1
|
Wanionok NE, Molinuevo MS, Fernández JM, Lucas B, Cortizo AM, Castillo EJ, Jiron JM, Claudia S, Leon S, Aguirre JI, McCarthy AD. Skeletal Effects of a Prolonged Oral Metformin Treatment in Adult Wistar Rats. Exp Clin Endocrinol Diabetes 2024; 132:547-556. [PMID: 38740375 DOI: 10.1055/a-2324-8661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
INTRODUCTION We previously showed that a 3-week oral metformin (MET) treatment enhances the osteogenic potential of bone marrow stromal cells (BMSCs) and improves several bone histomorphometric parameters in Wistar rats with metabolic syndrome (MetS). However, the skeletal effects of extended periods of MET need to be completely elucidated. Hence, in this study, the impact of a prolonged (3-month) MET treatment was investigated on bone architecture, histomorphometric and biomechanics variables, and osteogenic potential of BMSCs in Wistar rats with or without MetS. MATERIALS AND METHODS Young male Wistar rats (n=36) were randomized into four groups (n=9) that received either 20% fructose (F), MET (MET), F plus MET treatments (FMET), or drinking water alone (Veh). Rats were euthanized, blood was collected, and bones were dissected and processed for peripheral quantitative computed tomography (pQCT) analysis, static and dynamic histomorphometry, and bone biomechanics. In addition, BMSCs were isolated to determine their osteogenic potential. RESULTS MET affected trabecular and cortical bone, altering bone architecture and biomechanics. Furthermore, MET increased the pro-resorptive profile of BMSCs. In addition, fructose-induced MetS practically did not affect the the structural or mechanical variables of the skeleton. CONCLUSION A 3-month treatment with MET (with or without MetS) affects bone architecture and biomechanical variables in Wistar rats.
Collapse
Affiliation(s)
- Nahuel E Wanionok
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - María S Molinuevo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Juan M Fernández
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Besada Lucas
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Ana M Cortizo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Evelyn J Castillo
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Jessica M Jiron
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Sedlinsky Claudia
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Schurman Leon
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - José I Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Antonio D McCarthy
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| |
Collapse
|
2
|
Lasalvia S, Sedlinsky C, Schurman L, McCarthy AD, Wanionok NE. Metformin treatment prevents experimental metabolic syndrome-induced femoral bone marrow adiposity in rats. Rev Peru Med Exp Salud Publica 2024; 41:28-36. [PMID: 38808841 PMCID: PMC11149757 DOI: 10.17843/rpmesp.2024.411.13333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/28/2024] [Indexed: 05/30/2024] Open
Abstract
OBJECTIVE. Motivation for the study. Most research supports a negative association between metabolic syndrome and bone health, although there is an overall lack of consensus. Therefore, there is a need for research in this area to develop a better understanding. Main findings. Metabolic syndrome induced by a fructose-rich diet increases the adipogenic predisposition of bone marrow progenitor cells and femoral medullary adiposity in rats. Furthermore, this can be partially prevented by co-treatment with metformin. Implications. Experimental metabolic syndrome has negative effects on bone tissue and can be prevented by oral treatment with metformin as a normoglycemic drug. To determine the effect of metformin (MET) treatment on adipogenic predisposition of bone marrow progenitor cells (BMPC), bone marrow adiposity and bone biomechanical properties. MATERIALS AND METHODS. 20 young adult male Wistar rats were sorted into four groups. Each of the groups received the following in drinking water: 100% water (C); 20% fructose (F); metformin 100 mg/kg wt/day (M); or fructose plus metformin (FM). After five weeks the animals were sacrificed. Both humeri were dissected to obtain BMPC, and both femurs were dissected to evaluate medullary adiposity (histomorphometry) and biomechanical properties (3-point bending). BMPC were cultured in vitro in adipogenic medium to evaluate RUNX2, PPAR-γ and RAGE expression by RT-PCR, lipase activity and triglyceride accumulation. RESULTS. The fructose-rich diet (group F) caused an increase in both triglycerides in vitro, and medullary adiposity in vivo; being partially or totally prevented by co-treatment with metformin (group FM). No differences were found in femoral biomechanical tests in vivo, nor in lipase activity and RUNX2/PPAR-γ ratio in vitro. DRF increased RAGE expression in BMPC, being prevented by co-treatment with MET. CONCLUSIONS. Metabolic syndrome induced by a fructose-rich diet increases femoral medullary adiposity and, in part, the adipogenic predisposition of BMPC. In turn, this can be totally or partially prevented by oral co-treatment with MET.
Collapse
Affiliation(s)
- Siro Lasalvia
- Laboratory of Research on Osteopathies and Mineral Metabolism (LIOMM), Faculty of Exact Sciences, National University of La Plata. Buenos Aires, Argentina.National University of La PlataLaboratory of Research on Osteopathies and Mineral Metabolism (LIOMM)Faculty of Exact SciencesNational University of La PlataBuenos AiresArgentina
| | - Claudia Sedlinsky
- Laboratory of Research on Osteopathies and Mineral Metabolism (LIOMM), Faculty of Exact Sciences, National University of La Plata. Buenos Aires, Argentina.National University of La PlataLaboratory of Research on Osteopathies and Mineral Metabolism (LIOMM)Faculty of Exact SciencesNational University of La PlataBuenos AiresArgentina
| | - León Schurman
- Laboratory of Research on Osteopathies and Mineral Metabolism (LIOMM), Faculty of Exact Sciences, National University of La Plata. Buenos Aires, Argentina.National University of La PlataLaboratory of Research on Osteopathies and Mineral Metabolism (LIOMM)Faculty of Exact SciencesNational University of La PlataBuenos AiresArgentina
| | - Antonio Desmond McCarthy
- Laboratory of Research on Osteopathies and Mineral Metabolism (LIOMM), Faculty of Exact Sciences, National University of La Plata. Buenos Aires, Argentina.National University of La PlataLaboratory of Research on Osteopathies and Mineral Metabolism (LIOMM)Faculty of Exact SciencesNational University of La PlataBuenos AiresArgentina
| | - Nahuel Ezequiel Wanionok
- Laboratory of Research on Osteopathies and Mineral Metabolism (LIOMM), Faculty of Exact Sciences, National University of La Plata. Buenos Aires, Argentina.National University of La PlataLaboratory of Research on Osteopathies and Mineral Metabolism (LIOMM)Faculty of Exact SciencesNational University of La PlataBuenos AiresArgentina
| |
Collapse
|
3
|
Molinuevo MS, Cortizo AM, Sedlinsky C. Effects of advanced glycation end-products, diabetes and metformin on the osteoblastic transdifferentiation capacity of vascular smooth muscle cells: In vivo and in vitro studies. J Diabetes Complications 2023; 37:108626. [PMID: 37839167 DOI: 10.1016/j.jdiacomp.2023.108626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
AIMS Our objective was to study the vascular smooth muscle cells (VSMC) osteoblastic transdifferentiation in AGE exposed cells or those from diabetic animals, and its response to metformin treatment. METHODS VSMC were obtained from non-diabetic rats, grown with or without AGE; while VSMC of in vivo-ex vivo studies were obtained from non-diabetic control animals (C), diabetic (D), C treated with metformin (M) and D treated with metformin (D-M). We studied the osteoblastic differentiation by evaluating alkaline phosphatase (ALP), type I collagen (Col) and mineral deposit. RESULTS In vitro, AGE increased proliferation, migration, and osteoblastic differentiation of VSMC. Metformin cotreatment prevented the AGE induced proliferation and migration. Both AGE and metformin stimulated the expression of ALP and Col. AGE induced mineralization was prevented by metformin. VSMC from D expressed a higher production of Col and ALP. Those from D-M showed an ALP increase vs C and M, and a partial decrease vs D. Cultured in osteogenic medium, ALP, Col and mineralization increased in D vs C, remained unchanged in M, and were prevented in D-M animals. CONCLUSION Both AGE and DM favor VSMC differentiation towards the osteogenic phenotype and this effect can be prevented by metformin.
Collapse
Affiliation(s)
- María Silvina Molinuevo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina
| | - Ana María Cortizo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina.
| | - Claudia Sedlinsky
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina.
| |
Collapse
|
4
|
Hu J, Han J, Jin M, Jin J, Zhu J. Effects of metformin on bone mineral density and bone turnover markers: a systematic review and meta-analysis. BMJ Open 2023; 13:e072904. [PMID: 37355276 PMCID: PMC10314630 DOI: 10.1136/bmjopen-2023-072904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023] Open
Abstract
OBJECTIVES Metformin is associated with osteoblastogenesis and osteoclastogenesis. This study aims to investigate the impacts of metformin therapy on bone mineral density (BMD) and bone turnover markers. DESIGN Systematic review and meta-analysis of randomised controlled trials. METHODS Searches were carried out in PubMed, EMBASE, Web of science, Cochrane library, ClinicalTrials.gov from database inception to 26 September 2022. Two review authors assessed trial eligibility in accordance with established inclusion criteria. The risk of bias was assessed using the Cochrane Risk of Bias tool (RoB V.2.0). Data analysis was conducted with Stata Statistical Software V.16.0 and Review Manager Software V.5.3. RESULTS A total of 15 studies with 3394 participants were identified for the present meta-analysis. Our pooled results indicated that metformin had no statistically significant effects on BMD at lumbar spine (SMD=-0.05, 95% CI=-0.19 to 0.09, p=0.47, participants=810; studies=7), at femoral (MD=-0.01 g/cm2, 95% CI=-0.04 to 0.01 g/cm2, p=0.25, participants=601; studies=3) and at hip (MD=0.01 g/cm2, 95% CI=-0.02 to 0.03 g/cm2, p=0.56, participants=634; studies=4). Metformin did not lead to significant change in osteocalcin, osteoprotegerin and bone alkaline phosphatase. Metformin induced decreases in N-terminal propeptide of type I procollagen (MD=-6.09 µg/L, 95% CI=-9.38 to -2.81 µg/L, p=0.0003, participants=2316; studies=7) and C-terminal telopeptide of type I collagen (MD=-55.80 ng/L, 95% CI=-97.33 to -14.26 ng/L, p=0.008, participants=2325; studies=7). CONCLUSION This meta-analysis indicated that metformin had no significant effect on BMD. Metformin decreased some bone turnover markers as N-terminal propeptide of type I procollagen and C-terminal telopeptide of type I collagen. But the outcomes should be interpreted with caution due to several limitations.
Collapse
Affiliation(s)
- Jinhua Hu
- Department of Pharmacy, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, Shanghai, China
| | - Jingjie Han
- Department of Pharmacy, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, Shanghai, China
| | - Min Jin
- Department of Pharmacy, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, Shanghai, China
| | - Jing Jin
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, Shanghai, China
| | - Jialei Zhu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, Shanghai, China
| |
Collapse
|
5
|
A blast from the past: To tame time with metformin. Mech Ageing Dev 2022; 208:111743. [PMID: 36279989 DOI: 10.1016/j.mad.2022.111743] [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: 09/24/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
The strong evidence of metformin use in subjects affected by type 2 diabetes (T2DM) on health outcomes, together with data from pre-clinical studies, has led the gerontological research to study the therapeutic potential of such a drug as a slow-aging strategy. However, despite clinical use for over fifty years as an anti-diabetic drug, the mechanisms of action beyond glycemic control remain unclear. In this review, we have deeply examined the literature, doing a narrative review from the metformin story, through mechanisms of action to slow down aging potential, from lower organisms to humans. Based on the available evidence, we conclude that metformin, as shown in lower organisms and mice, may be effective in humans' longevity. A complete analysis and follow-up of ongoing clinical trials may provide more definitive answers as to whether metformin should be promoted beyond its use to treat T2DM as a drug that enhances both healthspan and lifespan.
Collapse
|
6
|
Huang X, Li S, Lu W, Xiong L. Metformin activates Wnt/β-catenin for the treatment of diabetic osteoporosis. BMC Endocr Disord 2022; 22:189. [PMID: 35869471 PMCID: PMC9306077 DOI: 10.1186/s12902-022-01103-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND With the deepening of social aging, the incidence rate of osteoporosis and diabetes continues to rise. More and more clinical studies show that diabetes is highly correlated with osteoporosis. Diabetes osteoporosis is considered as a metabolic bone disease of diabetes patients. This study aims to explore the role and mechanism of metformin (Met) in diabetic osteoporosis. METHODS Mouse MC3T3-E1 cells were treated with Met (0.5 mM) and exposed to high glucose (HG, 35 mM). The cells were cultured in an osteogenic medium for osteogenic differentiation, and the cell proliferation ability was determined using Cell Counting Kit-8; Alkaline phosphatase (ALP) activity detection and alizarin red staining were utilized to evaluate the effect of Met on MC3T3-E1 osteogenic differentiation. Western blot was used to detect the expressions of osteogenesis-related proteins (Runx2 and OCN) as well as Wnt/β-catenin signaling pathway-related proteins in MC3T3-E1 cells. RESULTS HG inhibited proliferation and calcification of MC3T3-E1 cells, down-regulated ALP activity, and the expression of Runx2 and OCN in MC3T3-E1 cells. Meanwhile, the activity of the Wnt/β-catenin signaling pathway was inhibited. Met treatment was found to significantly stimulate the proliferation and calcification of MC3T3-E1 cells under HG conditions, as well as increase the ALP activity and the protein expression level of Runx2 and OCN in the cells. As a result, osteogenic differentiation was promoted and osteoporosis was alleviated. Apart from this, Met also increased the protein expression level of Wnt1, β-catenin, and C-myc to activate the Wnt/β-catenin signaling pathway. CONCLUSION Met can stimulate the proliferation and osteogenic differentiation of MC3T3-E1 cells under HG conditions. Met may also treat diabetic osteoporosis through Wnt/β-catenin activation.
Collapse
Affiliation(s)
- Xiaopeng Huang
- Department of Orthopedics, Jiangxi Province Hospital of Integrated Chinese & Western Medicine, Nanchang, 330003, China
| | - Siyun Li
- Department of Orthopedics, Jiangxi Province Hospital of Integrated Chinese & Western Medicine, Nanchang, 330003, China
| | - Wenjie Lu
- Department of Orthopedics, Jiangxi Province Hospital of Integrated Chinese & Western Medicine, Nanchang, 330003, China
| | - Longjiang Xiong
- Department of Orthopedics, Jiangxi Province Hospital of Integrated Chinese & Western Medicine, Nanchang, 330003, China.
| |
Collapse
|
7
|
Effects of Metformin on Bone Mineral Density and Adiposity-Associated Pathways in Animal Models with Type 2 Diabetes Mellitus: A Systematic Review. J Clin Med 2022; 11:jcm11144193. [PMID: 35887957 PMCID: PMC9323116 DOI: 10.3390/jcm11144193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/02/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Recently, there have been investigations on metformin (Met) as a potential treatment for bone diseases such as osteoporosis, as researchers have outlined that type 2 diabetes mellitus (T2DM) poses an increased risk of fractures. Hence, this systematic review was conducted according to the 2020 PRISMA guidelines to evaluate the evidence that supports the bone-protective effects of metformin on male animal models with T2DM. Five databases—Google Scholar, PubMed, Wiley Online Library, SCOPUS, and ScienceDirect—were used to search for original randomized controlled trials published in English with relevant keywords. The search identified 18 articles that matched the inclusion criteria and illustrated the effects of Met on bone. This study demonstrates that Met improved bone density and reduced the effects of T2DM on adiposity formation in the animal models. Further research is needed to pinpoint the optimal dosage of Met required to exhibit these therapeutic effects.
Collapse
|
8
|
Song Y, Wu Z, Zhao P. The Function of Metformin in Aging-Related Musculoskeletal Disorders. Front Pharmacol 2022; 13:865524. [PMID: 35392559 PMCID: PMC8982084 DOI: 10.3389/fphar.2022.865524] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022] Open
Abstract
Metformin is a widely accepted first-line hypoglycemic agent in current clinical practice, and it has been applied to the clinic for more than 60 years. Recently, researchers have identified that metformin not only has an efficient capacity to lower glucose but also exerts anti-aging effects by regulating intracellular signaling molecules. With the accelerating aging process and mankind’s desire for a long and healthy life, studies on aging have witnessed an unprecedented boom. Osteoporosis, sarcopenia, degenerative osteoarthropathy, and frailty are age-related diseases of the musculoskeletal system. The decline in motor function is a problem that many elderly people have to face, and in serious cases, they may even fail to self-care, and their quality of life will be seriously reduced. Therefore, exploring potential treatments to effectively prevent or delay the progression of aging-related diseases is essential to promote healthy aging. In this review, we first briefly describe the origin of metformin and the aging of the movement system, and next review the evidence associated with its ability to extend lifespan. Furthermore, we discuss the mechanisms related to the modulation of aging in the musculoskeletal system by metformin, mainly its contribution to bone homeostasis, muscle aging, and joint degeneration. Finally, we analyze the protective benefits of metformin in aging-related diseases of the musculoskeletal system.
Collapse
Affiliation(s)
- Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Teissier T, Temkin V, Pollak RD, Cox LS. Crosstalk Between Senescent Bone Cells and the Bone Tissue Microenvironment Influences Bone Fragility During Chronological Age and in Diabetes. Front Physiol 2022; 13:812157. [PMID: 35388291 PMCID: PMC8978545 DOI: 10.3389/fphys.2022.812157] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/27/2022] [Indexed: 01/10/2023] Open
Abstract
Bone is a complex organ serving roles in skeletal support and movement, and is a source of blood cells including adaptive and innate immune cells. Structural and functional integrity is maintained through a balance between bone synthesis and bone degradation, dependent in part on mechanical loading but also on signaling and influences of the tissue microenvironment. Bone structure and the extracellular bone milieu change with age, predisposing to osteoporosis and increased fracture risk, and this is exacerbated in patients with diabetes. Such changes can include loss of bone mineral density, deterioration in micro-architecture, as well as decreased bone flexibility, through alteration of proteinaceous bone support structures, and accumulation of senescent cells. Senescence is a state of proliferation arrest accompanied by marked morphological and metabolic changes. It is driven by cellular stress and serves an important acute tumor suppressive mechanism when followed by immune-mediated senescent cell clearance. However, aging and pathological conditions including diabetes are associated with accumulation of senescent cells that generate a pro-inflammatory and tissue-destructive secretome (the SASP). The SASP impinges on the tissue microenvironment with detrimental local and systemic consequences; senescent cells are thought to contribute to the multimorbidity associated with advanced chronological age. Here, we assess factors that promote bone fragility, in the context both of chronological aging and accelerated aging in progeroid syndromes and in diabetes, including senescence-dependent alterations in the bone tissue microenvironment, and glycation changes to the tissue microenvironment that stimulate RAGE signaling, a process that is accelerated in diabetic patients. Finally, we discuss therapeutic interventions targeting RAGE signaling and cell senescence that show promise in improving bone health in older people and those living with diabetes.
Collapse
Affiliation(s)
- Thibault Teissier
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Vladislav Temkin
- Division of Medicine, Department of Endocrinology and Metabolism, The Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rivka Dresner Pollak
- Division of Medicine, Department of Endocrinology and Metabolism, The Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lynne S. Cox
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Patients with diabetes mellitus (DM) are at increased risk of developing osteopathogenesis and skeletal fragility. The role of the gut microbiota in both DM and osteopathy is not fully explored and may be involved in the pathology of both diseases. RECENT FINDINGS Gut microbiota alterations have been observed in DM and osteopathogenic disorders as compared with healthy controls, such as significantly lower abundance of Prevotella and higher abundance of Lactobacillus, with a diminished bacterial diversity. Other overlapping gastro-intestinal features include the loss of intestinal barrier function with translocation of bacterial metabolites to the blood stream, induction of immunological deficits and changes in hormonal and endocrinal signalling, which may lead to the development of diabetic osteopathy. Signalling pathways involved in both DM and osteopathy are affected by gut bacteria and their metabolites. Future studies should focus on gut microbiota involvement in both diseases.
Collapse
Affiliation(s)
- Julie Kristine Knudsen
- Centre for Clinical Research, North Denmark Regional Hospital, Bispensgade 37, 9800, Hjørring, Denmark.
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Denmark.
| | - Peter Leutscher
- Centre for Clinical Research, North Denmark Regional Hospital, Bispensgade 37, 9800, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Denmark
- Steno Diabetes Center North Jutland, Mølleparkvej 4, Aalborg, Denmark
| | - Suzette Sørensen
- Centre for Clinical Research, North Denmark Regional Hospital, Bispensgade 37, 9800, Hjørring, Denmark
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Denmark
- Steno Diabetes Center North Jutland, Mølleparkvej 4, Aalborg, Denmark
| |
Collapse
|
11
|
Palui R, Pramanik S, Mondal S, Ray S. Critical review of bone health, fracture risk and management of bone fragility in diabetes mellitus. World J Diabetes 2021; 12:706-729. [PMID: 34168723 PMCID: PMC8192255 DOI: 10.4239/wjd.v12.i6.706] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/08/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
The risk of fracture is increased in both type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). However, in contrast to the former, patients with T2DM usually possess higher bone mineral density. Thus, there is a considerable difference in the pathophysiological basis of poor bone health between the two types of diabetes. Impaired bone strength due to poor bone microarchitecture and low bone turnover along with increased risk of fall are among the major factors behind elevated fracture risk. Moreover, some antidiabetic medications further enhance the fragility of the bone. On the other hand, antiosteoporosis medications can affect the glucose homeostasis in these patients. It is also difficult to predict the fracture risk in these patients because conventional tools such as bone mineral density and Fracture Risk Assessment Tool score assessment can underestimate the risk. Evidence-based recommendations for risk evaluation and management of poor bone health in diabetes are sparse in the literature. With the advancement in imaging technology, newer modalities are available to evaluate the bone quality and risk assessment in patients with diabetes. The purpose of this review is to explore the pathophysiology behind poor bone health in diabetic patients. Approach to the fracture risk evaluation in both T1DM and T2DM as well as the pragmatic use and efficacy of the available treatment options have been discussed in depth.
Collapse
Affiliation(s)
- Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur 713212, West Bengal, India
| | - Subhodip Pramanik
- Department of Endocrinology, Neotia Getwel Healthcare Centre, Siliguri 734010, West Bengal, India
| | - Sunetra Mondal
- Department of Endocrinology, Institute of Post Graduate Medical Education and Research (IPGMER), Kolkata 700020, West Bengal, India
| | - Sayantan Ray
- Department of Endocrinology, Medica Superspeciality Hospital and Medica Clinic, Kolkata 700099, West Bengal, India
- Department of Endocrinology, Jagannath Gupta Institute of Medical Sciences and Hospital, Kolkata 700137, West Bengal, India
| |
Collapse
|
12
|
Shaik AR, Singh P, Shaik C, Kohli S, Vohora D, Ferrari SL. Metformin: Is It the Well Wisher of Bone Beyond Glycemic Control in Diabetes Mellitus? Calcif Tissue Int 2021; 108:693-707. [PMID: 33797562 DOI: 10.1007/s00223-021-00805-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/05/2021] [Indexed: 12/18/2022]
Abstract
Both diabetes mellitus and osteoporosis constitute a notable burden in terms of quality of life and healthcare costs. Diabetes mellitus affecting the skeletal system has been gaining attention in recent years and is now getting recognized as yet another complication of the disease, known as diabetic bone disease. As this condition with weaker bone strength increases fracture risk and reduces the quality of life, so much attention is being paid to investigate the molecular pathways through which both diabetes and its therapy are affecting bone metabolism. Out of many therapeutic agents currently available for managing diabetes mellitus, metformin is one of the most widely accepted first choices worldwide. The purpose of this review is to describe the effects of biguanide-metformin on bone metabolism in type 2 diabetes mellitus including its plausible mechanisms of action on the skeleton. In vitro studies suggest that metformin directly stimulates osteoblasts differentiation and may inhibit osteoclastogenesis by increasing osteoprotegerin expression, both through activation of the AMPK signaling pathway. Several studies in both preclinical and clinical settings report the favorable effects of metformin on bone microarchitecture, bone mineral density, bone turnover markers, and fracture risk. However, animal studies were not specific in terms of the diabetic models used and clinical studies were associated with several confounders. The review highlights some of these limitations and provide future recommendations for research in this area which is necessary to better understand the role of metformin on skeletal outcomes in diabetes.
Collapse
Affiliation(s)
- Abdul Rahaman Shaik
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Prabhjeet Singh
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Chandini Shaik
- Department of Pharmaceutical Analysis, University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Nagarjuna Nagar, Guntur, Andhra Pradesh, 522510, India
| | - Sunil Kohli
- Department of Medicine, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India.
| | - Serge Livio Ferrari
- Service and Laboratory of Bone Diseases, Department of Medicine, Geneva University Hospital and Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
13
|
Jiang LL, Liu L. Effect of metformin on stem cells: Molecular mechanism and clinical prospect. World J Stem Cells 2020; 12:1455-1473. [PMID: 33505595 PMCID: PMC7789120 DOI: 10.4252/wjsc.v12.i12.1455] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/28/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023] Open
Abstract
Metformin is a first-line medication for type II diabetes. Numerous studies have shown that metformin not only has hypoglycemic effects, but also modulates many physiological and pathological processes ranging from aging and cancer to fracture healing. During these different physiological activities and pathological changes, stem cells usually play a core role. Thus, many studies have investigated the effects of metformin on stem cells. Metformin affects cell differentiation and has promising applications in stem cell medicine. It exerts anti-aging effects and can be applied to gerontology and regenerative medicine. The potential anti-cancer stem cell effect of metformin indicates that it can be an adjuvant therapy for cancers. Furthermore, metformin has beneficial effects against many other diseases including cardiovascular and autoimmune diseases. In this review, we summarize the effects of metformin on stem cells and provide an overview of its molecular mechanisms and clinical prospects.
Collapse
Affiliation(s)
- Lin-Li Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lei Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China.
| |
Collapse
|
14
|
Aung M, Amin S, Gulraiz A, Gandhi FR, Pena Escobar JA, Malik BH. The Future of Metformin in the Prevention of Diabetes-Related Osteoporosis. Cureus 2020; 12:e10412. [PMID: 33062529 PMCID: PMC7550241 DOI: 10.7759/cureus.10412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As a worldwide aging population is on the rise, osteoporosis (OS) is becoming a global health burden. Therefore, many researchers and health authorities are looking into the potential prevention and treatment of OS. Although previously regarded as two separate pathological processes, diabetes (DM) and OS are now regarded as two conditions that can occur together. It is now believed that OS can develop as a complication of DM. This relationship is further evidenced through a reduction in bone mineral density in type-1 diabetes with a resulting increased risk of fracture. Although bone mineral density in type-2 diabetes mellitus is normal or increased, there is also increased fragility due to decreased bone quality. These abnormal bone qualities tend to occur through the production of reduced bone microvasculature and advanced glycation end product, AGE. Interestingly, one of the most common treatments for DM, metformin (MF), shows a promising result on the protection of diabetes and non-diabetes related bone turnover. It is believed that MF modulates its effect through the adenosine monophosphate-activated protein kinase (AMPK) pathway. Recent data regarded AMPK as a vital mediator of homeostasis. It is involved not only in glucose metabolism but also in osteogenesis. AMPK can directly influence the production of mature and good quality bone by decreasing osteoclasts, increasing osteoblast formation, and enhancing bone mineral deposition. As an activator of AMPK, MF also upregulates osteogenesis. Furthermore, MF can influence osteogenesis through a non-AMPK pathway, such as the fructose 1-6 phosphatase pathway, by reducing glucose levels. While already recognized as a safe and effective treatment for DM, this article discusses whether MF can be used for the prevention and treatment of OS.
Collapse
Affiliation(s)
- Myat Aung
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Emergency Department, Poole Hospital, Poole, GBR
| | - Saba Amin
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Azouba Gulraiz
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Fenil R Gandhi
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Julio A Pena Escobar
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bilal Haider Malik
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
15
|
Lin J, Xu R, Shen X, Jiang H, Du S. Metformin promotes the osseointegration of titanium implants under osteoporotic conditions by regulating BMSCs autophagy, and osteogenic differentiation. Biochem Biophys Res Commun 2020; 531:228-235. [PMID: 32800340 DOI: 10.1016/j.bbrc.2020.06.146] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/26/2020] [Indexed: 02/08/2023]
Abstract
Osteoporosis is a common bone disorder with adverse effects on oral osseointegration, and the effects of metformin on bone metabolism have received increasing attention. The aim of the present study was to test the hypothesis that metformin promoted osteogenesis of bone mesenchymal stem cells (BMSCs) and osseointegration of titanium implants. BMSCs were treated with metformin to assess autophagic capacity, reactive oxygen species (ROS) production, anti-aging ability, and osteogenic differentiation. To determine its potential application in peri-implant of the maxilla, metformin was injected around the implant each day, immediately after the implant was embedded into the tooth socket. The results showed that metformin increased the autophagic capacity and decreased ROS production of osteoporotic BMSCs under hypoxia and serum deprivation (H/SD) culturing conditions. Metformin treatment significantly enhanced stemness properties and mineralized nodule formation, and increased the expression of osteogenic markers, including runt related transcription factor 2 (Runx2), osteocalcin (OCN), and alkaline phosphatase (ALP). Moreover, metformin substantially accelerated the formation of new bone, ameliorated the bone microarchitecture and promoted osseointegration of the dental implant. Collectively, metformin induces an osteogenic effect around the implant. Considering the widespread use of metformin, the results of the present study might promote a novel understanding of the positive effects of local metformin delivery on alveolar ridge defect, and have potential clinical application for the acceleration of osseointegration.
Collapse
Affiliation(s)
- Jiating Lin
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Nanjing Medical University, Nanjing, 210029, China; Department of Stomatology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241000, China
| | - Rongyao Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiang Shen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hongbing Jiang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Shuhu Du
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
16
|
The Use of Metformin to Increase the Human Healthspan. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1260:319-332. [PMID: 32304040 DOI: 10.1007/978-3-030-42667-5_13] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metformin is a safe, effective and useful drug for glucose management in patients with diabetes. However in recent years, more attention has been paid to the possibility of using metformin as an anti-aging drug. It was shown to significantly increase the lifespan in some model organisms and delay the onset of age-associated declines. The current review summarizes advances in clinical research on the potential role of metformin in the field of lifespan and healthspan extension. Growing amounts of evidence from clinical trials suggest that metformin can effectively reduce the risk of many age-related diseases and conditions, including cardiometabolic disorders, neurodegeneration, chronic inflammation and frailty. Metformin also holds promise as a drug that could be repurposed for chemoprevention or adjuvant therapy for certain types of cancer. Moreover, metformin induces autophagy by activation of AMPK and can thus be potentially used to promote heathspan by hormesis-like mechanisms. Although long-term intake of metformin is associated with low risk of adverse events, well-designed clinical trials are still required to uncover the potential use of this drug as a geroprotector.
Collapse
|
17
|
Jiating L, Buyun J, Yinchang Z. Role of Metformin on Osteoblast Differentiation in Type 2 Diabetes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9203934. [PMID: 31886264 PMCID: PMC6899291 DOI: 10.1155/2019/9203934] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022]
Abstract
Metformin, an effective hypoglycemic, can modulate different points of malignant mass, polycystic ovary syndrome (PCOS), cardiovascular diseases, tuberculosis, and nerve regeneration. Recently, the effect of metformin on bone metabolism has been analyzed. Metformin relies on organic cation transporters (OCT1), a polyspecific cell membrane of the solute carrier 22A (SLC22A) gene family, to facilitate its intracellular uptake and action on complex I of the respiratory chain of mitochondria. These changes activate the cellular energy sensor AMP-activated protein kinase (AMPK). Thus, the increased cellular AMP/ATP ratio causes a dramatic and progressive activation of insulin and lysosomes, resulting in a decrease in intracellular glucose level, which promotes osteoblast proliferation and differentiation. AMPK also phosphorylates runt-related transcription factor 2 (Runx2) at S118, the lineage-specific transcriptional regulators, to promote osteogenesis. Metformin phosphorylates extracellular signal-regulated kinase (ERK), stimulates endothelial and inducible nitric oxide synthases (e/iNOS), inhibits the GSK3β/Wnt/β-catenin pathway, and promotes osteogenic differentiation of osteoblasts. The effect of metformin on hyperglycemia decreases intracellular reactive oxygen species (ROS) and advanced glycation end-products (AGEs) in collagen, and reduced serum levels of insulin-like growth factors (IGF-1) were beneficial for bone formation. Metformin has a certain effect on microangiopathy and anti-inflammation, which can induce osteoporosis, activate the activity of osteoclasts, and inhibit osteoblast activity, and has demonstrated extensive alteration in bone and mineral metabolism. The aim of this review was to elucidate the mechanisms of metformin on osteoblasts in insulin-deficient diabetes.
Collapse
Affiliation(s)
- Lin Jiating
- Department of Stomatology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province 241000, China
| | - Ji Buyun
- Department of Stomatology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province 241000, China
| | - Zhang Yinchang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province 241000, China
| |
Collapse
|
18
|
Zhao J, Li Y, Zhang H, Shi D, Li Q, Meng Y, Zuo L. Preventative effects of metformin on glucocorticoid-induced osteoporosis in rats. J Bone Miner Metab 2019; 37:805-814. [PMID: 30706148 DOI: 10.1007/s00774-019-00989-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 01/19/2019] [Indexed: 02/05/2023]
Abstract
This study evaluated the preventative effects of metformin (Met) on glucocorticoid (GC)-induced osteoporosis in a rat model, compared with alendronate (Aln). Twenty-eight 3-month-old female Sprague-Dawley rats were randomly assigned into four groups: normal control (Ctr), methylprednisolone (MP, 13 mg/kg/day, sc, 5 days per week), MP plus Aln orally (1 mg/kg/day), and MP plus Met orally (200 mg/kg/day). After 9 weeks, serum bone metabolic biochemistry, bone densitometry and histomorphometry were performed. The GC-induced osteoporosis model was characterized by decreased osteocalcin, increased tartrate-resistant acid phosphatase-5b (TRAP-5b), and decreased bone mineral density (BMD) in the femur and fifth lumbar vertebra (L5). Histomorphometrically, MP significantly decreased trabecular bone volume, decreased bone formation and increased bone resorption in proximal metaphysis, compared with the controls. Aln and Met increased the BMDs of femur (0.305 ± 0.011 vs. 0.280 ± 0.012, P < 0.05; 0.304 ± 0.019 vs. 0.280 ± 0.012, P < 0.05) and L5 (0.399 ± 0.029 vs. 0.358 ± 0.022, P < 0.05; 0.397 ± 0.022 vs. 0.358 ± 0.022, P < 0.05), compared with the model group. Met increased osteocalcin and decreased TRAP-5b, but Aln only decreased TRAP-5b, compared with model group. In histomorphometry of tibial proximal metaphysis, Aln and Met increased trabecular bone volume (39.21 ± 2.46 vs. 30.98 ± 5.83, P < 0.05; 38.97 ± 5.56 vs. 30.98 ± 5.83, P < 0.05), while Met increased the bone formation dynamic parameters and decreased bone resorption dynamic parameters, but Aln just decreased bone resorption dynamic parameters, compared with model group significantly. These findings suggest that metformin prevents GC-induced bone loss by suppressing bone resorption and stimulating bone formation in trabecular bone. The action mode of metformin was different from alendronate, which only suppressed bone resorption.
Collapse
Affiliation(s)
- Jianrong Zhao
- Department of Nephrology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Yingbin Li
- Guangdong Legend Pharmaceutical Technology Co., Ltd., Jiangmen, People's Republic of China
| | - Hao Zhang
- School of Dentistry, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Dongying Shi
- Department of Nephrology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Qingnan Li
- Guangdong Laboratory Animals Monitoring Institute and Key Laboratory of Guangdong Laboratory Animals, Guangzhou, People's Republic of China
- The Center for New Drug Function Research, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Yan Meng
- Department of Nephrology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| | - Li Zuo
- Department of Nephrology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, People's Republic of China.
| |
Collapse
|
19
|
Asadipooya K, Uy EM. Advanced Glycation End Products (AGEs), Receptor for AGEs, Diabetes, and Bone: Review of the Literature. J Endocr Soc 2019; 3:1799-1818. [PMID: 31528827 PMCID: PMC6734192 DOI: 10.1210/js.2019-00160] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022] Open
Abstract
Diabetes compromises bone cell metabolism and function, resulting in increased risk of fragility fracture. Advanced glycation end products (AGEs) interact with the receptor for AGEs (RAGE) and can make a meaningful contribution to bone cell metabolism and/or alter function. Searches in PubMed using the key words "advanced glycation end-product," "RAGE," "sRAGE," "bone," and "diabetes" were made to explain some of the clinical outcomes of diabetes in bone metabolism through the AGE-RAGE signaling pathway. All published clinical studies were included in tables. The AGE-RAGE signaling pathway participates in diabetic complications, including diabetic osteopathy. Some clinical results in diabetic patients, such as reduced bone density, suppressed bone turnover markers, and bone quality impairment, could be potentially due to AGE-RAGE signaling consequences. However, the AGE-RAGE signaling pathway has some helpful roles in the bone, including an increase in osteogenic function. Soluble RAGE (sRAGE), as a ligand decoy, may increase in either conditions of RAGE production or destruction, and then it cannot always reflect the AGE-RAGE signaling. Recombinant sRAGE can block the AGE-RAGE signaling pathway but is associated with some limitations, such as accessibility to AGEs, an increase in other RAGE ligands, and a long half-life (24 hours), which is associated with losing the beneficial effect of AGE/RAGE. As a result, sRAGE is not a helpful marker to assess activity of the RAGE signaling pathway. The recombinant sRAGE cannot be translated into clinical practice due to its limitations.
Collapse
Affiliation(s)
- Kamyar Asadipooya
- Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington, Kentucky
| | - Edilfavia Mae Uy
- Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
20
|
Abstract
Aging, as a physiological process mediated by numerous regulatory pathways and transcription factors, is manifested by continuous progressive functional decline and increasing risk of chronic diseases. There is an increasing interest to identify pharmacological agents for treatment and prevention of age-related disease in humans. Animal models play an important role in identification and testing of anti-aging compounds; this step is crucial before the drug will enter human clinical trial or will be introduced to human medicine. One of the main goals of animal studies is better understanding of mechanistic targets, therapeutic implications and side-effects of the drug, which may be later translated into humans. In this chapter, we summarized the effects of different drugs reported to extend the lifespan in model organisms from round worms to rodents. Resveratrol, rapamycin, metformin and aspirin, showing effectiveness in model organism life- and healthspan extension mainly target the master regulators of aging such as mTOR, FOXO and PGC1α, affecting autophagy, inflammation and oxidative stress. In humans, these drugs were demonstrated to reduce inflammation, prevent CVD, and slow down the functional decline in certain organs. Additionally, potential anti-aging pharmacologic agents inhibit cancerogenesis, interfering with certain aspects of cell metabolism, proliferation, angioneogenesis and apoptosis.
Collapse
|
21
|
Kalaitzoglou E, Fowlkes JL, Popescu I, Thrailkill KM. Diabetes pharmacotherapy and effects on the musculoskeletal system. Diabetes Metab Res Rev 2019; 35:e3100. [PMID: 30467957 PMCID: PMC6358500 DOI: 10.1002/dmrr.3100] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/14/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Persons with type 1 or type 2 diabetes have a significantly higher fracture risk than age-matched persons without diabetes, attributed to disease-specific deficits in the microarchitecture and material properties of bone tissue. Therefore, independent effects of diabetes drugs on skeletal integrity are vitally important. Studies of incretin-based therapies have shown divergent effects of different agents on fracture risk, including detrimental, beneficial, and neutral effects. The sulfonylurea class of drugs, owing to its hypoglycemic potential, is thought to amplify the risk of fall-related fractures, particularly in the elderly. Other agents such as the biguanides may, in fact, be osteo-anabolic. In contrast, despite similarly expected anabolic properties of insulin, data suggests that insulin pharmacotherapy itself, particularly in type 2 diabetes, may be a risk factor for fracture, negatively associated with determinants of bone quality and bone strength. Finally, sodium-dependent glucose co-transporter 2 inhibitors have been associated with an increased risk of atypical fractures in select populations, and possibly with an increase in lower extremity amputation with specific SGLT2I drugs. The role of skeletal muscle, as a potential mediator and determinant of bone quality, is also a relevant area of exploration. Currently, data regarding the impact of glucose lowering medications on diabetes-related muscle atrophy is more limited, although preclinical studies suggest that various hypoglycemic agents may have either aggravating (sulfonylureas, glinides) or repairing (thiazolidinediones, biguanides, incretins) effects on skeletal muscle atrophy, thereby influencing bone quality. Hence, the therapeutic efficacy of each hypoglycemic agent must also be evaluated in light of its impact, alone or in combination, on musculoskeletal health, when determining an individualized treatment approach. Moreover, the effect of newer medications (potentially seeking expanded clinical indication into the pediatric age range) on the growing skeleton is largely unknown. Herein, we review the available literature regarding effects of diabetes pharmacotherapy, by drug class and/or by clinical indication, on the musculoskeletal health of persons with diabetes.
Collapse
Affiliation(s)
- Evangelia Kalaitzoglou
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - John L Fowlkes
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Iuliana Popescu
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Kathryn M Thrailkill
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
22
|
Álvarez-Lloret P, Fernández JM, Molinuevo MS, Lino AB, Ferretti JL, Capozza RF, Cortizo AM, McCarthy AD. Multi-Scale Approach for the Evaluation of Bone Mineralization in Strontium Ranelate-Treated Diabetic Rats. Biol Trace Elem Res 2018; 186:457-466. [PMID: 29623650 DOI: 10.1007/s12011-018-1322-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/21/2018] [Indexed: 11/27/2022]
Abstract
Long-term diabetes mellitus can induce osteopenia and osteoporosis, an increase in the incidence of low-stress fractures, and/or delayed fracture healing. Strontium ranelate (SrR) is a dual-action anti-osteoporotic agent whose use in individuals with diabetic osteopathy has not been adequately evaluated. In this study, we studied the effects of an oral treatment with SrR and/or experimental diabetes on bone composition and biomechanics. Young male Wistar rats (half non-diabetic, half with streptozotocin/nicotinamide-induced diabetes) were either untreated or orally administered 625 mg/kg/day of SrR for 6 weeks. After sacrifice, femora from all animals were evaluated by a multi-scale approach (X-ray diffraction, Fourier transform infrared spectroscopy, inductively coupled plasma optical-emission spectrometry, static histomorphometry, pQCT, and mechanical testing) to determine chemical, crystalline, and biomechanical properties. Untreated diabetic animals (versus untreated non-diabetic) showed a decrease in femoral mineral carbonate content, in cortical thickness and BMC, in trabecular osteocyte density, in maximum load supported at rupture and at yield point, and in overall toughness at mid-shaft. Treatment of diabetic animals with SrR further affected several parameters of bone (some already impaired by diabetes): crystallinity index (indicating less mature apatite crystals); trabecular area, BMC, and vBMD; maximum load at yield point; and structural elastic rigidity. However, SrR was also able to prevent the diabetes-induced decreases in trabecular osteocyte density (completely) and in bone ultimate strength at rupture (partially). Our results indicate that SrR treatment can partially but significantly prevent some bone structural mechanical properties as previously affected by diabetes, but not others (which may even be worsened).
Collapse
Affiliation(s)
- Pedro Álvarez-Lloret
- Departament of Geology, University of Oviedo, C/Jesús Arias de Velasco, s/n, 33005, Oviedo, Spain
| | - Juan Manuel Fernández
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, 1900, La Plata, Argentina
| | - María Silvina Molinuevo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, 1900, La Plata, Argentina
| | - Agustina Berenice Lino
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, 1900, La Plata, Argentina
| | - José Luis Ferretti
- Centro de Estudios del Metabolismo Fosfocálcico (CeMFoC), Facultad de Medicina, Universidad Nacional de Rosario, 2000, Rosario, Argentina
| | - Ricardo Francisco Capozza
- Centro de Estudios del Metabolismo Fosfocálcico (CeMFoC), Facultad de Medicina, Universidad Nacional de Rosario, 2000, Rosario, Argentina
| | - Ana María Cortizo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, 1900, La Plata, Argentina
| | - Antonio Desmond McCarthy
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, 1900, La Plata, Argentina.
| |
Collapse
|
23
|
Metformin; an old antidiabetic drug with new potentials in bone disorders. Biomed Pharmacother 2018; 109:1593-1601. [PMID: 30551413 DOI: 10.1016/j.biopha.2018.11.032] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 01/15/2023] Open
Abstract
The prevalence of diabetes mellitus especially type 2 diabetes mellitus is increasing all over the world. In addition to cardiomyopathy and nephropathy, diabetics are at higher risk of mortality and morbidity due to greater risk of bone fractures and skeletal abnormalities. Patients with diabetes mellitus have lower bone quality in comparison to their non-diabetic counterparts mainly because of hyperglycemia, toxic effects of advanced glycosylation end-products (AGEs) on bone tissue, and impaired bone microvascular system. AGEs may also contribute to the development of osteoarthritis further to osteoporosis. Therefore, glycemic control in diabetic patients is vital for bone health. Metformin, a widely used antidiabetic drug, has been shown to improve bone quality and decrease the risk of fractures in patients with diabetes in addition to glycemic control and improving insulin sensitivity. AMP activated protein kinase (AMPK), the key molecule in metformin antidiabetic mechanism of action, is also effective in signaling pathways involved in bone physiology. This review, discusses the molecules linking diabetes and bone turnover, role of AMPK in bone metabolism, and the effect of metformin as an activator of AMPK on bone disorders and malignancies.
Collapse
|
24
|
Zheng L, Shen X, Ye J, Xie Y, Yan S. Metformin alleviates hyperglycemia-induced apoptosis and differentiation suppression in osteoblasts through inhibiting the TLR4 signaling pathway. Life Sci 2018; 216:29-38. [PMID: 30414431 DOI: 10.1016/j.lfs.2018.11.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/27/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023]
Abstract
AIMS Metformin was found to protect against hyperglycemia-induced injury in osteoblasts, but the cellular mechanisms involved remain unclear. Therefore, the aim of this study was to determine the effect of metformin on hyperglycemia-induced apoptosis and differentiation suppression in osteoblasts and to explore its relationships with the TLR4 signaling pathway. MAIN METHODS A mouse osteoblast cell line, MC3T3-E1, and a diabetic rat model were used to survey the protective effects of metformin on hyperglycemia-induced injury. TLR4 expression was altered using small interfering (si)RNA and lentivirus-mediated TLR4 overexpression. LPS was used as a specific TLR4 activator, and CLI-095 was used as a TLR4 inhibitor. KEY FINDINGS Metformin improved osteoblast differentiation, reduced apoptosis in hyperglycemic osteoblasts, and inhibited TLR4, MyD88 and NF-κB expression in a dose-dependent manner. Down-regulating the expression or inhibiting the activity of TLR4 enhanced these protective effects of metformin on osteoblast differentiation, cell viability and cell apoptosis in hyperglycemic conditions, whereas up-regulating the expression or activating the activity of TLR4 had the opposite effects. Activating NF-κB suppressed the protective effects of metformin, while inhibiting NF-κB activity had the opposite effects. Metformin increased ALP and OCN secretion, enhanced BMP-2 expression, improved bone mineral density (BMD), and decreased TLR4, MyD88 and NF-κB levels in the femur tissues of diabetic rats. SIGNIFICANCE Taken together our experimentation support the hypothesis that metformin may alleviate hyperglycemia-induced apoptosis and differentiation suppression in osteoblasts by inhibiting the TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lifeng Zheng
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Ximei Shen
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China; Diabetes Research Institute of Fujian Province, Fuzhou 350005, Fujian, China
| | - Junjian Ye
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Yun Xie
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Sunjie Yan
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China; Diabetes Research Institute of Fujian Province, Fuzhou 350005, Fujian, China.
| |
Collapse
|
25
|
Hua Y, Bi R, Zhang Y, Xu L, Guo J, Li Y. Different bone sites-specific response to diabetes rat models: Bone density, histology and microarchitecture. PLoS One 2018; 13:e0205503. [PMID: 30346963 PMCID: PMC6197850 DOI: 10.1371/journal.pone.0205503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/26/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Diabetes mellitus (DM) is the most common metabolic disorder that is characterized by hyperglycemia, it can be categorized by T1DM and T2DM. T1DM is also reported to cause bone loss. However, most reports regarding this aspect of T1DM have only investigated a single site; a comparison of bone loss from different areas of the body is still lacking. METHODS Thirty-five 12-week-old Sprague Dawley® (SD) rats were separated to seven groups. Five rats were euthanized without any surgery at 0 weeks for histological examination and determination of baseline characteristics. In 15 of the rats, DM was induced via Streptozotocin (STZ)-injection, and they were separated to 3 groups (4 weeks, 8 weeks and 12 weeks after STZ-injection). The remaining 15 rats were used as the control group (4 weeks, 8 weeks and 12 weeks after saline-injection). We tested bone-mass loss at four skeletal sites, the tibia, the femur greater trochanter, the spine, and the mandibular bones using micro-computed tomography (CT) and histological tests. RESULTS Tibia was influenced the most obvious(BV/TV decreased by 27.3%, 52.5%, and 81.2% at 4 weeks, 8 weeks, and 12 weeks, respectively. p<0.05). In contrast, the other three sites were influenced to a lesser extent and bone loss became prominent at a later time point according to the histological and micro-CT tests(Femur: BV/TV did not decrease significantly at the first month or second month. However, and decreased by 49.4% at the third month, P<0.05. Mandible: the BV/TV only decreased by 6.5% at 1 month after STZ-injection. There was still a significant difference between the second and third months. The BV/TV decreased by 47.0% and 68.1% at 2 months and 3 months, respectively, (p<0.05) Spine: the BV/TV only decreased by 6.7%. However, significant change was observed in the spine at the second month and third month after STZ injection. The BV/TV decreased by 45.4% and 64.3%, respectively, p<0.05). CONCLUSION The results indicate that T1DM can severely influence the bone structure of the 4 skeletal sites. Further, areas with dense trabecular bones were influenced less and at a later time point in comparison to the tibial region. CLINICAL RELEVANCE Our research can serve as a guide to help increase the success rate of implant treatment, and help decrease the fracture risk in different bone types with greater accuracy.
Collapse
Affiliation(s)
- Yunwei Hua
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Luchen Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiaoyang Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- * E-mail:
| |
Collapse
|
26
|
Piskovatska V, Stefanyshyn N, Storey KB, Vaiserman AM, Lushchak O. Metformin as a geroprotector: experimental and clinical evidence. Biogerontology 2018; 20:33-48. [PMID: 30255224 DOI: 10.1007/s10522-018-9773-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Apart from being a safe, effective and globally affordable glucose-lowering agent for the treatment of diabetes, metformin has earned much credit in recent years as a potential anti-aging formula. It has been shown to significantly increase lifespan and delay the onset of age-associated decline in several experimental models. The current review summarizes advances in clinical research on the potential role of metformin in the field of geroprotection, highlighting findings from pre-clinical studies on known and putative mechanisms behind its beneficial properties. A growing body of evidence from clinical trials demonstrates that metformin can effectively reduce the risk of many age-related diseases and conditions, including cardiometabolic disorders, neurodegeneration, cancer, chronic inflammation, and frailty. Metformin also holds promise as a drug that could be repurposed for chemoprevention or adjuvant therapy for certain cancer types. Moreover, due to the ability of metformin to induce autophagy by activation of AMPK, it is regarded as a potential hormesis-inducing agent with healthspan-promoting and pro-longevity properties. Long-term intake of metformin is associated with low risk of adverse events; however, well-designed clinical trials are still warranted to enable potential use of this therapeutic agent as a geroprotector.
Collapse
Affiliation(s)
- Veronika Piskovatska
- Clinic for Heart Surgery, University Clinic of the Martin Luther University, Halle, Germany
| | - Nadiya Stefanyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | | | | | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine.
| |
Collapse
|
27
|
Kheniser KG, Polanco Santos CM, Kashyap SR. The effects of diabetes therapy on bone: A clinical perspective. J Diabetes Complications 2018; 32:713-719. [PMID: 29747995 DOI: 10.1016/j.jdiacomp.2018.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/21/2018] [Accepted: 04/14/2018] [Indexed: 12/16/2022]
Abstract
The effects of diabetes and diabetes therapy on bone are less known among clinicians. Traditionally, the emphasis of diabetes therapy has been on reducing cardiovascular risk by facilitating reductions in weight, blood pressure, blood sugar, systemic inflammation, and lipid levels. Now, with ample research demonstrating that patients with diabetes are more susceptible to bone fractures relative to controls, there has been a greater or renewed interest in studying the effects of diabetes therapy on bone. Interestingly, the majority of antidiabetic agents positively affect bone, but a few have detrimental effects. Specifically, although insulin has been demonstrated to be anabolic to bone, the rate of hypoglycemic episodes are increased with exogenous infusion; consequently, there is an increased fall and fracture frequency. Other agents such as thiazolidinediones have more direct negative effects on bone through transcriptional regulation. Even metabolic surgery, to a varying operation-dependent extent, exacerbates bone strength and may heighten fracture rate. The remaining diabetes agents seem to have neutral or positive effects on bone. With the increasing incidence of diabetes, it is more pertinent than ever to fully comprehend the effects of diabetes-related therapeutic modalities.
Collapse
MESH Headings
- Bone Density/drug effects
- Bone Diseases, Metabolic/etiology
- Bone Diseases, Metabolic/pathology
- Bone Diseases, Metabolic/prevention & control
- Bone and Bones/drug effects
- Bone and Bones/physiology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Fractures, Bone/etiology
- Fractures, Bone/pathology
- Humans
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Osteoporosis/etiology
- Osteoporosis/metabolism
- Osteoporosis/pathology
- Risk Factors
Collapse
Affiliation(s)
- Karim G Kheniser
- Department of Endocrinology and Metabolism, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States.
| | - Carmen M Polanco Santos
- Department of Endocrinology and Metabolism, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States.
| | - Sangeeta R Kashyap
- Department of Endocrinology and Metabolism, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States.
| |
Collapse
|
28
|
de Oliveira GJPL, Basso TLD, Fontanari LA, Faloni APDS, Marcantonio É, Orrico SRP. Glycemic control protects against trabecular bone microarchitectural damage in a juvenile male rat model of streptozotocin-induced diabetes. Endocr Res 2017; 42:171-179. [PMID: 28281839 DOI: 10.1080/07435800.2017.1292521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE To determine which features of the bone microarchitecture are affected by established diabetes mellitus (DM) and the effectiveness of glycemic control in the protection of bone tissue. MATERIAL AND METHODS Sixty juvenile Wistar male rats were divided into three groups of 20 animals: a control group (C) that included healthy animals, a diabetic group (D) that included animals with induced diabetes, and a controlled diabetic group (CD) that included animals with induced diabetes that were treated with insulin. The animals were euthanized at the periods of 6 and 8 weeks after the induction of diabetes (10 animals per group/period). Vertebral L4 specimens were submitted to μCT analysis to assess the following parameters of the bone microarchitecture: bone volume fraction (BV/TV), trabecular thickness (Tb.Th), trabecular number (Tb.N), and trabecular spacing (Tb.Sp). RESULTS The D group exhibited lower values of BV/TV (%) and numbers of trabeculae compared with the C group at 6 and 8 weeks and compared with the CD group at 8 weeks. The CD group exhibited higher trabecular thickness values compared with the D group at 8 weeks. There were no differences between the groups regarding the spaces between the trabeculae. CONCLUSION Induced diabetes affected the microarchitecture of the trabecular bone of the vertebrae by reducing the values of the majority of the parameters in relation to those of the control group. Glycemic control with insulin appears to protect bones from the effects of the hyperglycemia.
Collapse
Affiliation(s)
| | - Túlio Luiz Durigan Basso
- a Department of Diagnosis and Surgery , Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry , Araraquara , Brazil
| | - Lucas Amaral Fontanari
- a Department of Diagnosis and Surgery , Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry , Araraquara , Brazil
| | - Ana Paula de Souza Faloni
- b Department of Histology, School of Dentistry , Araraquara University (UNIARA) , Araraquara , Brazil
| | - Élcio Marcantonio
- a Department of Diagnosis and Surgery , Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry , Araraquara , Brazil
| | - Silvana Regina Perez Orrico
- a Department of Diagnosis and Surgery , Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry , Araraquara , Brazil
| |
Collapse
|
29
|
Tan N, Liu X, Cai Y, Zhang S, Jian B, Zhou Y, Xu X, Ren S, Wei H, Song Y. The influence of direct laser metal sintering implants on the early stages of osseointegration in diabetic mini-pigs. Int J Nanomedicine 2017; 12:5433-5442. [PMID: 28814861 PMCID: PMC5546787 DOI: 10.2147/ijn.s138615] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background High failure rates of oral implants have been reported in diabetic patients due to the disruption of osseointegration. The aim of this study was to investigate whether direct laser metal sintering (DLMS) could improve osseointegration in diabetic animal models. Methods Surface characterizations were carried out on two types of implants. Cell morphology and the osteogenic-related gene expression of MG63 cells were observed under conditions of DLMS and microarc oxidation (MAO). A diabetes model in mini-pigs was established by intravenous injection of streptozotocin (150 mg/kg), and a total of 36 implants were inserted into the mandibular region. Micro-computed tomography (micro-CT) and histologic evaluations were performed 3 and 6 months after implantation. Results The Ra (the average of the absolute height of all points) of MAO surface was 2.3±0.3 µm while the DLMS surface showed the Ra of 27.4±1.1 µm. The cells on DLMS implants spread out more podia than those on MAO implants through cell morphology analysis. Osteogenic-related gene expression was also dramatically increased in the DLMS group. Obvious improvement was observed in the micro-CT and Van Gieson staining analyses of DLMS implants compared with MAO at 3 months, although this difference disappeared by 6 months. DLMS implants showed a higher bone–implant contact percentage (33.2%±11.2%) at 3 months compared with MAO group (18.9%±7.3%) while similar results were showed at 6 months between DLMS group (42.8%±10.1%) and MAO group (38.3%±10.8%). Conclusion The three-dimensional environment of implant surfaces with highly porous and fully interconnected channel and pore architectures can improve cell spreading and accelerate the progress of osseointegration in diabetic mini-pigs.
Collapse
Affiliation(s)
- Naiwen Tan
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Stomatology, Hospital 463 of PLA, Xi'an, Shaanxi, China
| | - Xiangwei Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yanhui Cai
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sijia Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bo Jian
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuchao Zhou
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaoru Xu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuai Ren
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongbo Wei
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yingliang Song
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Xi'an, Shaanxi, China.,Department of Implant Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
30
|
Molinuevo MS, Fernández JM, Cortizo AM, McCarthy AD, Schurman L, Sedlinsky C. Advanced glycation end products and strontium ranelate promote osteogenic differentiation of vascular smooth muscle cells in vitro: Preventive role of vitamin D. Mol Cell Endocrinol 2017; 450:94-104. [PMID: 28456475 DOI: 10.1016/j.mce.2017.04.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 04/19/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023]
Abstract
Advanced glycation end products (AGE) have been demonstrated to induce the osteogenic trans-differentiation of vascular smooth muscle cells (VSMC). Strontium ranelate (SR) is an anti-osteoporotic agent that has both anti-catabolic and anabolic actions on bone tissue. However, in the last years SR has been associated with an increase of cardiovascular risk. We hypothesize that SR can increase the osteoblastic trans-differentiation of VSMC and the induction of extracellular calcifications, an effect that could be potentiated in the presence of AGE and inhibited by simultaneous administration of vitamin D. The present results of our in vitro experiments demonstrate that AGE and SR alone or in combination, stimulate L-type calcium channels, causing an increase in reactive oxygen species and activation of both ERK and NFkB, with the final effect of promoting the osteogenic shift of VSMC. Importantly, these in vitro effects of AGE and/or SR can be prevented by co-incubation with vitamin D.
Collapse
Affiliation(s)
- María Silvina Molinuevo
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. 47 y 115, (1900) La Plata, Argentina
| | - Juan Manuel Fernández
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. 47 y 115, (1900) La Plata, Argentina
| | - Ana María Cortizo
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. 47 y 115, (1900) La Plata, Argentina
| | - Antonio Desmond McCarthy
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. 47 y 115, (1900) La Plata, Argentina
| | - León Schurman
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. 47 y 115, (1900) La Plata, Argentina
| | - Claudia Sedlinsky
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. 47 y 115, (1900) La Plata, Argentina.
| |
Collapse
|
31
|
Park YE, Musson DS, Naot D, Cornish J. Cell–cell communication in bone development and whole-body homeostasis and pharmacological avenues for bone disorders. Curr Opin Pharmacol 2017; 34:21-35. [DOI: 10.1016/j.coph.2017.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/07/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
|
32
|
Felice JI, Schurman L, McCarthy AD, Sedlinsky C, Aguirre JI, Cortizo AM. Effects of fructose-induced metabolic syndrome on rat skeletal cells and tissue, and their responses to metformin treatment. Diabetes Res Clin Pract 2017; 126:202-213. [PMID: 28259010 DOI: 10.1016/j.diabres.2017.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/06/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022]
Abstract
AIMS Deleterious effects of metabolic syndrome (MS) on bone are still controversial. In this study we evaluated the effects of a fructose-induced MS, and/or an oral treatment with metformin on the osteogenic potential of bone marrow mesenchymal stromal cells (MSC), as well as on bone formation and architecture. METHODS 32 male 8week-old Wistar rats were assigned to four groups: control (C), control plus oral metformin (CM), rats receiving 10% fructose in drinking water (FRD), and FRD plus metformin (FRDM). Samples were collected to measure blood parameters, and to perform pQCT analysis and static and dynamic histomorphometry. MSC were isolated to determine their osteogenic potential. RESULTS Metformin improved blood parameters in FRDM rats. pQCT and static and dynamic histomorphometry showed no significant differences in trabecular and cortical bone parameters among groups. FRD reduced TRAP expression and osteocyte density in trabecular bone and metformin only normalized osteocyte density. FRD decreased the osteogenic potential of MSC and metformin administration could revert some of these parameters. CONCLUSIONS FRD-induced MS shows reduction in MSC osteogenic potential, in osteocyte density and in TRAP activity. Oral metformin treatment was able to prevent trabecular osteocyte loss and the reduction in extracellular mineralization induced by FRD-induced MS.
Collapse
Affiliation(s)
- Juan Ignacio Felice
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - León Schurman
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Antonio Desmond McCarthy
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Claudia Sedlinsky
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - José Ignacio Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Ana María Cortizo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina.
| |
Collapse
|
33
|
Abstract
Diabetes be it type 1 or type 2 is associated with an increased risk of fragility fractures. The mechanisms underlying this increased risk are just being elucidated. Anti-diabetes medications are crucial for maintaining glucose control and for preventing micro- and macrovascular complications in diabetes. However, they may modulate fracture risk in diabetes in different ways. Thiazolidinediones have demonstrated an unfavorable effect on the skeleton, while metformin and sulfonylureas may have a neutral if not beneficial effect on bone. The use of insulin has been associated with an increased risk of fragility fractures though it is not clear whether it is due to direct influence of insulin or whether it is mediated through hypoglycemia and increased falls risk. The overall effect of incretin mimetics appears to be beneficial; however, this has to be elucidated further. The bone effects of pramlintide, a synthetic analog of amylin, have not been explored fully. Finally, issues regarding bone safety of SGLT2 (sodium-dependent glucose transporter 2) inhibitors, the newest anti-diabetic medications on the market are of concern. The purpose of this review is to provide a comprehensive overview of the effect of these medications on bone metabolism and the studies exploring the risk or lack thereof of these medications on bone loss and fragility fractures.
Collapse
Affiliation(s)
- Manju Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, ACADEMIA, 20 College Road, Singapore, 169856, Singapore.
| |
Collapse
|
34
|
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]
|
35
|
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: 75] [Impact Index Per Article: 8.3] [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
|
36
|
Alendronate Can Improve Bone Alterations in Experimental Diabetes by Preventing Antiosteogenic, Antichondrogenic, and Proadipocytic Effects of AGEs on Bone Marrow Progenitor Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5891925. [PMID: 27840829 PMCID: PMC5093246 DOI: 10.1155/2016/5891925] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/03/2016] [Indexed: 01/29/2023]
Abstract
Bisphosphonates such as alendronate are antiosteoporotic drugs that inhibit the activity of bone-resorbing osteoclasts and secondarily promote osteoblastic function. Diabetes increases bone-matrix-associated advanced glycation end products (AGEs) that impair bone marrow progenitor cell (BMPC) osteogenic potential and decrease bone quality. Here we investigated the in vitro effect of alendronate and/or AGEs on the osteoblastogenic, adipogenic, and chondrogenic potential of BMPC isolated from nondiabetic untreated rats. We also evaluated the in vivo effect of alendronate (administered orally to rats with insulin-deficient Diabetes) on long-bone microarchitecture and BMPC multilineage potential. In vitro, the osteogenesis (Runx2, alkaline phosphatase, type 1 collagen, and mineralization) and chondrogenesis (glycosaminoglycan production) of BMPC were both decreased by AGEs, while coincubation with alendronate prevented these effects. The adipogenesis of BMPC (PPARγ, intracellular triglycerides, and lipase) was increased by AGEs, and this was prevented by coincubation with alendronate. In vivo, experimental Diabetes (a) decreased femoral trabecular bone area, osteocyte density, and osteoclastic TRAP activity; (b) increased bone marrow adiposity; and (c) deregulated BMPC phenotypic potential (increasing adipogenesis and decreasing osteogenesis and chondrogenesis). Orally administered alendronate prevented all these Diabetes-induced effects on bone. Thus, alendronate could improve bone alterations in diabetic rats by preventing the antiosteogenic, antichondrogenic, and proadipocytic effects of AGEs on BMPC.
Collapse
|
37
|
Yu JW, Deng YP, Han X, Ren GF, Cai J, Jiang GJ. Metformin improves the angiogenic functions of endothelial progenitor cells via activating AMPK/eNOS pathway in diabetic mice. Cardiovasc Diabetol 2016; 15:88. [PMID: 27316923 PMCID: PMC4912824 DOI: 10.1186/s12933-016-0408-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Background Endothelial dysfunction has been suggested as a possible causal link between hyperglycemia and microvascular complications in diabetes mellitus. The effect of metformin on endothelial progenitor cells (EPCs) is still unclear. This study was designed to test the hypothesis that metformin could accelerate wound healing by improving the impaired EPC functions in streptozotocin-induced diabetic mice. Methods Streptozotocin (STZ, 60 mg/kg/d × 5 d, i.p.) was injected to induce type 1 diabetes in male C57BL/6 mice. Mice were treated with metformin (250 mg/kg/d, i.g.) for consecutive 14 days. Wound closure was evaluated by wound area and number of CD31 stained capillaries. Functions of bone marrow-endothelial progenitor cells (BM-EPCs) were assessed by tube formation and migration assays, and expression of AMP-activated protein kinase (AMPK) and endothelial nitric oxide synthase (eNOS) was determined by western blot analysis. Results Metformin accelerated wound closure and stimulated angiogenesis in diabetic mice. The number of circulating EPCs was increased significantly in metformin treated diabetic mice. Abilities of tube formation and migration of BM-EPCs were impaired in diabetic mice, which were improved by metformin. Expression of both phosphorylated-AMPK and phosphorylated-eNOS was significantly increased, and nitric oxide (NO) production was enhanced by metformin in BM-EPCs of diabetic mice. In vitro, metformin improved impaired BM-EPC functions, and increased phosphorylated-eNOS expression and NO production in cultured BM-EPCs caused by high glucose, which was prevented by the AMPK inhibitor compound C. Conclusions Our results suggest that metformin could improve BM-EPC functions in STZ-induced diabetic mice, which was possibly dependent on the AMPK/eNOS pathway. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0408-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jia-Wen Yu
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311202, Zhejiang, China
| | - Ya-Ping Deng
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311202, Zhejiang, China
| | - Xue Han
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311202, Zhejiang, China
| | - Guo-Fei Ren
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311202, Zhejiang, China
| | - Jian Cai
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311202, Zhejiang, China
| | - Guo-Jun Jiang
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311202, Zhejiang, China.
| |
Collapse
|
38
|
Suh KS, Chon S, Choi EM. Luteolin alleviates methylglyoxal-induced cytotoxicity in osteoblastic MC3T3-E1 cells. Cytotechnology 2016; 68:2539-2552. [PMID: 27221336 DOI: 10.1007/s10616-016-9977-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/22/2016] [Indexed: 01/01/2023] Open
Abstract
Methylglyoxal (MG), a reactive sugar-derived metabolite, exerts harmful effects by inducing oxidative stress, which aggravates a series of diabetic complications, including osteoporosis. The present study was performed to examine the effects of luteolin, a dietary polyphenolic flavonoid, on MG-induced cytotoxicity in MC3T3-E1 osteoblastic cells. Pretreatment of MC3T3-E1 osteoblastic cells with luteolin prevented MG-induced cell death and production of tumor necrosis factor-alpha, intracellular reactive oxygen species, mitochondrial superoxide, and cardiolipin peroxidation. In addition, luteolin increased the levels of glutathione and nuclear factor erythroid 2-related factor 2 (Nrf2) and decreased the inhibition of heme oxygenase-1 activity by MG. Pretreatment with luteolin prior to MG exposure reduced MG-induced mitochondrial dysfunction and increased the peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) and nitric oxide levels, suggesting that luteolin may induce mitochondrial biogenesis. Taken together, these observations indicated that luteolin has potential as a preventive agent against the development of diabetic osteopathy related to MG-induced oxidative stress in diabetes.
Collapse
Affiliation(s)
- Kwang Sik Suh
- Research Institute of Endocrinology, Kyung Hee University Hospital, 1, Hoegi-dong, Dongdaemun-gu, Seoul, 130-702, South Korea
| | - Suk Chon
- Department of Endocrinology and Metabolism, School of Medicine, Kyung Hee University, 1, Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, South Korea
| | - Eun Mi Choi
- Department of Endocrinology and Metabolism, School of Medicine, Kyung Hee University, 1, Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, South Korea.
| |
Collapse
|
39
|
McCarthy AD, Cortizo AM, Sedlinsky C. Metformin revisited: Does this regulator of AMP-activated protein kinase secondarily affect bone metabolism and prevent diabetic osteopathy. World J Diabetes 2016; 7:122-133. [PMID: 27022443 PMCID: PMC4807302 DOI: 10.4239/wjd.v7.i6.122] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 12/24/2015] [Accepted: 01/31/2016] [Indexed: 02/05/2023] Open
Abstract
Patients with long-term type 1 and type 2 diabetes mellitus (DM) can develop skeletal complications or “diabetic osteopathy”. These include osteopenia, osteoporosis and an increased incidence of low-stress fractures. In this context, it is important to evaluate whether current anti-diabetic treatments can secondarily affect bone metabolism. Adenosine monophosphate-activated protein kinase (AMPK) modulates multiple metabolic pathways and acts as a sensor of the cellular energy status; recent evidence suggests a critical role for AMPK in bone homeostasis. In addition, AMPK activation is believed to mediate most clinical effects of the insulin-sensitizer metformin. Over the past decade, several research groups have investigated the effects of metformin on bone, providing a considerable body of pre-clinical (in vitro, ex vivo and in vivo) as well as clinical evidence for an anabolic action of metformin on bone. However, two caveats should be kept in mind when considering metformin treatment for a patient with type 2 DM at risk for diabetic osteopathy. In the first place, metformin should probably not be considered an anti-osteoporotic drug; it is an insulin sensitizer with proven macrovascular benefits that can secondarily improve bone metabolism in the context of DM. Secondly, we are still awaiting the results of randomized placebo-controlled studies in humans that evaluate the effects of metformin on bone metabolism as a primary endpoint.
Collapse
|
40
|
Berberine in Combination with Insulin Has Additive Effects on Titanium Implants Osseointegration in Diabetes Mellitus Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:824259. [PMID: 26783411 PMCID: PMC4691484 DOI: 10.1155/2015/824259] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/09/2015] [Accepted: 11/25/2015] [Indexed: 02/05/2023]
Abstract
This study evaluated the effects of berberine in combination with insulin on early osseointegration of implants in diabetic rats. Fifty male Sprague-Dawley rats were randomly divided into 5 groups: healthy rats were used as control (HC), and streptozotocin-induced diabetic rats were treated with insulin, berberine, berberine + insulin (IB), or no treatment. Each rat received one machined-surface cp-Ti implant into the right tibia and was given insulin injection and/or gavage feeding with berberine daily for 8 weeks until being sacrificed. Serum levels of alkaline phosphatase (ALP) and bone gamma-carboxyglutamic acid-containing protein (BGP) were analyzed in each group. Peri-implant mineral apposition was marked by fluorochrome double-labeling and osseointegration was histomorphologically examined. The ALP and BGP levels decreased in diabetic rats but were successfully corrected by insulin and berberine combined treatment. Moreover, untreated diabetic rats had less labeled mineral apposition and impaired osseointegration. In contrast, Groups I, B, and IB were observed with increased peri-implant bone formation. The combination treatment of insulin and berberine was more effective than each administrated as a monotherapy. These results suggest that berberine combined with insulin could promote osseointegration in diabetic rats, thereby highlighting its potential application to patients, though further studies are needed.
Collapse
|
41
|
Wang Y, Zhu G, Li N, Song J, Wang L, Shi X. Small molecules and their controlled release that induce the osteogenic/chondrogenic commitment of stem cells. Biotechnol Adv 2015; 33:1626-40. [PMID: 26341834 DOI: 10.1016/j.biotechadv.2015.08.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/21/2015] [Accepted: 08/23/2015] [Indexed: 12/17/2022]
Abstract
Stem cell-based tissue engineering plays a significant role in skeletal system repair and regenerative therapies. However, stem cells must be differentiated into specific mature cells prior to implantation (direct implantation may lead to tumour formation). Natural or chemically synthesised small molecules provide an efficient, accurate, reversible, and cost-effective way to differentiate stem cells compared with bioactive growth factors and gene-related methods. Thus, investigating the influences of small molecules on the differentiation of stem cells is of great significance. Here, we review a series of small molecules that can induce or/and promote the osteogenic/chondrogenic commitment of stem cells. The controlled release of these small molecules from various vehicles for stem cell-based therapies and tissue engineering applications is also discussed. The extensive studies in this field represent significant contributions to stem cell-based tissue engineering research and regenerative medicine.
Collapse
Affiliation(s)
- Yingjun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Guanglin Zhu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Nanying Li
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Juqing Song
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Lin Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510640, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, PR China.
| |
Collapse
|
42
|
Qian C, Zhu C, Yu W, Jiang X, Zhang F. High-Fat Diet/Low-Dose Streptozotocin-Induced Type 2 Diabetes in Rats Impacts Osteogenesis and Wnt Signaling in Bone Marrow Stromal Cells. PLoS One 2015; 10:e0136390. [PMID: 26296196 PMCID: PMC4546646 DOI: 10.1371/journal.pone.0136390] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/04/2015] [Indexed: 01/11/2023] Open
Abstract
Bone regeneration disorders are a significant problem in patients with type 2 diabetes mellitus. Bone marrow stromal cells (BMSCs) are recognized as ideal seed cells for tissue engineering because they can stimulate osteogenesis during bone regeneration. Therefore, the aim of this study was to investigate the osteogenic potential of BMSCs derived from type 2 diabetic rats and the pathogenic characteristics of dysfunctional BMSCs that affect osteogenesis. BMSCs were isolated from normal and high-fat diet+streptozotocin-induced type 2 diabetic rats. Cell metabolic activity, alkaline phosphatase (ALP) activity, mineralization and osteogenic gene expression were reduced in the type 2 diabetic rat BMSCs. The expression levels of Wnt signaling genes, such as β-catenin, cyclin D1 and c-myc, were also significantly decreased in the type 2 diabetic rat BMSCs, but the expression of GSK3β remained unchanged. The derived BMSCs were cultured on calcium phosphate cement (CPC) scaffolds and placed subcutaneously into nude mice for eight weeks; they were detected at a low level in newly formed bone. The osteogenic potential of the type 2 diabetic rat BMSCs was not impaired by the culture environment, but it was impaired by inhibition of the Wnt signaling pathway, likely due to an insufficient accumulation of β-catenin rather than because of GSK3β stimulation. Using BMSCs derived from diabetic subjects could offer an alternative method of regenerating bone together with the use of supplementary growth factors to stimulate the Wnt signaling pathway.
Collapse
MESH Headings
- Alkaline Phosphatase/genetics
- Alkaline Phosphatase/metabolism
- Animals
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- Calcium Phosphates/chemistry
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Gene Expression Regulation
- Glycogen Synthase Kinase 3/genetics
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- Male
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mesenchymal Stem Cells/pathology
- Mice
- Mice, Nude
- Osteogenesis/genetics
- Primary Cell Culture
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Rats
- Rats, Sprague-Dawley
- Streptozocin
- Transplantation, Heterologous
- Wnt Signaling Pathway
Collapse
Affiliation(s)
- Chao Qian
- Department of Prosthodontics, School of Stomatology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology. Shanghai, 200011, People’s Republic of China
| | - Chenyuan Zhu
- Department of Prosthodontics, School of Stomatology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology. Shanghai, 200011, People’s Republic of China
| | - Weiqiang Yu
- Department of Prosthodontics, School of Stomatology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology. Shanghai, 200011, People’s Republic of China
| | - Xinquan Jiang
- Department of Prosthodontics, School of Stomatology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology. Shanghai, 200011, People’s Republic of China
- * E-mail: (FZ); (XJ)
| | - Fuqiang Zhang
- Department of Prosthodontics, School of Stomatology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology. Shanghai, 200011, People’s Republic of China
- * E-mail: (FZ); (XJ)
| |
Collapse
|
43
|
Palermo A, D'Onofrio L, Eastell R, Schwartz AV, Pozzilli P, Napoli N. Oral anti-diabetic drugs and fracture risk, cut to the bone: safe or dangerous? A narrative review. Osteoporos Int 2015; 26:2073-89. [PMID: 25910746 DOI: 10.1007/s00198-015-3123-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022]
Abstract
Fracture risk is higher in older adults with type 2 diabetes and may be influenced by treatments for diabetes. Oral anti-diabetic drugs have different effects on bone metabolism. The purpose of this review is to describe the effects of these drugs on bone metabolism and fracture risk. Osteoporosis is a progressive skeletal disorder that is characterized by compromised bone strength and increased risk of fracture. This condition has become an important global health problem, affecting approximately 200 million people worldwide. Another chronic and highly prevalent condition is diabetes mellitus, which affects more than 380 million people; both type 1 and type 2 diabetes are risk factors for fracture. Type 2 diabetes, in particular, is associated with impaired bone strength, although it is characterized by normal or elevated bone mineral density. Several therapeutic strategies are available to achieve the best outcomes in the management of diabetes mellitus but these have different effects on bone metabolism. The purpose of this narrative review is to describe the effects of oral hypoglycemic agents (metformin, sulfonylureas, thiazolidinediones, meglitinides, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists and sodium-dependent glucose transporter 2 inhibitors) on bone metabolism and on the risk of developing fragility fractures in patients with type 2 diabetes. Both diabetes and osteoporosis represent a significant burden in terms of healthcare costs and quality of life. It is very important to choose therapies for diabetes that ensure good metabolic control whilst preserving skeletal health.
Collapse
Affiliation(s)
- A Palermo
- Department of Endocrinology and Diabetes, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 21-00128, Rome, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Silva JC, Sampaio P, Fernandes MH, Gomes PS. The Osteogenic Priming of Mesenchymal Stem Cells is Impaired in Experimental Diabetes. J Cell Biochem 2015; 116:1658-67. [DOI: 10.1002/jcb.25126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/03/2015] [Indexed: 02/01/2023]
Affiliation(s)
- J. C. Silva
- Laboratory for Bone Metabolism and Regeneration; Faculty of Dental Medicine; University of Porto; Rua Dr. Manuel Pereira da Silva Porto 4200-393 Portugal
| | - P. Sampaio
- Institute for Molecular Cell Biology (IBMC); Porto Portugal
| | - M. H. Fernandes
- Laboratory for Bone Metabolism and Regeneration; Faculty of Dental Medicine; University of Porto; Rua Dr. Manuel Pereira da Silva Porto 4200-393 Portugal
| | - P. S. Gomes
- Laboratory for Bone Metabolism and Regeneration; Faculty of Dental Medicine; University of Porto; Rua Dr. Manuel Pereira da Silva Porto 4200-393 Portugal
| |
Collapse
|
45
|
Hegazy SK. Evaluation of the anti-osteoporotic effects of metformin and sitagliptin in postmenopausal diabetic women. J Bone Miner Metab 2015; 33:207-12. [PMID: 24633493 DOI: 10.1007/s00774-014-0581-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/13/2014] [Indexed: 12/21/2022]
Abstract
Osteoporosis is the most important metabolic bone disease in patients with diabetes mellitus. Several studies have documented that metformin is osteogenic in vitro. In contrast, others showed no effect of metformin on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells. Incretin hormones have received much attention because of their beneficial effects beyond glycemia, including on bone health. The study evaluated the anti-osteoporotic effect of metformin and sitagliptin in postmenopausal diabetic women. Forty postmenopausal diabetic women were randomly divided into two equal groups. Group 1 received metformin (Glucophage(®) 500 mg) 1 tablet twice daily, and group 2 received sitagliptin (Januvia(®) 100 mg) 1 tablet/day, for 12 weeks. Fasting blood and urine samples were collected for measurement of serum total alkaline phosphatase (ALP), osteocalcin, and urinary deoxypyridinoline (DPD). Laboratory tests were measured at baseline, after 4 and 8 weeks, and at the end of the study. Bone mineral density of the anterior posterior lumbar spine was measured by dual energy X-ray absorptiometry at baseline and after 12 weeks of the intervention. In the metformin-treated group, the mean values for all markers of bone turnover at 12 weeks of treatment were not significantly different from baseline. In group 2, the mean serum total ALP was significantly decreased, serum osteocalcin levels were non-significantly decreased gradually by 10% at 12 weeks, while urinary DPD decreased significantly and was then maintained at 28% decrease at 12 weeks. In conclusion, metformin is neither osteogenic nor has anti-osteoporotic effect, while sitagliptin could positively regulate bone metabolism.
Collapse
|
46
|
Illien-Jünger S, Lu Y, Qureshi SA, Hecht AC, Cai W, Vlassara H, Striker GE, Iatridis JC. Chronic ingestion of advanced glycation end products induces degenerative spinal changes and hypertrophy in aging pre-diabetic mice. PLoS One 2015; 10:e0116625. [PMID: 25668621 PMCID: PMC4323205 DOI: 10.1371/journal.pone.0116625] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022] Open
Abstract
Intervertebral disc (IVD) degeneration and pathological spinal changes are major causes of back pain, which is the top cause of global disability. Obese and diabetic individuals are at increased risk for back pain and musculoskeletal complications. Modern diets contain high levels of advanced glycation end products (AGEs), cyto-toxic components which are known contributors to obesity, diabetes and accelerated aging pathologies. There is little information about potential effects of AGE rich diet on spinal pathology, which may be a contributing cause for back pain which is common in obese and diabetic individuals. This study investigated the role of specific AGE precursors (e.g. methylglyoxal-derivatives (MG)) on IVD and vertebral pathologies in aging C57BL6 mice that were fed isocaloric diets with standard (dMG+) or reduced amounts of MG derivatives (dMG-; containing 60-70% less dMG). dMG+ mice exhibited a pre-diabetic phenotype, as they were insulin resistant but not hyperglycemic. Vertebrae of dMG+ mice displayed increased cortical-thickness and cortical-area, greater MG-AGE accumulation and ectopic calcification in vertebral endplates. IVD morphology of dMG+ mice exhibited ectopic calcification, hypertrophic differentiation and glycosaminoglycan loss relative to dMG- mice. Overall, chronic exposure to dietary AGEs promoted age-accelerated IVD degeneration and vertebral alterations involving ectopic calcification which occurred in parallel with insulin resistance, and which were prevented with dMG- diet. This study described a new mouse model for diet-induced spinal degeneration, and results were in support of the hypothesis that chronic AGE ingestion could be a factor contributing to a pre-diabetic state, ectopic calcifications in spinal tissues, and musculoskeletal complications that are more generally known to occur with chronic diabetic conditions.
Collapse
Affiliation(s)
- Svenja Illien-Jünger
- Leni & Peter W. May Department of Orthopaedics/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Young Lu
- Leni & Peter W. May Department of Orthopaedics/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Sheeraz A. Qureshi
- Leni & Peter W. May Department of Orthopaedics/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Andrew C. Hecht
- Leni & Peter W. May Department of Orthopaedics/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Weijing Cai
- Department of Geriatrics and Palliative Care, Division of Experimental Diabetes and Aging/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Helen Vlassara
- Department of Geriatrics and Palliative Care, Division of Experimental Diabetes and Aging/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Gary E. Striker
- Department of Geriatrics and Palliative Care, Division of Experimental Diabetes and Aging, and Division of Nephrology, Department of Medicine/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - James C. Iatridis
- Leni & Peter W. May Department of Orthopaedics/Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
47
|
Abstract
Type 2 diabetes mellitus (T2DM) is a common chronic disease that may be associated with an increased risk of fracture. Evidence that thiazolidinediones (TZDs) increase fracture risk in women with T2DM has focused attention on the skeletal effects of treatments for diabetes. Only scant, low-quality evidence is available for non-TZD diabetes medications and bone health, but it suggests that there are no clinically important effects.
Collapse
Affiliation(s)
- Andrew Grey
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, New Zealand,
| |
Collapse
|
48
|
Felice JI, Gangoiti MV, Molinuevo MS, McCarthy AD, Cortizo AM. Effects of a metabolic syndrome induced by a fructose-rich diet on bone metabolism in rats. Metabolism 2014; 63:296-305. [PMID: 24355623 DOI: 10.1016/j.metabol.2013.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 10/22/2013] [Accepted: 11/03/2013] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aims of this study were: first, to evaluate the possible effects of a fructose rich diet (FRD)-induced metabolic syndrome (MS) on different aspects of long bone histomorphometry in young male rats; second, to investigate the effects of this diet on bone tissue regeneration; and third, to correlate these morphometric alterations with changes in the osteogenic/adipogenic potential and expression of specific transcription factors, of marrow stromal cells (MSC) isolated from rats with fructose-induced MS. MATERIALS/METHODS MS was induced in rats by treatment with a FRD for 28 days. Halfway through treatment, a parietal wound was made and bone healing was evaluated 14 days later. After treatments, histomorphometric analysis was performed in dissected femoral and parietal bones. MSC were isolated from the femora of control or fructose-treated rats and differentiated either to osteoblasts (evaluated by type 1 collagen, Alkaline phosphatase and extracellular nodule mineralization) or to adipocytes (evaluated by intracellular triglyceride accumulation). Expression of Runx2 and PPARγ was assessed by Western blot. RESULTS Fructose-induced MS induced deleterious effects on femoral metaphysis microarchitecture and impaired bone regeneration. Fructose treatment decreased the osteogenic potential of MSC and Runx2 expression. In addition, it increased the adipogenic commitment of MSC and PPARγ expression. CONCLUSIONS Fructose-induced MS is associated with deleterious effects on bone microarchitecture and with a decrease in bone repair. These alterations could be due to a deviation in the adipogenic/osteogenic commitment of MSC, probably by modulation of the Runx2/PPARγ ratio.
Collapse
Affiliation(s)
- Juan Ignacio Felice
- LIOMM (Laboratorio de Investigación en Osteopatías y Metabolismo Mineral), Department of Biological Sciences, School of Exact Sciences, National University of La Plata, La Plata, Argentina
| | - María Virginia Gangoiti
- LIOMM (Laboratorio de Investigación en Osteopatías y Metabolismo Mineral), Department of Biological Sciences, School of Exact Sciences, National University of La Plata, La Plata, Argentina
| | - María Silvina Molinuevo
- LIOMM (Laboratorio de Investigación en Osteopatías y Metabolismo Mineral), Department of Biological Sciences, School of Exact Sciences, National University of La Plata, La Plata, Argentina
| | - Antonio Desmond McCarthy
- LIOMM (Laboratorio de Investigación en Osteopatías y Metabolismo Mineral), Department of Biological Sciences, School of Exact Sciences, National University of La Plata, La Plata, Argentina
| | - Ana María Cortizo
- LIOMM (Laboratorio de Investigación en Osteopatías y Metabolismo Mineral), Department of Biological Sciences, School of Exact Sciences, National University of La Plata, La Plata, Argentina.
| |
Collapse
|