1
|
Lozano D, Gortazar AR, Portal-Núñez S. Osteostatin, a peptide for the future treatment of musculoskeletal diseases. Biochem Pharmacol 2024; 223:116177. [PMID: 38552853 DOI: 10.1016/j.bcp.2024.116177] [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: 12/30/2023] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 04/08/2024]
Abstract
Nowadays, the treatment of musculoskeletal diseases represents a major challenge in the developed world. Diseases such as osteoporosis, osteoarthritis and arthritis have a high incidence and prevalence as a consequence of population aging, and they are also associated with a socioeconomic burden. Many efforts have been made to find a treatment for these diseases with various levels of success, but new approaches are still needed to deal with these pathologies. In this context, one peptide derived for the C-terminal extreme of the Parathormone related Peptide (PTHrP) called Osteostatin can be useful to treat musculoskeletal diseases. This pentapeptide (TRSAW) has demonstrated both in different in vitro and in vivo models, its role as a molecule with anti-resorptive, anabolic, anti-inflammatory, and anti-antioxidant properties. Our aim with this work is to review the Osteostatin main features, the knowledge of its mechanisms of action as well as its possible use for the treatment of osteoporosis, bone regeneration and fractures and against arthritis given its anti-inflammatory properties.
Collapse
Affiliation(s)
- Daniel Lozano
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Arancha R Gortazar
- Grupo de Fisiopatología Ósea, Departamento de Ciencias Médicas Básicas, Instituto de Medicina Aplicada de la Universidad San Pablo-CEU, Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe s/n, 28925 Madrid, Spain
| | - Sergio Portal-Núñez
- Grupo de Fisiopatología Ósea, Departamento de Ciencias Médicas Básicas, Instituto de Medicina Aplicada de la Universidad San Pablo-CEU, Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe s/n, 28925 Madrid, Spain.
| |
Collapse
|
2
|
Marino S, Ozgurel SU, McAndrews K, Cregor M, Villaseñor A, Mamani-Huanca M, Barbas C, Gortazar A, Sato AY, Bellido T. Abaloparatide is more potent than teriparatide in restoring bone mass and strength in type 1 diabetic male mice. Bone 2024; 181:117042. [PMID: 38360197 DOI: 10.1016/j.bone.2024.117042] [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: 12/19/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/17/2024]
Abstract
This study investigated the efficacy of the two FDA-approved bone anabolic ligands of the parathyroid hormone receptor 1 (PTH1R), teriparatide or human parathyroid hormone 1-34 (PTH) and abaloparatide (ABL), to restoring skeletal health using a preclinical murine model of streptozotocin-induced T1-DM. Intermittent daily subcutaneous injections of equal molar doses (12 pmoles/g/day) of PTH (50 ng/g/day), ABL (47.5 ng/g/day), or vehicle, were administered for 28 days to 5-month-old C57Bl/6 J male mice with established T1-DM or control (C) mice. ABL was superior to PTH in increasing or restoring bone mass in control or T1-MD mice, respectively, which was associated with superior stimulation of trabecular and periosteal bone formation, upregulation of osteoclastic/osteoblastic gene expression, and increased circulating bone remodeling markers. Only ABL corrected the reduction in ultimate load, which is a measure of bone strength, induced by T1-DM, and it also increased energy to ultimate load. In addition, bones from T1-DM mice treated with PTH or ABL exhibited increased ultimate stress, a material index, compared to T1-DM mice administered with vehicle. And both PTH and ABL prevented the increased expression of the Wnt antagonist Sost/sclerostin displayed by T1-DM mice. Further, PTH and ABL increased to a similar extent the circulating bone resorption marker CTX and the bone formation marker P1NP in T1-DM after 2 weeks of treatment; however, only ABL sustained these increases after 4 weeks of treatment. We conclude that at equal molar doses, ABL is more effective than PTH in increasing bone mass and restoring the cortical and trabecular bone lost with T1-DM, due to higher and longer-lasting increases in bone remodeling.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Central Arkansas Veterans Healthcare System, John L. McClellan Little Rock, AR, USA.
| | - Serra Ucer Ozgurel
- Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| | - Kevin McAndrews
- Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| | - Meloney Cregor
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Central Arkansas Veterans Healthcare System, John L. McClellan Little Rock, AR, USA; Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| | - Alma Villaseñor
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Madrid, Spain.
| | - Maricuz Mamani-Huanca
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Madrid, Spain.
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Madrid, Spain.
| | - Arancha Gortazar
- Bone Physiopathology laboratory, Applied Molecular Medicine Institute (IMMA), Universidad San Pablo-CEU, CEU Universities, Campus Monteprincipe, 28925 Alcorcón, Madrid, Spain.
| | - Amy Y Sato
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Central Arkansas Veterans Healthcare System, John L. McClellan Little Rock, AR, USA; Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| | - Teresita Bellido
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Central Arkansas Veterans Healthcare System, John L. McClellan Little Rock, AR, USA; Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Milovanovic P, Busse B. Micropetrosis: Osteocyte Lacunar Mineralization in Aging and Disease. Curr Osteoporos Rep 2023; 21:750-757. [PMID: 37917286 DOI: 10.1007/s11914-023-00832-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
PURPOSE OF REVIEW As the importance of osteocytes for bone mineral homeostasis is increasingly recognized, there is growing interest in osteocyte cell death as a relevant indicator in various physiological and pathological conditions. Micropetrosis is an established term used to describe osteocyte lacunae that are filled with minerals following osteocyte death. While the early reports of micropetrosis were purely descriptive, there is now an increasing body of literature showing quantitative data on micropetrosis in various conditions such as aging, osteoporosis, immobilization, and diabetes, and in osteoporosis treatment (denosumab and bisphosphonates). This review summarizes quantitative findings on micropetrosis, with a particular emphasis on the recent advances in the field. RECENT FINDINGS There is growing evidence that micropetrosis is more common in older, osteoporotic, and immobilized individuals, as well as in individuals with type 1 or type 2 diabetes. Denosumab and bisphosphonates seem to affect lacunar mineralization differently, where specifically bisphosphonates have been shown to prolong osteocyte viability and reduce micropetrosis. Despite continuous proceedings in the field of osteocyte-lacunar-network characteristics, more studies are necessary to further clarify the mechanisms of lacunar mineralization, the inter-site variability of micropetrosis accumulation, the relevance of micropetrosis in various diseases and conditions, and whether micropetrosis could be an indicator of bone fragility or a target for treatment.
Collapse
Affiliation(s)
- Petar Milovanovic
- Center of Bone Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Librizzi M, Naselli F, Abruscato G, Luparello C, Caradonna F. Parathyroid Hormone Related Protein (PTHrP)-Associated Molecular Signatures in Tissue Differentiation and Non-Tumoral Diseases. BIOLOGY 2023; 12:950. [PMID: 37508381 PMCID: PMC10376784 DOI: 10.3390/biology12070950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/14/2023] [Accepted: 07/01/2023] [Indexed: 07/30/2023]
Abstract
Parathyroid-hormone-related protein (PTHrP) is encoded by the PTHLH gene which, via alternative promoter usage and splicing mechanisms, can give rise to at least three isoforms of 139, 141, and 173 amino acids with distinct C-terminals. PTHrP is subjected to different post-translational processing that generates smaller bioactive forms, comprising amino terminus, mid-region (containing a nuclear/nucleolar targeting signal), and carboxy terminus peptides. Both the full-length protein and the discrete peptides are key controllers of viability, proliferation, differentiation, and apoptosis in diverse normal and pathological biological systems via the reprogramming of gene expression and remodulation of PKA or PKC-mediated signalization mechanisms. The aim of this review is to pick up selected studies on PTHrP-associated signatures as revealed by molecular profiling assays, focusing on the available data about exemplary differentiating, differentiated, or nontumoral cell and tissue models. In particular, the data presented relate to adipose, bone, dental, cartilaginous, and skin tissues, as well as intestinal, renal, hepatic, pulmonary, and pancreatic epithelia, with a focus on hepatic fibrosis-, pancreatitis-, and diabetes-related changes as diseased states. When reported, the biochemical and/or physiological aspects associated with the specific molecular modulation of gene expression and signal transduction pathways in the target model systems under examination are also briefly described.
Collapse
Affiliation(s)
- Mariangela Librizzi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Flores Naselli
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Giulia Abruscato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Fabio Caradonna
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| |
Collapse
|
5
|
Dragoun Kolibová S, Wölfel EM, Hemmatian H, Milovanovic P, Mushumba H, Wulff B, Neidhardt M, Püschel K, Failla AV, Vlug A, Schlaefer A, Ondruschka B, Amling M, Hofbauer LC, Rauner M, Busse B, Jähn-Rickert K. Osteocyte apoptosis and cellular micropetrosis signify skeletal aging in type 1 diabetes. Acta Biomater 2023; 162:254-265. [PMID: 36878337 DOI: 10.1016/j.actbio.2023.02.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/03/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Bone fragility is a profound complication of type 1 diabetes mellitus (T1DM), increasing patient morbidity. Within the mineralized bone matrix, osteocytes build a mechanosensitive network that orchestrates bone remodeling; thus, osteocyte viability is crucial for maintaining bone homeostasis. In human cortical bone specimens from individuals with T1DM, we found signs of accelerated osteocyte apoptosis and local mineralization of osteocyte lacunae (micropetrosis) compared with samples from age-matched controls. Such morphological changes were seen in the relatively young osteonal bone matrix on the periosteal side, and micropetrosis coincided with microdamage accumulation, implying that T1DM drives local skeletal aging and thereby impairs the biomechanical competence of the bone tissue. The consequent dysfunction of the osteocyte network hampers bone remodeling and decreases bone repair mechanisms, potentially contributing to the enhanced fracture risk seen in individuals with T1DM. STATEMENT OF SIGNIFICANCE: Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease that causes hyperglycemia. Increased bone fragility is one of the complications associated with T1DM. Our latest study on T1DM-affected human cortical bone identified the viability of osteocytes, the primary bone cells, as a potentially critical factor in T1DM-bone disease. We linked T1DM with increased osteocyte apoptosis and local accumulation of mineralized lacunar spaces and microdamage. Such structural changes in bone tissue suggest that T1DM speeds up the adverse effects of aging, leading to the premature death of osteocytes and potentially contributing to diabetes-related bone fragility.
Collapse
Affiliation(s)
- Sofie Dragoun Kolibová
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Eva Maria Wölfel
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany; Interdisciplinary Competence Center for Interface Research, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Haniyeh Hemmatian
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Petar Milovanovic
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany; University of Belgrade, Faculty of Medicine, Center of Bone Biology, Institute of Anatomy, Belgrade, Serbia
| | - Herbert Mushumba
- University Medical Center Hamburg Eppendorf, Institute of Legal Medicine, Hamburg, Germany
| | - Birgit Wulff
- University Medical Center Hamburg Eppendorf, Institute of Legal Medicine, Hamburg, Germany
| | - Maximilian Neidhardt
- Institute of Medical Technology and Intelligent Systems, Hamburg University of Technology, Am-Schwarzenberg-Campus 3, Hamburg 21073, Germany; Interdisciplinary Competence Center for Interface Research, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Klaus Püschel
- University Medical Center Hamburg Eppendorf, Institute of Legal Medicine, Hamburg, Germany
| | - Antonio Virgilio Failla
- University Medical Center Hamburg Eppendorf, UKE microscopy Imaging facility, Hamburg, Germany
| | - Annegreet Vlug
- Leiden University Medical Center (LUMC), Centre for Bone Quality, Leiden, the Netherlands
| | - Alexander Schlaefer
- Institute of Medical Technology and Intelligent Systems, Hamburg University of Technology, Am-Schwarzenberg-Campus 3, Hamburg 21073, Germany; Interdisciplinary Competence Center for Interface Research, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Benjamin Ondruschka
- University Medical Center Hamburg Eppendorf, Institute of Legal Medicine, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology, Department of Medicine III, Technische Universität Dresden Medical Center, Diabetes, and Metabolic Bone Diseases, Dresden, Germany
| | - Martina Rauner
- Division of Endocrinology, Department of Medicine III, Technische Universität Dresden Medical Center, Diabetes, and Metabolic Bone Diseases, Dresden, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany; Interdisciplinary Competence Center for Interface Research, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| | - Katharina Jähn-Rickert
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany; Mildred Scheel Cancer Career Center Hamburg, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| |
Collapse
|
6
|
Yao S, Du Z, Xiao L, Yan F, Ivanovski S, Xiao Y. Morphometric Changes of Osteocyte Lacunar in Diabetic Pig Mandibular Cancellous Bone. Biomolecules 2022; 13:biom13010049. [PMID: 36671434 PMCID: PMC9856050 DOI: 10.3390/biom13010049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Osteocytes play an important role in bone metabolism. The interactions of osteocytes with the surrounding microenvironment can alter cellular and lacunar morphological changes. However, objective quantification of osteocyte lacunae is challenging due to their deep location in the bone matrix. This project established a novel method for the analytical study of osteocytes/lacunae, which was then used to evaluate the osteocyte morphological changes in diabetic pig mandibular bone. Eight miniature pigs were sourced, and diabetes was randomly induced in four animals using streptozotocin (STZ) administration. The mandibular tissues were collected and processed. The jawbone density was evaluated with micro-CT. Osteocyte lacunae were effectively acquired and identified using backscattered electron scanning microscopy (BSE). A significantly decreased osteocyte lacunae size was found in the diabetic group. Using the acid etching method, it was demonstrated that the area of osteocyte and lacunae, and the pericellular areas were both significantly reduced in the diabetes group. In conclusion, a standard and relatively reliable method for analyzing osteocyte/lacunae morphological changes under compromised conditions has been successfully established. This method demonstrates that diabetes can significantly decrease osteocyte/lacunae size in a pig's mandibular cancellous bone.
Collapse
Affiliation(s)
- Sheng Yao
- School of Mechanical, Medical and Process Engineering, Center of Biomedical Technology, Queensland University of Technology, Brisbane, QLD 4059, Australia
- The First Hospital of Wuhan, Wuhan 430033, China
| | - Zhibin Du
- School of Mechanical, Medical and Process Engineering, Center of Biomedical Technology, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Lan Xiao
- School of Mechanical, Medical and Process Engineering, Center of Biomedical Technology, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Saso Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Yin Xiao
- School of Mechanical, Medical and Process Engineering, Center of Biomedical Technology, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Medicine and Dentistry & Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia
- Correspondence:
| |
Collapse
|
7
|
Hatch JM, Segvich DM, Kohler R, Wallace JM. Skeletal manifestations in a streptozotocin-induced C57BL/6 model of Type 1 diabetes. Bone Rep 2022; 17:101609. [PMID: 35941910 PMCID: PMC9356200 DOI: 10.1016/j.bonr.2022.101609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/27/2022] [Accepted: 07/30/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetes Mellitus is a metabolic disease which profoundly affects many organ systems in the body, including the skeleton. As is often the case with biology, there are inherent differences between the sexes when considering skeletal development and disease progression and outcome. Therefore, the aim of this study was to develop a protocol to reliably induce diabetes in both sexes of the C57BL/6 mouse utilizing streptozotocin (STZ) and to characterize the resulting bone phenotype. We hypothesized that destruction of the β-cells in the pancreatic islet by STZ would result in a diabetic state with downstream skeletal manifestations. Beginning at 8 weeks of age, mice were injected for 5 consecutive days with STZ (65 mg/kg males, 90 mg/kg females) dissolved in a citrate buffer. The diabetic state of the mice was monitored for 5 weeks to ensure persistent hyperglycemia and mice were euthanized at 15 weeks of age. Diabetes was confirmed through blood glucose monitoring, glucose and insulin tolerance testing, HbA1c measurement, and histological staining of the pancreas. The resulting bone phenotype was characterized using microcomputed tomography to assess bone structure, and whole bone mechanical testing to assess bone functional integrity. Mice from both sexes experienced loss of β-cell mass and increased glycation of hemoglobin, as well as reduced trabecular thickness and trabecular tissues mineral density (TMD), and reduced cortical thickness and cortical bone area fraction. In female mice the change area fraction was driven by a reduction in overall bone size while in male mice, the change was driven by increased marrow area. Males also experienced reduced cortical TMD. Mechanical bending tests of the tibiae showed significant results in females with a reduction in yield force and ultimate force driving lower work to yield and total work and a roughly 40 % reduction of stiffness. When tissue level parameters were estimated using beam theory, there was a significant reduction in yield and ultimate stresses as well as elastic modulus. The previously reported mechanistic similarity in the action of STZ on murine animals, as well as the ease of STZ administration via IP injection make this model is a strong candidate for future exploration of osteoporotic bone disease, Diabetes Mellitus, and the link between estrogen and glucose sensitivity.
Collapse
Affiliation(s)
- Jennifer M. Hatch
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Dyann M. Segvich
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Rachel Kohler
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
8
|
Ebrahim N, El-Halim HEA, Helal OK, El-Azab NEE, Badr OAM, Hassouna A, Saihati HAA, Aborayah NH, Emam HT, El-Wakeel HS, Aljasir M, El-Sherbiny M, Sarg NAS, Shaker GA, Mostafa O, Sabry D, Fouly MAK, Forsyth NR, Elsherbiny NM, Salim RF. Effect of bone marrow mesenchymal stem cells-derived exosomes on diabetes-induced retinal injury: Implication of Wnt/ b-catenin signaling pathway. Biomed Pharmacother 2022; 154:113554. [PMID: 35987163 DOI: 10.1016/j.biopha.2022.113554] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a serious microvascular complication of diabetes mellitus. Mesenchymal stem cells are currently studied as therapeutic strategy for management of DR. Exosomes, considered as a promising cell-free therapy option, display biological functions similar to those of their parent cells. In retinal development, Wnt/b-catenin signaling provides key cues for functional progression. The present study aimed to evaluate the potential efficacy of bone marrow-derived mesenchymal stem cell-derived exosomes (BM-MSCs-Ex) in diabetes-induced retinal injury via modulation of the Wnt/ b-catenin signaling pathway. METHODS Eighty-one rats were allocated into 6 groups (control, DR, DR + DKK1, DR + exosomes, DR + Wnt3a and DR + exosomes+Wnt3a). Evaluation of each group was via histopathological examination, assessment of gene and/or protein expression concerned with oxidative stress (SOD1, SOD2, Nox2, Nox4, iNOS), inflammation (TNF-α, ICAM-1, NF-κB) and angiogenesis (VEGF, VE-cadherin). RESULTS Results demonstrated that exosomes blocked the wnt/b-catenin pathway in diabetic retina concomitant with significant reduction of features of DR as shown by downregulation of retinal oxidants, upregulation of antioxidant enzymes, suppression of retinal inflammatory and angiogenic markers. These results were further confirmed by histopathological results, fundus examination and optical coherence tomography. Additionally, exosomes ameliorative effects abrogated wnt3a-triggered retinal injury in DR. CONCLUSION Collectively, these data demonstrated that exosomes ameliorated diabetes-induced retinal injury via suppressing Wnt/ b-catenin signaling with subsequent reduction of oxidative stress, inflammation and angiogenesis.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt; Stem Cell Unit, Faculty of Medicine, Benha University, Egypt.
| | | | - Omayma Kamel Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt
| | | | - Omnia A M Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Egypt.
| | - Amira Hassouna
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland, New Zealand.
| | - Hajir A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Saudi Arabia.
| | | | - Hanan Tawfeek Emam
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Egypt.
| | - Hend S El-Wakeel
- Department of Physiology, Faculty of Medicine, Benha University, Egypt.
| | - Mohammad Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Naglaa A S Sarg
- Department of Anatomy, Benha Faculty of Medicine, Benha University, Egypt.
| | - Gehan Ahmed Shaker
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt.
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University, Cairo 11562, Egypt.
| | | | - Nicholas Robert Forsyth
- Guy Hilton Research Laboratories, School of Pharmacy and Bioengineering, Faculty of Medicine and Health Sciences, Keele University, Newcastle ST5 5BG, UK.
| | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Biochemistry department, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Egypt.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Better understanding of the mechanisms underlying skeletal dysfunction in the context of diabetes is needed to guide the development of therapeutic interventions to reduce the burden of diabetic fractures. Osteocytes, the 'master regulators' of bone remodeling, have emerged as key culprits in the pathogenesis of diabetes-related skeletal fragility. RECENT FINDINGS Both type 1 diabetes and type 2 diabetes cause chronic hyperglycemia that, over time, reduces bone quality and bone formation. In addition to acting as mechanosensors, osteocytes are important regulators of osteoblast and osteoclast activities; however, diabetes leads to osteocyte dysfunction. Indeed, diabetes causes the accumulation of advanced glycation end-products and senescent cells that can affect osteocyte viability and functions via increased receptor for advanced glycation endproducts (RAGE) signaling or the production of a pro-inflammatory senescence-associated secretory phenotype. These changes may increase osteocyte-derived sclerostin production and decrease the ability of osteocytes to sense mechanical stimuli thereby contributing to poor bone quality in humans with diabetes. SUMMARY Osteocyte dysfunction exists at the nexus of diabetic skeletal disease. Therefore, interventions targeting the RAGE signaling pathway, senescent cells, and those that inhibit sclerostin or mechanically stimulate osteocytes may alleviate the deleterious effects of diabetes on osteocytes and bone quality.
Collapse
Affiliation(s)
| | | | - Joshua N. Farr
- Correspondence: Joshua N. Farr, , Mayo Clinic, Guggenheim 7-11D, 200 First Street SW, Rochester, MN 55905, Telephone: 507-538-0085
| |
Collapse
|
10
|
Jeddi S, Yousefzadeh N, Kashfi K, Ghasemi A. Role of nitric oxide in type 1 diabetes-induced osteoporosis. Biochem Pharmacol 2021; 197:114888. [PMID: 34968494 DOI: 10.1016/j.bcp.2021.114888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D)-induced osteoporosis is characterized by decreased bone mineral density, bone quality, rate of bone healing, bone formation, and increased bone resorption. Patients with T1D have a 2-7-fold higher risk of osteoporotic fracture. The mechanisms leading to increased risk of osteoporotic fracture in T1D include insulin deficiency, hyperglycemia, insulin resistance, lower insulin-like growth factor-1, hyperglycemia-induced oxidative stress, and inflammation. In addition, a higher probability of falling, kidney dysfunction, weakened vision, and neuropathy indirectly increase the risk of osteoporotic fracture in T1D patients. Decreased nitric oxide (NO) bioavailability contributes to the pathophysiology of T1D-induced osteoporotic fracture. This review discusses the role of NO in osteoblast-mediated bone formation and osteoclast-mediated bone resorption in T1D. In addition, the mechanisms involved in reduced NO bioavailability and activity in type 1 diabetic bones as well as NO-based therapy for T1D-induced osteoporosis are summarized. Available data indicates that lower NO bioavailability in diabetic bones is due to disruption of phosphatidylinositol 3‑kinase/protein kinase B/endothelial NO synthases and NO/cyclic guanosine monophosphate/protein kinase G signaling pathways. Thus, NO bioavailability may be boosted directly or indirectly by NO donors. As NO donors with NO-like effects in the bone, inorganic nitrate and nitrite can potentially be used as novel therapeutic agents for T1D-induced osteoporosis. Inorganic nitrites and nitrates can decrease the risk for osteoporotic fracture probably directly by decreasing osteoclast activity, decreasing fat accumulation in the marrow cavity, increasing osteoblast activity, and increasing bone perfusion or indirectly, by improving hyperglycemia, insulin resistance, and reducing body weight.
Collapse
Affiliation(s)
- Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA.
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Leanza G, Fontana F, Lee SY, Remedi MS, Schott C, Ferron M, Hamilton-Hall M, Alippe Y, Strollo R, Napoli N, Civitelli R. Gain-of-Function Lrp5 Mutation Improves Bone Mass and Strength and Delays Hyperglycemia in a Mouse Model of Insulin-Deficient Diabetes. J Bone Miner Res 2021; 36:1403-1415. [PMID: 33831261 PMCID: PMC8360087 DOI: 10.1002/jbmr.4303] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/21/2021] [Accepted: 03/28/2021] [Indexed: 01/26/2023]
Abstract
High fracture rate and high circulating levels of the Wnt inhibitor, sclerostin, have been reported in diabetic patients. We studied the effects of Wnt signaling activation on bone health in a mouse model of insulin-deficient diabetes. We introduced the sclerostin-resistant Lrp5A214V mutation, associated with high bone mass, in mice carrying the Ins2Akita mutation (Akita), which results in loss of beta cells, insulin deficiency, and diabetes in males. Akita mice accrue less trabecular bone mass with age relative to wild type (WT). Double heterozygous Lrp5A214V /Akita mutants have high trabecular bone mass and cortical thickness relative to WT animals, as do Lrp5A214V single mutants. Likewise, the Lrp5A214V mutation prevents deterioration of biomechanical properties occurring in Akita mice. Notably, Lrp5A214V /Akita mice develop fasting hyperglycemia and glucose intolerance with a delay relative to Akita mice (7 to 8 vs. 5 to 6 weeks, respectively), despite lack of insulin production in both groups by 6 weeks of age. Although insulin sensitivity is partially preserved in double heterozygous Lrp5A214V /Akita relative to Akita mutants up to 30 weeks of age, insulin-dependent phosphorylated protein kinase B (pAKT) activation in vitro is not altered by the Lrp5A214V mutation. Although white adipose tissue depots are equally reduced in both compound and Akita mice, the Lrp5A214V mutation prevents brown adipose tissue whitening that occurs in Akita mice. Thus, hyperactivation of Lrp5-dependent signaling fully protects bone mass and strength in prolonged hyperglycemia and improves peripheral glucose metabolism in an insulin independent manner. Wnt signaling activation represents an ideal therapeutic approach for diabetic patients at high risk of fracture. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Giulia Leanza
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Medicine, Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome, Italy
| | - Francesca Fontana
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Seung-Yon Lee
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Maria S Remedi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Céline Schott
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada.,Molecular Biology Programs & Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Mathieu Ferron
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada.,Molecular Biology Programs & Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Malcolm Hamilton-Hall
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yael Alippe
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Rocky Strollo
- Department of Medicine, Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome, Italy
| | - Nicola Napoli
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Medicine, Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome, Italy
| | - Roberto Civitelli
- Division of Bone and Mineral Diseases, Department of Medicine, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
12
|
Hildebrandt N, Colditz J, Dutra C, Goes P, Salbach-Hirsch J, Thiele S, Hofbauer LC, Rauner M. Role of osteogenic Dickkopf-1 in bone remodeling and bone healing in mice with type I diabetes mellitus. Sci Rep 2021; 11:1920. [PMID: 33479403 PMCID: PMC7820472 DOI: 10.1038/s41598-021-81543-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is associated with low bone mass and a higher risk for fractures. Dickkopf-1 (Dkk1), which inhibits Wnt signaling, osteoblast function, and bone formation, has been found to be increased in the serum of patients with T1DM. Here, we investigated the functional role of Dkk1 in T1DM-induced bone loss in mice. T1DM was induced in 10-week-old male mice with Dkk1-deficiency in late osteoblasts/osteocytes (Dkk1f/f;Dmp1-Cre, cKO) and littermate control mice by 5 subsequent injections of streptozotocin (40 mg/kg). Age-matched, non-diabetic control groups received citrate buffer instead. At week 12, calvarial defects were created in subgroups of each cohort. After a total of 16 weeks, weight, fat, the femoral bone phenotype and the area of the bone defect were analyzed using µCT and dynamic histomorphometry. During the experiment, diabetic WT and cKO mice did not gain body weight compared to control mice. Further they lost their perigonadal and subcutaneous fat pads. Diabetic mice had highly elevated serum glucose levels and impaired glucose tolerance, regardless of their Dkk1 levels. T1DM led to a 36% decrease in trabecular bone volume in Cre− negative control animals, whereas Dkk1 cKO mice only lost 16%. Of note, Dkk1 cKO mice were completely protected from T1DM-induced cortical bone loss. T1DM suppressed the bone formation rate, the number of osteoblasts at trabecular bone, serum levels of P1NP and bone defect healing in both, Dkk1-deficient and sufficient, mice. This may be explained by increased serum sclerostin levels in both genotypes and the strict dependence on bone formation for bone defect healing. In contrast, the number of osteoclasts and TRACP 5b serum levels only increased in diabetic control mice, but not in Dkk1 cKO mice. In summary, Dkk1 derived from osteogenic cells does not influence the development of T1DM but plays a crucial role in T1DM-induced bone loss in male mice by regulating osteoclast numbers.
Collapse
Affiliation(s)
- Nick Hildebrandt
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Juliane Colditz
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103, Leipzig, Germany
| | - Caio Dutra
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,Post-Graduation Program in Morphological Science, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Paula Goes
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,Department of Pathology and Legal Medicine, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Juliane Salbach-Hirsch
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
13
|
Yang M, Song B, Liu J, Bing Z, Wang Y, Yu L. Gene signature for prognosis in comparison of pancreatic cancer patients with diabetes and non-diabetes. PeerJ 2020; 8:e10297. [PMID: 33240632 PMCID: PMC7666560 DOI: 10.7717/peerj.10297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/13/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) has much weaker prognosis, which can be divided into diabetes and non-diabetes. PC patients with diabetes mellitus will have more opportunities for physical examination due to diabetes, while pancreatic cancer patients without diabetes tend to have higher risk. Identification of prognostic markers for diabetic and non-diabetic pancreatic cancer can improve the prognosis of patients with both types of pancreatic cancer. METHODS Both types of PC patients perform differently at the clinical and molecular levels. The Cancer Genome Atlas (TCGA) is employed in this study. The gene expression of the PC with diabetes and non-diabetes is used for predicting their prognosis by LASSO (Least Absolute Shrinkage and Selection Operator) Cox regression. Furthermore, the results are validated by exchanging gene biomarker with each other and verified by the independent Gene Expression Omnibus (GEO) and the International Cancer Genome Consortium (ICGC). The prognostic index (PI) is generated by a combination of genetic biomarkers that are used to rank the patient's risk ratio. Survival analysis is applied to test significant difference between high-risk group and low-risk group. RESULTS An integrated gene prognostic biomarker consisted by 14 low-risk genes and six high-risk genes in PC with non-diabetes. Meanwhile, and another integrated gene prognostic biomarker consisted by five low-risk genes and three high-risk genes in PC with diabetes. Therefore, the prognostic value of gene biomarker in PC with non-diabetes and diabetes are all greater than clinical traits (HR = 1.102, P-value < 0.0001; HR = 1.212, P-value < 0.0001). Gene signature in PC with non-diabetes was validated in two independent datasets. CONCLUSIONS The conclusion of this study indicated that the prognostic value of genetic biomarkers in PCs with non-diabetes and diabetes. The gene signature was validated in two independent databases. Therefore, this study is expected to provide a novel gene biomarker for predicting prognosis of PC with non-diabetes and diabetes and improving clinical decision.
Collapse
Affiliation(s)
- Mingjun Yang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, Gansu, China
| | - Boni Song
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, Gansu, China
- Institute of Modern Physics of Chinese Academy of Sciences, Lanzhou, China
| | - Juxiang Liu
- Gansu Key Laboratory of Endocrine and metabolism, Department of Endocrinology, Gansu Provincial People’s Hospital, Lanzhou, Gansu, China
| | - Zhitong Bing
- Institute of Modern Physics of Chinese Academy of Sciences, Lanzhou, China
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University,, Lanzhou, China
| | - Yonggang Wang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, Gansu, China
| | - Linmiao Yu
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, Gansu, China
| |
Collapse
|
14
|
Wang Y, Han B, Ding J, Qiu C, Wang W. Endoplasmic reticulum stress mediates osteocyte death under oxygen-glucose deprivation in vitro. Acta Histochem 2020; 122:151577. [PMID: 32778239 DOI: 10.1016/j.acthis.2020.151577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/06/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
As a vascularized organ, bone is known to be susceptible to ischemia. Ischemic osteonecrosis or skeletal unloading lead to ischemia in bone microenvironment that causes osteocytes to suffer hypoxia and nutrition deprivation. OBJECTIVE To explore the effects of Oxygen-glucose deprivation (OGD) on osteocytes and the potential mechanism. METHODS OGD model was established in cultured MLO-Y4 cell. Cell damage, intracellular oxidative stress and cell apoptosis were detected at different OGD times (0, 2, 4, 8, 12, 24 h), and the changes in endoplasmic reticulum (ER) stress-related indicators were observed. Furthermore, cells were treated with 4-phenylbutyrate sodium (4-PBA) to inhibit ER stress, and cell damage and oxidative stress level were detected. RESULTS The cell viability under OGD exhibited a significantly reduced in a time-dependent manner, and the level of intracellular reactive oxygen species (ROS) were increased, cell apoptosis and ER stress was induced. Inhibition of ER stress can reduce cell death and intracellular ROS levels. CONCLUSION Our study demonstrated that ER stress regulates OGD-induced apoptotic cell death in MLO-Y4 cells via intracellular ROS.
Collapse
|
15
|
Abstract
Parathyroid hormone (PTH), PTH-related peptide (PTHrP), PTHR, and their cognate G protein-coupled receptor play defining roles in the regulation of extracellular calcium and phosphate metabolism and in controlling skeletal growth and repair. Acting through complex signaling mechanisms that in many instances proceed in a tissue-specific manner, precise control of these processes is achieved. A variety of direct and indirect disease processes, along with genetic anomalies, can cause these schemes to become dysfunctional. Here, we review the basic components of this regulatory network and present both the well-established elements and emerging findings and concepts with the overall objective to provide a framework for understanding the elementary aspects of how PTH and PTHrP behave and as a call to encourage further investigation that will yield more comprehensive understanding of the physiological and pathological steps at play, with a goal toward novel therapeutic interventions.
Collapse
|
16
|
Costantini S, Conte C. Bone health in diabetes and prediabetes. World J Diabetes 2019; 10:421-445. [PMID: 31523379 PMCID: PMC6715571 DOI: 10.4239/wjd.v10.i8.421] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/03/2019] [Accepted: 07/20/2019] [Indexed: 02/05/2023] Open
Abstract
Bone fragility has been recognized as a complication of diabetes, both type 1 diabetes (T1D) and type 2 diabetes (T2D), whereas the relationship between prediabetes and fracture risk is less clear. Fractures can deeply impact a diabetic patient's quality of life. However, the mechanisms underlying bone fragility in diabetes are complex and have not been fully elucidated. Patients with T1D generally exhibit low bone mineral density (BMD), although the relatively small reduction in BMD does not entirely explain the increase in fracture risk. On the contrary, patients with T2D or prediabetes have normal or even higher BMD as compared with healthy subjects. These observations suggest that factors other than bone mass may influence fracture risk. Some of these factors have been identified, including disease duration, poor glycemic control, presence of diabetes complications, and certain antidiabetic drugs. Nevertheless, currently available tools for the prediction of risk inadequately capture diabetic patients at increased risk of fracture. Aim of this review is to provide a comprehensive overview of bone health and the mechanisms responsible for increased susceptibility to fracture across the spectrum of glycemic status, spanning from insulin resistance to overt forms of diabetes. The management of bone fragility in diabetic patient is also discussed.
Collapse
Affiliation(s)
- Silvia Costantini
- Department of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, Milan 20123, Italy
- Epatocentro Ticino, Lugano 6900, Switzerland
| | - Caterina Conte
- Department of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, Milan 20123, Italy
- IRCCS Ospedale San Raffaele, Internal Medicine and Transplantation, Milan 20123, Italy
| |
Collapse
|
17
|
Chen S, Liu D, He S, Yang L, Bao Q, Qin H, Liu H, Zhao Y, Zong Z. Differential effects of type 1 diabetes mellitus and subsequent osteoblastic β-catenin activation on trabecular and cortical bone in a mouse model. Exp Mol Med 2018; 50:1-14. [PMID: 30518745 PMCID: PMC6281645 DOI: 10.1038/s12276-018-0186-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/27/2018] [Accepted: 09/09/2018] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a pathological condition associated with osteopenia. WNT/β-catenin signaling is implicated in this process. Trabecular and cortical bone respond differently to WNT/β-catenin signaling in healthy mice. We investigated whether this signaling has different effects on trabecular and cortical bone in T1DM. We first established a streptozotocin-induced T1DM mouse model and then constitutively activated β-catenin in osteoblasts in the setting of T1DM (T1-CA). The extent of bone loss was greater in trabecular bone than that in cortical bone in T1DM mice, and this difference was consistent with the reduction in the expression of β-catenin signaling in the two bone compartments. Further experiments demonstrated that in T1DM mice, trabecular bone showed lower levels of insulin-like growth factor-1 receptor (IGF-1R) than the levels in cortical bone, leading to lower WNT/β-catenin signaling activity through the inhibition of the IGF-1R/Akt/glycogen synthase kinase 3β (GSK3β) pathway. After β-catenin was activated in T1-CA mice, the bone mass and bone strength increased to substantially greater extents in trabecular bone than those in cortical bone. In addition, the cortical bone of the T1-CA mice displayed an unexpected increase in bone porosity, with increased bone resorption. The downregulated expression of WNT16 might be responsible for these cortical bone changes. In conclusion, we found that although the activation of WNT/β-catenin signaling increased the trabecular bone mass and bone strength in T1DM mice, it also increased the cortical bone porosity, impairing the bone strength. These findings should be considered in the future treatment of T1DM-related osteopenia.
Collapse
Affiliation(s)
- Sixu Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China.,Department of Orthopedics, The 118th Hospital of the Chinese People's Liberation Army, 325000, Wenzhou, Zhejiang, China
| | - Daocheng Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China.,Department of Emergency, Xinqiao Hospital, Army Medical University, 400037, ChongQing, China
| | - Sihao He
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China.,Department of Emergency, Xinqiao Hospital, Army Medical University, 400037, ChongQing, China
| | - Lei Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China.,Department of Emergency, Xinqiao Hospital, Army Medical University, 400037, ChongQing, China
| | - Quanwei Bao
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China.,Department of Emergency, Xinqiao Hospital, Army Medical University, 400037, ChongQing, China
| | - Hao Qin
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China.,Department of Emergency, Xinqiao Hospital, Army Medical University, 400037, ChongQing, China
| | - Huayu Liu
- Department of Trauma Surgery, Daping Hospital, Army Medical University, 400042, ChongQing, China
| | - Yufeng Zhao
- Department of Trauma Surgery, Daping Hospital, Army Medical University, 400042, ChongQing, China
| | - Zhaowen Zong
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of War Wound Rescue Skills Training, Base of Army Health Service Training, Army Medical University, 400038, ChongQing, China. .,Department of Emergency, Xinqiao Hospital, Army Medical University, 400037, ChongQing, China.
| |
Collapse
|
18
|
Napoli N, Strollo R, Defeudis G, Leto G, Moretti C, Zampetti S, D'Onofrio L, Campagna G, Palermo A, Greto V, Manfrini S, Hawa MI, Leslie RD, Pozzilli P, Buzzetti R. Serum Sclerostin and Bone Turnover in Latent Autoimmune Diabetes in Adults. J Clin Endocrinol Metab 2018; 103:1921-1928. [PMID: 29506222 DOI: 10.1210/jc.2017-02274] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/26/2018] [Indexed: 01/15/2023]
Abstract
PURPOSE Bone formation is impaired in both type 1 diabetes and type 2 diabetes (T2D), whereas sclerostin, an antagonist of bone formation, is increased in T2D only. No data are available on latent autoimmune diabetes in adults (LADA), an autoimmune type of diabetes that may clinically resemble T2D at diagnosis. We evaluated serum sclerostin and bone turnover markers in LADA compared with those in T2D and whether metabolic syndrome (MetS) affects sclerostin in T2D or LADA. METHODS This cross-sectional study included 98 patients with T2D and 89 with LADA from the Action LADA and Non Insulin Requiring Autoimmune Diabetes cohorts. Patients were further divided according to MetS status. Nondiabetic participants (n = 53) were used as controls. Serum sclerostin, bone formation (pro-collagen type 1 N-terminal propeptide [P1NP]), and bone resorption (C-terminal telopeptide of type I collagen [CTX]) were analyzed. RESULTS Patients with T2D had higher sclerostin than did those with LADA [P = 0.0008, adjusted for sex and body mass index (BMI)], even when analysis was restricted to patients with MetS (adjusted P = 0.03). Analysis of T2D and LADA groups separately showed that sclerostin was similar between those with and those without MetS. However, a positive trend between sclerostin and number of MetS features was seen with T2D (P for trend = 0.001) but not with LADA. Patients with T2D or LADA had lower CTX than did controls (P = 0.0003) and did not have significantly reduced P1NP. Sclerostin was unrelated to age or hemoglobin A1c but was correlated with BMI (ρ = 0.29; P = 0.0001), high-density lipoprotein (ρ = -0.23; P = 0.003), triglycerides (ρ = 0.19; P = 0.002), and time since diagnosis (ρ = 0.32; P < 0.0001). CONCLUSIONS Patients with LADA presented lower bone resorption than did controls, similar to patients with T2D. Sclerostin is increased in T2D but not in LADA, suggesting possible roles on bone metabolism in T2D only.
Collapse
Affiliation(s)
- Nicola Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
- I.R.C.C.S. Istituto Ortopedico Galeazzi, Milan, Italy
| | - Rocky Strollo
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, London, United Kingdom
| | - Giuseppe Defeudis
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
| | - Gaetano Leto
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Chiara Moretti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Simona Zampetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Luca D'Onofrio
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Giuseppe Campagna
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Andrea Palermo
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
| | - Valentina Greto
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
| | - Silvia Manfrini
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
| | - Mohammed I Hawa
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, London, United Kingdom
| | - R David Leslie
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, London, United Kingdom
| | - Paolo Pozzilli
- Unit of Endocrinology and Diabetes, Department of Medicine, Universitá Campus Bio-Medico di Roma, Rome, Italy
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, London, United Kingdom
| | - Raffaella Buzzetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | | |
Collapse
|
19
|
Ma XY, Wen XX, Yang XJ, Zhou DP, Wu Q, Feng YF, Ding HJ, Lei W, Yu HL, Liu B, Xiang LB, Wang TS. Ophiopogonin D improves osteointegration of titanium alloy implants under diabetic conditions by inhibition of ROS overproduction via Wnt/β-catenin signaling pathway. Biochimie 2018; 152:31-42. [PMID: 29705132 DOI: 10.1016/j.biochi.2018.04.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 04/24/2018] [Indexed: 01/24/2023]
Abstract
A high failure rate of titanium implants in diabetic patients has been indicated in clinical evidences. Excessive oxidative stress at the bone-implant interface plays an important role in the impaired osteointegration under diabetic conditions. While the underlying mechanisms remain unknown and the targeted treatments are urgently needed. Ophiopogonin D (OP-D), isolated from Chinese herbal Radix Ophiopogon japonicus, is generally reported to be a potent antioxidant agent. In the present study, we hypothesized that OP-D exerted promotive effects on osteointegration against oxidative stress, and investigated the underlying mechanisms associated with alteration of Wnt/β-catenin signaling pathway. Rabbit osteoblasts incubated on titanium alloy implant were co-cultured with normal serum (NS), diabetic serum (DS), DS + OP-D, DS + NAC (a potent ROS inhibitor) and DS + OP-D + Dkk1 (a Wnt inhibitor) for examinations of osteoblast behaviors. For in vivo study, titanium alloy implants were implanted into the femoral condyle defects on diabetic rabbits. Results demonstrated that diabetes-induced oxidative stress resulted in osteoblast dysfunctions and apoptotic injury at the bone-implant interface, concomitant with the inactivation of Wnt/β-catenin signaling. Importantly, OP-D administration attenuated oxidative stress, directly reactivating Wnt/β-catenin signaling. Osteoblast dysfunctions were thus reversed as evidenced by improved osteoblast adhesion, proliferation and differentiation, and ameliorated apoptotic injury, exerting similar effects to NAC treatment. In addition, the positive effects afforded by OP-D were confirmed by improved osteointegration and oetogenesis within the titanium alloy implants in vivo by Micro-CT and histological analyses. Furthermore, the pro-osteogenic effects of OP-D were almost completely abolished by the Wnt inhibitor Dkk1. These results demonstrated, for the first time, OP-D administration alleviated the damaged osteointegration of titanium alloy implants under diabetic conditions by means of inhibiting oxidative stress via a Wnt/β-catenin-dependent mechanism. The OP-D administration would become a reliable treatment strategy for implant failure therapy in diabetics due to the optimal anti-oxidative and pro-osteogenic properties.
Collapse
Affiliation(s)
- Xiang-Yu Ma
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang, Liaoning, 110016, China; Department of Orthopedics of the 463 Hospital of PLA, Shenyang, Liaoning, 110042, China.
| | - Xin-Xin Wen
- Department of Orthopedics of the 463 Hospital of PLA, Shenyang, Liaoning, 110042, China
| | - Xiao-Jiang Yang
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Da-Peng Zhou
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang, Liaoning, 110016, China
| | - Qiong Wu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shengyang, 110164, China
| | - Ya-Fei Feng
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hai-Jiao Ding
- Department of Orthopedics of the 463 Hospital of PLA, Shenyang, Liaoning, 110042, China
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hai-Long Yu
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang, Liaoning, 110016, China
| | - Bing Liu
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang, Liaoning, 110016, China
| | - Liang-Bi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang, Liaoning, 110016, China.
| | - Tian-Sheng Wang
- Department of Orthopedics of the 463 Hospital of PLA, Shenyang, Liaoning, 110042, China.
| |
Collapse
|
20
|
Portal-Núñez S, Ardura JA, Lozano D, Martínez de Toda I, De la Fuente M, Herrero-Beaumont G, Largo R, Esbrit P. Parathyroid hormone-related protein exhibits antioxidant features in osteoblastic cells through its N-terminal and osteostatin domains. Bone Joint Res 2018; 7:58-68. [PMID: 29330344 PMCID: PMC5805825 DOI: 10.1302/2046-3758.71.bjr-2016-0242.r2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objectives Oxidative stress plays a major role in the onset and progression of involutional osteoporosis. However, classical antioxidants fail to restore osteoblast function. Interestingly, the bone anabolism of parathyroid hormone (PTH) has been shown to be associated with its ability to counteract oxidative stress in osteoblasts. The PTH counterpart in bone, which is the PTH-related protein (PTHrP), displays osteogenic actions through both its N-terminal PTH-like region and the C-terminal domain. Methods We examined and compared the antioxidant capacity of PTHrP (1-37) with the C-terminal PTHrP domain comprising the 107-111 epitope (osteostatin) in both murine osteoblastic MC3T3-E1 cells and primary human osteoblastic cells. Results We showed that both N- and C-terminal PTHrP peptides at 100 nM decreased reactive oxygen species production and forkhead box protein O activation following hydrogen peroxide (H2O2)-induced oxidation, which was related to decreased lipid oxidative damage and caspase-3 activation in these cells. This was associated with their ability to restore the deleterious effects of H2O2 on cell growth and alkaline phosphatase activity, as well as on the expression of various osteoblast differentiation genes. The addition of Rp-cyclic 3′,5′-hydrogen phosphorothioate adenosine triethylammonium salt (a cyclic 3',5'-adenosine monophosphate antagonist) and calphostin C (a protein kinase C inhibitor), or a PTH type 1 receptor antagonist, abrogated the effects of N-terminal PTHrP, whereas protein phosphatase 1 (an Src kinase activity inhibitor), SU1498 (a vascular endothelial growth factor receptor 2 inhibitor), or an anti osteostatin antiserum, inhibited the effects of C-terminal PTHrP. Conclusion These findings indicate that the antioxidant properties of PTHrP act through its N- and C-terminal domains and provide novel insights into the osteogenic action of PTHrP. Cite this article: S. Portal-Núñez, J. A. Ardura, D. Lozano, I. Martínez de Toda, M. De la Fuente, G. Herrero-Beaumont, R. Largo, P. Esbrit. Parathyroid hormone-related protein exhibits antioxidant features in osteoblastic cells through its N-terminal and osteostatin domains. Bone Joint Res 2018;7:58–68. DOI: 10.1302/2046-3758.71.BJR-2016-0242.R2.
Collapse
Affiliation(s)
- S Portal-Núñez
- Bone and Joint Research Unit, The Institution of Health Research (IIS)-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - J A Ardura
- The Institution of Applied Molecular Medicine (IMMA), Universidad San Pablo CEU Madrid, Spain
| | - D Lozano
- Department of Inorganic and Bioinorganic Chemistry, Complutense University, Madrid, Spain
| | - I Martínez de Toda
- Animal Physiology II. Faculty of Biology, Complutense University, Madrid, Spain
| | - M De la Fuente
- Animal Physiology II. Faculty of Biology, Complutense University, Madrid, Spain
| | - G Herrero-Beaumont
- Bone and Joint Research Unit, The Institution of Health Research (IIS)-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - R Largo
- Bone and Joint Research Unit, The Institution of Health Research (IIS)-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - P Esbrit
- Bone and Joint Research Unit, The Institution of Health Research (IIS)-Fundación Jiménez Díaz, UAM, Madrid, Spain
| |
Collapse
|
21
|
López-Herradón A, Fujikawa R, Gómez-Marín M, Stedile-Lovatel JP, Mulero F, Ardura JA, Ruiz P, Muñoz I, Esbrit P, Mahíllo-Fernández I, Ortega-de Mues A. Impact of Chiropractic Manipulation on Bone and Skeletal Muscle of Ovariectomized Rats. Calcif Tissue Int 2017; 101:519-529. [PMID: 28755011 DOI: 10.1007/s00223-017-0304-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/12/2017] [Indexed: 01/01/2023]
Abstract
Evidence suggests that chiropractic manipulation might exert positive effects in osteoporotic patients. The aim of this study was to evaluate the effects of chiropractic manipulation on bone structure and skeletal muscle in rats with bone loss caused by ovariectomy (OVX). The 6-month old Sprague-Dawley rats at 10 weeks following OVX or sham operation (Sh) did not suffer chiropractic manipulation (NM group) or were submitted to true chiropractic manipulation using the chiropractic adjusting instrument Activator V® three times/week for 6 weeks as follows: Force 1 setting was applied onto the tibial tubercle of the rat right hind limb (TM group), whereas the corresponding left hind limb received a false manipulation (FM group) consisting of ActivatorV® firing in the air and slightly touching the tibial tubercle. Bone mineral density (BMD) and bone mineral content (BMC) were determined in long bones and L3-L4 vertebrae in all rats. Femora and tibia were analyzed by μCT. Mechano growth factor (MGF) was detected in long bones and soleus, quadriceps and tibial muscles by immunohistochemistry and Western blot. The decrease of BMD and BMC as well as trabecular bone impairment in the long bones of OVX rats vs Sh controls was partially reversed in the TM group versus FM or NM rats. This bone improvement by chiropractic manipulation was associated with an increased MGF expression in the quadriceps and the anterior tibial muscle in OVX rats. These findings support the notion that chiropractic manipulation can ameliorate osteoporotic bone at least partly by targeting skeletal muscle.
Collapse
Affiliation(s)
- A López-Herradón
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - R Fujikawa
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain
| | - M Gómez-Marín
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain
| | - J P Stedile-Lovatel
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain
| | - F Mulero
- Unidad de Imagen Molecular, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - J A Ardura
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - P Ruiz
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain
| | - I Muñoz
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain
| | - P Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - I Mahíllo-Fernández
- Unidad de Epidemiología y Bioestadística, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - A Ortega-de Mues
- Madrid College of Chiropractic, Real Centro Universitario Escorial-María Cristina, Paseo de los Alamillos, 2, San Lorenzo de El Escorial, 28200, Madrid, Spain.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The goal of this paper is to evaluate critically the literature published over the past 3 years regarding the Wnt signaling pathway. The Wnt pathway was found to be involved in bone biology in 2001-2002 with the discovery of a (G171V) mutation in the lipoprotein receptor-related protein 5 (LRP5) that resulted in high bone mass and another mutation that completely inactivated Lrp5 function and resulted in osteoporosis pseudoglioma syndrome (OPPG). The molecular biology has been complex, and very interesting. It has provided many opportunities for exploitation to develop new clinical treatments, particularly for osteoporosis. More clinical possibilities include: treatments for fracture healing, corticosteroid osteoporosis, osteogenesis imperfecta, and others. In addition, we wish to provide historical information coming from distant publications (~350 years ago) regarding bone biology that have been confirmed by study of Wnt signaling. RECENT FINDINGS A recent finding is the development of an antibody to sclerostin that is under study as a treatment for osteoporosis. Development of treatments for other forms of osteoporosis, such as corticosteroid osteoporosis, is also underway. The full range of the applications of the work is not yet been achieved.
Collapse
Affiliation(s)
- Mark L Johnson
- Department of Oral and Craniofacial Sciences, UMKC School of Dentistry, 650 East 25th Street, Kansas City, MO, 64108, USA
| | - Robert R Recker
- Creighton University, 601 N 30th St., Ste 4841, Omaha, NE, 68131, USA.
| |
Collapse
|
23
|
Maycas M, McAndrews KA, Sato AY, Pellegrini GG, Brown DM, Allen MR, Plotkin LI, Gortazar AR, Esbrit P, Bellido T. PTHrP-Derived Peptides Restore Bone Mass and Strength in Diabetic Mice: Additive Effect of Mechanical Loading. J Bone Miner Res 2017; 32:486-497. [PMID: 27683064 DOI: 10.1002/jbmr.3007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 01/17/2023]
Abstract
There is an unmet need to understand the mechanisms underlying skeletal deterioration in diabetes mellitus (DM) and to develop therapeutic approaches to treat bone fragility in diabetic patients. We demonstrate herein that mice with type 1 DM induced by streptozotocin exhibited low bone mass, inferior mechanical and material properties, increased bone resorption, decreased bone formation, increased apoptosis of osteocytes, and increased expression of the osteocyte-derived bone formation inhibitor Sost/sclerostin. Further, short treatment of diabetic mice with parathyroid hormone related protein (PTHrP)-derived peptides corrected these changes to levels undistinguishable from non-diabetic mice. In addition, diabetic mice exhibited reduced bone formation in response to mechanical stimulation, which was corrected by treatment with the PTHrP peptides, and higher prevalence of apoptotic osteocytes, which was reduced by loading or by the PTHrP peptides alone and reversed by a combination of loading and PTHrP peptide treatment. In vitro experiments demonstrated that the PTHrP peptides or mechanical stimulation by fluid flow activated the survival kinases ERKs and induced nuclear translocation of the canonical Wnt signaling mediator β-catenin, and prevented the increase in osteocytic cell apoptosis induced by high glucose. Thus, PTHrP-derived peptides cross-talk with mechanical signaling pathways to reverse skeletal deterioration induced by DM in mice. These findings suggest a crucial role of osteocytes in the harmful effects of diabetes on bone and raise the possibility of targeting these cells as a novel approach to treat skeletal deterioration in diabetes. Moreover, our study suggests the potential therapeutic efficacy of combined pharmacological and mechanical stimuli to promote bone accrual and maintenance in diabetic subjects. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marta Maycas
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
| | - Kevin A McAndrews
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Amy Y Sato
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gretel G Pellegrini
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Drew M Brown
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Arancha R Gortazar
- Instituto de Medicina Molecular Aplicada-Universidad San Pablo CEU, Madrid, Spain
| | - Pedro Esbrit
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.,Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
24
|
Tsentidis C, Gourgiotis D, Kossiva L, Marmarinos A, Doulgeraki A, Karavanaki K. Increased levels of Dickkopf-1 are indicative of Wnt/β-catenin downregulation and lower osteoblast signaling in children and adolescents with type 1 diabetes mellitus, contributing to lower bone mineral density. Osteoporos Int 2017; 28:945-953. [PMID: 27766367 DOI: 10.1007/s00198-016-3802-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/03/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED Higher levels of Dickkopf-1, which is an inhibitor of Wnt/β-catenin bone metabolic pathway, could be indicative of downregulated Wnt system, with possible lower osteoblast activation and higher osteoclast signaling in type 1 diabetes mellitus children and adolescents. Dickkopf-1 could significantly contribute to diabetes osteopathy. INTRODUCTION Increased fracture risk and elevated Dickkopf-1 levels, which is an inhibitor of Wnt/β-catenin bone metabolic pathway, have been documented in adult patients with type 2 diabetes mellitus (T2D), while no relevant data exist on childhood type 1 diabetes (T1D). Our aim was to study plasma Dickkopf-1 distribution in children and adolescents with T1D and to correlate Dickkopf-1 with metabolic bone markers and bone mineral density (BMD). METHODS We evaluated 40 children and adolescents with T1D (mean ± SD age 13.04 ± 3.53 years, T1D duration 5.15 ± 3.33 years) and 40 healthy age-matched and gender-matched controls (age 12.99 ± 3.3 years). Dickkopf-1 and bone metabolic markers were measured, while total body and lumbar spine BMD were evaluated with dual-energy X-ray absorptiometry (DXA). RESULTS Dickkopf-1 demonstrated a Gaussian distribution, with higher levels in T1D patients (13.56 ± 5.34 vs 11.35 ± 3.76 pmol/L, p = 0.024). Higher values were found in boys and in prepubertal children. Dickkopf-1 correlated positively with osteoprotegerin and fasting glucose in patients, while positive correlation with sclerostin and total soluble receptor activator of nuclear factor-kappaB ligand (s-RANKL) was found in controls. Positive correlations with C-telopeptide cross-links (CTX), osteocalcin, alkaline phosphatase, phosphate, and insulin-like growth factor 1 (IGF1) were documented in both groups. Lumbar spine Z-score was positively associated with Dickkopf-1 in controls, while a negative trend was found in patients. CONCLUSIONS Higher levels of Dickkopf-1 could indicate a downregulated Wnt/β-catenin system with possible lower osteoblast activation and higher osteoclast signaling in T1D children and adolescents. Dickkopf-1 could possibly be a significant contributor of T1D osteopathy. Future therapies could focus on Wnt/β-catenin metabolic pathway.
Collapse
Affiliation(s)
- C Tsentidis
- Diabetes Clinic, 2nd Department of Pediatrics, Athens University Medical School, "P&A Kyriakou" Children's Hospital, Thivon & Livadias, 11527 Ampelokipi, Athens, Greece.
| | - D Gourgiotis
- Laboratory of Clinical Biochemistry-Molecular Diagnostics, 2nd Department of Pediatrics, Athens University Medical School, "P & A Kyriakou" Children's Hospital, Athens, Greece
| | - L Kossiva
- Diabetes Clinic, 2nd Department of Pediatrics, Athens University Medical School, "P&A Kyriakou" Children's Hospital, Thivon & Livadias, 11527 Ampelokipi, Athens, Greece
| | - A Marmarinos
- Laboratory of Clinical Biochemistry-Molecular Diagnostics, 2nd Department of Pediatrics, Athens University Medical School, "P & A Kyriakou" Children's Hospital, Athens, Greece
| | - A Doulgeraki
- Department of Bone and Mineral Metabolism, Institute of Child Health, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - K Karavanaki
- Diabetes Clinic, 2nd Department of Pediatrics, Athens University Medical School, "P&A Kyriakou" Children's Hospital, Thivon & Livadias, 11527 Ampelokipi, Athens, Greece
| |
Collapse
|
25
|
Osagie-Clouard L, Sanghani A, Coathup M, Briggs T, Bostrom M, Blunn G. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017; 6:14-21. [PMID: 28062525 PMCID: PMC5227055 DOI: 10.1302/2046-3758.61.bjr-2016-0085.r1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022] Open
Abstract
Intermittently administered parathyroid hormone (PTH 1-34) has been shown to promote bone formation in both human and animal studies. The hormone and its analogues stimulate both bone formation and resorption, and as such at low doses are now in clinical use for the treatment of severe osteoporosis. By varying the duration of exposure, parathyroid hormone can modulate genes leading to increased bone formation within a so-called 'anabolic window'. The osteogenic mechanisms involved are multiple, affecting the stimulation of osteoprogenitor cells, osteoblasts, osteocytes and the stem cell niche, and ultimately leading to increased osteoblast activation, reduced osteoblast apoptosis, upregulation of Wnt/β-catenin signalling, increased stem cell mobilisation, and mediation of the RANKL/OPG pathway. Ongoing investigation into their effect on bone formation through 'coupled' and 'uncoupled' mechanisms further underlines the impact of intermittent PTH on both cortical and cancellous bone. Given the principally catabolic actions of continuous PTH, this article reviews the skeletal actions of intermittent PTH 1-34 and the mechanisms underlying its effect. CITE THIS ARTICLE L. Osagie-Clouard, A. Sanghani, M. Coathup, T. Briggs, M. Bostrom, G. Blunn. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017;6:14-21. DOI: 10.1302/2046-3758.61.BJR-2016-0085.R1.
Collapse
Affiliation(s)
- L Osagie-Clouard
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - A Sanghani
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Coathup
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - T Briggs
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Bostrom
- Hospital for Special Surgery, New York, New York, USA
| | - G Blunn
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| |
Collapse
|
26
|
Pereira M, Gohin S, Lund N, Hvid A, Smitham PJ, Oddy MJ, Reichert I, Farlay D, Roux JP, Cleasby ME, Chenu C. Sclerostin does not play a major role in the pathogenesis of skeletal complications in type 2 diabetes mellitus. Osteoporos Int 2017; 28:309-320. [PMID: 27468901 PMCID: PMC5206261 DOI: 10.1007/s00198-016-3718-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED In contrast to previously reported elevations in serum sclerostin levels in diabetic patients, the present study shows that the impaired bone microarchitecture and cellular turnover associated with type 2 diabetes mellitus (T2DM)-like conditions in ZDF rats are not correlated with changes in serum and bone sclerostin expression. INTRODUCTION T2DM is associated with impaired skeletal structure and a higher prevalence of bone fractures. Sclerostin, a negative regulator of bone formation, is elevated in serum of diabetic patients. We aimed to relate changes in bone architecture and cellular activities to sclerostin production in the Zucker diabetic fatty (ZDF) rat. METHODS Bone density and architecture were measured by micro-CT and bone remodelling by histomorphometry in tibiae and femurs of 14-week-old male ZDF rats and lean Zucker controls (n = 6/group). RESULTS ZDF rats showed lower trabecular bone mineral density and bone mass compared to controls, due to decreases in bone volume and thickness, along with impaired bone connectivity and cortical bone geometry. Bone remodelling was impaired in diabetic rats, demonstrated by decreased bone formation rate and increased percentage of tartrate-resistant acid phosphatase-positive osteoclastic surfaces. Serum sclerostin levels (ELISA) were higher in ZDF compared to lean rats at 9 weeks (+40 %, p < 0.01), but this difference disappeared as their glucose control deteriorated and by week 14, ZDF rats had lower sclerostin levels than control rats (-44 %, p < 0.0001). Bone sclerostin mRNA (qPCR) and protein (immunohistochemistry) were similar in ZDF, and lean rats at 14 weeks and genotype did not affect the number of empty osteocytic lacunae in cortical and trabecular bone. CONCLUSION T2DM results in impaired skeletal architecture through altered remodelling pathways, but despite altered serum levels, it does not appear that sclerostin contributes to the deleterious effect of T2DM in rat bone.
Collapse
Affiliation(s)
- M Pereira
- Department of Comparative Biomedical sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| | - S Gohin
- Department of Comparative Biomedical sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - N Lund
- Department of Comparative Biomedical sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - A Hvid
- Department of Comparative Biomedical sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - P J Smitham
- University College London, London, UK
- The University of Adelaide, Adelaide, Australia
| | - M J Oddy
- University College Hospital, London, UK
| | | | - D Farlay
- INSERM UMR1033 and Université de Lyon, Lyon, France
| | - J P Roux
- INSERM UMR1033 and Université de Lyon, Lyon, France
| | - M E Cleasby
- Department of Comparative Biomedical sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - C Chenu
- Department of Comparative Biomedical sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
27
|
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
|
28
|
Epstein S, Defeudis G, Manfrini S, Napoli N, Pozzilli P. Diabetes and disordered bone metabolism (diabetic osteodystrophy): time for recognition. Osteoporos Int 2016; 27:1931-51. [PMID: 26980458 DOI: 10.1007/s00198-015-3454-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/07/2015] [Indexed: 02/06/2023]
Abstract
Diabetes and osteoporosis are rapidly growing diseases. The link between the high fracture incidence in diabetes as compared with the non-diabetic state has recently been recognized. While this review cannot cover every aspect of diabetic osteodystrophy, it attempts to incorporate current information from the First International Symposium on Diabetes and Bone presentations in Rome in 2014. Diabetes and osteoporosis are fast-growing diseases in the western world and are becoming a major problem in the emerging economic nations. Aging of populations worldwide will be responsible for an increased risk in the incidence of osteoporosis and diabetes. Furthermore, the economic burden due to complications of these diseases is enormous and will continue to increase unless public awareness of these diseases, the curbing of obesity, and cost-effective measures are instituted. The link between diabetes and fractures being more common in diabetics than non-diabetics has been widely recognized. At the same time, many questions remain regarding the underlying mechanisms for greater bone fragility in diabetic patients and the best approach to risk assessment and treatment to prevent fractures. Although it cannot cover every aspect of diabetic osteodystrophy, this review will attempt to incorporate current information particularly from the First International Symposium on Diabetes and Bone presentations in Rome in November 2014.
Collapse
Affiliation(s)
- S Epstein
- Division of Endocrinology, Mount Sinai School of Medicine, New York, NY, USA
| | - G Defeudis
- Unit of Endocrinology and Diabetes, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128, Rome, Italy.
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - S Manfrini
- Unit of Endocrinology and Diabetes, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128, Rome, Italy
| | - N Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128, Rome, Italy
| | - P Pozzilli
- Unit of Endocrinology and Diabetes, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128, Rome, Italy
| | | |
Collapse
|
29
|
Tsentidis C, Gourgiotis D, Kossiva L, Marmarinos A, Doulgeraki A, Karavanaki K. Sclerostin distribution in children and adolescents with type 1 diabetes mellitus and correlation with bone metabolism and bone mineral density. Pediatr Diabetes 2016; 17:289-99. [PMID: 26094958 DOI: 10.1111/pedi.12288] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 03/28/2015] [Accepted: 05/08/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Sclerostin is an inhibitor of the Wnt/beta-catenin bone metabolic pathway. Increased sclerostin levels and reduced bone mineral density (BMD) have been documented in adult patients with diabetes mellitus (DM), predominantly in those with type 2 diabetes mellitus (T2DM). No relative data exist on childhood type 1 diabetes mellitus (T1DM). Our objective was to study plasma sclerostin in T1DM children and adolescents and controls and its correlations with metabolic bone markers and BMD. SUBJECTS AND METHODS This was a cross-sectional study that was conducted at an outpatient clinical center. Forty T1DM children and adolescents were evaluated (mean ± SD age: 13.04 ± 3.53 yr, T1DM duration: 5.15 ± 3.33 yr), along with 40 healthy matched controls (age 12.99 ± 3.3 yr). Sclerostin, soluble receptor activator of nuclear factor-kappaB ligand (s-RANKL), osteoprotegerin, osteocalcin, C-telopeptide crosslinks, electrolytes, parathyroid hormone (PTH), and total 25(OH)D were measured. Lumbar and subcranial total body BMD were evaluated with dual energy X-ray absorptiometry (DXA). RESULTS Sclerostin levels demonstrated a Gaussian distribution, with no significant difference between patients and controls (51.56 ± 12.05 vs. 50.98 ± 13.55 pmol/L, p = 0.84). Significantly lower values were found in girls and prepubertal children. Sclerostin values were significantly and gradually increased in children through pubertal Tanner stages 1-3, were reduced at stage 4 and increased again at pubertal stage 5. Sclerostin levels were positively correlated with logCTX (logarithm of C-terminal telopeptide crosslinks of type I collagen), logOsteocalcin (logarithm of Osteocalcin), magnesium, total body, and L1-L4 BMD z-score. CONCLUSIONS T1DM patients had similar levels of sclerostin with controls. Sclerostin correlated with bone resorption and formation markers and also with bone mass indices, gender, and pubertal stage. The decrease in sclerostin values observed in pubertal stage 4 adolescents coincides with the concurrent growth spurt, and is consistent with sclerostin physiology as an inhibiting signal.
Collapse
Affiliation(s)
- Charalampos Tsentidis
- Diabetic Clinic, 2nd Department of Pediatrics, Athens University Medical School, 'P&A Kyriakou' Children's Hospital, Athens, Greece
| | - Dimitrios Gourgiotis
- Laboratory of Clinical Biochemistry - Molecular Diagnostics, 2nd Department of Pediatrics, Athens University Medical School, 'P & A Kyriakou' Children's Hospital, Athens, Greece
| | - Lydia Kossiva
- Diabetic Clinic, 2nd Department of Pediatrics, Athens University Medical School, 'P&A Kyriakou' Children's Hospital, Athens, Greece
| | - Antonios Marmarinos
- Laboratory of Clinical Biochemistry - Molecular Diagnostics, 2nd Department of Pediatrics, Athens University Medical School, 'P & A Kyriakou' Children's Hospital, Athens, Greece
| | - Artemis Doulgeraki
- Department of Bone and Mineral Metabolism, Institute of Child Health, 'Aghia Sophia' Children's Hospital, Athens, Greece
| | - Kyriaki Karavanaki
- Diabetic Clinic, 2nd Department of Pediatrics, Athens University Medical School, 'P&A Kyriakou' Children's Hospital, Athens, Greece
| |
Collapse
|
30
|
Esbrit P, Herrera S, Portal-Núñez S, Nogués X, Díez-Pérez A. Parathyroid Hormone-Related Protein Analogs as Osteoporosis Therapies. Calcif Tissue Int 2016; 98:359-69. [PMID: 26259869 DOI: 10.1007/s00223-015-0050-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/03/2015] [Indexed: 12/14/2022]
Abstract
The only bone anabolic agent currently available for osteoporosis treatment is parathyroid hormone (PTH)-either its N-terminal 1-34 fragment or the whole molecule of 1-84 aminoacids-whose intermittent administration stimulates new bone formation by targeting osteoblastogenesis and osteoblast survival. PTH-related protein (PTHrP) is an abundant factor in bone which shows N-terminal homology with PTH and thus exhibits high affinity for the same PTH type 1 receptor in osteoblasts. Therefore, it is not surprising that intermittently administered N-terminal PTHrP peptides induce bone anabolism in animals and humans. Furthermore, the C-terminal region of PTHrP also elicits osteogenic features in vitro in osteoblastic cells and in various animal models of osteoporosis. In this review, we discuss the current concepts about the cellular and molecular mechanisms whereby PTHrP may induce anabolic actions in bone. Pre-clinical studies and clinical data using N-terminal PTHrP analogs are also summarized, pointing to PTHrP as a promising alternative to current bone anabolic therapies.
Collapse
Affiliation(s)
- Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Avda. Reyes Católicos, 2, 28040, Madrid, Spain.
- Universidad Autónoma de Madrid, Madrid, Spain.
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Instituto de Salud Carlos III, Madrid, Spain.
| | - Sabina Herrera
- Hospital del Mar-IMIM, Universidad Autónoma de Barcelona, Barcelona, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Portal-Núñez
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Nogués
- Hospital del Mar-IMIM, Universidad Autónoma de Barcelona, Barcelona, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Instituto de Salud Carlos III, Madrid, Spain
| | - Adolfo Díez-Pérez
- Hospital del Mar-IMIM, Universidad Autónoma de Barcelona, Barcelona, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
The Effects of Chinese Medicine on Activation of Wnt/β-Catenin Signal Pathway under High Glucose Condition. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:295135. [PMID: 26495008 PMCID: PMC4606185 DOI: 10.1155/2015/295135] [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: 02/03/2015] [Revised: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 11/17/2022]
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by chronic hyperglycemia and a series of complications. The Wnt/β-catenin signaling pathway is a complex protein interaction network, which is also a key regulator of cell proliferation and differentiation. Many scholars have found that high glucose can activate the Wnt signaling pathway. However, the effects of activation of this pathway in the presence of high glucose levels during the progression of diabetes still remained unclear. Here, we provide a review of the study on the effects of high glucose state on the Wnt/β-catenin signal pathway and the influence of Chinese medicine on it.
Collapse
|
32
|
Adverse Effects of Diabetes Mellitus on the Skeleton of Aging Mice. J Gerontol A Biol Sci Med Sci 2015; 71:290-9. [DOI: 10.1093/gerona/glv160] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/19/2015] [Indexed: 01/15/2023] Open
|
33
|
Zhukouskaya VV, Eller-Vainicher C, Shepelkevich AP, Dydyshko Y, Cairoli E, Chiodini I. Bone health in type 1 diabetes: focus on evaluation and treatment in clinical practice. J Endocrinol Invest 2015; 38:941-50. [PMID: 25863666 DOI: 10.1007/s40618-015-0284-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/31/2015] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Type 1 diabetes (T1D) is an autoimmune disease with chronic hyperglycemic state, which incidence has been globally rising during the past decades. Besides the well-known diabetic complications such as retinopathy, nephropathy and neuropathy, T1D is characterized also by poor bone health. The reduced bone mineralization, quality and strength lead to vertebral and hip fractures as the most important clinical manifestations. Suppressed bone turnover is the main characteristic of T1D-associated bone disorder. RESULTS This is thought to be due to hyperglycemia, hypoinsulinemia, autoimmune inflammation, low levels of insulin-like growth factor-1 and vitamin D. Young age of T1D manifestation, chronic poor glycemic control, high daily insulin dose, low body mass index, reduced renal function and the presence of diabetic complications are clinical factors useful for identifying T1D patients at risk of reduced bone mineral density. Although the clinical risk factors for fracture risk are still unknown, chronic poor glycemic control and the presence of diabetic complications might raise the suspicion of elevated fracture risk in T1D. In the presence of the above-mentioned risk factors, the assessment of bone mineral density by dual-energy X-ray absorptiometry and the search of asymptomatic vertebral fracture by vertebral fracture assessment or lateral X-ray radiography of thorax-lumbar spine should be recommended. CONCLUSION There is no consensus about the treatment of diabetic bone disorder. However, the improvement of glycemic control has been suggested to have a beneficial effect on bone in T1D. Recently, several experiments showed promising results on using anabolic pharmacological agents in diabetic rodents with bone disorder. Therefore, randomized clinical trials are needed to test the possible use of the bone anabolic therapies in humans with T1D.
Collapse
Affiliation(s)
- V V Zhukouskaya
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy,
| | | | | | | | | | | |
Collapse
|
34
|
Coe LM, Tekalur SA, Shu Y, Baumann MJ, McCabe LR. Bisphosphonate treatment of type I diabetic mice prevents early bone loss but accentuates suppression of bone formation. J Cell Physiol 2015; 230:1944-53. [PMID: 25641511 DOI: 10.1002/jcp.24929] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
Type I (T1) diabetes is an autoimmune and metabolic disease associated with bone loss. Previous studies demonstrate that T1-diabetes decreases osteoblast activity and viability. Bisphosphonate therapy, commonly used to treat osteoporosis, is demonstrated to inhibit osteoclast activity as well as osteoblast apoptosis. Therefore, we examined the effect of weekly alendronate treatments on T1-diabetes induced osteoblast apoptosis and bone loss. Bone TUNEL assays identified that alendronate therapy prevents the diabetes-induced osteoblast death observed during early stages of diabetes development. Consistent with this, alendronate treatment for 40 days was able to prevent diabetes-induced trabecular bone loss. Alendronate was also able to reduce marrow adiposity in both control diabetic mice compared to untreated mice. Mechanical testing indicated that 40 days of alendronate treatment increased bone stiffness but decreased the work required for fracture in T1-diabetic and alendronate treated mice. Of concern at this later time point, bone formation rate and osteoblast markers, which were already decreased in diabetic mice, were further suppressed in alendronate-treated diabetic mice. Taken together, our results suggest that short-term alendronate treatment can prevent T1-diabetes-induced bone loss in mice, possibly in part by inhibiting diabetes onset associated osteoblast death, while longer treatment enhanced bone density but at the cost of further suppressing bone formation in diabetic mice.
Collapse
Affiliation(s)
- Lindsay M Coe
- Department of Physiology, Biomedical Imaging Research Center, Michigan State University, East Lansing, Michigan
| | | | | | | | | |
Collapse
|
35
|
Roy S, Bae E, Amin S, Kim D. Extracellular matrix, gap junctions, and retinal vascular homeostasis in diabetic retinopathy. Exp Eye Res 2015; 133:58-68. [PMID: 25819455 DOI: 10.1016/j.exer.2014.08.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022]
Abstract
The vascular basement membrane (BM) contains extracellular matrix (ECM) proteins that assemble in a highly organized manner to form a supportive substratum for cell attachment facilitating myriad functions that are vital to cell survival and overall retinal homeostasis. The BM provides a microenvironment in which bidirectional signaling through integrins regulates cell attachment, turnover, and functionality. In diabetic retinopathy, the BM undergoes profound structural and functional changes, and recent studies have brought to light the implications of such changes. Thickened vascular BM in the retinal capillaries actively participate in the development and progression of characteristic changes associated with diabetic retinopathy. High glucose (HG)-induced compromised cell-cell communication via gap junctions (GJ) in retinal vascular cells may disrupt homeostasis in the retinal microenvironment. In this review, the role of altered ECM synthesis, compromised GJ activity, and disturbed retinal homeostasis in the development of retinal vascular lesions in diabetic retinopathy are discussed.
Collapse
Affiliation(s)
- Sayon Roy
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA.
| | - Edward Bae
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| | - Shruti Amin
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| | - Dongjoon Kim
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
36
|
Type 1 diabetes and osteoporosis: from molecular pathways to bone phenotype. J Osteoporos 2015; 2015:174186. [PMID: 25874154 PMCID: PMC4385591 DOI: 10.1155/2015/174186] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/09/2015] [Indexed: 01/06/2023] Open
Abstract
The link between type 1 diabetes mellitus (DM1) and osteoporosis, identified decades ago, has gained attention in recent years. While a number of cellular mechanisms have been postulated to mediate this association, it is now established that defects in osteoblast differentiation and activity are the main culprits underlying bone fragility in DM1. Other contributing factors include an accumulation of advanced glycation end products (AGEs) and the development of diabetes complications (such as neuropathy and hypoglycemia), which cause further decline in bone mineral density (BMD), worsening geometric properties within bone, and increased fall risk. As a result, patients with DM1 have a 6.9-fold increased incidence of hip fracture compared to controls. Despite this increased fracture risk, bone fragility remains an underappreciated complication of DM1 and is not addressed in most diabetes guidelines. There is also a lack of data regarding the efficacy of therapeutic strategies to treat osteoporosis in this patient population. Together, our current understanding of bone fragility in DM1 calls for an update of diabetes guidelines, better screening tools, and further research into the use of therapeutic strategies in this patient population.
Collapse
|
37
|
Zhou KK, Benyajati S, Le Y, Cheng R, Zhang W, Ma JX. Interruption of Wnt signaling in Müller cells ameliorates ischemia-induced retinal neovascularization. PLoS One 2014; 9:e108454. [PMID: 25271989 PMCID: PMC4182699 DOI: 10.1371/journal.pone.0108454] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 08/29/2014] [Indexed: 01/26/2023] Open
Abstract
Retinal Müller cells are major producers of inflammatory and angiogenic cytokines which contribute to diabetic retinopathy (DR). Over-activation of the Wnt/β-catenin pathway has been shown to play an important pathogenic role in DR. However, the roles of Müller cell-derived Wnt/β-catenin signaling in retinal neovascularization (NV) and DR remain undefined. In the present study, mice with conditional β-catenin knockout (KO) in Müller cells were generated and subjected to oxygen-induced retinopathy (OIR) and streptozotocin (STZ)-induced diabetes. Wnt signaling was evaluated by measuring levels of β-catenin and expression of its target genes using immunoblotting. Retinal vascular permeability was measured using Evans blue as a tracer. Retinal NV was visualized by angiography and quantified by counting pre-retinal nuclei. Retinal pericyte loss was evaluated using retinal trypsin digestion. Electroretinography was performed to examine visual function. No abnormalities were detected in the β-catenin KO mice under normal conditions. In OIR, retinal levels of β-catenin and VEGF were significantly lower in the β-catenin KO mice than in littermate controls. The KO mice also had decreased retinal NV and vascular leakage in the OIR model. In the STZ-induced diabetic model, disruption of β-catenin in Müller cells attenuated over-expression of inflammatory cytokines and ameliorated pericyte dropout in the retina. These findings suggest that Wnt signaling activation in Müller cells contributes to retinal NV, vascular leakage and inflammation and represents a potential therapeutic target for DR.
Collapse
Affiliation(s)
- Kelu Kevin Zhou
- Department of Physiology, Harold Hamm Diabetes Center, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Siribhinya Benyajati
- Department of Physiology, Harold Hamm Diabetes Center, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Yun Le
- Department of Medicine Endocrinology, Harold Hamm Diabetes Center, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Rui Cheng
- Department of Physiology, Harold Hamm Diabetes Center, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jian-xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
38
|
Ma XY, Feng YF, Ma ZS, Li X, Wang J, Wang L, Lei W. The promotion of osteointegration under diabetic conditions using chitosan/hydroxyapatite composite coating on porous titanium surfaces. Biomaterials 2014; 35:7259-70. [DOI: 10.1016/j.biomaterials.2014.05.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/13/2014] [Indexed: 12/13/2022]
|
39
|
Abstract
Type 1 diabetes (T1D) is autoimmune disease with chronic hyperglycaemic state. Besides diabetic retinopathy, nephropathy, and neuropathy, T1D is characterized by poor bone health. The reduced bone mineralization and quality/strength, due to hyperglycemia, hypoinsulinemia, autoimmune inflammation, low levels of insulin growth factor-1 (IGF-1), and vitamin D, lead to vertebral/hip fractures. Young age of T1D manifestation, chronic poor glycemic control, high daily insulin dose, low BMI, reduced renal function, and the presence of complications can be helpful in identifying T1D patients at risk of reduced bone mineral density. Although risk factors for fracture risk are still unknown, chronic poor glycemic control and presence of diabetic complications might raise the suspicion of elevated fracture risk in T1D. In the presence of the risk factors, the assessment of bone mineral density by dual-energy X-ray absorptiometry and the search of asymptomatic vertebral fracture by lateral X-ray radiography of thorax-lumbar spine should be recommended. The improvement of glycemic control may have a beneficial effect on bone in T1D. Several experiments showed promising results on using anabolic pharmacological agents (recombinant IGF-1 and parathyroid hormone) in diabetic rodents with bone disorder. Randomized clinical trials are needed in order to test the possible use of bone anabolic therapies in humans with T1D.
Collapse
|
40
|
Danks JA, Freeman AN, Martin TJ. Historical Perspective and Evolutionary Origins of Parathyroid Hormone-Related Protein. Clin Rev Bone Miner Metab 2014. [DOI: 10.1007/s12018-014-9163-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
41
|
Pérez-Castrillón JL, Riancho JA, de Luis D, Gonzalez-Sagrado M, Domingo-Andres M, Dueñas-Laita A. Expression of genes related to energy metabolism (osteocalcin, FOXO1, insulin receptor, and SOST) in bone cells of Goto-Kakizaki rats and response to bariatric surgery. Surg Obes Relat Dis 2014; 10:299-303. [DOI: 10.1016/j.soard.2013.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 07/23/2013] [Accepted: 08/10/2013] [Indexed: 12/31/2022]
|
42
|
Rodríguez-de la Rosa L, López-Herradón A, Portal-Núñez S, Murillo-Cuesta S, Lozano D, Cediel R, Varela-Nieto I, Esbrit P. Treatment with N- and C-terminal peptides of parathyroid hormone-related protein partly compensate the skeletal abnormalities in IGF-I deficient mice. PLoS One 2014; 9:e87536. [PMID: 24503961 PMCID: PMC3913635 DOI: 10.1371/journal.pone.0087536] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 12/31/2013] [Indexed: 11/18/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) deficiency causes growth delay, and IGF-I has been shown to partially mediate bone anabolism by parathyroid hormone (PTH). PTH-related protein (PTHrP) is abundant in bone, and has osteogenic features by poorly defined mechanisms. We here examined the capacity of PTHrP (1-36) and PTHrP (107-111) (osteostatin) to reverse the skeletal alterations associated with IGF-I deficiency. Igf1-null mice and their wild type littermates were treated with each PTHrP peptide (80 µg/Kg/every other day/2 weeks; 2 males and 4 females for each genotype) or saline vehicle (3 males and 3 females for each genotype). We found that treatment with either PTHrP peptide ameliorated trabecular structure in the femur in both genotypes. However, these peptides were ineffective in normalizing the altered cortical structure at this bone site in Igf1-null mice. An aberrant gene expression of factors associated with osteoblast differentiation and function, namely runx2, osteoprotegerin/receptor activator of NF-κB ligand ratio, Wnt3a , cyclin D1, connexin 43, catalase and Gadd45, as well as in osteocyte sclerostin, was found in the long bones of Igf1-null mice. These mice also displayed a lower amount of trabecular osteoblasts and osteoclasts in the tibial metaphysis than those in wild type mice. These alterations in Igf1-null mice were only partially corrected by each PTHrP peptide treatment. The skeletal expression of Igf2, Igf1 receptor and Irs2 was increased in Igf1-null mice, and this compensatory profile was further improved by treatment with each PTHrP peptide related to ERK1/2 and FoxM1 activation. In vitro, PTHrP (1-36) and osteostatin were effective in promoting bone marrow stromal cell mineralization in normal mice but not in IGF-I-deficient mice. Collectively, these findings indicate that PTHrP (1-36) and osteostatin can exert several osteogenic actions even in the absence of IGF-I in the mouse bone.
Collapse
Affiliation(s)
- Lourdes Rodríguez-de la Rosa
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Ana López-Herradón
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Portal-Núñez
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Murillo-Cuesta
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Daniel Lozano
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Cediel
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Centro Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Unidad 761, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
- Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
43
|
Autophagy impairment aggravates the inhibitory effects of high glucose on osteoblast viability and function. Biochem J 2014; 455:329-37. [PMID: 23981124 DOI: 10.1042/bj20130562] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Autophagy is a highly regulated homoeostatic process involved in the lysosomal degradation of damaged cell organelles and proteins. This process is considered an important pro-survival mechanism under diverse stress conditions. A diabetic milieu is known to hamper osteoblast viability and function. In the present study, we explored the putative protective role of autophagy in osteoblastic cells exposed to an HG (high glucose) medium. HG was found to increase protein oxidation and triggered autophagy by a mechanism dependent on reactive oxygen species overproduction in osteoblastic MC3T3-E1 cells. MC3T3-E1 cell survival was impaired by HG and worsened by chemical or genetic inhibition of autophagy. These findings were mimicked by H2O2-induced oxidative stress in these cells. Autophagy impairment led to both defective mitochondrial morphology and decreased bioenergetic machinery and inhibited further osteoblast differentiation in MC3T3-E1 cells upon exposure to HG. These novel findings indicate that autophagy is an essential mechanism to maintain osteoblast viability and function in an HG environment.
Collapse
|
44
|
Napoli N, Strollo R, Paladini A, Briganti SI, Pozzilli P, Epstein S. The alliance of mesenchymal stem cells, bone, and diabetes. Int J Endocrinol 2014; 2014:690783. [PMID: 25140176 PMCID: PMC4124651 DOI: 10.1155/2014/690783] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 06/11/2014] [Indexed: 12/15/2022] Open
Abstract
Bone fragility has emerged as a new complication of diabetes. Several mechanisms in diabetes may influence bone homeostasis by impairing the action between osteoblasts, osteoclasts, and osteocytes and/or changing the structural properties of the bone tissue. Some of these mechanisms can potentially alter the fate of mesenchymal stem cells, the initial precursor of the osteoblast. In this review, we describe the main factors that impair bone health in diabetic patients and their clinical impact.
Collapse
Affiliation(s)
- Nicola Napoli
- Division of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy
- Division of Bone and Mineral Diseases, Washington University in St Louis, St Louis, MO, USA
- *Nicola Napoli:
| | - Rocky Strollo
- Division of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Angela Paladini
- Division of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Silvia I. Briganti
- Division of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Paolo Pozzilli
- Division of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy
- Centre for Diabetes, The Blizard Building, Barts and The London School of Medicine, Queen Mary, University of London, London, UK
| | - Sol Epstein
- Division of Endocrinology, Mount Sinai School of Medicine, New York, USA
| |
Collapse
|
45
|
Tolosa MJ, Chuguransky SR, Sedlinsky C, Schurman L, McCarthy AD, Molinuevo MS, Cortizo AM. Insulin-deficient diabetes-induced bone microarchitecture alterations are associated with a decrease in the osteogenic potential of bone marrow progenitor cells: preventive effects of metformin. Diabetes Res Clin Pract 2013; 101:177-86. [PMID: 23806481 DOI: 10.1016/j.diabres.2013.05.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/21/2013] [Accepted: 05/30/2013] [Indexed: 12/15/2022]
Abstract
AIMS Diabetes mellitus is associated with metabolic bone disease and increased low-impact fractures. The insulin-sensitizer metformin possesses in vitro, in vivo and ex vivo osteogenic effects, although this has not been adequately studied in the context of diabetes. We evaluated the effect of insulin-deficient diabetes and/or metformin on bone microarchitecture, on osteogenic potential of bone marrow progenitor cells (BMPC) and possible mechanisms involved. METHODS Partially insulin-deficient diabetes was induced in rats by nicotinamide/streptozotocin-injection, with or without oral metformin treatment. Femoral metaphysis micro-architecture, ex vivo osteogenic potential of BMPC, and BMPC expression of Runx-2, PPARγ and receptor for advanced glycation endproducts (RAGE) were investigated. RESULTS Histomorphometric analysis of diabetic femoral metaphysis demonstrated a slight decrease in trabecular area and a significant reduction in osteocyte density, growth plate height and TRAP (tartrate-resistant acid phosphatase) activity in the primary spongiosa. BMPC obtained from diabetic animals showed a reduction in Runx-2/PPARγ ratio and in their osteogenic potential, and an increase in RAGE expression. Metformin treatment prevented the diabetes-induced alterations in bone micro-architecture and BMPC osteogenic potential. CONCLUSION Partially insulin-deficient diabetes induces deleterious effects on long-bone micro-architecture that are associated with a decrease in BMPC osteogenic potential, which could be mediated by a decrease in their Runx-2/PPARγ ratio and up-regulation of RAGE. These diabetes-induced alterations can be totally or partially prevented by oral administration of metformin.
Collapse
Affiliation(s)
- María José Tolosa
- Laboratorio de Investigación en Osteopatías y Metabolismo Mineral, Department of Biological Sciences, School of Exact Sciences, National University of La Plata, Argentina
| | | | | | | | | | | | | |
Collapse
|
46
|
López-Herradón A, Portal-Núñez S, García-Martín A, Lozano D, Pérez-Martínez FC, Ceña V, Esbrit P. Inhibition of the canonical Wnt pathway by high glucose can be reversed by parathyroid hormone-related protein in osteoblastic cells. J Cell Biochem 2013; 114:1908-16. [DOI: 10.1002/jcb.24535] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 02/28/2013] [Indexed: 01/17/2023]
|
47
|
Esbrit P, Alcaraz MJ. Current perspectives on parathyroid hormone (PTH) and PTH-related protein (PTHrP) as bone anabolic therapies. Biochem Pharmacol 2013; 85:1417-23. [PMID: 23500550 DOI: 10.1016/j.bcp.2013.03.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 03/01/2013] [Accepted: 03/01/2013] [Indexed: 12/19/2022]
Abstract
Osteoporosis is characterized by low bone mineral density and/or poor bone microarchitecture leading to an increased risk of fractures. The skeletal alterations in osteoporosis are a consequence of a relative deficit of bone formation compared to bone resorption. Osteoporosis therapies have mostly relied on antiresorptive drugs. An alternative therapeutic approach for osteoporosis is currently available, based on the intermittent administration of parathyroid hormone (PTH). Bone anabolism caused by PTH therapy is mainly accounted for by the ability of PTH to increase osteoblastogenesis and osteoblast survival. PTH and PTH-related protein (PTHrP)-an abundant local factor in bone- interact with the common PTH type 1 receptor with similar affinities in osteoblasts. Studies mainly in osteoporosis rodent models and limited data in postmenopausal women suggest that N-terminal PTHrP peptides might be considered a promising bone anabolic therapy. In addition, putative osteogenic actions of PTHrP might be ascribed not only to its N-terminal domain but also to its PTH-unrelated C-terminal region. In this review, we discuss the underlying cellular and molecular mechanisms of the anabolic actions of PTH and the similar potential of PTH-related protein (PTHrP) to increase bone mass and improve bone regeneration.
Collapse
Affiliation(s)
- Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria-IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain.
| | | |
Collapse
|
48
|
Hiremath M, Dann P, Fischer J, Butterworth D, Boras-Granic K, Hens J, Van Houten J, Shi W, Wysolmerski J. Parathyroid hormone-related protein activates Wnt signaling to specify the embryonic mammary mesenchyme. Development 2012; 139:4239-49. [PMID: 23034629 DOI: 10.1242/dev.080671] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Parathyroid hormone-related protein (PTHrP) regulates cell fate and specifies the mammary mesenchyme during embryonic development. Loss of PTHrP or its receptor (Pthr1) abolishes the expression of mammary mesenchyme markers and allows mammary bud cells to revert to an epidermal fate. By contrast, overexpression of PTHrP in basal keratinocytes induces inappropriate differentiation of the ventral epidermis into nipple-like skin and is accompanied by ectopic expression of Lef1, β-catenin and other markers of the mammary mesenchyme. In this study, we document that PTHrP modulates Wnt/β-catenin signaling in the mammary mesenchyme using a Wnt signaling reporter, TOPGAL-C. Reporter expression is completely abolished by loss of PTHrP signaling and ectopic reporter activity is induced by overexpression of PTHrP. We also demonstrate that loss of Lef1, a key component of the Wnt pathway, attenuates the PTHrP-induced abnormal differentiation of the ventral skin. To characterize further the contribution of canonical Wnt signaling to embryonic mammary development, we deleted β-catenin specifically in the mammary mesenchyme. Loss of mesenchymal β-catenin abolished expression of the TOPGAL-C reporter and resulted in mammary buds with reduced expression of mammary mesenchyme markers and impaired sexual dimorphism. It also prevented the ectopic, ventral expression of mammary mesenchyme markers caused by overexpression of PTHrP in basal keratinocytes. Therefore, we conclude that a mesenchymal, canonical Wnt pathway mediates the PTHrP-dependent specification of the mammary mesenchyme.
Collapse
Affiliation(s)
- Minoti Hiremath
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ke HZ, Richards WG, Li X, Ominsky MS. Sclerostin and Dickkopf-1 as therapeutic targets in bone diseases. Endocr Rev 2012; 33:747-83. [PMID: 22723594 DOI: 10.1210/er.2011-1060] [Citation(s) in RCA: 304] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The processes of bone growth, modeling, and remodeling determine the structure, mass, and biomechanical properties of the skeleton. Dysregulated bone resorption or bone formation may lead to metabolic bone diseases. The Wnt pathway plays an important role in bone formation and regeneration, and expression of two Wnt pathway inhibitors, sclerostin and Dickkopf-1 (DKK1), appears to be associated with changes in bone mass. Inactivation of sclerostin leads to substantially increased bone mass in humans and in genetically manipulated animals. Studies in various animal models of bone disease have shown that inhibition of sclerostin using a monoclonal antibody (Scl-Ab) increases bone formation, density, and strength. Additional studies show that Scl-Ab improves bone healing in models of bone repair. Inhibition of DKK1 by monoclonal antibody (DKK1-Ab) stimulates bone formation in younger animals and to a lesser extent in adult animals and enhances fracture healing. Thus, sclerostin and DKK1 are emerging as the leading new targets for anabolic therapies to treat bone diseases such as osteoporosis and for bone repair. Clinical trials are ongoing to evaluate the effects of Scl-Ab and DKK1-Ab in humans for the treatment of bone loss and for bone repair.
Collapse
Affiliation(s)
- Hua Zhu Ke
- Metabolic Disorders Research, Amgen Inc., One Amgen Center Drive, MS 29-M-B, Thousand Oaks, California 91320, USA.
| | | | | | | |
Collapse
|
50
|
Lozano D, Trejo CG, Gómez-Barrena E, Manzano M, Doadrio JC, Salinas AJ, Vallet-Regí M, García-Honduvilla N, Esbrit P, Buján J. Osteostatin-loaded onto mesoporous ceramics improves the early phase of bone regeneration in a rabbit osteopenia model. Acta Biomater 2012; 8:2317-23. [PMID: 22414621 DOI: 10.1016/j.actbio.2012.03.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 03/01/2012] [Accepted: 03/06/2012] [Indexed: 01/03/2023]
Abstract
Parathyroid hormone-related protein (PTHrP) is an important modulator of bone formation. Recently, we reported that PTHrP (107-111) (osteostatin) coating onto mesoporous ceramics confers osteogenic activity to these materials. Bone repair is dramatically compromised in osteopenia/osteoporosis. Thus, we examined the efficacy of unmodified and organically modified SBA15 ceramics loaded with osteostatin in promoting bone repair in an osteoporotic rabbit model. Osteoporosis was induced in New Zealand rabbits by methylprednisolone administration, and healthy rabbits were used as controls. Tested materials were implanted into a femoral cavitary defect, and animals were sacrificed at 2 weeks post-implantation. At this time, implants were encapsulated by a variable layer of fibrotic tissue with no evidence of inflammation. Similarly to observations in normal rabbits, both types of osteostatin-loaded bioceramics induced tissue regeneration associated with increased staining for PCNA, Runx2, osteopontin, and/or vascular endothelial growth factor in osteoporotic rabbits. Our present findings demonstrate that these osteostatin-bearing bioceramics increase the early repair response not only in normal bone but also in osteoporotic bone after a local injury.
Collapse
|