1
|
Mohsin S, Brock F, Kaimala S, Greenwood C, Sulaiman M, Rogers K, Adeghate E. A pilot study: effect of irisin on trabecular bone in a streptozotocin-induced animal model of type 1 diabetic osteopathy utilizing a micro-CT. PeerJ 2023; 11:e16278. [PMID: 37868046 PMCID: PMC10588705 DOI: 10.7717/peerj.16278] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
Background Osteoporosis is a significant co-morbidity of type 1 diabetes mellitus (DM1) leading to increased fracture risk. Exercise-induced hormone 'irisin' in low dosage has been shown to have a beneficial effect on bone metabolism by increasing osteoblast differentiation and reducing osteoclast maturation, and inhibiting apoptosis and inflammation. We investigated the role of irisin in treating diabetic osteopathy by observing its effect on trabecular bone. Methods DM1 was induced by intraperitoneal injection of streptozotocin 60 mg/kg body weight. Irisin in low dosage (5 µg twice a week for 6 weeks I/P) was injected into half of the control and 4-week diabetic male Wistar rats. Animals were sacrificed six months after induction of diabetes. The trabecular bone in the femoral head and neck was analyzed using a micro-CT technique. Bone turnover markers were measured using ELISA, Western blot, and RT-PCR techniques. Results It was found that DM1 deteriorates the trabecular bone microstructure by increasing trabecular separation (Tb-Sp) and decreasing trabecular thickness (Tb-Th), bone volume fraction (BV/TV), and bone mineral density (BMD). Irisin treatment positively affects bone quality by increasing trabecular number p < 0.05 and improves the BMD, Tb-Sp, and BV/TV by 21-28%. The deterioration in bone microarchitecture is mainly attributed to decreased bone formation observed as low osteocalcin and high sclerostin levels in diabetic bone samples p < 0.001. The irisin treatment significantly suppressed the serum and bone sclerostin levels p < 0.001, increased the serum CTX1 levels p < 0.05, and also showed non-significant improvement in osteocalcin levels. Conclusions This is the first pilot study to our knowledge that shows that a low dose of irisin marginally improves the trabecular bone in DM1 and is an effective peptide in reducing sclerostin levels.
Collapse
Affiliation(s)
- Sahar Mohsin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abudhabi, United Arab Emirates
| | - Fiona Brock
- Cranfield Forensic Institute, Cranfield University, Shrivenham, United Kingdom
| | - Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abudhabi, United Arab Emirates
| | - Charlene Greenwood
- School of Chemical and Physical Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Mohsin Sulaiman
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abudhabi, United Arab Emirates
| | - Keith Rogers
- Cranfield Forensic Institute, Cranfield University, Shrivenham, United Kingdom
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abudhabi, United Arab Emirates
| |
Collapse
|
2
|
Yang S, Sun Y, Kapilevich L, Zhang X, Huang Y. Protective effects of curcumin against osteoporosis and its molecular mechanisms: a recent review in preclinical trials. Front Pharmacol 2023; 14:1249418. [PMID: 37790808 PMCID: PMC10544586 DOI: 10.3389/fphar.2023.1249418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Osteoporosis (OP) is one of the most common metabolic skeletal disorders and is commonly seen in the elderly population and postmenopausal women. It is mainly associated with progressive loss of bone mineral density, persistent deterioration of bone microarchitecture, and increased fracture risk. To date, drug therapy is the primary method used to prevent and treat osteoporosis. However, long-term drug therapy inevitably leads to drug resistance and specific side effects. Therefore, researchers are constantly searching for new monomer compounds from natural plants. As a candidate for the treatment of osteoporosis, curcumin (CUR) is a natural phenolic compound with various pharmacological and biological activities, including antioxidant, anti-apoptotic, and anti-inflammatory. This compound has gained research attention for maintaining bone health in various osteoporosis models. We reviewed preclinical and clinical studies of curcumin in preventing and alleviating osteoporosis. These results suggest that if subjected to rigorous pharmacological and clinical trials, naturally-derived curcumin could be used as a complementary and alternative medicine for the treatment of osteoporosis by targeting osteoporosis-related mechanistic pathways. This review summarizes the mechanisms of action and potential therapeutic applications of curcumin in the prevention and mitigation of osteoporosis and provides reference for further research and development of curcumin.
Collapse
Affiliation(s)
- Shenglei Yang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yuying Sun
- School of Stomatology, Binzhou Medical College, Yantai, China
| | - Leonid Kapilevich
- Faculty of Physical Education, Nаtionаl Reseаrch Tomsk Stаte University, Tomsk, Russiа
| | - Xin’an Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yue Huang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
3
|
Effect of chronic lithium on mechanical sensitivity and trabecular bone loss induced by type-1 diabetes mellitus in mice. Biometals 2022; 35:1033-1042. [PMID: 35849260 DOI: 10.1007/s10534-022-00421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/04/2022] [Indexed: 11/02/2022]
Abstract
Type-1 diabetes mellitus (T1DM) is a chronic condition characterized by long-term hyperglycemia that results in several complications such as painful peripheral neuropathy, bone deterioration, and increased risk of bone fractures. Lithium, a first-line therapy for bipolar disorder, has become an attractive agent for attenuating peripheral neuropathy and menopause-induced bone loss. Therefore, our aim was to determine the effect of chronic lithium treatment on mechanical hypersensitivity and trabecular bone loss induced by T1DM in mice. T1DM was induced in male C57BL/6J mice by intraperitoneal injection of streptozotocin (STZ, 50 mg/kg/day, for 5 consecutive days). 12 weeks after T1DM-induction, mice received a daily intraperitoneal injection of vehicle, 30 or 60 mg/kg lithium (as LiCl) for 6 weeks. Throughout the treatment period, blood glucose levels and mechanical sensitivity were evaluated every 2 weeks. After lithium treatment, the femur and L5 vertebra were harvested for microcomputed tomography (microCT) analysis. T1DM mice showed significant hyperglycemia, mechanical hypersensitivity, and significant trabecular bone loss as compared with the control group. Chronic lithium treatment did not revert the hindpaw mechanical hypersensitivity nor hyperglycemia associated to T1DM induced by STZ. In contrast, microCT analysis revealed that lithium reverted, in a dose-dependent manner, the loss of trabecular bone associated to T1DM induced by STZ at both the distal femur and L5 vertebra. Lithium treatment by itself did not affect any trabecular bone parameter in non-diabetic mice.
Collapse
|
4
|
Jeddi S, Yousefzadeh N, Kashfi K, Ghasemi A. Role of nitric oxide in type 1 diabetes-induced osteoporosis. Biochem Pharmacol 2021; 197:114888. [PMID: 34968494 DOI: 10.1016/j.bcp.2021.114888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D)-induced osteoporosis is characterized by decreased bone mineral density, bone quality, rate of bone healing, bone formation, and increased bone resorption. Patients with T1D have a 2-7-fold higher risk of osteoporotic fracture. The mechanisms leading to increased risk of osteoporotic fracture in T1D include insulin deficiency, hyperglycemia, insulin resistance, lower insulin-like growth factor-1, hyperglycemia-induced oxidative stress, and inflammation. In addition, a higher probability of falling, kidney dysfunction, weakened vision, and neuropathy indirectly increase the risk of osteoporotic fracture in T1D patients. Decreased nitric oxide (NO) bioavailability contributes to the pathophysiology of T1D-induced osteoporotic fracture. This review discusses the role of NO in osteoblast-mediated bone formation and osteoclast-mediated bone resorption in T1D. In addition, the mechanisms involved in reduced NO bioavailability and activity in type 1 diabetic bones as well as NO-based therapy for T1D-induced osteoporosis are summarized. Available data indicates that lower NO bioavailability in diabetic bones is due to disruption of phosphatidylinositol 3‑kinase/protein kinase B/endothelial NO synthases and NO/cyclic guanosine monophosphate/protein kinase G signaling pathways. Thus, NO bioavailability may be boosted directly or indirectly by NO donors. As NO donors with NO-like effects in the bone, inorganic nitrate and nitrite can potentially be used as novel therapeutic agents for T1D-induced osteoporosis. Inorganic nitrites and nitrates can decrease the risk for osteoporotic fracture probably directly by decreasing osteoclast activity, decreasing fat accumulation in the marrow cavity, increasing osteoblast activity, and increasing bone perfusion or indirectly, by improving hyperglycemia, insulin resistance, and reducing body weight.
Collapse
Affiliation(s)
- Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA.
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Brunetti G, D'Amato G, De Santis S, Grano M, Faienza MF. Mechanisms of altered bone remodeling in children with type 1 diabetes. World J Diabetes 2021; 12:997-1009. [PMID: 34326950 PMCID: PMC8311475 DOI: 10.4239/wjd.v12.i7.997] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/17/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Bone loss associated with type 1 diabetes mellitus (T1DM) begins at the onset of the disease, already in childhood, determining a lower bone mass peak and hence a greater risk of osteoporosis and fractures later in life. The mechanisms underlying diabetic bone fragility are not yet completely understood. Hyperglycemia and insulin deficiency can affect the bone cells functions, as well as the bone marrow fat, thus impairing the bone strength, geometry, and microarchitecture. Several factors, like insulin and growth hormone/insulin-like growth factor 1, can control bone marrow mesenchymal stem cell commitment, and the receptor activator of nuclear factor-κB ligand/osteoprotegerin and Wnt-b catenin pathways can impair bone turnover. Some myokines may have a key role in regulating metabolic control and improving bone mass in T1DM subjects. The aim of this review is to provide an overview of the current knowledge of the mechanisms underlying altered bone remodeling in children affected by T1DM.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University "A. Moro" of Bari, Bari 70125, Italy
| | - Gabriele D'Amato
- Department of Women’s and Children’s Health, ASL Bari, Neonatal Intensive Care Unit, Di Venere Hospital, Bari 70124, Italy
| | - Stefania De Santis
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, Bari 70126, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, Univ Bari, Bari 70124, Italy
| | - Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University "A.Moro", Bari 70124, Italy
| |
Collapse
|
6
|
Bollavaram K, Leeman TH, Lee MW, Kulkarni A, Upshaw SG, Yang J, Song H, Platt MO. Multiple sites on SARS-CoV-2 spike protein are susceptible to proteolysis by cathepsins B, K, L, S, and V. Protein Sci 2021; 30:1131-1143. [PMID: 33786919 PMCID: PMC8138523 DOI: 10.1002/pro.4073] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/15/2021] [Accepted: 03/24/2021] [Indexed: 12/23/2022]
Abstract
SARS‐CoV‐2 is the coronavirus responsible for the COVID‐19 pandemic. Proteases are central to the infection process of SARS‐CoV‐2. Cleavage of the spike protein on the virus's capsid causes the conformational change that leads to membrane fusion and viral entry into the target cell. Since inhibition of one protease, even the dominant protease like TMPRSS2, may not be sufficient to block SARS‐CoV‐2 entry into cells, other proteases that may play an activating role and hydrolyze the spike protein must be identified. We identified amino acid sequences in all regions of spike protein, including the S1/S2 region critical for activation and viral entry, that are susceptible to cleavage by furin and cathepsins B, K, L, S, and V using PACMANS, a computational platform that identifies and ranks preferred sites of proteolytic cleavage on substrates, and verified with molecular docking analysis and immunoblotting to determine if binding of these proteases can occur on the spike protein that were identified as possible cleavage sites. Together, this study highlights cathepsins B, K, L, S, and V for consideration in SARS‐CoV‐2 infection and presents methodologies by which other proteases can be screened to determine a role in viral entry. This highlights additional proteases to be considered in COVID‐19 studies, particularly regarding exacerbated damage in inflammatory preconditions where these proteases are generally upregulated. PDB Code(s): 6VYB, 4Z2A, 5F02, 4P6E, 5TUN, 2IPP and 3H6S;
Collapse
Affiliation(s)
- Keval Bollavaram
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Tiffanie H Leeman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Maggie W Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Akhil Kulkarni
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Sophia G Upshaw
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Jiabei Yang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA.,Biomedical Engineering, Peking University, Beijing, China
| | - Hannah Song
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Merlotti D, Cosso R, Eller-Vainicher C, Vescini F, Chiodini I, Gennari L, Falchetti A. Energy Metabolism and Ketogenic Diets: What about the Skeletal Health? A Narrative Review and a Prospective Vision for Planning Clinical Trials on this Issue. Int J Mol Sci 2021; 22:ijms22010435. [PMID: 33406758 PMCID: PMC7796307 DOI: 10.3390/ijms22010435] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
The existence of a common mesenchymal cell progenitor shared by bone, skeletal muscle, and adipocytes cell progenitors, makes the role of the skeleton in energy metabolism no longer surprising. Thus, bone fragility could also be seen as a consequence of a “poor” quality in nutrition. Ketogenic diet was originally proven to be effective in epilepsy, and long-term follow-up studies on epileptic children undergoing a ketogenic diet reported an increased incidence of bone fractures and decreased bone mineral density. However, the causes of such negative impacts on bone health have to be better defined. In these subjects, the concomitant use of antiepileptic drugs and the reduced mobilization may partly explain the negative effects on bone health, but little is known about the effects of diet itself, and/or generic alterations in vitamin D and/or impaired growth factor production. Despite these remarks, clinical studies were adequately designed to investigate bone health are scarce and bone health related aspects are not included among the various metabolic pathologies positively influenced by ketogenic diets. Here, we provide not only a narrative review on this issue, but also practical advice to design and implement clinical studies on ketogenic nutritional regimens and bone health outcomes. Perspectives on ketogenic regimens, microbiota, microRNAs, and bone health are also included.
Collapse
Affiliation(s)
- Daniela Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (L.G.)
| | - Roberta Cosso
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
| | - Cristina Eller-Vainicher
- Unit of Endocrinology, Fondazione IRCCS Cà Granda-Ospedale Maggiore Policlinico Milano, 20122 Milano, Italy;
| | - Fabio Vescini
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia of Udine, 33100 Udine, Italy;
| | - Iacopo Chiodini
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milano, Italy
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (L.G.)
| | - Alberto Falchetti
- Istituto Auxologico Italiano “Scientific Institute for Hospitalisation and Care”, 20100 Milano, Italy; (R.C.); (I.C.)
- Correspondence:
| |
Collapse
|
8
|
Yu J, Shi YC, Ping F, Li W, Zhang HB, He SL, Zhao Y, Xu LL, Li YX. Liraglutide Inhibits Osteoclastogenesis and Improves Bone Loss by Downregulating Trem2 in Female Type 1 Diabetic Mice: Findings From Transcriptomics. Front Endocrinol (Lausanne) 2021; 12:763646. [PMID: 34975749 PMCID: PMC8715718 DOI: 10.3389/fendo.2021.763646] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The mechanisms of bone fragility in type 1 diabetes (T1D) are not fully understood. Whether glucagon-like peptide-1 receptor (GLP-1R) agonists could improve bone quality in T1D context also remains elusive. AIMS We aimed to explore the possible mechanisms of bone loss in T1D and clarify whether liraglutide has effects on bone quality of T1D mice using transcriptomics. METHODS Female streptozotocin-induced diabetic C57BL/6J mice were randomly divided into four groups and received the following treatments daily for 8 weeks: saline as controls, insulin, liraglutide, and liraglutide combined with insulin. These groups were also compared with non-STZ-treated normal glucose tolerance (NGT) group. Trunk blood and bone tissues were collected for analysis. Three tibia from each of the NGT, saline-treated, and liraglutide-treated groups were randomly selected for transcriptomics. RESULTS Compared with NGT mice, saline-treated T1D mice manifested markedly hyperglycemia and weight loss, and micro-CT revealed significantly lower bone mineral density (BMD) and deficient microarchitectures in tibias. Eight weeks of treatment with liraglutide alone or combined with insulin rescued the decreased BMD and partly corrected the compromised trabecular microarchitectures. Transcriptomics analysis showed there were 789 differentially expressed genes mainly mapped to osteoclastogenesis and inflammation pathways. The RT-qPCR verified that the gene expression of Trem2, Nfatc1, Trap, and Ctsk were significantly increased in the tibia of T1D compared with those in the NGT group. Liraglutide treatment alone or combined with insulin could effectively suppress osteoclastogenesis by downregulating the gene expression of Trem2, Nfatc1, Ctsk, and Trap. CONCLUSIONS Taken together, increased osteoclastogenesis with upregulated expression of Trem2 played an important role in bone loss of T1D mice. Liraglutide provided protective effects on bone loss in T1D mice by suppressing osteoclastogenesis.
Collapse
Affiliation(s)
- Jie Yu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan-Chuan Shi
- Group of Neuroendocrinology, Diabetes and Metabolism Division, Garvan Institute of Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- Faculty of Medicine, UNSW, Sydney, NSW, Australia
| | - Fan Ping
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Li
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hua-Bing Zhang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shu-Li He
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuan Zhao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ling-Ling Xu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Ling-Ling Xu, ; Yu-Xiu Li,
| | - Yu-Xiu Li
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Ling-Ling Xu, ; Yu-Xiu Li,
| |
Collapse
|
9
|
Polak D, Sanui T, Nishimura F, Shapira L. Diabetes as a risk factor for periodontal disease-plausible mechanisms. Periodontol 2000 2020; 83:46-58. [PMID: 32385872 DOI: 10.1111/prd.12298] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The present narrative review examines the scientific evidence of the biological mechanisms that may link periodontitis and diabetes, as a source of comorbidity. Publications regarding periodontitis and diabetes, in human, animals, and in vitro were screened for their relevance. Periodontal microbiome studies indicate a possible association between altered glucose metabolism in prediabetes and diabetes and changes in the periodontal microbiome. Coinciding with this, hyperglycemia enhances expression of pathogen receptors, which enhance host response to the dysbiotic microbiome. Hyperglycemia also promotes pro-inflammatory response independently or via the advanced glycation end product/receptor for advanced glycation end product pathway. These processes excite cellular tissue destruction functions, which further enhance pro-inflammatory cytokines expression and alteration in the RANKL/osteoprotegerin ratio, promoting formation and activation of osteoclasts. The evidence supports the role of several pathogenic mechanisms in the path of true causal comorbidity between poorly controlled diabetes and periodontitis. However, further research is needed to better understand these mechanisms and to explore other mechanisms.
Collapse
Affiliation(s)
- David Polak
- Department of Periodontology, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Terukazu Sanui
- Section of Periodontology, Division of Oral Rehabilitation, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| | - Fusanori Nishimura
- Section of Periodontology, Division of Oral Rehabilitation, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| | - Lior Shapira
- Department of Periodontology, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| |
Collapse
|
10
|
Sasso GRDS, Florencio-Silva R, da Fonseca CCN, Cezar LC, Carbonel AAF, Gil CD, Simões MDJ, Girão MJBC. Effects of estrogen deficiency followed by streptozotocin-induced diabetes on periodontal tissues of female rats. J Mol Histol 2020; 51:353-365. [PMID: 32488735 DOI: 10.1007/s10735-020-09885-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/29/2020] [Indexed: 01/18/2023]
Abstract
Although both estrogen deficiency and diabetes contribute to periodontal tissue deterioration, the combined effects of these conditions on periodontium is unknown. Thus, we analyzed the combined effects of ovariectomy followed by streptozotocin (STZ)-induced diabetes on periodontal tissues of rats. Twenty adult rats were ovariectomized (OVX) or SHAM-operated (SHAM). After 3 weeks, the rats received an intraperitoneal injection of STZ (60 mg/kg/body weight) to induce diabetes or vehicle (blank) solution. The groups were assigned as follows (n = 5): SHAM-vehicle (SHAM), OVX-vehicle (OVX), SHAM + STZ (SHAM-Di), and OVX + STZ (OVX-Di). Seven weeks post-diabetes induction, the rats were euthanized. Blood samples were collected for glucose measurements and maxillae were processed for paraffin embedding. Sections stained with hematoxylin/eosin, Masson's trichrome, and picrosirius-red were used for alveolar bone loss and collagen fiber analysis in the lamina propria. Immunohistochemistry was performed for runt-related transcription factor 2 (Runx2), matrix metalloproteinase 9 (MMP-9), and tryptase detection. Alveolar bone loss and fewer collagen fibers were observed in the OVX-Di group, collagen fibers with irregular organization, and MMP-9 immunoreactivity were more evident in diabetic groups, and MMP-9-positive osteoclasts on alveolar bone surface were noticed in all groups. The OVX-Di group showed lower Runx2 immunoreactivity (osteoblast formation marker), and more tryptase-positive cells (mast cell marker) in the alveolar bone marrow. Our results indicate that estrogen depletion, followed by STZ-induced diabetes, promotes periodontal tissue deterioration that is more evident than both interventions applied alone. Furthermore, our results points to a possible participation of bone-derived mast cells in this process.
Collapse
Affiliation(s)
- Gisela Rodrigues da Silva Sasso
- Departamento de Ginecologia, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil.
- Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil.
| | - Rinaldo Florencio-Silva
- Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
| | - Caio Cesar Navarrete da Fonseca
- Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
| | - Luana Carvalho Cezar
- Faculdade de Medicina Veterinária e Zootecnia, Patologia Experimental e Comparada, Universidade de São Paulo, São Paulo, Brazil
| | - Adriana Aparecida Ferraz Carbonel
- Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
| | - Cristiane Damas Gil
- Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
| | - Manuel de Jesus Simões
- Departamento de Ginecologia, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
- Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brazil
| | | |
Collapse
|
11
|
Yang J, Chen S, Zong Z, Yang L, Liu D, Bao Q, Du W. The increase in bone resorption in early-stage type I diabetic mice is induced by RANKL secreted by increased bone marrow adipocytes. Biochem Biophys Res Commun 2020; 525:433-439. [PMID: 32102755 DOI: 10.1016/j.bbrc.2020.02.079] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Bone marrow adipose tissue (BMAT) has recently been found to induce osteoclastogenesis by secreting RANKL. Although Type 1 diabetes mellitus (T1DM) has been reported to be associated with increased BMAT and bone loss, little is known about the relationship between BMAT and osteoclasts in T1DM. We studied the role of BMAT in the alterations of osteoclast activities in early-stage T1DM, by using a streptozotocin-induced T1DM mouse model. Our results showed that osteoclast activity was enhanced in the long bones of T1DM mice, accompanied by increased protein expression of RANKL. However, RANKL mRNA levels in bone tissues of T1DM mice remained unchanged. Meanwhile, we found that BMAT was significantly increased in the long bones of T1DM mice, and both mRNA and protein levels of RANKL were elevated in the diabetic BMAT. More importantly, RANKL protein was mainly expressed on the cell membranes of the increased adipocytes, most of which were located next to the metaphyseal region. These results suggest that the enhanced bone resorption in early-stage diabetic mice is induced by RANKL derived from BMAT rather than the bone tissue itself.
Collapse
Affiliation(s)
- Jiazhi Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China; Department of Orthopedics, Xinqiao Hospital, Army Medical University, ChongQing, 400037, PR China
| | - Sixu Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China; Department of Orthopedics, The 906th Hospital of the Chinese People's Liberation Army, Wenzhou, Zhejiang, 325000, PR China
| | - Zhaowen Zong
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China; Department of Orthopedics, Xinqiao Hospital, Army Medical University, ChongQing, 400037, PR China.
| | - Lei Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China; Department of Orthopedics, Xinqiao Hospital, Army Medical University, ChongQing, 400037, PR China
| | - Daocheng Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China; Department of Orthopedics, Xinqiao Hospital, Army Medical University, ChongQing, 400037, PR China
| | - Quanwei Bao
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China; Department of Emergency, Xinqiao Hospital, Army Medical University, ChongQing, 400037, PR China
| | - Wenqiong Du
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, ChongQing, 400038, PR China
| |
Collapse
|
12
|
Eller-Vainicher C, Cairoli E, Grassi G, Grassi F, Catalano A, Merlotti D, Falchetti A, Gaudio A, Chiodini I, Gennari L. Pathophysiology and Management of Type 2 Diabetes Mellitus Bone Fragility. J Diabetes Res 2020; 2020:7608964. [PMID: 32566682 PMCID: PMC7262667 DOI: 10.1155/2020/7608964] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
Individuals with type 2 diabetes mellitus (T2DM) have an increased risk of bone fragility fractures compared to nondiabetic subjects. This increased fracture risk may occur despite normal or even increased values of bone mineral density (BMD), and poor bone quality is suggested to contribute to skeletal fragility in this population. These concepts explain why the only evaluation of BMD could not be considered an adequate tool for evaluating the risk of fracture in the individual T2DM patient. Unfortunately, nowadays, the bone quality could not be reliably evaluated in the routine clinical practice. On the other hand, getting further insight on the pathogenesis of T2DM-related bone fragility could consent to ameliorate both the detection of the patients at risk for fracture and their appropriate treatment. The pathophysiological mechanisms underlying the increased risk of fragility fractures in a T2DM population are complex. Indeed, in T2DM, bone health is negatively affected by several factors, such as inflammatory cytokines, muscle-derived hormones, incretins, hydrogen sulfide (H2S) production and cortisol secretion, peripheral activation, and sensitivity. All these factors may alter bone formation and resorption, collagen formation, and bone marrow adiposity, ultimately leading to reduced bone strength. Additional factors such as hypoglycemia and the consequent increased propensity for falls and the direct effects on bone and mineral metabolism of certain antidiabetic medications may contribute to the increased fracture risk in this population. The purpose of this review is to summarize the literature evidence that faces the pathophysiological mechanisms underlying bone fragility in T2DM patients.
Collapse
Affiliation(s)
- C. Eller-Vainicher
- Unit of Endocrinology, Fondazione IRCCS Cà Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - E. Cairoli
- Istituto Auxologico Italiano, IRCCS, Unit for Bone Metabolism Diseases and Diabetes & Lab of Endocrine and Metabolic Research, Italy
- Dept. of Clinical Sciences & Community Health, University of Milan, Milan, Italy
| | - G. Grassi
- Unit of Endocrinology, Fondazione IRCCS Cà Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Dept. of Clinical Sciences & Community Health, University of Milan, Milan, Italy
| | - F. Grassi
- Ramses Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - A. Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - D. Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - A. Falchetti
- Istituto Auxologico Italiano, IRCCS, Unit for Bone Metabolism Diseases and Diabetes & Lab of Endocrine and Metabolic Research, Italy
| | - A. Gaudio
- Department of Clinical and Experimental Medicine, University of Catania, University Hospital ‘G. Rodolico', Catania, Italy
| | - I. Chiodini
- Istituto Auxologico Italiano, IRCCS, Unit for Bone Metabolism Diseases and Diabetes & Lab of Endocrine and Metabolic Research, Italy
- Dept. of Clinical Sciences & Community Health, University of Milan, Milan, Italy
| | - L. Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| |
Collapse
|
13
|
Costantini S, Conte C. Bone health in diabetes and prediabetes. World J Diabetes 2019; 10:421-445. [PMID: 31523379 PMCID: PMC6715571 DOI: 10.4239/wjd.v10.i8.421] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/03/2019] [Accepted: 07/20/2019] [Indexed: 02/05/2023] Open
Abstract
Bone fragility has been recognized as a complication of diabetes, both type 1 diabetes (T1D) and type 2 diabetes (T2D), whereas the relationship between prediabetes and fracture risk is less clear. Fractures can deeply impact a diabetic patient's quality of life. However, the mechanisms underlying bone fragility in diabetes are complex and have not been fully elucidated. Patients with T1D generally exhibit low bone mineral density (BMD), although the relatively small reduction in BMD does not entirely explain the increase in fracture risk. On the contrary, patients with T2D or prediabetes have normal or even higher BMD as compared with healthy subjects. These observations suggest that factors other than bone mass may influence fracture risk. Some of these factors have been identified, including disease duration, poor glycemic control, presence of diabetes complications, and certain antidiabetic drugs. Nevertheless, currently available tools for the prediction of risk inadequately capture diabetic patients at increased risk of fracture. Aim of this review is to provide a comprehensive overview of bone health and the mechanisms responsible for increased susceptibility to fracture across the spectrum of glycemic status, spanning from insulin resistance to overt forms of diabetes. The management of bone fragility in diabetic patient is also discussed.
Collapse
Affiliation(s)
- Silvia Costantini
- Department of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, Milan 20123, Italy
- Epatocentro Ticino, Lugano 6900, Switzerland
| | - Caterina Conte
- Department of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, Milan 20123, Italy
- IRCCS Ospedale San Raffaele, Internal Medicine and Transplantation, Milan 20123, Italy
| |
Collapse
|
14
|
Liu C, Zhu R, Liu H, Li L, Chen B, Jia Q, Wang L, Ma R, Tian S, Wang M, Fu M, Niu J, Orekhov AN, Gao S, Zhang D, Zhao B. Aqueous Extract of Mori Folium Exerts Bone Protective Effect Through Regulation of Calcium and Redox Homeostasis via PTH/VDR/CaBP and AGEs/RAGE/Nox4/NF-κB Signaling in Diabetic Rats. Front Pharmacol 2018; 9:1239. [PMID: 30459613 PMCID: PMC6233025 DOI: 10.3389/fphar.2018.01239] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/11/2018] [Indexed: 12/16/2022] Open
Abstract
Purpose: The present study is aimed to explore whether the aqueous extract of Mori Folium (MF) exhibits bone protective effect by regulating calcium and redox homeostasis in diabetic rats, and to identify the signaling pathways involved in this process. Methods: Diabetic rats were established using high-sugar and high-fat diet and streptozotocin (STZ) (30 mg/kg for 3 consecutive days). The serum levels of osteocalcin (OC), insulin-like growth factor-1 (IGF-1), tartrate-resistant acid phosphatase (TRAP), phosphorus (P), calcium (Ca), 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], parathormone (PTH), advanced glycation end products (AGEs), superoxide dismutase (SOD), and malondialdehyde (MDA), total antioxidant capacity (TAC), 8-hydroxy-2'-deoxyguanosine (8-OH-dG), and interleukin 6 (IL-6) were determined by ELISA or biochemical assays. Histopathological alterations in the femurs were evaluated by the stainings of hematoxylin-eosin (H&E) and alizarin red S. In addition, femoral strength was detected by a three-point bending assay, bone microstructure was detected with micro-computer tomography. Bone material properties were examined by Fourier-transform infrared spectroscopy. Furthermore, the expressions of IGF-1, runt-related transcription factor 2 (Runx2), osteoprotegerin (OPG), receptor activator of nuclear factor kappa-B ligand (RANKL), cathepsin K, AGEs, receptor of advanced glycation end products (RAGE), NADPH oxidase 4 (Nox4), and nuclear factor kappa-B (NF-κB) in the femurs and tibias, and the alterations in the levels of calcium-binding protein-28k (CaBP-28k), transient receptor potential V6 (TRPV6), and vitamin D receptor (VDR) in the kidneys and duodenums were determined by western blot and immunohistochemical analysis. Results: Treatment of diabetic rats with MF aqueous extract induces an increase in the levels of OC and IGF-1 as well as a decrease in TRAP level in serum. MF treatment also upregulates the expression of OPG, downregulates the expressions of AGEs, RAGE, Nox4, NF-κB, and RANKL, which leads to improve bone microstructure and strength exhibited by an increase in cortical area ratio, cortical thickness, and trabecular area ratio as well as ultimate load, elastic modulus, and bending stress in the femurs and tibias of diabetic rats. In addition, MF aqueous extract preserves bone material properties by decreasing the ratio of fatty acid/collagen and increasing the ratio of mineral/matrix in the femurs of diabetic rats. Moreover, MF treatment increases the levels of P, Ca, and 1,25(OH)2D3, and decreases the level of PTH in the serum, as well as upregulates the expressions of TRPV6 and VDR in the duodenums and CaBP-28k in the kidneys of diabetic rats. Additionally, MF has ability of rebuilding redox homeostasis and eliminating inflammatory stress by increasing the levels of SOD and TAC as well as decreasing the levels of IL-6, AGEs, MDA, and 8-OH-dG. Conclusions: MF treatment may improve bone quality through maintenance of calcium homeostasis via regulating the PTH/VDR/CaBP signaling, and elimination of oxidative stress via regulating the AGEs/RAGE/Nox4/NF-κB signaling. These results may suggest the potential of MF in preventing the development of diabetic osteoporosis.
Collapse
Affiliation(s)
- Chenyue Liu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Ruyuan Zhu
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Haixia Liu
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Lin Li
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Beibei Chen
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Qiangqiang Jia
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Lili Wang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Rufeng Ma
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Simin Tian
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Min Wang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Min Fu
- The Research Institute of McGill University Health Center, Montreal, QC, Canada
| | - Jianzhao Niu
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Sihua Gao
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Dongwei Zhang
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Abstract
The adult human skeleton is a multifunctional organ undergoing constant remodeling through the opposing activities of the bone-resorbing osteoclast and the bone-forming osteoblast. The exquisite balance between bone resorption and bone formation is responsible for bone homeostasis in healthy adults. However, evidence has emerged that such a balance is likely disrupted in diabetes where systemic glucose metabolism is dysregulated, resulting in increased bone frailty and osteoporotic fractures. These findings therefore underscore the significance of understanding the role and regulation of glucose metabolism in bone under both normal and pathological conditions. Recent studies have shed new light on the metabolic plasticity and the critical functions of glucose metabolism during osteoclast and osteoblast differentiation. Moreover, these studies have begun to identify intersections between glucose metabolism and the growth factors and transcription factors previously known to regulate osteoblasts and osteoclasts. Here we summarize the current knowledge in the nascent field, and suggest that a fundamental understanding of glucose metabolic pathways in the critical bone cell types may open new avenues for developing novel bone therapeutics.
Collapse
Affiliation(s)
- Courtney M Karner
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Fanxin Long
- Department of Orthopaedic Surgery, Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63131, USA.
| |
Collapse
|
16
|
Cao GL, Tian FM, Liu GY, Song HP, Yuan LL, Geng LD, Bei MJ, Zheng ZY, Zhang L. Strontium Ranelate Combined with Insulin Is as Beneficial as Insulin Alone in Treatment of Fracture Healing in Ovariectomized Diabetic Rats. Med Sci Monit 2018; 24:6525-6536. [PMID: 30221634 PMCID: PMC6154119 DOI: 10.12659/msm.911573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) and estrogen deficiency both predispose fracture patients to increased risk of delayed union or nonunion. The present study investigated the effects of strontium ranelate (SR) on fracture healing in ovariectomized (OVX) diabetic rats. Material/Methods A mid-shaft fracture was established in female normal control (CF), diabetic (DF), and OVX diabetic (DOF) rats. Treated DOF rats received either insulin alone (DOFI) or combined with SR (DOFIS). All rats were euthanized at 2 or 3 weeks after fracture. Fracture healing was evaluated using radiological, histological, immunohistochemical, and micro-computed tomography analyses. Results At 3 weeks after fracture, radiological and histological evaluations demonstrated delayed fracture healing in the DF group compared with the CF group, which was exacerbated by OVX, as indicated by the significantly lower X-ray score, BMD, BV/TV, and Md.Ar/Ps.Cl.Ar, and the markedly decreased OCN and Col I expression in the DOF group. All these changes were prevented by insulin alone or combined with SR treatment. In comparison with the DOFI group, DOFIS rats displayed markedly higher OCN expression at 2 weeks after fracture and Col I expression at 2 and 3 weeks after fracture. Conclusions These results demonstrated delayed fracture healing with preexisting estrogen deficiency and T2DM. While insulin alone and combined with SR were both effective in promoting bone fracture healing in this model, their combined treatment showed significant improvement in promoting osteogenic marker expression, but not of the radiological appearance, compared with insulin alone.
Collapse
Affiliation(s)
- Guo-Long Cao
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Fa-Ming Tian
- Medical Research Center, North China University of Science and Technology, Tangshan, Hebei, China (mainland).,International Science and Technology Cooperation Base of Geriatric Medicine, Department of International Cooperation, Ministry of Science and Technology of China, Tangshan, Hebei, China (mainland)
| | - Guang-Yuan Liu
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Hui-Ping Song
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Lei-Liang Yuan
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Lin-Dan Geng
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Ming-Jian Bei
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Zhi-Yuan Zheng
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Mine Medical Security Center, Meitan General Hospital, Beijing, China (mainland)
| |
Collapse
|
17
|
Cai J, Li W, Sun T, Li X, Luo E, Jing D. Pulsed electromagnetic fields preserve bone architecture and mechanical properties and stimulate porous implant osseointegration by promoting bone anabolism in type 1 diabetic rabbits. Osteoporos Int 2018. [PMID: 29523929 DOI: 10.1007/s00198-018-4392-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED The effects of exogenous pulsed electromagnetic field (PEMF) stimulation on T1DM-associated osteopathy were investigated in alloxan-treated rabbits. We found that PEMF improved bone architecture, mechanical properties, and porous titanium (pTi) osseointegration by promoting bone anabolism through a canonical Wnt/β-catenin signaling-associated mechanism, and revealed the clinical potential of PEMF stimulation for the treatment of T1DM-associated bone complications. INTRODUCTION Type 1 diabetes mellitus (T1DM) is associated with deteriorated bone architecture and impaired osseous healing potential; nonetheless, effective methods for resisting T1DM-associated osteopenia/osteoporosis and promoting bone defect/fracture healing are still lacking. PEMF, as a safe and noninvasive method, have proven to be effective for promoting osteogenesis, whereas the potential effects of PEMF on T1DM osteopathy remain poorly understood. METHODS We herein investigated the effects of PEMF stimulation on bone architecture, mechanical properties, bone turnover, and its potential molecular mechanisms in alloxan-treated diabetic rabbits. We also developed novel nontoxic Ti2448 pTi implants with closer elastic modulus with natural bone and investigated the impacts of PEMF on pTi osseointegration for T1DM bone-defect repair. RESULTS The deteriorations of cancellous and cortical bone architecture and tissue-level mechanical strength were attenuated by 8-week PEMF stimulation. PEMF also promoted osseointegration and stimulated more adequate bone ingrowths into the pore spaces of pTi in T1DM long-bone defects. Moreover, T1DM-associated reduction of bone formation was significantly attenuated by PEMF, whereas PEMF exerted no impacts on bone resorption. We also found PEMF-induced activation of osteoblastogenesis-related Wnt/β-catenin signaling in T1DM skeletons, but PEMF did not alter osteoclastogenesis-associated RANKL/RANK signaling gene expression. CONCLUSION We reveal that PEMF improved bone architecture, mechanical properties, and pTi osseointegration by promoting bone anabolism through a canonical Wnt/β-catenin signaling-associated mechanism. This study enriches our basic knowledge for understanding skeletal sensitivity in response to external electromagnetic signals, and also opens new treatment alternatives for T1DM-associated osteopenia/osteoporosis and osseous defects in an easy and highly efficient manner.
Collapse
MESH Headings
- Animals
- Biomechanical Phenomena/physiology
- Bone Diseases, Metabolic/etiology
- Bone Diseases, Metabolic/physiopathology
- Bone Diseases, Metabolic/prevention & control
- Bone Remodeling/physiology
- Bone and Bones/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/physiopathology
- Implants, Experimental
- Magnetic Field Therapy/methods
- Male
- Osseointegration/physiology
- Porosity
- Rabbits
- Titanium
- Wnt Signaling Pathway/physiology
- X-Ray Microtomography
Collapse
Affiliation(s)
- J Cai
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xi'an-Xianyang New Economic Zone, Xianyang, 712046, China.
- Department of Biomedical Engineering, Fourth Military Medical University, 17 West Changle Road, Xi'an, 710032, China.
| | - W Li
- Department of Biomedical Engineering, Fourth Military Medical University, 17 West Changle Road, Xi'an, 710032, China
| | - T Sun
- Department of Biomedical Engineering, Fourth Military Medical University, 17 West Changle Road, Xi'an, 710032, China
| | - X Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - E Luo
- Department of Biomedical Engineering, Fourth Military Medical University, 17 West Changle Road, Xi'an, 710032, China
| | - D Jing
- Department of Biomedical Engineering, Fourth Military Medical University, 17 West Changle Road, Xi'an, 710032, China.
| |
Collapse
|
18
|
Ferreira ECS, Bortolin RH, Freire-Neto FP, Souza KSC, Bezerra JF, Ururahy MAG, Ramos AMO, Himelfarb ST, Abreu BJ, Didone TVN, Pedrosa LFC, Medeiros AC, Doi SQ, Brandão-Neto J, Hirata RDC, Rezende LA, Almeida MG, Hirata MH, Rezende AA. Zinc supplementation reduces RANKL/OPG ratio and prevents bone architecture alterations in ovariectomized and type 1 diabetic rats. Nutr Res 2017; 40:48-56. [PMID: 28473060 DOI: 10.1016/j.nutres.2017.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/16/2017] [Accepted: 03/09/2017] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes mellitus (T1DM) and estrogen deficiency are associated with several alterations in bone turnover. Zinc (Zn) is required for growth, development, and overall health. Zinc has been used in complementary therapy against bone loss in several diseases. We hypothesized that Zn supplementation represents a potential therapy against severe bone loss induced by the combined effect of estrogen deficiency and T1DM. We evaluated the protective effect of Zn against bone alterations in a chronic model of these disorders. Female Wistar rats were ramdomized into 3 groups (5 rats each): control, OVX/T1DM (ovariectomized rats with streptozotocin-induced T1DM), and OVX/T1DM+Zn (OVX/T1DM plus daily Zn supplementation). Serum biochemical, bone histomorphometric, and molecular analyses were performed. Histomorphometric parameters were similar between the control and OVX/T1DM+Zn groups, suggesting that Zn prevents bone architecture alterations. In contrast, the OVX/T1DM group showed significantly lower trabecular width and bone area as well as greater trabecular separation than the control. The OVX/T1DM and OVX/T1DM+Zn groups had significantly higher serum alkaline phosphatase activity than the control. The supplemented group had higher levels of serum-ionized calcium and phosphorus than the nonsupplemented group. The RANKL/OPG ratio was similar between the control and OVX/T1DM+Zn groups, whereas it was higher in the OVX/T1DM group. In conclusion, Zn supplementation prevents bone alteration in chronic OVX/T1DM rats, as demonstrated by the reduced RANKL/OPG ratio and preservation of bone architecture. The findings may represent a novel therapeutic approach to preventing OVX/T1DM-induced bone alterations.
Collapse
Affiliation(s)
- Elaine C S Ferreira
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Raul H Bortolin
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Francisco P Freire-Neto
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Karla S C Souza
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - João F Bezerra
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Marcela A G Ururahy
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Ana M O Ramos
- Department of Clinical Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Silvia T Himelfarb
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Bento J Abreu
- Department of Morphology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Thiago V N Didone
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Lucia F C Pedrosa
- Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Aldo C Medeiros
- Department of Clinical Medicine, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Sonia Q Doi
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - José Brandão-Neto
- Department of Clinical Medicine, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Rosário D C Hirata
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Luciana A Rezende
- Department of Chemistry, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Maria G Almeida
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Mario H Hirata
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Adriana A Rezende
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
19
|
Palermo A, D'Onofrio L, Buzzetti R, Manfrini S, Napoli N. Pathophysiology of Bone Fragility in Patients with Diabetes. Calcif Tissue Int 2017; 100:122-132. [PMID: 28180919 DOI: 10.1007/s00223-016-0226-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 12/20/2016] [Indexed: 02/07/2023]
Abstract
It has been well established that bone fragility is one of the chronic complications of diabetes mellitus, and both type 1 and type 2 diabetes are risk factors for fragility fractures. Diabetes may negatively affect bone health by unbalancing several pathways: bone formation, bone resorption, collagen formation, inflammatory cytokine, muscular and incretin system, bone marrow adiposity and calcium metabolism. The purpose of this narrative review is to explore the current understanding of pathophysiological pathways underlying bone fragility in diabetics. In particular, the review will focus on the peculiar cellular and molecular system impairment that may lead to increased risk of fracture in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Andrea Palermo
- Diabetes and Bone network, Department Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
| | - Luca D'Onofrio
- Department of Experimental Medicine, Polo Pontino, Sapienza University of Rome, Rome, Italy
| | - Raffaella Buzzetti
- Department of Experimental Medicine, Polo Pontino, Sapienza University of Rome, Rome, Italy
| | - Silvia Manfrini
- Diabetes and Bone network, Department Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
| | - Nicola Napoli
- Diabetes and Bone network, Department Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21 - 00128, Rome, Italy.
- Division of Bone and Mineral Diseases, Washington University in St Louis, St Louis, USA.
| |
Collapse
|
20
|
Iyer S, Han L, Ambrogini E, Yavropoulou M, Fowlkes J, Manolagas SC, Almeida M. Deletion of FoxO1, 3, and 4 in Osteoblast Progenitors Attenuates the Loss of Cancellous Bone Mass in a Mouse Model of Type 1 Diabetes. J Bone Miner Res 2017; 32:60-69. [PMID: 27491024 PMCID: PMC5492385 DOI: 10.1002/jbmr.2934] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes is associated with osteopenia and increased fragility fractures, attributed to reduced bone formation. However, the molecular mechanisms mediating these effects remain unknown. Insulin promotes osteoblast formation and inhibits the activity of the FoxO transcription factors. FoxOs, on the other hand, inhibit osteoprogenitor proliferation and bone formation. Here, we investigated whether FoxOs play a role in the low bone mass associated with type 1 diabetes, using mice lacking FoxO1, 3, and 4 in osteoprogenitor cells (FoxO1,3,4ΔOsx1-Cre ). Streptozotocin-induced diabetes caused a reduction in bone mass and strength in FoxO-intact mice. In contrast, cancellous bone was unaffected in diabetic FoxO1,3,4ΔOsx1-Cre mice. The low bone mass in the FoxO-intact diabetic mice was associated with decreased osteoblast number and bone formation, as well as decreased expression of the anti-osteoclastogenic cytokine osteoprotegerin (OPG) and increased osteoclast number. FoxO deficiency did not alter the effects of diabetes on bone formation; however, it did prevent the decrease in OPG and the increase in osteoclast number. Addition of high glucose to osteoblastic cell cultures decreased OPG mRNA, indicating that hyperglycemia in and of itself contributes to diabetic bone loss. Taken together, these results suggest that FoxOs exacerbate the loss of cancellous bone mass associated with type 1 diabetes and that inactivation of FoxOs might ameliorate the adverse effects of insulin deficiency. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Srividhya Iyer
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Li Han
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Maria Yavropoulou
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - John Fowlkes
- Barnstable Brown Diabetes and Obesity Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| |
Collapse
|
21
|
Histological evidence that metformin reverses the adverse effects of diabetes on orthodontic tooth movement in rats. J Mol Histol 2016; 48:73-81. [DOI: 10.1007/s10735-016-9707-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
|
22
|
Kalaitzoglou E, Popescu I, Bunn RC, Fowlkes JL, Thrailkill KM. Effects of Type 1 Diabetes on Osteoblasts, Osteocytes, and Osteoclasts. Curr Osteoporos Rep 2016; 14:310-319. [PMID: 27704393 PMCID: PMC5106298 DOI: 10.1007/s11914-016-0329-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW To describe the effects of type 1 diabetes on bone cells. RECENT FINDINGS Type 1 diabetes (T1D) is associated with low bone mineral density, increased risk of fractures, and poor fracture healing. Its effects on the skeleton were primarily attributed to impaired bone formation, but recent data suggests that bone remodeling and resorption are also compromised. The hyperglycemic and inflammatory environment associated with T1D impacts osteoblasts, osteocytes, and osteoclasts. The mechanisms involved are complex; insulinopenia, pro-inflammatory cytokine production, and alterations in gene expression are a few of the contributing factors leading to poor osteoblast activity and survival and, therefore, poor bone formation. In addition, the observed sclerostin level increase accompanied by decreased osteocyte number and enhanced osteoclast activity in T1D results in uncoupling of bone remodeling. T1D negatively impacts osteoblasts and osteocytes, whereas its effects on osteoclasts are not well characterized, although the limited studies available indicate increased osteoclast activity, favoring bone resorption.
Collapse
Affiliation(s)
- Evangelia Kalaitzoglou
- UK Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, 830 S. Limestone St., Lexington, KY, 40536, USA.
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.
| | - Iuliana Popescu
- UK Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, 830 S. Limestone St., Lexington, KY, 40536, USA
| | - R Clay Bunn
- UK Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, 830 S. Limestone St., Lexington, KY, 40536, USA
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - John L Fowlkes
- UK Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, 830 S. Limestone St., Lexington, KY, 40536, USA
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Kathryn M Thrailkill
- UK Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, 830 S. Limestone St., Lexington, KY, 40536, USA
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| |
Collapse
|
23
|
Seref-Ferlengez Z, Suadicani SO, Thi MM. A new perspective on mechanisms governing skeletal complications in type 1 diabetes. Ann N Y Acad Sci 2016; 1383:67-79. [PMID: 27571221 DOI: 10.1111/nyas.13202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/11/2016] [Accepted: 07/18/2016] [Indexed: 12/29/2022]
Abstract
This review focuses on bone mechanobiology in type 1 diabetes (T1D), an area of research on diabetes-associated skeletal complications that is still in its infancy. We first provide a brief overview of the deleterious effects of diabetes on the skeleton and of the knowledge gained from studies with rodent models of T1D. Second, we discuss two specific hallmarks of T1D, low insulin and high glucose, and address the extent to which they affect skeletal health. Third, we highlight the mechanosensitive nature of bone tissue and the importance of mechanical loading for bone health. We also summarize recent advances in bone mechanobiology that implicate osteocytes as the mechanosensors and major regulatory cells in the bone. Finally, we discuss recent evidence indicating that the diabetic bone is "deaf" to mechanical loading and that osteocytes are central players in mechanisms that lead to bone loss in T1D.
Collapse
Affiliation(s)
- Zeynep Seref-Ferlengez
- Department of Orthopaedic Surgery.,Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE)
| | - Sylvia O Suadicani
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE).,Department of Neuroscience.,Department of Urology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
| | - Mia M Thi
- Department of Orthopaedic Surgery.,Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE).,Department of Neuroscience
| |
Collapse
|
24
|
Reni C, Mangialardi G, Meloni M, Madeddu P. Diabetes Stimulates Osteoclastogenesis by Acidosis-Induced Activation of Transient Receptor Potential Cation Channels. Sci Rep 2016; 6:30639. [PMID: 27468810 PMCID: PMC4965751 DOI: 10.1038/srep30639] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 07/07/2016] [Indexed: 01/10/2023] Open
Abstract
Patients with type 1 diabetes have lower bone mineral density and higher risk of fractures. The role of osteoblasts in diabetes-related osteoporosis is well acknowledged whereas the role of osteoclasts (OCLs) is still unclear. We hypothesize that OCLs participate in pathological bone remodeling. We conducted studies in animals (streptozotocin-induced type 1 diabetic mice) and cellular models to investigate canonical and non-canonical mechanisms underlying excessive OCL activation. Diabetic mice show an increased number of active OCLs. In vitro studies demonstrate the involvement of acidosis in OCL activation and the implication of transient receptor potential cation channel subfamily V member 1 (TRPV1). In vivo studies confirm the establishment of local acidosis in the diabetic bone marrow (BM) as well as the ineffectiveness of insulin in correcting the pH variation and osteoclast activation. Conversely, treatment with TRPV1 receptor antagonists re-establishes a physiological OCL availability. These data suggest that diabetes causes local acidosis in the BM that in turn increases osteoclast activation through the modulation of TRPV1. The use of clinically available TRPV1 antagonists may provide a new means to combat bone problems associated with diabetes.
Collapse
Affiliation(s)
- Carlotta Reni
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Giuseppe Mangialardi
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Marco Meloni
- Vascular Pathology and Regeneration, Bristol Heart Institute, University of Bristol, UK.,University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| |
Collapse
|
25
|
Peng J, Hui K, Hao C, Peng Z, Gao QX, Jin Q, Lei G, Min J, Qi Z, Bo C, Dong QN, Bing ZH, Jia XY, Fu DL. Low bone turnover and reduced angiogenesis in streptozotocin-induced osteoporotic mice. Connect Tissue Res 2016; 57:277-89. [PMID: 27028715 DOI: 10.3109/03008207.2016.1171858] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
It is known that type 1 diabetes (T1D) reduces bone mass and increases the risk for fragility fractures, an effect that has been largely ascribed to decreased bone formation. However, the potential role of decreased angiogenesis as a factor in osteogenesis reduction has not been extensively studied. Furthermore, there is controversy surrounding the effect of T1D on bone resorption. This study characterized bone microstructure, bone strength, and bone turnover of streptozotocin (STZ)-induced diabetic mice (T1D mice) and explored the role of angiogenesis in the pathogenesis of T1D-induced osteoporosis. Results demonstrate that T1D deteriorated trabecular microarchitecture and led to reduced bone strength. Furthermore, T1D mice showed reduced osteoblast number/bone surface (N.Ob/BS), mineral apposition rate, mineral surface/BS, and bone formation rate/BS, suggesting attenuated bone formation. Decreased angiogenesis was shown by a reduced number of blood vessels in the femur and decreased expression of platelet endothelial cell adhesion molecule (CD31), nerve growth factor, hypoxia-inducible factor-1α, and vascular endothelial growth factor was observed. On the other hand, reduced bone resorption, an effect that could lead to impaired osteogenesis, was demonstrated by lower osteoclast number/BS and decreased tartrate-resistant acid phosphatase and cathepsin K mRNA levels. Reduced number of osteoblasts and decreased expression of receptor activator for nuclear factor-κB ligand could be responsible for compromised bone resorption in T1D mice. In conclusion, T1D mice display reduced bone formation and bone resorption, suggesting decreased bone turnover. Furthermore, this study points to impairments in angiogenesis as a pivotal cause of decreased bone formation.
Collapse
Affiliation(s)
- Jia Peng
- a Department of Orthopaedics , The Second Affiliated Hospital of Soochow University , Suzhou , Jiangsu Province , China.,b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Kang Hui
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Chen Hao
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Zhao Peng
- c Intensive Care Unit , The Second Affiliated Hospital of Soochow University , Suzhou , Jiangsu Province , China
| | - Qian Xing Gao
- d Department of Rehabilitation, 359th Hospital of Chinese People's Liberation Army , Zhen Jiang , Jiangsu Province , China
| | - Qi Jin
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Guo Lei
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Jiang Min
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Zhou Qi
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Chen Bo
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Qian Nian Dong
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Zhou Han Bing
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Xu You Jia
- a Department of Orthopaedics , The Second Affiliated Hospital of Soochow University , Suzhou , Jiangsu Province , China
| | - Deng Lian Fu
- b Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
26
|
Guo Y, Wang L, Ma R, Mu Q, Yu N, Zhang Y, Tang Y, Li Y, Jiang G, Zhao D, Mo F, Gao S, Yang M, Kan F, Ma Q, Fu M, Zhang D. JiangTang XiaoKe granule attenuates cathepsin K expression and improves IGF-1 expression in the bone of high fat diet induced KK-Ay diabetic mice. Life Sci 2016; 148:24-30. [PMID: 26892148 DOI: 10.1016/j.lfs.2016.02.056] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 01/25/2016] [Accepted: 02/13/2016] [Indexed: 12/19/2022]
Abstract
AIM To assess the beneficial effects of JiangTang XiaoKe (JTXK) granule on the bone metabolism in high fat diet (HFD) fed KK-Ay diabetic mice. MATERIALS AND METHODS The KK-Ay mice were used as a diabetic model, while C57BL/6 mice were utilized as the non-diabetic control. The left tibia was used for determining bone mineral density (BMD) and bone ash coefficient. The HE and alizarin red S staining of femur were employed to evaluate bone pathology and calcium deposition. The expressions of alkaline phosphatase (ALP), insulin growth factor 1 (IGF-1) and cathepsin K were assessed by western blotting and immunohistochemical staining. KEY FINDINGS JTXK granule significantly improved the bone ash coefficient, the distribution of trabecular bone and the calcification nodules deposition in KK-Ay mice with diabetes. IGF-1 and ALP expressions were significantly decreased, and cathepsin K expression was dramatically increased in the HFD fed KK-Ay diabetic model mice, which can be reversed by JTXK granule treatment. JTXK granule at medium or high dosage was more efficient in improving diabetic bone quality when compared with that in mice with a low dosage. However, the BMD values in each group of KK-Ay diabetic mice were not significantly different. SIGNIFICANCE We demonstrate that cathepsin K expression is increased in KK-Ay diabetic mouse model. JTXK granule treatment inhibits osteoclastic bone resorption and promotes the new bone formation by decreasing cathepsin K activity and increasing IGF-1 and ALP levels. These changes may contribute to the increase of bone strength and thus reducing the risk of bone fractures.
Collapse
Affiliation(s)
- Yubo Guo
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lili Wang
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Rufeng Ma
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qianqian Mu
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Na Yu
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi Zhang
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuqing Tang
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yu Li
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guangjian Jiang
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dandan Zhao
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fangfang Mo
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Sihua Gao
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Meijuan Yang
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Feifei Kan
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qun Ma
- Chinese Material Medical School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Min Fu
- McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Dongwei Zhang
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
27
|
Diedrich J, Gusky HC, Podgorski I. Adipose tissue dysfunction and its effects on tumor metabolism. Horm Mol Biol Clin Investig 2015; 21:17-41. [PMID: 25781550 DOI: 10.1515/hmbci-2014-0045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/14/2015] [Indexed: 12/12/2022]
Abstract
Growing by an alarming rate in the Western world, obesity has become a condition associated with a multitude of diseases such as diabetes, metabolic syndrome and various cancers. Generally viewed as an abnormal accumulation of hypertrophied adipocytes, obesity is also a poor prognostic factor for recurrence and chemoresistance in cancer patients. With more than two-thirds of the adult population in the United States considered clinically overweight or obese, it is critical that the relationship between obesity and cancer is further emphasized and elucidated. Adipocytes are highly metabolically active cells, which, through release of adipokines and cytokines and activation of endocrine and paracrine pathways, affect processes in neighboring and distant cells, altering their normal homeostasis. This work will examine specifically how adipocyte-derived factors regulate the cellular metabolism of malignant cells within the tumor niche. Briefly, tumor cells undergo metabolic pressure towards a more glycolytic and hypoxic state through a variety of metabolic regulators and signaling pathways, i.e., phosphoinositol-3 kinase (PI3K), hypoxia-inducible factor-1 alpha (HIF-1α), and c-MYC signaling. Enhanced glycolysis and high lactate production are hallmarks of tumor progression largely because of a process known as the Warburg effect. Herein, we review the latest literature pertaining to the body of work on the interactions between adipose and tumor cells, and underlining the changes in cancer cell metabolism that have been targeted by the currently available treatments.
Collapse
|
28
|
Diabetes mellitus related bone metabolism and periodontal disease. Int J Oral Sci 2015; 7:63-72. [PMID: 25857702 PMCID: PMC4817554 DOI: 10.1038/ijos.2015.2] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus and periodontal disease are chronic diseases affecting a large number of populations worldwide. Changed bone metabolism is one of the important long-term complications associated with diabetes mellitus. Alveolar bone loss is one of the main outcomes of periodontitis, and diabetes is among the primary risk factors for periodontal disease. In this review, we summarise the adverse effects of diabetes on the periodontium in periodontitis subjects, focusing on alveolar bone loss. Bone remodelling begins with osteoclasts resorbing bone, followed by new bone formation by osteoblasts in the resorption lacunae. Therefore, we discuss the potential mechanism of diabetes-enhanced bone loss in relation to osteoblasts and osteoclasts.
Collapse
|
29
|
Increase in osteoclastogenesis in an obese Otsuka Long-Evans Tokushima fatty rat model. Mol Med Rep 2015; 12:3874-3880. [DOI: 10.3892/mmr.2015.3811] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 03/03/2015] [Indexed: 11/05/2022] Open
|
30
|
Lee HP, Lin YY, Duh CY, Huang SY, Wang HM, Wu SF, Lin SC, Jean YH, Wen ZH. Lemnalol attenuates mast cell activation and osteoclast activity in a gouty arthritis model. ACTA ACUST UNITED AC 2014; 67:274-85. [PMID: 25557511 DOI: 10.1111/jphp.12331] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 09/21/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVES In this study, we investigated the effects of a soft coral-derived anti-inflammatory compound, lemnalol, on mast cell (MC) function and osteoclast activity in rats with monosodium urate (MSU) crystal-induced gouty arthritis. METHODS In this study, we examined the therapeutic effects of lemnalol on intra-articular injection of MSU induces gouty arthritis with the measurement of ankle oedema. Toluidine blue staining were used to analyse the infiltration and the percentage degranulation MCs. Immunohistochemical analysis showed CD117, transforming growth factor beta 1 (TGF-β1), matrix metalloproteinase 9 (MMP-9), the osteoclast markers cathepsin K and tartrate-resistant acid phosphatase (TRAP) protein expression in ankle tissue. KEY FINDINGS We found that both infiltration and degranulation of MCs increased at 24 h after MSU injection in the ankle joint. Immunohistochemical analysis showed that MSU induced upregulation of TGF-β1, MMP-9, the osteoclast markers cathepsin K and TRAP in ankle tissues. Administration of lemnalol ameliorated MSU-induced TGF-β1, MMP-9, cathepsin K and TRAP protein expression. CONCLUSIONS Taken together, our results show that MSU-induced gouty arthritis is accompanied by osteoclast-related protein upregulation and that lemnalol treatment may be beneficial for the attenuation of MC infiltration and degranulation and for suppressing osteoclast activation in gouty arthritis.
Collapse
Affiliation(s)
- Hsin-Pai Lee
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, Ping-Tung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Prates TP, Taira TM, Holanda MC, Bignardi LA, Salvador SL, Zamboni DS, Cunha FQ, Fukada SY. NOD2 contributes to Porphyromonas gingivalis-induced bone resorption. J Dent Res 2014; 93:1155-62. [PMID: 25239844 DOI: 10.1177/0022034514551770] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The NOD-like receptors are cytoplasmic proteins that sense microbial by-products released by invasive bacteria. Although NOD1 and NOD2 are functionally expressed in cells from oral tissues and play a role triggering immune responses, the role of NOD2 receptor in the bone resorption and in the modulation of osteoclastogenesis is still unclear. We show that in an experimental model of periodontitis with Porphyromonas gingivalis W83, NOD2(-/-) mice showed lower bone resorption when compared to wild type. Quantitative polymerase chain reaction analysis revealed that wild-type infected mice showed an elevated RANKL/OPG ratio when compared to NOD2(-/-) infected mice. Moreover, the expression of 2 osteoclast activity markers-cathepsin K and matrix metalloproteinase 9-was significantly lower in gingival tissue from NOD2(-/-) infected mice compared to WT infected ones. The in vitro study reported an increase in the expression of the NOD2 receptor 24 hr after stimulation of hematopoietic bone marrow cells with M-CSF and RANKL. We also evaluated the effect of direct activation of NOD2 receptor on osteoclastogenesis, by the activation of this receptor in preosteoclasts culture, with different concentrations of muramyl dipeptide. The results show no difference in the number of TRAP-positive cells. Although it did not alter the osteoclasts differentiation, the activation of NOD2 receptor led to a significant increase of cathepsin K expression. We confirm that this enzyme was active, since the osteoclasts resorption capacity was enhanced by muramyl dipeptide stimulation, evaluated in osteoassay plate. These results show that the lack of NOD2 receptor impairs the bone resorption, suggesting that NOD2 receptor could contribute to the progression of bone resorption in experimental model of periodontitis. The stimulation of NOD2 by its agonist, muramyl dipeptide, did not affect osteoclastogenesis, but it does favor the bone resorption capacity identified by increased osteoclast activity.
Collapse
Affiliation(s)
- T P Prates
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - T M Taira
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo
| | - M C Holanda
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo
| | - L A Bignardi
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - S L Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo
| | - D S Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo
| | - F Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo
| | - S Y Fukada
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo
| |
Collapse
|
32
|
Lapmanee S, Charoenphandhu N, Aeimlapa R, Suntornsaratoon P, Wongdee K, Tiyasatkulkovit W, Kengkoom K, Chaimongkolnukul K, Seriwatanachai D, Krishnamra N. High dietary cholesterol masks type 2 diabetes-induced osteopenia and changes in bone microstructure in rats. Lipids 2014; 49:975-86. [PMID: 25200330 DOI: 10.1007/s11745-014-3950-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes mellitus (T2DM) often occurs concurrently with high blood cholesterol or dyslipidemia. Although T2DM has been hypothesized to impair bone microstructure, several investigations showed that, when compared to age-matched healthy individuals, T2DM patients had normal or relatively high bone mineral density (BMD). Since cholesterol and lipids profoundly affect the function of osteoblasts and osteoclasts, it might be cholesterol that obscured the changes in BMD and bone microstructure in T2DM. The present study, therefore, aimed to determine bone elongation, epiphyseal histology, and bone microstructure in non-obese T2DM Goto-Kakizaki rats treated with normal (GK-ND) and high cholesterol diet. We found that volumetric BMD was lower in GK-ND rats than the age-matched wild-type controls. In histomorphometric study of tibial metaphysis, T2DM evidently suppressed osteoblast function as indicated by decreases in osteoblast surface, mineral apposition rate, and bone formation rate in GK-ND rats. Meanwhile, the osteoclast surface and eroded surface were increased in GK-ND rats, thus suggesting an activation of bone resorption. T2DM also impaired bone elongation, presumably by retaining the chondrogenic precursor cells in the epiphyseal resting zone. Interestingly, several bone changes in GK rats (e.g., increased osteoclast surface) disappeared after high cholesterol treatment as compared to wild-type rats fed high cholesterol diet. In conclusion, high cholesterol diet was capable of masking the T2DM-induced osteopenia and changes in several histomorphometric parameters that indicated bone microstructural defect. Cholesterol thus explained, in part, why a decrease in BMD was not observed in T2DM, and hence delayed diagnosis of the T2DM-associated bone disease.
Collapse
Affiliation(s)
- Sarawut Lapmanee
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Izumi Y, Hayashi M, Morimoto R, Cheng XW, Wu H, Ishii H, Yasuda Y, Yoshikawa D, Izawa H, Matsuo S, Oiso Y, Murohara T. Impact of circulating cathepsin K on the coronary calcification and the clinical outcome in chronic kidney disease patients. Heart Vessels 2014; 31:6-14. [DOI: 10.1007/s00380-014-0570-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 08/15/2014] [Indexed: 12/26/2022]
|
34
|
Zinc supplementation inhibits the increase in osteoclastogenesis and decrease in osteoblastogenesis in streptozotocin-induced diabetic rats. Eur J Pharmacol 2013; 714:41-7. [PMID: 23735664 DOI: 10.1016/j.ejphar.2013.05.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 05/03/2013] [Accepted: 05/20/2013] [Indexed: 01/16/2023]
Abstract
Zinc (Zn) has been shown to stimulate bone formation and inhibit osteoclastic bone resorption and osteoclastogenesis. However, the effects of Zn on bone metabolism in diabetic animals remain to be clarified in vivo. Here, the effects of Zn supplementation on bone metabolism, including osteoclastogenesis and osteoblastogenesis, were investigated using streptozotocine (STZ)-induced diabetic rats. Zn-supplemented water (7.5 mg/L) was given for 1 week to diabetic rats injected with STZ (30 mg/kg body weight) 1 week earlier. The Zn supplement prevented a decrease in the activity and mRNA of alkaline phosphatase (ALP), osteocalcin mRNA, and hydroxyproline and calcium levels, and an increase in the activity and mRNA of tartrate-resistant acid phosphatase (TRAP) and cathepsin K in the proximal tibia of diabetic rats. Histological analysis revealed that the Zn supplement inhibited the diabetes-induced increase and decrease in the number of osteoclasts and osteoblasts, respectively, in the metaphysis of the proximal tibia. The increase in mRNA levels of receptor for activation of NF-κB (RANK), c-fos, c-jun, TRAP, and cathepsin K and decrease in the expression of Runx2, Dlx5, osterix, ALP, osteocalcin, and collagen were prevented by the supplement. The decrease in β-catenin, phosphorylated GSK3β, phosphorylated Akt, insulin-like growth factor 1 (IGF-1), and IGF-1 receptor (IGF-1R) protein levels in diabetic rats was also inhibited, although Zn did not affect the diabetes-increased gene and protein expression of Sost and Dkk1. These results suggested that Zn prevented the diabetes-induced increase in osteoclastogenesis and decrease in osteoblastogenesis by inhibiting RANK expression and stimulating IGF-1/IGF-1R/Akt/GSK3β/β-catenin signaling, respectively.
Collapse
|
35
|
NAKANISHI ATSUKO, IITSUKA NATSUMI, TSUKAMOTO IKUYO. Fish oil suppresses bone resorption by inhibiting osteoclastogenesis through decreased expression of M-CSF, PU.1, MITF and RANK in ovariectomized rats. Mol Med Rep 2013; 7:1896-903. [DOI: 10.3892/mmr.2013.1446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 04/12/2013] [Indexed: 11/05/2022] Open
|
36
|
NAKANISHI ATSUKO, HIE MAMIKO, IITSUKA NATSUMI, TSUKAMOTO IKUYO. A crucial role for reactive oxygen species in macrophage colony-stimulating factor-induced RANK expression in osteoclastic differentiation. Int J Mol Med 2013; 31:874-80. [DOI: 10.3892/ijmm.2013.1258] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 12/07/2012] [Indexed: 11/06/2022] Open
|
37
|
Hie M, Iitsuka N, Otsuka T, Nakanishi A, Tsukamoto I. Zinc deficiency decreases osteoblasts and osteoclasts associated with the reduced expression of Runx2 and RANK. Bone 2011; 49:1152-9. [PMID: 21893222 DOI: 10.1016/j.bone.2011.08.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 07/25/2011] [Accepted: 08/16/2011] [Indexed: 12/22/2022]
Abstract
The effects of Zinc(Zn)-deficiency on the function and differentiation of osteoblasts and osteoclasts were investigated in vivo using rats, which were fed a Zn-adequate (control) or Zn-free diet (ZD) or pair-fed a Zn-adequate diet (PF) for 3 weeks. Levels of Zn, insulin, insulin-like growth factor I (IGF-I), and osteoclacin in serum and the activities and numbers of osteoblasts and osteoclasts in bone decreased in ZD rats compared with the control and PF rats. The frequency analyses showed that the precursors of osteoblasts and osteoclasts decreased in bone marrow of ZD, but not PF, rats. The expression of receptor for activation of NF-κB (RANK) decreased with the Zn-deficiency, although RANK ligand, osteoprotegerin, macrophage colony-stimulating factor, and c-fms levels were unaltered. The protein level of a transcription factor MITF, but not PU.1, decreased. The expression of Runx2 decreased associated with the decrease in β-catenin protein and the suppression of glycogen synthase kinase 3β (GSK3β) inhibition and Akt activation. The gene expression of the insulin receptor, IGF-I and the IGF-I receptor was decreased with a reduced level of transcription factor SP-1. These results suggested that a deficiency of Zn decreased osteoclastogenesis associated with the reduced expression of RANK through a decrease in MITF protein, and osteoblastogenesis associated with the reduced expression of Runx2 through the inhibition of Wnt/β-catenin signaling via the suppression of GSK3β inhibition and Akt activation preceded by the reduced level of SP-1 protein.
Collapse
Affiliation(s)
- Mamiko Hie
- Department of Food Science and Nutrition, Nara Women's University, Nara 630, Japan
| | | | | | | | | |
Collapse
|
38
|
Hie M, Tsukamoto I. Administration of zinc inhibits osteoclastogenesis through the suppression of RANK expression in bone. Eur J Pharmacol 2011; 668:140-6. [PMID: 21806983 DOI: 10.1016/j.ejphar.2011.07.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 06/20/2011] [Accepted: 07/14/2011] [Indexed: 01/08/2023]
Abstract
Zinc (Zn) has been known to inhibit osteoclastic bone resorption and stimulate osteoblastic bone formation. However, the mechanisms responsible for these effects have not been well characterized in vivo. Here, the effects of a dietary administration of Zn on osteoclastogenesis and osteoblastogenesis were investigated in Zn-adequate rats. The administration of Zn decreased the activities of bone tartrate-resistant acid phosphatase (TRAP) and cathepsin K, without affecting the serum osteocalcin level. Histological analysis showed a decrease in the number of osteoclasts with a normal number of osteoblasts in the metaphysis of the proximal tibia. The mRNA levels of receptor for activation of NF-κB (RANK), c-fos, c-jun, TRAP and cathepsin K were significantly decreased, although those of RANK ligand, macrophage colony-stimulating factor and c-fms were unaltered. The gene expression of bone morphogenic protein-2, Runx2, Dlx5, osterix, alkaline phosphatase, osteocalcin and collagen was not affected. The level of the RANK protein decreased, while the levels of the Runx2 and β-catenin proteins were unchanged. Further, the osteoclastic differentiation of precursor cells in vitro was suppressed. The suppressed osteoclastogenesis was associated with decreased levels of reactive oxygen species, extracellular signal-regulated kinase (ERK) activation and RANK expression. A lower lipid peroxide level and a higher glutathione level were also observed. These results suggested that Zn-administration did not affect osteoblastogenesis but decreased osteoclastogenesis by inhibiting RANK expression through suppression of the production of reactive oxygen species and ERK activation in Zn-adequate rats.
Collapse
Affiliation(s)
- Mamiko Hie
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | | |
Collapse
|
39
|
Jing D, Cai J, Shen G, Huang J, Li F, Li J, Lu L, Luo E, Xu Q. The preventive effects of pulsed electromagnetic fields on diabetic bone loss in streptozotocin-treated rats. Osteoporos Int 2011; 22:1885-95. [PMID: 20976595 DOI: 10.1007/s00198-010-1447-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 09/28/2010] [Indexed: 01/26/2023]
Abstract
UNLABELLED The present study was the first report demonstrating that pulsed electromagnetic field (PEMF) could partially prevent bone strength and architecture deterioration and improve the impaired bone formation in streptozotocin-induced diabetic rats. The findings indicated that PEMF might become a potential additive method for inhibiting diabetic osteopenia or osteoporosis. INTRODUCTION Diabetes mellitus (DM) can cause various musculoskeletal abnormalities. Optimal therapeutic methods for diabetic bone complication are still lacking. It is essential to develop more effective and safe therapeutic methods for diabetic bone disorders. Pulsed electromagnetic field (PEMF) as an alternative noninvasive method has proven to be effective for treating fracture healing and osteoporosis in non-diabetic conditions. However, the issue about the therapeutic effects of PEMF on diabetic bone complication has not been previously investigated. METHODS We herein systematically evaluated the preventive effects of PEMF on diabetic bone loss in streptozotocin-treated rats. Two similar experiments were conducted. In each experiment, 16 diabetic and eight non-diabetic rats were equally assigned to the control, DM, and DM + PEMF group. DM + PEMF group was subjected to daily 8-h PEMF exposure for 8 weeks. RESULTS In experiment 1, three-point bending test suggested that PEMF improved the biomechanical quality of diabetic bone tissues, evidenced by increased maximum load, stiffness, and energy absorption. Microcomputed tomography analysis demonstrated that DM-induced bone architecture deterioration was partially reversed by PEMF, evidenced by increased Tb.N, Tb.Th, BV/TV, and Conn.D and reduced Tb.Sp and SMI. Serum OC analysis indicated that PEMF partially prevented DM-induced decrease in bone formation. In experiment 2, no significant difference in the bone resorption marker TRACP5b was observed. These biochemical findings were further supported by the dynamic bone histomorphometric parameters BFR/BS and Oc.N/BS. CONCLUSIONS The results demonstrated that PEMF could partially prevent DM-induced bone strength and architecture deterioration and improve the impaired bone formation. PEMF might become a potential additive method for inhibiting diabetic osteoporosis.
Collapse
Affiliation(s)
- D Jing
- Faculty of Biomedical Engineering, Fourth Military Medical University, 17 West Changle Road, Xi'an 710032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhang Y, Papasian CJ, Deng HW. Alteration of vitamin D metabolic enzyme expression and calcium transporter abundance in kidney involved in type 1 diabetes-induced bone loss. Osteoporos Int 2011; 22:1781-8. [PMID: 20878391 PMCID: PMC4537183 DOI: 10.1007/s00198-010-1404-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 08/17/2010] [Indexed: 12/24/2022]
Abstract
UNLABELLED This study aimed to delineate the mechanism involved in type 1 diabetes-induced bone loss. The results revealed the alteration of vitamin D metabolic enzyme expression and the downregulation of renal calcium transporter abundance in type 1 diabetic mice. INTRODUCTION The purpose of this study was to investigate the changes of the expression of vitamin D metabolic enzymes and transcellular calcium-transporting proteins in kidneys from mice with experimentally induced diabetes. METHODS Male DBA/2J mice were injected with either vehicle (control) or streptozotocin (STZ) daily for five consecutive days. Bone mineral density was measured by peripheral quantitative computerized tomography, and bone histomorphology was analyzed by Safranin O staining. Real-time PCR and Western blotting were applied to determine the expression of target genes and proteins. RESULTS Type 1 diabetes produced high urinary calcium excretion and loss of trabecular bone measured at the proximal metaphysis of the tibia and the distal femur. Bone loss was associated with deterioration of trabecular bone microstructure. Quantified PCR results showed that mRNA expression level in the kidney of diabetic mice for 25-hydroxyvitamin D-24-hydroxylase was downregulated at week 10, while those for 25-hydroxyvitamin D-1α-hydroxylase were upregulated at week 20. In addition, mRNA expression levels for renal transient receptor potential V6, plasma membrane Ca-ATPase (PMCA)1b, and vitamin D receptor (VDR) genes were decreased in STZ-treated mice. Western blot analysis showed that protein expression of PMCA1b and VDR was significantly decreased in kidneys from STZ-treated mice compared to that of controls. CONCLUSIONS The limitation in this study is the lack of vitamin D, parathyroid hormone, and phosphorus levels in serum. However, the present study supports the conclusion that the underlying mechanism contributing to type 1 diabetes-associated bone loss may be alterations of vitamin D metabolic enzyme expression and associated decreases in expression of renal calcium transporters.
Collapse
Affiliation(s)
- Y Zhang
- Center of System Biomedical Sciences, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, People's Republic of China.
| | | | | |
Collapse
|
41
|
Braga SMG, Taddei SRDA, Andrade I, Queiroz-Junior CM, Garlet GP, Repeke CE, Teixeira MM, da Silva TA. Effect of diabetes on orthodontic tooth movement in a mouse model. Eur J Oral Sci 2011; 119:7-14. [PMID: 21244505 DOI: 10.1111/j.1600-0722.2010.00793.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Orthodontic tooth movement is achieved by the remodeling of alveolar bone in response to mechanical loading. Type 1 diabetes results in bone remodeling, suggesting that this disease might affect orthodontic tooth movement. The present study investigated the effects of the diabetic state on orthodontic tooth movement. An orthodontic appliance was placed in normoglycemic (NG), streptozotocin-induced diabetes (DB), and insulin-treated DB (IT) C57BL6/J mice. Histomorphometric analysis and quantitative PCR of periodontium were performed. The DB mice exhibited greater orthodontic tooth movement and had a higher number of tartrate-resistant acid phosphate (TRAP) -positive osteoclasts than NG mice. This was associated with increased expression of factors involved in osteoclast activity and recruitment (Rankl, Csf1, Ccl2, Ccl5, and Tnfa) in DB mice. The expression of osteoblastic markers (Runx2, Ocn, Col1, and Alp) was decreased in DB mice. Reversal of the diabetic state by insulin treatment resulted in morphological findings similar to those of NG mice. These results suggest that the diabetic state up-regulates osteoclast migration and activity and down-regulates osteoblast differentiation, resulting in greater orthodontic tooth movement.
Collapse
Affiliation(s)
- Sarah M G Braga
- Department of Orthodontics, Faculty of Dentistry, Pontifícia Universidade Católica de Minas Gerais (PUC-Minas), Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Bone response to orthodontic forces in diabetic Wistar rats. Am J Orthod Dentofacial Orthop 2011; 139:S76-82. [PMID: 21435542 DOI: 10.1016/j.ajodo.2010.06.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Revised: 06/01/2010] [Accepted: 06/01/2010] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Patients with type 1 diabetes have shown decreased bone mineral density (BMD) values. The need for orthodontic treatment in diabetic patients is usually associated with occlusal problems and the occurrence of abnormalities in the development of the jaws. The aim of this study was to analyze bone response of insulin-treated and untreated diabetic rats after applying orthodontic forces. METHODS Wistar rats were divided into 3 groups: experimental orthodontics, experimental diabetes and orthodontics, and experimental diabetes treated with insulin and experimental orthodontics. Orthodontic forces were applied the first day of the seventh week. Forty-eight hours after placement, all the animals were killed, and the maxillae were excised and processed using routine histologic techniques. RESULTS Bone activity in the periodontal cortex of the dental alveolus showed a significant decrease in bone formation and erosive areas in diabetic animals as compared with controls. A recovery of these parameters could be observed in the group with experimental diabetes treated with insulin and experimental orthodontics. Bone volume in the interradicular bone showed no significant differences among groups. CONCLUSIONS People with diabetes should not receive orthodontic treatment until their metabolic status normalizes. Bone response to orthodontic forces in insulin-treated diabetic subjects does not differ significantly from that observed in healthy subjects.
Collapse
|
43
|
Hie M, Tsukamoto I. Vitamin C-deficiency stimulates osteoclastogenesis with an increase in RANK expression. J Nutr Biochem 2011; 22:164-71. [DOI: 10.1016/j.jnutbio.2010.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2009] [Revised: 12/15/2009] [Accepted: 01/04/2010] [Indexed: 11/28/2022]
|
44
|
Zhang Y, Li Q, Wan HY, Xiao HH, Lai WP, Yao XS, Wong MS. Study of the mechanisms by which Sambucus williamsii HANCE extract exert protective effects against ovariectomy-induced osteoporosis in vivo. Osteoporos Int 2011; 22:703-9. [PMID: 20414641 DOI: 10.1007/s00198-010-1240-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 03/11/2010] [Indexed: 11/25/2022]
Abstract
UNLABELLED The purpose of this study is to investigate the dose-dependent effects of SWH on bone properties and the mechanism involved in mediating the osteoprotective actions of SWH. The results indicated that SWH could improve bone properties by inhibiting the process of bone resorption and stimulating the process of bone formation. INTRODUCTION Our previous study showed that Sambucus williamsii HANCE (SWH) improved trabecular bone mass and cortical bone strength in ovariectomized (OVX) rats. The purpose of this study is to investigate the dose-dependent effects of SWH on bone properties and the mechanism involved in mediating the osteoprotective actions of SWH. METHODS Three-month-old C57BL/6J mice were fed a phytoestrogen-free diet and subjected to either ovariectomy or sham operation. OVX mice were treated with genistein (50 mg/kg), or a low (200 mg/kg), medium (500 mg/kg), or high (1,000 mg/kg) dose of SWH extract. RESULTS SWH could dose-dependently decrease urinary Ca excretion and increase serum Ca level in OVX mice. It could increase tibial bone mineral density and exert beneficial effects on the microarchitecture of trabecular bone in the OVX mice. SWH suppressed the ovariectomy-induced expression of Cbfa1 mRNA and cathepsin K mRNA and enhanced the ratio of OPG/RANKL mRNA expression in the tibia. In vitro study showed that SWH dramatically reduced the number of TRAP-positive cells in RANKL-induced RAW 264.7 cells. CONCLUSIONS The present study indicated that SWH could improve bone properties by inhibiting the process of bone resorption and stimulating the process of bone formation.
Collapse
Affiliation(s)
- Y Zhang
- Center of System Biomedical Sciences, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
45
|
Ariffin SHZ, Abidin IZZ, Yazid MD, Wahab RMA. Differentiation analyses of adult suspension mononucleated peripheral blood cells of Mus musculus. Cell Commun Signal 2010; 8:29. [PMID: 20969794 PMCID: PMC2984458 DOI: 10.1186/1478-811x-8-29] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 10/23/2010] [Indexed: 11/14/2022] Open
Abstract
Background The purpose of this study is to determine whether isolated suspension mouse peripheral mononucleated blood cells have the potential to differentiate into two distinct types of cells, i.e., osteoblasts and osteoclasts. Results Differentiation into osteoblast cells was concomitant with the activation of the Opn gene, increment of alkaline phosphatase (ALP) activity and the existence of bone nodules, whereas osteoclast cells activated the Catk gene, increment of tartrate resistant acid phosphatase (TRAP) activity and showed resorption activities via resorption pits. Morphology analyses showed the morphology of osteoblast and osteoclast cells after von Kossa and May-Grunwald-Giemsa staining respectively. Conclusions In conclusion, suspension mononucleated cells have the potentiality to differentiate into mature osteoblasts and osteoclasts, and hence can be categorized as multipotent stem cells.
Collapse
Affiliation(s)
- Shahrul Hisham Zainal Ariffin
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia.
| | | | | | | |
Collapse
|
46
|
Kayal RA, Siqueira M, Alblowi J, McLean J, Krothapalli N, Faibish D, Einhorn TA, Gerstenfeld LC, Graves DT. TNF-alpha mediates diabetes-enhanced chondrocyte apoptosis during fracture healing and stimulates chondrocyte apoptosis through FOXO1. J Bone Miner Res 2010; 25:1604-15. [PMID: 20200974 PMCID: PMC3154002 DOI: 10.1002/jbmr.59] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
To gain insight into the effect of diabetes on fracture healing, experiments were carried out focusing on chondrocyte apoptosis during the transition from cartilage to bone. Type 1 diabetes was induced in mice by multiple low-dose streptozotocin injections, and simple transverse fractures of the tibia or femur was carried out. Large-scale transcriptional profiling and gene set enrichment analysis were performed to examine apoptotic pathways on total RNA isolated from fracture calluses on days 12, 16, and 22, a period of endochondral bone formation when cartilage is resorbed and chondrocyte numbers decrease. Tumor necrosis factor alpha (TNF-alpha) protein levels were assessed by ELISA and caspase-3 by bioactivity assay. The role of TNF was examined by treating mice with the TNF-specific inhibitor pegsunercept. In vitro studies investigated the proapoptotic transcription factor FOXO1 in regulating TNF-induced apoptosis of chondrogenic ATDC5 and C3H10T1/2 cells as representative of differentiated chondrocytes, which are important during endochondral ossification. mRNA profiling revealed an upregulation of gene sets related to apoptosis in the diabetic group on day 16 when cartilage resorption is active but not day 12 or day 22. This coincided with elevated TNF-alpha protein levels, chondrocyte apoptosis, enhanced caspase-3 activity, and increased FOXO1 nuclear translocation (p < .05). Inhibition of TNF significantly reduced these parameters in the diabetic mice but not in normoglycemic control mice (p < .05). Silencing FOXO1 using siRNA in vitro significantly reduced TNF-induced apoptosis and caspase activity in differentiated chondrocytes. The mRNA levels of the proapoptotic genes caspase-3, caspase-8, caspase-9, and TRAIL were significantly reduced with silencing of FOXO1 in chondrocytic cells. Inhibiting caspase-8 and caspase-9 significantly reduced TNF-induced apoptosis in chondrogenic cells. These results suggest that diabetes causes an upregulation of proapoptotic genes during the transition from cartilage to bone in fracture healing. Diabetes increased chondrocyte apoptosis through a mechanism that involved enhanced production of TNF-alpha, which stimulates chondrocyte apoptosis and upregulates mRNA levels of apoptotic genes through FOXO1 activation.
Collapse
Affiliation(s)
- Rayyan A Kayal
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Erythrina variegata extract exerts osteoprotective effects by suppression of the process of bone resorption. Br J Nutr 2010; 104:965-71. [DOI: 10.1017/s0007114510001789] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Our previous study showed that Erythrina variegata L. (EV) inhibited bone loss and improved bone properties in ovariectomised rats. The purpose of the present study is to investigate the potential mechanism involved in mediating the osteoprotective actions of EV. Female Sprague–Dawley rats were fed a phyto-oestrogen-free diet and subjected to either ovariectomy or a sham operation. Ovariectomised rats were treated with genistein (40 mg/kg) as well as low (200 mg/kg), medium (500 mg/kg) or high (1000 mg/kg) doses of EV extract. Bone properties and mRNA expressions were evaluated by micro-computed tomography and quantitative RT-PCR, respectively. Osteoclast differentiation in RAW 264·7 cells was studied by tartrate-resistant acid phosphatase (TRAP) staining. High doses of EV could decrease urinary Ca and P excretion, maintain serum Ca and P level, and exert beneficial effects on the micro-structure and morphology of trabecular bone and cortical bone in ovariectomised rats. EV suppressed the up-regulation of cathepsin K mRNA and the down-regulation of osteoprotegrin mRNA in the tibia of ovariectomised rats. TRAP-positive cell numbers were significantly decreased in receptor activator of nuclear factor-κB ligand (RANKL)-induced RAW 264·7 cells when co-cultured with EV extracts. The present study indicated that the protective effects of EV on bone properties in ovariectomised rats are likely to be mediated by its inhibitory actions on the process of bone resorption via the suppression of osteoclast differentiation and maturation.
Collapse
|
48
|
Abstract
Diabetes mellitus and, in particular, type 1 diabetes has been associated with impaired osseous wound healing properties. The scope of the present review is to discuss the clinical evidence supporting a higher rate of complications during fracture healing in diabetic patients and the histological evidence indicating impaired potential for intramembranous and endochondral ossification in the presence of uncontrolled experimental diabetes. The article further provides a synthesis of our current understanding of the plausible molecular mechanisms underlying the diabetic bone healing pathophysiology and of the role of insulin treatment in promoting osseous healing in the diabetic status.
Collapse
Affiliation(s)
- M Retzepi
- Periodontology Unit, Clinical Research Division, UCL Eastman Dental Institute, London, UK.
| | | |
Collapse
|
49
|
Garg G, Pradeep AR, Thorat MK. Effect of nonsurgical periodontal therapy on crevicular fluid levels of Cathepsin K in periodontitis. Arch Oral Biol 2009; 54:1046-51. [PMID: 19782963 DOI: 10.1016/j.archoralbio.2009.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 08/19/2009] [Accepted: 08/26/2009] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Cathepsin K (CTSK), predominantly expressed in osteoclasts, is a potent extracellular matrix degrading enzyme that plays a critical role in osteoclast-mediated bone resorption. Its increased gingival crevicular fluid (GCF) levels in periodontal disease have been reported in a previous study. The present study has been carried out to assess the role of CTSK in periodontal disease and to determine the effect of periodontal treatment on CTSK concentration in GCF. DESIGN 60 subjects were divided into three groups (n=20) based on gingival index (GI), probing pocket depth (PPD) and clinical attachment loss (CAL): healthy (group I), gingivitis (group II) and chronic periodontitis (group III). A fourth group (group IV) consisted of 20 subjects from group III, 6-8 weeks after nonsurgical periodontal therapy (scaling and root planing). GCF samples collected from each patient were quantified for CTSK using ELISA. RESULTS The mean CTSK concentration in GCF was found to be the highest in group III, i.e. 55.55 pmol/l. The mean CTSK concentration in GCF in group I and group II was 5.95 pmol/l and 6.90 pmol/l respectively. The mean CTSK concentration in GCF in group IV decreased to 11.15 pmol/l, slightly more than that in groups I and II. CONCLUSIONS GCF CTSK levels increased in periodontitis and correlated negatively with clinical parameters like GI, PPD and CAL. CTSK levels decreased after nonsurgical treatment of periodontitis. Thus, CTSK can be considered as a 'marker of osteoclastic activity' in periodontal disease and also deserves further consideration as a therapeutic target.
Collapse
Affiliation(s)
- Garima Garg
- Department of Periodontics, Government Dental College and Research Institute, Fort, Bangalore 560002, Karnataka, India.
| | | | | |
Collapse
|
50
|
Hie M, Yamazaki M, Tsukamoto I. Curcumin suppresses increased bone resorption by inhibiting osteoclastogenesis in rats with streptozotocin-induced diabetes. Eur J Pharmacol 2009; 621:1-9. [PMID: 19699734 DOI: 10.1016/j.ejphar.2009.08.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Accepted: 08/11/2009] [Indexed: 10/20/2022]
Abstract
Curcumin is a potent inhibitor of the transcription factor activator protein-1 which plays an essential role in osteoclastogenesis. However, the effects of curcumin on bone metabolism have not been clarified in vivo. We reported herein the inhibitory effects of curcumin on the stimulated osteoclastic activity in insulin-dependent diabetes mellitus using rats with streptozotocin-induced diabetes. A dietary supplement of curcumin reversed the increase in levels of activity and mRNA of tartrate-resistant acid phosphatase (TRAP) and cathepsin K to control values. A histochemical analysis showed that the increase in TRAP-positive cells in the distal femur of the diabetic rats was reduced to the control level by the supplement. These results suggested that curcumin reduced diabetes-stimulated bone resorptive activity and the number of osteoclasts. When bone marrow cells were cultured with macrophage colony stimulating factor and receptor activator NF-kappaB ligand (RANKL), the increased activity to form TRAP-positive multinucleated cells and the increased levels of mRNA and protein of c-fos and c-jun in the cultured cells from diabetic rats decreased to control levels in the curcumin-supplemented rats. Similarly, the increased expression of c-fos and c-jun in the distal femur of the diabetic rats was significantly reduced by the supplement. These results suggested that curcumin suppressed the increased bone resorptive activity through the prevention of osteoclastogenesis associated with inhibition of the expression of c-fos and c-jun in the diabetic rats.
Collapse
Affiliation(s)
- Mamiko Hie
- Department of Food Science and Nutrition, Nara Women's University, Nara, Japan
| | | | | |
Collapse
|