(REG) induction by osteoclasts by repressing Notch ligand expression in osteoclasts, but this effect can be counteracted by addition of RANKL. Ovariectomized mice treated with low-dose RANKL induced Tc(REG) that suppressed bone resorption, decreased T(EFF) levels, and increased bone formation. High-dose RANKL had the expected osteolytic effect. Low-dose RANKL administration in ovariectomized mice lacking CD8 T cells was also osteolytic, confirming that Tc(REG) mediate this bone anabolic effect. Our results show that although RANKL directly stimulates osteoclasts to resorb bone, it also controls the osteoclasts' ability to induce regulatory T cells, engaging an important negative feedback loop. In addition to the conceivable clinical relevance to treatment of osteoporosis, these observations have potential relevance to induction of tolerance and autoimmune diseases.
Collapse
Affiliation(s)
- Zachary S. Buchwald
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| | - Chang Yang
- Division of Bone and Mineral Disease, Department of Medicine, Washington University in St. Louis
| | - Suman Nellore
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| | - Elena V. Shashkova
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| | - Jennifer L. Davis
- Division of Bone and Mineral Disease, Department of Medicine, Washington University in St. Louis
| | - Anna Cline
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| | - Je Ko
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| | - Deborah V. Novack
- Division of Bone and Mineral Disease, Department of Medicine, Washington University in St. Louis
| | - Richard DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| | - Rajeev Aurora
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine
| |
Collapse
|
90
|
Lai N, Zhang Z, Wang B, Miao X, Guo Y, Yao C, Wang Z, Wang L, Ma R, Li X, Jiang G. Regulatory effect of traditional Chinese medicinal formula Zuo-Gui-Wan on the Th17/Treg paradigm in mice with bone loss induced by estrogen deficiency. JOURNAL OF ETHNOPHARMACOLOGY 2015; 166:228-239. [PMID: 25824592 DOI: 10.1016/j.jep.2015.03.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/15/2015] [Accepted: 03/09/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bone loss is a common pathological condition induced by estrogen deficiency. The Th17/Treg paradigm, which can be skewed by estrogen, plays an important role in regulating bone metabolism AIM OF THE STUDY The purpose of this study was to determine the role of the Th17/Treg shift in estrogen deficiency-induced bone loss in mouse models and to elucidate the immunopharmacologic mechanism of Zuo-Gui-Wan (ZGW) in preventing bone loss in this process by regulating Th17/Treg paradigm. MATERIALS AND METHODS Splenocytes of ovariectomized (Ovx) mice and naturally aged mice were isolated and Flow cytometry was used to detect the Th17/Treg subsets. In addition, serum estrodiol (E2) and serum C-terminal telopeptides of type Ι collagen (CTx) were detected by ELISA assay. Bone mineral density (BMD) of the left tibiae was measured by dual-energy X-ray absorptiometry. Moreover, Ovx mice were administrated with different doses of ZGW for 12 weeks, and BMD and Th17/Treg subsets were determined. Bone histomorphometry was observed by Hematoxylin and eosin (H&E) staining and serum protein levels of IL-6 were analyzed by ELISA assay. In addition, the mRNA and protein expression of RORγt and Foxp3 were detected by RT-PCR and Western blot respectively. RESULT The Th17/Treg paradigm shifted to Th17 in estrogen-deficient mice both in the Ovx mice and the naturally aged mice. BMD and E2 levels negatively correlated with the Th17/Treg ratio. After ZGW administration, the BMD was enhanced markedly in the Ovx mice as well as in the naturally aged mice. Both the mRNA and protein expressions of IL-6 and RORγt were decreased, whereas those of Foxp3 were increased significantly after ZGW administration. CONCLUSION Th17/Treg shift involved in the bone loss induced by estrogen deficiency. ZGW prevented bone loss efficiently and skewed Th17/Treg paradigm towards Treg without enhancing E2.
Collapse
Affiliation(s)
- Nannan Lai
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Laboratory for Immunopharmacology of State Administration of Traditional Chinese Medicine, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China
| | - Zhirong Zhang
- Department of reproductive center, Yantai Yuhuangding Hospital, Shandong University, 44 Wenhua xi Road, Jinan 250012, China
| | - Bin Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 42 Wenhua xi Road, Jinan 250011, China
| | - Xiuming Miao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 42 Wenhua xi Road, Jinan 250011, China
| | - Yuqi Guo
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Laboratory for Immunopharmacology of State Administration of Traditional Chinese Medicine, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China
| | - Chengfang Yao
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Laboratory for Immunopharmacology of State Administration of Traditional Chinese Medicine, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China
| | - Zhaoxia Wang
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Laboratory for Immunopharmacology of State Administration of Traditional Chinese Medicine, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China
| | - Li Wang
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Laboratory for Immunopharmacology of State Administration of Traditional Chinese Medicine, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China
| | - Ruiping Ma
- Qianfo Hospital of Shandong Province, 16766 Jingshi Road, Jinan 250014, China
| | - Xia Li
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China.
| | - Guosheng Jiang
- Key Laboratory for Tumor Immunology and Traditional Chinese Medicine Immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory for Rare & Uncommon Disease, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China; Key Laboratory of Biotechnology Drugs of the Ministry of Public Health, Institute of Basic Medicine, Shandong Academy of Medical Sciences, 18877 Jingshi Road, Jinan 250062, China.
| |
Collapse
|
91
|
The Impact of Immune System in Regulating Bone Metastasis Formation by Osteotropic Tumors. J Immunol Res 2015; 2015:143526. [PMID: 26064994 PMCID: PMC4433688 DOI: 10.1155/2015/143526] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/02/2014] [Indexed: 01/31/2023] Open
Abstract
Bone metastases are frequent and debilitating consequence for many tumors, such as breast, lung, prostate, and kidney cancer. Many studies report the importance of the immune system in the pathogenesis of bone metastasis. Indeed, bone and immune system are strictly linked to each other because bone regulates the hematopoietic stem cells from which all cells of the immune system derive, and many immunoregulatory cytokines influence the fate of bone cells. Furthermore, both cytokines and factors produced by immune and bone cells promote the growth of tumor cells in bone, contributing to supporting the vicious cycle of bone metastasis. This review summarizes the current knowledge on the interactions among bone, immune, and tumor cells aiming to provide an overview of the osteoimmunology field in bone metastasis from solid tumors.
Collapse
|
92
|
Mori G, D'Amelio P, Faccio R, Brunetti G. Bone-immune cell crosstalk: bone diseases. J Immunol Res 2015; 2015:108451. [PMID: 26000310 PMCID: PMC4427089 DOI: 10.1155/2015/108451] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 01/14/2023] Open
Abstract
Bone diseases are associated with great morbidity; thus, the understanding of the mechanisms leading to their development represents a great challenge to improve bone health. Recent reports suggest that a large number of molecules produced by immune cells affect bone cell activity. However, the mechanisms are incompletely understood. This review aims to shed new lights into the mechanisms of bone diseases involving immune cells. In particular, we focused our attention on the major pathogenic mechanism underlying periodontal disease, psoriatic arthritis, postmenopausal osteoporosis, glucocorticoid-induced osteoporosis, metastatic solid tumors, and multiple myeloma.
Collapse
Affiliation(s)
- Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Patrizia D'Amelio
- Department of Medical Science, Section of Gerontology and Bone Metabolism Diseases, University of Torino, 10126 Torino, Italy
| | - Roberta Faccio
- Department of Orthopedics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari, 70124 Bari, Italy
| |
Collapse
|
93
|
Wang L, Liu S, Zhao Y, Liu D, Liu Y, Chen C, Karray S, Shi S, Jin Y. Osteoblast-induced osteoclast apoptosis by fas ligand/FAS pathway is required for maintenance of bone mass. Cell Death Differ 2015; 22:1654-64. [PMID: 25744024 DOI: 10.1038/cdd.2015.14] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/05/2015] [Accepted: 01/19/2015] [Indexed: 12/29/2022] Open
Abstract
The interplay between osteoblasts and osteoclasts has a crucial role in maintaining bone homeostasis. In this study, we reveal that osteoblasts are capable of inducing osteoclast apoptosis by FAS ligand (FASL)/FAS signaling. Conditional knockout of FASL in osteoblasts results in elevated osteoclast numbers and activity, along with reduced bone mass, suggesting that osteoblast-produced FASL is required to maintain physiological bone mass. More interestingly, we show that osteoblasts from ovariectomized (OVX) osteoporotic mice exhibit decreased FASL expression that results from the IFN-γ- and TNF-α-activated NF-κB pathway, leading to reduced osteoclast apoptosis and increased bone resorption. Systemic administration of either IFN-γ or TNF-α ameliorates the osteoporotic phenotype in OVX mice and rescues FASL expression in osteoblasts. In addition, ovariectomy induces more significant bone loss in FASL conditional knockout mice than in control group with increased osteoclast activity in which the levels of RANKL and OPG remain unchanged. Taken together, this study suggests that osteoblast-induced osteoclast apoptosis via FASL/FAS signaling is a previously unrecognized mechanism that has an important role in the maintenance of bone mass in both physiological conditions and OVX osteoporosis.
Collapse
Affiliation(s)
- L Wang
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - S Liu
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Zhao
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - D Liu
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Liu
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S Karray
- INSERM U753, Institut Gustave Roussy, Villejuif Cedex, France
| | - S Shi
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Jin
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
94
|
Nagy V, Penninger JM. The RANKL-RANK Story. Gerontology 2015; 61:534-42. [PMID: 25720990 DOI: 10.1159/000371845] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022] Open
Abstract
Receptor activator of nuclear factor x03BA;B (RANK) and its ligand (RANKL) have originally been described for their key roles in bone metabolism and the immune system. Subsequently, it has been shown that the RANKL-RANK system is critical in the formation of mammary epithelia in lactating females and the thermoregulation of the central nervous system. RANKL and RANK are under the tight control of the female sex hormones estradiol and progesterone. A reduction of the circulating female sex hormones leading to an increase in RANKL-RANK signaling is the leading cause of osteoporosis in postmenopausal women. Denosumab, a human monoclonal anti-RANKL antibody, has been approved for the treatment of postmenopausal osteoporosis, where it is showing great promise. In addition, RANKL-RANK signaling also plays a critical role in other bone pathologies, bone metastasis or hormone-driven breast cancer. This review will highlight some of the functions of RANKL-RANK in bone turnover, the immune system and brain with a focus on the regulatory role of the female sex hormones.
Collapse
Affiliation(s)
- Vanja Nagy
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
| | | |
Collapse
|
95
|
Liu Y, Wang L, Liu S, Liu D, Chen C, Xu X, Chen X, Shi S. Transplantation of SHED prevents bone loss in the early phase of ovariectomy-induced osteoporosis. J Dent Res 2014; 93:1124-32. [PMID: 25252877 DOI: 10.1177/0022034514552675] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Stem cells from human exfoliated deciduous teeth (SHED) are a unique postnatal stem cell population, possessing multipotent differentiation capacity and immunomodulatory properties. However, the mechanism by which SHED treat immune diseases is not fully understood. Here we show that systemic transplantation of SHED via the tail vein ameliorates ovariectomy (OVX)-induced osteopenia by reducing T-helper 1 (Th1) and T-helper 17 (Th17) cell numbers in the recipient OVX mice. Mechanistically, SHED transplantation induces activated T-cell apoptosis in OVX mice via Fas ligand (FasL)-mediated Fas pathway activation, leading to up-regulation of regulatory T-cells (Tregs) and down-regulation of Th1 and Th17 cells. This SHED-mediated immunomodulation rescues OVX-induced impairment of bone marrow mesenchymal stem cells (BMMSCs) and activation of osteoclastogenesis, resulting in increased bone mass. In summary, SHED-mediated T-cell apoptosis via a FasL/Fas pathway results in immune tolerance and ameliorates the osteopenia phenotype in OVX mice.
Collapse
Affiliation(s)
- Y Liu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA Department of Pediatric Dentistry, School of Stomatology, China Medical University, Shenyang, China
| | - L Wang
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - S Liu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - D Liu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| | - C Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| | - X Xu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - X Chen
- Department of Pediatric Dentistry, School of Stomatology, China Medical University, Shenyang, China
| | - S Shi
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
96
|
Barbour KE, Lui LY, Ensrud KE, Hillier TA, LeBlanc ES, Ing SW, Hochberg MC, Cauley JA. Inflammatory markers and risk of hip fracture in older white women: the study of osteoporotic fractures. J Bone Miner Res 2014; 29:2057-64. [PMID: 24723386 PMCID: PMC4336950 DOI: 10.1002/jbmr.2245] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/04/2014] [Accepted: 03/17/2014] [Indexed: 11/12/2022]
Abstract
Hip fractures are the most devastating consequence of osteoporosis and impact 1 in 6 white women leading to a two- to threefold increased mortality risk in the first year. Despite evidence of inflammatory markers in the pathogenesis of osteoporosis, few studies have examined their effect on hip fracture. To determine if high levels of inflammation increase hip fracture risk and to explore mediation pathways, a case-cohort design nested in a cohort of 4709 white women from the Study of Osteoporotic Fractures was used. A random sample of 1171 women was selected as the subcohort (mean age 80.1 ± 4.2 years) plus the first 300 women with incident hip fracture. Inflammatory markers interleukin-6 (IL-6) and soluble receptors (SR) for IL-6 (IL-6 SR) and tumor necrosis factor (TNF SR1 and TNF SR2) were measured, and participants were followed for a median (interquartile range) of 6.3 (3.7, 6.9) years. In multivariable models, the hazard ratio (HR) of hip fracture for women in the highest inflammatory marker level (quartile 4) was 1.64 (95% confidence interval [CI], 1.09-2.48, p trend = 0.03) for IL-6 and 2.05 (95% CI, 1.35-3.12, p trend <0.01) for TNF SR1 when compared with women in the lowest level (quartile 1). Among women with 2 and 3-4 inflammatory markers in the highest quartile, the HR of hip fracture was 1.51 (95% CI, 1.07-2.14) and 1.42 (95% CI, 0.87-2.31) compared with women with zero to one marker(s) in the highest quartile (p trend = 0.03). After individually adjusting for seven potential mediators, cystatin-C (a biomarker of renal function) and bone mineral density (BMD) attenuated HRs among women with the highest inflammatory burden by 64% and 50%, respectively, suggesting a potential mediating role. Older white women with high inflammatory burden are at increased risk of hip fracture in part due to poor renal function and low BMD.
Collapse
|
97
|
Tyagi AM, Mansoori MN, Srivastava K, Khan MP, Kureel J, Dixit M, Shukla P, Trivedi R, Chattopadhyay N, Singh D. Enhanced immunoprotective effects by anti-IL-17 antibody translates to improved skeletal parameters under estrogen deficiency compared with anti-RANKL and anti-TNF-α antibodies. J Bone Miner Res 2014; 29:1981-92. [PMID: 24677326 DOI: 10.1002/jbmr.2228] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/07/2014] [Accepted: 03/13/2014] [Indexed: 12/19/2022]
Abstract
Activated T cell has a key role in the interaction between bone and immune system. T cells produce proinflammatory cytokines, including receptor activator of NF-κB ligand (RANKL), tumor necrosis factor α (TNF-α), and interleukin 17 (IL-17), all of which augment osteoclastogenesis. RANKL and TNF-α are targeted by inhibitors such as denosumab, a human monoclonal RANKL antibody, and infliximab, which neutralizes TNF-α. IL-17 is also an important mediator of bone loss, and an antibody against IL-17 is undergoing phase II clinical trial for rheumatoid arthritis. Although there are a few studies showing suppression of Th17 cell differentiation and induction of regulatory T cells (Tregs) by infliximab, the effect of denosumab remains poorly understood. In this study, we investigated the effects of anti-TNF-α, anti-RANKL, or anti-IL-17 antibody administration to estrogen-deficient mice on CD4(+) T-cell proliferation, CD28 loss, Th17/Treg balance and B lymphopoesis, and finally, the translation of these immunomodulatory effects on skeletal parameters. Adult Balb/c mice were treated with anti-RANKL/-TNF-α/-IL-17 subcutaneously, twice a week, postovariectomy (Ovx) for 4 weeks. Animals were then autopsied; bone marrow cells were collected for FACS and RNA analysis and serum collected for ELISA. Bones were dissected for static and dynamic histomorphometry studies. We observed that although anti-RANKL and anti-TNF-α therapies had no effect on Ovx-induced CD4(+) T-cell proliferation and B lymphopoesis, anti-IL-17 effectively suppressed both events with concomitant reversal of CD28 loss. Anti-IL-17 antibody reduced proinflammatory cytokine production and induced Tregs. All three antibodies restored trabecular microarchitecture with comparable efficacy; however, cortical bone parameters, bone biomechanical properties, and histomorphometry were best preserved by anti-IL-17 antibody, likely attributable to its inhibitory effect on osteoblast apoptosis and increased number of bone lining cells and Wnt10b expression. Based on the superior immunoprotective effects of anti-IL-17, which appears to translate to a better skeletal preservation, we propose beginning clinical trials using a humanized antibody against IL-17 for treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Abdul M Tyagi
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Apalset EM, Gjesdal CG, Ueland PM, Øyen J, Meyer K, Midttun Ø, Eide GE, Tell GS. Interferon gamma (IFN-γ)-mediated inflammation and the kynurenine pathway in relation to risk of hip fractures: the Hordaland Health Study. Osteoporos Int 2014; 25:2067-75. [PMID: 24817202 PMCID: PMC4099528 DOI: 10.1007/s00198-014-2720-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/14/2014] [Indexed: 01/19/2023]
Abstract
UNLABELLED The cytokine interferon gamma (IFN-γ) stimulates neopterin release and tryptophan degradation into kynurenines through the kynurenine pathway. High levels of neopterin were associated with increased hip fracture risk, as were some of the kynurenines, suggesting a role of IFN-γ-mediated inflammation in the processes leading to hip fracture. INTRODUCTION Low-grade systemic inflammation has been associated with bone loss and risk of fractures. Interferon gamma (IFN-γ) initiates macrophage release of neopterin and also stimulates degradation of tryptophan along the kynurenine pathway as part of cell-mediated immune activation. Plasma neopterin and the kynurenine/tryptophan ratio (KTR) are thus markers of IFN-γ-mediated inflammation. Risk of hip fracture was investigated in relation to markers of inflammation and metabolites in the kynurenine pathway (kynurenines). METHODS Participants (71 to 74 years, N = 3,311) in the community-based Hordaland Health Study (HUSK) were followed for hip fractures from enrolment (1998-2000) until 31 December 2009. Plasma C-reactive protein (CRP), neopterin, KTR, and six kynurenines were investigated as predictors of hip fracture, using Cox proportional hazards regression analyses. RESULTS A hazard ratio (HR) of 1.9 (95% confidence interval (CI) 1.3-2.7) for hip fracture was found in the highest compared to the lowest quartile of neopterin (p trend across quartiles <0.001). CRP and KTR were not related to hip fracture risk. Among the kynurenines, a higher risk of fracture was found in the highest compared to the lowest quartiles of anthranilic acid and 3-hydroxykynurenine. For subjects in the highest quartiles of neopterin, CRP, and KTR compared to those in no top quartiles, HR was 2.5 (95% CI 1.6-4.0). CONCLUSIONS This may indicate a role for low-grade immune activation in the pathogenic processes leading to hip fracture.
Collapse
Affiliation(s)
- E. M. Apalset
- Department of Global Public Health and Primary Care, University of Bergen, Kalfarveien 31, 5018 Bergen, Norway
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - C. G. Gjesdal
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - P. M. Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
| | - J. Øyen
- Department of Global Public Health and Primary Care, University of Bergen, Kalfarveien 31, 5018 Bergen, Norway
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - K. Meyer
- Bevital A/S, Laboratoriebygget, Bergen, Norway
| | - Ø. Midttun
- Bevital A/S, Laboratoriebygget, Bergen, Norway
| | - G. E. Eide
- Department of Global Public Health and Primary Care, University of Bergen, Kalfarveien 31, 5018 Bergen, Norway
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
| | - G. S. Tell
- Department of Global Public Health and Primary Care, University of Bergen, Kalfarveien 31, 5018 Bergen, Norway
| |
Collapse
|
99
|
Adeel S, Singh K, Vydareny KH, Kumari M, Shah E, Weitzmann MN, Tangpricha V. Bone loss in surgically ovariectomized premenopausal women is associated with T lymphocyte activation and thymic hypertrophy. J Investig Med 2014; 61:1178-83. [PMID: 24141238 DOI: 10.2310/jim.0000000000000016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Postmenopausal osteoporosis is associated with estrogen deficiency and rapid bone loss. The mechanism by which estrogen deficiency results in bone loss has not been fully explained. Studies in mice rendered acutely estrogen deficient by ovariectomy have suggested that estrogen deficiency results in an activated T-lymphocyte phenotype and increased production of pro-osteoclastic cytokines. The aim of this study was to translate these findings from mouse models that suggest that the T lymphocyte plays an important role in the etiology of postmenopausal osteoporosis. We recruited premenopausal women who underwent ovariectomy for benign gynecologic conditions or for prophylaxis against ovarian cancer and a group of matched control women without ovariectomy (OVX). Subjects provided blood samples to characterize T-lymphocyte phenotype by flow cytometry and for T-lymphocyte culture and collection of conditioned media. Bone mineral density at the lumbar spine and left femoral neck was performed annually for 2 years, and volumetric measurements by computed tomography (CT) of the thymus were obtained during the first 6 months. We enrolled 6 patients who underwent OVX and 13 control women. The OVX subjects had a significant loss of bone mineral density at the lumbar spine and left femoral neck. The volumetric thymus measurements suggested an increase in thymus size in the OVX subjects but did not reach statistical significance owing to the small sample size. The T-lymphocyte phenotype in the OVX subjects demonstrated increased T-lymphocyte activation by flow cytometry compared to the control subjects. Our findings support the hypothesis that estrogen deficiency leads to an activated T-lymphocyte phenotype, which may contribute to the bone loss seen in estrogen deficiency. Larger clinical studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Saira Adeel
- From the *Division of Endocrinology, Metabolism and Lipids, Department of Medicine, †Department of Surgery, and ‡Department of Radiology, Emory University School of Medicine, Atlanta; and §Atlanta VA Medical Center, Decatur, GA
| | | | | | | | | | | | | |
Collapse
|
100
|
Wang L, Zhao Y, Liu Y, Akiyama K, Chen C, Qu C, Jin Y, Shi S. IFN-γ and TNF-α synergistically induce mesenchymal stem cell impairment and tumorigenesis via NFκB signaling. Stem Cells 2014; 31:1383-95. [PMID: 23553791 DOI: 10.1002/stem.1388] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/18/2013] [Accepted: 03/04/2013] [Indexed: 12/25/2022]
Abstract
An inflammatory microenvironment may cause organ degenerative diseases and malignant tumors. However, the precise mechanisms of inflammation-induced diseases are not fully understood. Here, we show that the proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor α (TNF-α) synergistically impair self-renewal and differentiation of mesenchymal stem cells (MSCs) via nuclear factor κB (NFκB)-mediated activation of mothers against decapentaplegic homolog 7 (SMAD7) in ovariectomized (OVX) mice. More interestingly, a long-term elevated levels of IFN-γ and TNF-α result in significantly increased susceptibility to malignant transformation in MSCs through NFκB-mediated upregulation of the oncogenes c-Fos and c-Myc. Depletion of either IFN-γ or TNF-α in OVX mice abolishes MSC impairment and the tendency toward malignant transformation with no NFκB-mediated oncogene activation. Systemic administration of aspirin, which significantly reduces the levels of IFN-γ and TNF-α, results in blockage of MSC deficiency and tumorigenesis by inhibition of NFκB/SMAD7 and NFκB/c-FOS and c-MYC pathways in OVX mice. In summary, this study reveals that inflammation factors, such as IFN-γ and TNF-α, synergistically induce MSC deficiency via NFκB/SMAD7 signaling and tumorigenesis via NFκB-mediated oncogene activation.
Collapse
Affiliation(s)
- Lei Wang
- University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|