1
|
Tsadaris SA, Komatsu DE, Grubisic V, Ramos RL, Hadjiargyrou M. A GCaMP reporter mouse with chondrocyte specific expression of a green fluorescent calcium indicator. Bone 2024; 188:117234. [PMID: 39147354 PMCID: PMC11392458 DOI: 10.1016/j.bone.2024.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
One of the major processes occurring during the healing of a fractured long bone is chondrogenesis, leading to the formation of the soft callus, which subsequently undergoes endochondral ossification and ultimately bridges the fracture site. Thus, understanding the molecular mechanisms of chondrogenesis can enhance our knowledge of the fracture repair process. One such molecular process is calciun (Ca++) signaling, which is known to play a critical role in the development and regeneration of multiple tissues, including bone, in response to external stimuli. Despite the existence of various mouse models for studying Ca++ signaling, none of them were designed to specifically examine the skeletal system or the various musculoskeletal cell types. As such, we generated a genetically engineered mouse model that is specific to cartilage (crossed with Col2a1 Cre mice) to study chondrocytes. Herein, we report on the characterization of this transgenic mouse line using conditional expression of GCaMP6f, a Ca++-indicator protein. Specifically, this mouse line exhibits increased GCaMP6f fluorescence following Ca++ binding in chondrocytes. Using this model, we show real-time Ca++ signaling in embryos, newborn and adult mice, as well as in fracture calluses. Further, robust expression of GCaMP6f in chondrocytes can be easily detected in embryos, neonates, adults, and fracture callus tissue sections. Finally, we also report on Ca++ signaling pathway gene expression, as well as real-time Ca++ transient measurements in fracture callus chondrocytes. Taken together, these mice provide a new experimental tool to study chondrocyte-specific Ca++ signaling during skeletal development and regeneration, as well as various in vitro perturbations.
Collapse
Affiliation(s)
- Sotirios A Tsadaris
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY, USA
| | - Vladimir Grubisic
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA; Center for Biomedical Innovation, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Raddy L Ramos
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Michael Hadjiargyrou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA.
| |
Collapse
|
2
|
Chen X, Du J, Zhan W, Shao B, Jiang H, Chen Z, Wang C. Polyene phosphatidylcholine promotes tibial fracture healing in rats by stimulating angiogenesis dominated by the VEGFA/VEGFR2 signaling pathway. Biochem Biophys Res Commun 2024; 719:150100. [PMID: 38763043 DOI: 10.1016/j.bbrc.2024.150100] [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: 03/09/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
One of the factors that predispose to fractures is liver damage. Interestingly, fractures are sometimes accompanied by abnormal liver function. Polyene phosphatidylcholine (PPC) is an important liver repair drug. We wondered if PPC had a role in promoting fracture healing. A rat model of tibial fracture was developed using the modified Einhorn model method. X-rays were used to detect the progression of fracture healing. Progress of ossification and angiogenesis at the fracture site were analyzed by Safranin O/fast green staining and CD31 immunohistochemistry. To investigate whether PPC has a direct angiogenesis effect, HUVECs were used. We performed MTT, wound healing, Transwell migration, and tube formation assays. Finally, RT-qPCR and Western blot analysis were used to study the underlying mechanism. The results showed that PPC significantly shortened the apparent recovery time of mobility in rats. PPC treatment significantly promoted the formation of cartilage callus, endochondral ossification, and angiogenesis at the fracture site. In vitro, PPC promoted the proliferative viability of HUVECs, their ability to heal wounds, and their ability to penetrate membranes in the Transwell apparatus and increased the tube formation of cells. The transcription of VEGFA, VEGFR2, PLCγ, RAS, ERK1/2 and MEK1/2 was significantly up regulated by PPC. Further, the protein level results demonstrated a significant increase in the expression of VEGFA, VEGFR2, MEK1/2, and ERK1/2 proteins. In conclusion, our findings suggest that PPC promotes angiogenesis by activating the VEGFA/VEGFR2 and downstream signaling pathway, thereby accelerating fracture healing.
Collapse
Affiliation(s)
- Xing Chen
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jinge Du
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenxuan Zhan
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Binghao Shao
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huaying Jiang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhaolong Chen
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chunmei Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
3
|
Ganse B. Methods to accelerate fracture healing - a narrative review from a clinical perspective. Front Immunol 2024; 15:1384783. [PMID: 38911851 PMCID: PMC11190092 DOI: 10.3389/fimmu.2024.1384783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/14/2024] [Indexed: 06/25/2024] Open
Abstract
Bone regeneration is a complex pathophysiological process determined by molecular, cellular, and biomechanical factors, including immune cells and growth factors. Fracture healing usually takes several weeks to months, during which patients are frequently immobilized and unable to work. As immobilization is associated with negative health and socioeconomic effects, it would be desirable if fracture healing could be accelerated and the healing time shortened. However, interventions for this purpose are not yet part of current clinical treatment guidelines, and there has never been a comprehensive review specifically on this topic. Therefore, this narrative review provides an overview of the available clinical evidence on methods that accelerate fracture healing, with a focus on clinical applicability in healthy patients without bone disease. The most promising methods identified are the application of axial micromovement, electromagnetic stimulation with electromagnetic fields and direct electric currents, as well as the administration of growth factors and parathyroid hormone. Some interventions have been shown to reduce the healing time by up to 20 to 30%, potentially equivalent to several weeks. As a combination of methods could decrease the healing time even further than one method alone, especially if their mechanisms of action differ, clinical studies in human patients are needed to assess the individual and combined effects on healing progress. Studies are also necessary to determine the ideal settings for the interventions, i.e., optimal frequencies, intensities, and exposure times throughout the separate healing phases. More clinical research is also desirable to create an evidence base for clinical guidelines. To make it easier to conduct these investigations, the development of new methods that allow better quantification of fracture-healing progress and speed in human patients is needed.
Collapse
Affiliation(s)
- Bergita Ganse
- Innovative Implant Development (Fracture Healing), Clinics and Institutes of Surgery, Saarland University, Homburg, Germany
- Department of Trauma, Hand and Reconstructive Surgery, Clinics and Institutes of Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
4
|
Zidrou C, Kapetanou A, Rizou S. The effect of drugs on implant osseointegration- A narrative review. Injury 2023; 54:110888. [PMID: 37390787 DOI: 10.1016/j.injury.2023.110888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 07/02/2023]
Abstract
OBJECTIVE This narrative review aims to investigate the effects of drugs on implant osseointegration, analyzing their potential positive or negative impact on the direct structural and functional connection between bone and load-carrying implants. BACKGROUND The review seeks to provide a comprehensive understanding of osseointegration, which refers to the successful integration of an implant with living bone, resulting in no progressive relative movement between them. Exploring the effects of drugs on implant osseointegration is crucial for optimizing outcomes and enhancing patient care in orthopedic implant procedures. METHODS Relevant studies on the effects of drugs on implant osseointegration were identified through a literature search. Electronic databases, including PubMed, Embase, and Google Scholar, were utilized, employing appropriate keywords and MeSH terms related to osseointegration, implants, and drug interventions. The search was limited to English studies. DISCUSSION This overview presents a detailed analysis of the effects of drugs on implant osseointegration. It explores drugs such as bisphosphonates, teriparatide, statins, angiotensin-converting enzyme inhibitors, beta-blockers, nitrites, and thiazide diuretics as promoters of osseointegration. Conversely, loop diuretics, non-steroidal anti-inflammatory drugs, corticosteroids, cyclosporine A, cisplatin, methotrexate, antibiotics, proton pump inhibitors (PPIs), antiepileptics, selective serotonin reuptake inhibitors (SSRIs), and anticoagulants are discussed as inhibitors of the process. The role of vitamin D3 remains uncertain. The complex relationship between drugs and the biology of implant osseointegration is emphasized, underscoring the need for further in vitro and in vivo studies to validate their effects CONCLUSION: This narrative review contributes to the literature by providing an overview of the effects of drugs on implant osseointegration. It highlights the complexity of the subject and emphasizes the necessity for more extensive and sophisticated studies in the future. Based on the synthesis of the reviewed literature, certain drugs, such as bisphosphonates and teriparatide, show potential for promoting implant osseointegration, while others, including loop diuretics and certain antibiotics, may impede the process. However, additional research is required to solidify these conclusions and effectively inform clinical practice.
Collapse
Affiliation(s)
- Christiana Zidrou
- 2nd Orthopaedic Department, G. Papageorgiou General Hospital, Thessaloniki, Greece.
| | | | - Stavroula Rizou
- National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
5
|
Diagnostic Imaging Studies on Local and Systemic Erythropoietin Application for Promoting Bone Regeneration in Rat Calvarial Defects. Vet Sci 2022; 9:vetsci9100578. [PMID: 36288191 PMCID: PMC9607163 DOI: 10.3390/vetsci9100578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/06/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022] Open
Abstract
The purpose of this study was to compare the effects of local and systemic application of recombinant human erythropoietin (rhEPO) on the healing of rat calvarial defects. Twenty-four male skeletally-mature Wistar rats were used. Two bone 5 mm critical size defects were created in calvarial bones of each rat. In rats from experimental group I (n = 12), EPO was applied locally on a collagen cone in left defects, whereas a collagen cone soaked with physiological saline was placed in right defects. The rats from experimental group II were injected once intraperitoneally with 4900 IU/kg EPO; a collagen cone was only placed in left defects, whereas the right defects were left empty. The systemic effect of EPO treatment was monitored by haematological analyses on days 0, 30 and 90. Bone healing was monitored via radiography and computed tomography on the same time intervals. The results demonstrated that local EPO application had no significant effect on haemopoiesis, unlike the systemic application. At the same time, it resulted in new bone formation and therefore, could be successfully used as a means of promoting bone regeneration.
Collapse
|
6
|
Besleaga C, Nan B, Popa AC, Balescu LM, Nedelcu L, Neto AS, Pasuk I, Leonat L, Popescu-Pelin G, Ferreira JMF, Stan GE. Sr and Mg Doped Bi-Phasic Calcium Phosphate Macroporous Bone Graft Substitutes Fabricated by Robocasting: A Structural and Cytocompatibility Assessment. J Funct Biomater 2022; 13:jfb13030123. [PMID: 36135559 PMCID: PMC9502687 DOI: 10.3390/jfb13030123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/29/2022] Open
Abstract
Bi-phasic calcium phosphates (BCPs) are considered prominent candidate materials for the fabrication of bone graft substitutes. Currently, supplemental cation-doping is suggested as a powerful path to boost biofunctionality, however, there is still a lack of knowledge on the structural role of such substituents in BCPs, which in turn, could influence the intensity and extent of the biological effects. In this work, pure and Mg- and Sr-doped BCP scaffolds were fabricated by robocasting from hydrothermally synthesized powders, and then preliminarily tested in vitro and thoroughly investigated physically and chemically. Collectively, the osteoblast cell culture assays indicated that all types of BCP scaffolds (pure, Sr- or Sr–Mg-doped) delivered in vitro performances similar to the biological control, with emphasis on the Sr–Mg-doped ones. An important result was that double Mg–Sr doping obtained the ceramic with the highest β-tricalcium phosphate (β-TCP)/hydroxyapatite mass concentration ratio of ~1.8. Remarkably, Mg and Sr were found to be predominantly incorporated in the β-TCP lattice. These findings could be important for the future development of BCP-based bone graft substitutes since the higher dissolution rate of β-TCP enables an easier release of the therapeutic ions. This may pave the road toward medical devices with more predictable in vivo performance.
Collapse
Affiliation(s)
- Cristina Besleaga
- National Institute of Materials Physics, RO-077125 Magurele, Romania
| | - Bo Nan
- Department of Materials and Ceramics Engineering, CICECO, University of Aveiro, 3810-193 Aveiro, Portugal
| | | | | | - Liviu Nedelcu
- National Institute of Materials Physics, RO-077125 Magurele, Romania
| | - Ana Sofia Neto
- Department of Materials and Ceramics Engineering, CICECO, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Iuliana Pasuk
- National Institute of Materials Physics, RO-077125 Magurele, Romania
| | - Lucia Leonat
- National Institute of Materials Physics, RO-077125 Magurele, Romania
| | - Gianina Popescu-Pelin
- National Institute for Lasers, Plasma and Radiation Physics, RO-077125 Magurele, Romania
| | - José M. F. Ferreira
- Department of Materials and Ceramics Engineering, CICECO, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: (J.M.F.F.); (G.E.S.)
| | - George E. Stan
- National Institute of Materials Physics, RO-077125 Magurele, Romania
- Correspondence: (J.M.F.F.); (G.E.S.)
| |
Collapse
|
7
|
Effect of chronic lithium on mechanical sensitivity and trabecular bone loss induced by type-1 diabetes mellitus in mice. Biometals 2022; 35:1033-1042. [PMID: 35849260 DOI: 10.1007/s10534-022-00421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/04/2022] [Indexed: 11/02/2022]
Abstract
Type-1 diabetes mellitus (T1DM) is a chronic condition characterized by long-term hyperglycemia that results in several complications such as painful peripheral neuropathy, bone deterioration, and increased risk of bone fractures. Lithium, a first-line therapy for bipolar disorder, has become an attractive agent for attenuating peripheral neuropathy and menopause-induced bone loss. Therefore, our aim was to determine the effect of chronic lithium treatment on mechanical hypersensitivity and trabecular bone loss induced by T1DM in mice. T1DM was induced in male C57BL/6J mice by intraperitoneal injection of streptozotocin (STZ, 50 mg/kg/day, for 5 consecutive days). 12 weeks after T1DM-induction, mice received a daily intraperitoneal injection of vehicle, 30 or 60 mg/kg lithium (as LiCl) for 6 weeks. Throughout the treatment period, blood glucose levels and mechanical sensitivity were evaluated every 2 weeks. After lithium treatment, the femur and L5 vertebra were harvested for microcomputed tomography (microCT) analysis. T1DM mice showed significant hyperglycemia, mechanical hypersensitivity, and significant trabecular bone loss as compared with the control group. Chronic lithium treatment did not revert the hindpaw mechanical hypersensitivity nor hyperglycemia associated to T1DM induced by STZ. In contrast, microCT analysis revealed that lithium reverted, in a dose-dependent manner, the loss of trabecular bone associated to T1DM induced by STZ at both the distal femur and L5 vertebra. Lithium treatment by itself did not affect any trabecular bone parameter in non-diabetic mice.
Collapse
|
8
|
Chen K, Gao H, Yao Y. Prospects of cell chemotactic factors in bone and cartilage tissue engineering. Expert Opin Biol Ther 2022; 22:883-893. [PMID: 35668707 DOI: 10.1080/14712598.2022.2087471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Ke Chen
- Department of Joint Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangdong Key Laboratory of Orthopedic Technology and Implant Materials
| | - Hui Gao
- Department of Joint Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangdong Key Laboratory of Orthopedic Technology and Implant Materials
| | - Yongchang Yao
- Department of Joint Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangdong Key Laboratory of Orthopedic Technology and Implant Materials
| |
Collapse
|
9
|
Aslroosta H, Yaghobee S, Akbari S, Kanounisabet N. The effects of topical erythropoietin on non-surgical treatment of periodontitis: a preliminary study. BMC Oral Health 2021; 21:240. [PMID: 33957902 PMCID: PMC8101234 DOI: 10.1186/s12903-021-01607-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/23/2021] [Indexed: 11/19/2022] Open
Abstract
Background The purpose of periodontal treatments is to reduce inflammation, restore gingival health and clinical attachment level gain by controlling microbial plaque formation and other etiological factors. One of the drugs that has been tested in many areas and shown good anti-inflammatory properties is erythropoietin (EPO). We evaluated the effect of this drug on the improvement of periodontitis after the phase I treatment. Methods This study was conducted on 30 patients with stage III periodontitis who had at least two bilateral teeth with CAL of ≥ 5 mm and PPD ≥ 6 mm at ≥ 2 non‐adjacent teeth and bleeding on probing. After oral hygiene instruction and scaling and root planning (SRP), EPO gel containing a solution of 4000 units was applied deeply in the test group and placebo gel was deeply administered in the control pockets (5 times, every other day). The clinical parameters of the plaque index (PI), gingival index (GI), clinical attachment level (CAL), probing depth (PD) and bleeding index (BI) were measured at baseline and after three months of follow up. The P-value was set at 0.05. Results All clinical variables improved after treatment in both groups. The BI and GI scores (which reflects the degree of gingival inflammation) showed statistically more reduction in test group. The CAL decreased from 5.1 ± 4.1 to 3.40 ± 2.71 mm; and 5.67 ± 4.32 to 4.33 ± 3.19 mm in test and control group, respectively (P < 0.00). After the treatment, there was a significant greater reduction in CAL and also PD values in test group (P < 0.01). Conclusion Local application of EPO gel in adjunct to SRP can improve clinical inflammation and CAL gain in periodontitis. Trial registration: This study was registered at 2017-11-06 in IRCT. All procedures performed in this study were approved with ID number of IR.TUMS.DENTISTRY.REC.1396.3139 in Tehran University of medical science. Supplementary Information The online version contains supplementary material available at 10.1186/s12903-021-01607-y.
Collapse
Affiliation(s)
- Hoori Aslroosta
- Department of Periodontics, School of Dentistry, Faculty of Dentistry, Tehran University of Medical Sciences, North Kargar Street, Tehran, Iran
| | - Siamak Yaghobee
- Department of Periodontics, School of Dentistry, Faculty of Dentistry, Tehran University of Medical Sciences, North Kargar Street, Tehran, Iran
| | - Solmaz Akbari
- Department of Periodontics, School of Dentistry, Faculty of Dentistry, Tehran University of Medical Sciences, North Kargar Street, Tehran, Iran
| | - Negar Kanounisabet
- Department of Periodontics, School of Dentistry, Faculty of Dentistry, Tehran University of Medical Sciences, North Kargar Street, Tehran, Iran.
| |
Collapse
|
10
|
Vasileva R, Chaprazov T. Preclinical studies on pleiotropic functions of erythropoietin on bone healing. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2020-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Erythropoietin (ЕPО) is a glycoprotein hormone, mainly known for its haemopoietic function. For orthopaedics, its pleiotropic effects – osteogenic and angiogenic potential, are of primary interest. The exact mechanism of EPO action is still unclear. The effects of EPO on bone healing were investigated through experiments with rats, mice, rabbits and pigs. Each of used models for experimental bone defects (calvarial models, long bone segmental defects, posterolateral spinal fusion and corticosteroid-induced femoral head osteonecrosis) has specific advantages and flaws. Obtaining specific and correct results is largely dependent on the used model. The brief evaluation of models could serve for standardisation of preclinical studies on bone regeneration.
Collapse
|
11
|
Trivedi R, Adhikary S, Kothari P, Ahmad N, Mittapelly N, Pandey G, Shukla M, Kumar S, Dev K, Choudhary D, Maurya R, Lal J, Mishra PR. Self-emulsifying formulation of Spinacia oleracea reduces the dose and escalates bioavailability of bioactive compounds to accelerate fracture repair in rats. CLINICAL PHYTOSCIENCE 2020. [DOI: 10.1186/s40816-020-00190-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract
Background
Spinach (Spinacia oleracea) is a rich source of flavonoids and therefore widely used therapeutically as an antioxidant agent in traditional medicine. The present study was undertaken to study the bone regenerating property of dried Spinacia oleracea extract (DSE) and self-emulsifying formulation of the extract (FDSE) on drill-hole model of fracture repair in rats.
Methods
0.8 mm hole was drilled in the diaphyseal region of femur in adult SD rats. DSE and formulated extract (FDSE) was administered orally and fractured femur was collected after treatment regimen. Micro-CT, transcriptional analysis and measurement of calcein intensity of callus formed at the injured site was performed to study the efficacy of the extract and formulation on bone regeneration. Further, compounds from extract were assessed for in-vitro osteoblast activity.
Results
Micro-architecture of the regenerated bone at injured site exhibited 26% (p < 0.001) and 35% (p < 0.01) increased BV/TV (bone volume /tissue volume) and Tb.N. (trabecular number) for DSE (500 mg.kg− 1). Further, FDSE exhibited similar augmentation in BV/TV (p < 0.01) and Tb. N (p < 0.01) parameters at dose of 250 mg.kg− 1. Analogous results were obtained from transcriptional analysis and calcein intensity at the fractured site. 3-O-Methylpatuletin, one of the compound isolated from the extract stimulated the differentiation and mineralization of primary osteoblast and depicted concentration dependent antagonizing effect of H2O2 in osteoblast apparently, minimizing ROS generation thus affectivity in fracture repair.
Conclusions
The present study showed that bone regenerating property of spinach was augmented by formulating extract to deliverable form and can be further studied to develop as therapeutic agent for fracture repair.
Graphical abstract
Collapse
|
12
|
Xue H, Tao D, Weng Y, Fan Q, Zhou S, Zhang R, Zhang H, Yue R, Wang X, Wang Z, Sun Y. Glycosylation of dentin matrix protein 1 is critical for fracture healing via promoting chondrogenesis. Front Med 2019; 13:575-589. [DOI: 10.1007/s11684-019-0693-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/25/2019] [Indexed: 10/26/2022]
|
13
|
Klontzas ME, Reakasame S, Silva R, Morais JC, Vernardis S, MacFarlane RJ, Heliotis M, Tsiridis E, Panoskaltsis N, Boccaccini AR, Mantalaris A. Oxidized alginate hydrogels with the GHK peptide enhance cord blood mesenchymal stem cell osteogenesis: A paradigm for metabolomics-based evaluation of biomaterial design. Acta Biomater 2019; 88:224-240. [PMID: 30772514 DOI: 10.1016/j.actbio.2019.02.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023]
Abstract
Oxidized alginate hydrogels are appealing alternatives to natural alginate due to their favourable biodegradability profiles and capacity to self-crosslink with amine containing molecules facilitating functionalization with extracellular matrix cues, which enable modulation of stem cell fate, achieve highly viable 3-D cultures, and promote cell growth. Stem cell metabolism is at the core of cellular fate (proliferation, differentiation, death) and metabolomics provides global metabolic signatures representative of cellular status, being able to accurately identify the quality of stem cell differentiation. Herein, umbilical cord blood mesenchymal stem cells (UCB MSCs) were encapsulated in novel oxidized alginate hydrogels functionalized with the glycine-histidine-lysine (GHK) peptide and differentiated towards the osteoblastic lineage. The ADA-GHK hydrogels significantly improved osteogenic differentiation compared to gelatin-containing control hydrogels, as demonstrated by gene expression, alkaline phosphatase activity and bone extracellular matrix deposition. Metabolomics revealed the high degree of metabolic heterogeneity in the gelatin-containing control hydrogels, captured the enhanced osteogenic differentiation in the ADA-GHK hydrogels, confirmed the similar metabolism between differentiated cells and primary osteoblasts, and elucidated the metabolic mechanism responsible for the function of GHK. Our results suggest a novel paradigm for metabolomics-guided biomaterial design and robust stem cell bioprocessing. STATEMENT OF SIGNIFICANCE: Producing high quality engineered bone grafts is important for the treatment of critical sized bone defects. Robust and sensitive techniques are required for quality assessment of tissue-engineered constructs, which result to the selection of optimal biomaterials for bone graft development. Herein, we present a new use of metabolomics signatures in guiding the development of novel oxidised alginate-based hydrogels with umbilical cord blood mesenchymal stem cells and the glycine-histidine-lysine peptide, demonstrating that GHK induces stem cell osteogenic differentiation. Metabolomics signatures captured the enhanced osteogenesis in GHK hydrogels, confirmed the metabolic similarity between differentiated cells and primary osteoblasts, and elucidated the metabolic mechanism responsible for the function of GHK. In conclusion, our results suggest a new paradigm of metabolomics-driven design of biomaterials.
Collapse
|
14
|
Zubareva EV, Nadezhdin SV, Burda YE, Nadezhdina NA, Gashevskaya A. Pleiotropic effects of Erythropoietin. Influence of Erythropoietin on processes of mesenchymal stem cells differentiation. RESEARCH RESULTS IN PHARMACOLOGY 2019. [DOI: 10.3897/rrpharmacology.5.33457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Structure and synthesis of Erythropoietin: Erythropoietin (EPO) is a glycoprotein hormone.Recombinant Erythropoietin (Epoetin): Human recombinant erythropoietin is characterised as a factor which stimulates differentiation and proliferation of erythroid precursor cells, and as a tissue protective factor.Anti-ischemic effects of recombinant Erythropoietin: Erythropoietin is one of the most perspective humoral agents which are involved in the preconditioning phenomenon.Erythropoietin receptors and signal transduction pathways: Erythropoietin effects on cells through their interconnection with erythropoietin receptors, which triggers complex intracellular signal cascades, such as JAK2/STAT signaling pathway, phosphatidylinositol 3-kinase (PI3K), protein kinase C, mitogen-activated protein kinase (MAPK), and nuclear factor (NF)-κB signaling pathways.Mechanisms of the effect of Erythropoietin on hematopoietic and non-hematopoietic cells and tissues: In addition to regulation of haemopoiesis, erythropoietin mediates bone formation as it has an effect on hematopoietic stem cells and osteoblastic niche, and this illustrates connection between the processes of haematopoiesis and osteopoiesis which take place in the red bone marrow.The effect of Erythropoietin on mesenchymal stem cells and process of bone tissue formation: Erythropoietin promotes mesenchymal stem cells proliferation, migration and differentiation in osteogenic direction. The evidence of which is expression of bone phenotype by cells under the influence of EPO, including activation of bone specific transcription factors Runx2, osteocalcin and bone sialoprotein.Conclusion: Erythropoietin has a pleiotropic effect on various types of cells and tissues. But the mechanisms which are involved in the process of bone tissue restoration via erythropoietin are still poorly understood.
Collapse
|
15
|
Composite Hydrogels with the Simultaneous Release of VEGF and MCP-1 for Enhancing Angiogenesis for Bone Tissue Engineering Applications. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8122438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rapid new microvascular network induction was critical for bone regeneration, which required the spatiotemporal delivery of growth factors and transplantation of endothelial cells. In this study, the linear poly(d,l-lactic-co-glycolic acid)-b-methoxy poly(ethylene glycol) (PLGA-mPEG) block copolymer microspheres were prepared for simultaneously delivering vascular endothelial growth factor (VEGF) and monocyte chemotactic protein-1 (MCP-1). Then, vascular endothelial cells (VECs) with growth factor loaded microspheres were composited into a star-shaped PLGA-mPEG block copolymer solution. After this, composite hydrogel (microspheres ratio: 5 wt%) was formed by increasing the temperature to 37 °C. The release profiles of VEGF and MCP-1 from composite hydrogels in 30 days were investigated to confirm the different simultaneous delivery systems. The VECs exhibited a good proliferation in the composite hydrogels, which proved that the composite hydrogels had a good cytocompatibility. Furthermore, in vivo animal experiments showed that the vessel density and the mean vessel diameters increased over weeks after the composite hydrogels were implanted into the necrosis site of the rabbit femoral head. The above results suggested that the VECs-laden hydrogel composited with the dual-growth factor simultaneous release system has the potential to enhance angiogenesis in bone tissue engineering.
Collapse
|
16
|
Tite T, Popa AC, Balescu LM, Bogdan IM, Pasuk I, Ferreira JMF, Stan GE. Cationic Substitutions in Hydroxyapatite: Current Status of the Derived Biofunctional Effects and Their In Vitro Interrogation Methods. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E2081. [PMID: 30355975 PMCID: PMC6266948 DOI: 10.3390/ma11112081] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/13/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
High-performance bioceramics are required for preventing failure and prolonging the life-time of bone grafting scaffolds and osseous implants. The proper identification and development of materials with extended functionalities addressing socio-economic needs and health problems constitute important and critical steps at the heart of clinical research. Recent findings in the realm of ion-substituted hydroxyapatite (HA) could pave the road towards significant developments in biomedicine, with an emphasis on a new generation of orthopaedic and dentistry applications, since such bioceramics are able to mimic the structural, compositional and mechanical properties of the bone mineral phase. In fact, the fascinating ability of the HA crystalline lattice to allow for the substitution of calcium ions with a plethora of cationic species has been widely explored in the recent period, with consequent modifications of its physical and chemical features, as well as its functional mechanical and in vitro and in vivo biological performance. A comprehensive inventory of the progresses achieved so far is both opportune and of paramount importance, in order to not only gather and summarize information, but to also allow fellow researchers to compare with ease and filter the best solutions for the cation substitution of HA-based materials and enable the development of multi-functional biomedical designs. The review surveys preparation and synthesis methods, pinpoints all the explored cation dopants, and discloses the full application range of substituted HA. Special attention is dedicated to the antimicrobial efficiency spectrum and cytotoxic trade-off concentration values for various cell lines, highlighting new prophylactic routes for the prevention of implant failure. Importantly, the current in vitro biological tests (widely employed to unveil the biological performance of HA-based materials), and their ability to mimic the in vivo biological interactions, are also critically assessed. Future perspectives are discussed, and a series of recommendations are underlined.
Collapse
Affiliation(s)
- Teddy Tite
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
| | - Adrian-Claudiu Popa
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
- Army Centre for Medical Research, RO-010195 Bucharest, Romania.
| | | | | | - Iuliana Pasuk
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
| | - José M F Ferreira
- Department of Materials and Ceramics Engineering, CICECO, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - George E Stan
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
| |
Collapse
|
17
|
Vachhani K, Whyne C, Wang Y, Burns DM, Nam D. Low-dose lithium regimen enhances endochondral fracture healing in osteoporotic rodent bone. J Orthop Res 2018; 36:1783-1789. [PMID: 29106746 DOI: 10.1002/jor.23799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/26/2017] [Indexed: 02/04/2023]
Abstract
Osteoporotic bone fractures are highly prevalent and involve lengthy recovery. Lithium, commonly used in psychiatric medicine, inhibits glycogen synthase kinase-3β in the Wnt/β-catenin pathway, leading to up-regulation of osteogenesis. Our recent preclinical work demonstrated that a 20 mg/kg lithium dose administered beginning 7 days post-fracture for 14 days optimally improved femoral fracture healing in healthy rats at 4 weeks post fracture (46% higher torsional strength). In this study, lithium treatment was evaluated for healing of osteoporotic bone fractures. Six-month-old ovariectomized rats were subjected to closed, load-drop induced femoral diaphyseal fracture. Two regimens involving treatment initiation on day 7 and day 10, respectively, 20 mg/kg/day oral dose and 14 days duration were evaluated. Femurs of lithium- vs. saline- treated rats were analyzed at 4 weeks (for day 7 onset regimen) or 6 weeks (for day 10 onset regimen) post-fracture by stereology and torsional mechanical testing. Initiation on day 10 led to a significant 50% higher maximum yield torque (primary outcome measure) at 6 weeks (309 vs. 206 N-mm, p = 0.005; n = 7, 7). Initiation on day 7 suggested a trend toward a more modest improvement in maximum yield torque (13%) evaluated at 4 weeks post-fracture (234 vs. 206 N-mm, p = 0.10; n = 10, 13). Qualitatively, lithium-treated femurs demonstrated better periosteal and mineralized callus bridging in the day 10 initiation group. Lithium is a widely-available, orally administered, low-cost drug, which represents a feasible pharmacological intervention for both healthy and osteoporotic fracture healing. This study provides important guidelines for future clinical evaluation of lithium in osteoporotic fracture patients. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1783-1789, 2018.
Collapse
Affiliation(s)
- Kathak Vachhani
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Cari Whyne
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Division of Orthopedic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Yufa Wang
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Ontario, Canada
| | - David M Burns
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Division of Orthopedic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Diane Nam
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Ontario, Canada.,Division of Orthopedic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Leucht P, Lee S, Yim N. Wnt signaling and bone regeneration: Can't have one without the other. Biomaterials 2018; 196:46-50. [PMID: 29573821 DOI: 10.1016/j.biomaterials.2018.03.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 12/25/2022]
Abstract
Advances in the understanding of the complexities of the Wnt signaling pathway during development and tissue homeostasis have made the Wnt pathway one of the prime candidates for translational applications during tissue regeneration. Wnts are key components of the stem cell niche and are short range signaling molecules responsible for cellular decisions such as proliferation and differentiation. Systemic treatment using biologics targeting the Wnt signaling pathway have shown promising early results and will likely enter the clinical arena in the near future. This comprehensive review summarizes the intricacies how Wnts function in the context of the bone regeneration.
Collapse
Affiliation(s)
- Philipp Leucht
- NYU Langone Health, Departments of Orthopaedic Surgery and Cell Biology, New York, NY, USA.
| | - Sooyeon Lee
- NYU Langone Health, Departments of Orthopaedic Surgery and Cell Biology, New York, NY, USA
| | - Nury Yim
- NYU Langone Health, Departments of Orthopaedic Surgery and Cell Biology, New York, NY, USA
| |
Collapse
|
19
|
Lin TK, Liou YS, Lin CH, Chou P, Jong GP. High-potency statins but not all statins decrease the risk of new-onset osteoporotic fractures: a nationwide population-based longitudinal cohort study. Clin Epidemiol 2018; 10:159-165. [PMID: 29403315 PMCID: PMC5779306 DOI: 10.2147/clep.s145311] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background Statins have been linked to new-onset osteoporotic fractures (NOFs), and different statins may alter the risk for the development of NOFs. Aim In this study, we investigated the association between different statins and the development of NOFs. Patients and methods This was a longitudinal cohort study performed using data from claim forms submitted to the Taiwan Bureau of National Health Insurance, including case patients with NOFs from January 2004 to December 2013 and non-NOF subjects. We estimated the hazard ratios (HRs) of NOFs associated with statin use. Nonuser subjects served as the reference group. Results A total of 44,405 patients with NOFs were identified from among 170,533 patients with hyperlipidemia during the study period. The risk of developing NOFs after adjusting for age, sex, comorbidities, and concurrent medication use was lower among users of atorvastatin (HR, 0.77; 95% CI, 0.71-0.84) and rosuvastatin (HR, 0.72; 95% CI, 0.64-0.81) than among simvastatin users. Lovastatin, pravastatin, fluvastatin, and pitavastatin were not associated with the risk of developing NOFs compared with simvastatin users. Conclusion This study supports previous reports regarding a beneficial effect of statin use and NOF risk, but not all statins. Patients taking atorvastatin or rosuvastatin were at lower risk of developing NOFs compared with simvastatin users during the 10-year follow-up. Other statins such as pravastatin, fluvastatin, lovastatin, and pitavastatin were not associated with NOFs. This study also highlighted that high-potency statin has a dose-response effect on lower NOF risk.
Collapse
Affiliation(s)
- Tsung-Kun Lin
- Department of Pharmacy, Taoyuan Armed Forces General Hospital, Taoyuan.,Institute of Public Health and Community Medicine Research Center, National Yang-Ming University, Taipei
| | - Yi-Sheng Liou
- Department of Family Medicine and Geriatrics, Taichung Veteran General Hospital, Taichung.,School of Public Health, National Defense Medical Center, Taipei
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung
| | - Pesus Chou
- Institute of Public Health and Community Medicine Research Center, National Yang-Ming University, Taipei
| | - Gwo-Ping Jong
- Division of Internal Cardiology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
20
|
Salmon B, Liu B, Shen E, Chen T, Li J, Gillette M, Ransom RC, Ezran M, Johnson CA, Castillo AB, Shen WJ, Kraemer FB, Smith AA, Helms JA. WNT-activated bone grafts repair osteonecrotic lesions in aged animals. Sci Rep 2017; 7:14254. [PMID: 29079746 PMCID: PMC5660190 DOI: 10.1038/s41598-017-14395-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/10/2017] [Indexed: 02/05/2023] Open
Abstract
The Wnt pathway is a new target in bone therapeutic space. WNT proteins are potent stem cell activators and pro-osteogenic agents. Here, we gained insights into the molecular and cellular mechanisms responsible for liposome-reconstituted recombinant human WNT3A protein (L-WNT3A) efficacy to treat osteonecrotic defects. Skeletal injuries were coupled with cryoablation to create non-healing osteonecrotic defects in the diaphysis of the murine long bones. To replicate clinical therapy, osteonecrotic defects were treated with autologous bone graft, which were simulated by using bone graft material from syngeneic ACTB-eGFP-expressing mice. Control osteonecrotic defects received autografts alone; test sites received autografts treated ex vivo with L-WNT3A. In vivo µCT monitored healing over time and immunohistochemistry were used to track the fate of donor cells and assess their capacity to repair osteonecrotic defects according to age and WNT activation status. Collectively, analyses demonstrated that cells from the autograft directly contributed to repair of an osteonecrotic lesion, but this contribution diminished as the age of the donor increased. Pre-treating autografts from aged animals with L-WNT3A restored osteogenic capacity to autografts back to levels observed in autografts from young animals. A WNT therapeutic approach may therefore have utility in the treatment of osteonecrosis, especially in aged patients.
Collapse
Affiliation(s)
- B Salmon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
- Paris Descartes University - Sorbonne Paris Cité, EA 2496 - Orofacial Pathologies, Imaging and Biotherapies Lab and Dental Medicine Department, Bretonneau Hospital, HUPNVS, AP-HP, Paris, France
| | - B Liu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
| | - E Shen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
| | - T Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J Li
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - M Gillette
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
| | - R C Ransom
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
| | - M Ezran
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
| | - C A Johnson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
- Department of Plastic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - A B Castillo
- Department of Mechanical and Aerospace Engineering, New York University Polytechnic School of Engineering, Brooklyn, NY, USA
| | - W J Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - F B Kraemer
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - A A Smith
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA
| | - J A Helms
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
21
|
Sphingosine 1-phosphate (S1P) signalling: Role in bone biology and potential therapeutic target for bone repair. Pharmacol Res 2017; 125:232-245. [PMID: 28855094 DOI: 10.1016/j.phrs.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
Abstract
The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.
Collapse
|
22
|
ZOFKOVA I, BLAHOS J. New Molecules Modulating Bone Metabolism – New Perspectives in the Treatment of Osteoporosis. Physiol Res 2017; 66:S341-S347. [DOI: 10.33549/physiolres.933720] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this review the authors outline traditional antiresorptive pharmaceuticals, such as bisphosphonates, monoclonal antibodies against RANKL, SERMs, as well as a drug with an anabolic effect on the skeleton, parathormone. However, there is also a focus on non-traditional strategies used in therapy for osteolytic diseases. The newest antiosteoporotic pharmaceuticals increase osteoblast differentiation via BMP signaling (harmine), or stimulate osteogenic differentiation of mesenchymal stem cells through Wnt/β-catenin (icarrin, isoflavonoid caviunin, or sulfasalazine). A certain promise in the treatment of osteoporosis is shown by molecules targeting non-coding microRNAs (which are critical for osteoclastogenesis) or those stimulating osteoblast activity via epigenetic mechanisms. Vitamin D metabolites have specific antiosteoporotic potencies, modulating the skeleton not only via mineralization, but markedly also through the direct effects on the bone microstructure.
Collapse
Affiliation(s)
- I. ZOFKOVA
- Institute of Endocrinology, Prague, Czech Republic
| | | |
Collapse
|
23
|
Klontzas ME, Vernardis SI, Heliotis M, Tsiridis E, Mantalaris A. Metabolomics Analysis of the Osteogenic Differentiation of Umbilical Cord Blood Mesenchymal Stem Cells Reveals Differential Sensitivity to Osteogenic Agents. Stem Cells Dev 2017; 26:723-733. [PMID: 28418785 PMCID: PMC5439454 DOI: 10.1089/scd.2016.0315] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) of fetal origin, such as umbilical cord blood MSCs (UCB MSCs), have emerged as a promising cell source for musculoskeletal tissue regeneration because of their higher proliferation potential, lack of donor site morbidity, and their off-the-shelf potential. MSCs differentiated toward the osteogenic lineage exhibit a specific metabolic phenotype characterized by reliance to oxidative phosphorylation for energy production and reduced glycolytic rates. Currently, limited information exists on the metabolic transitions at different stages of the osteogenic process after osteoinduction with different agents. Herein, the osteoinduction efficiency of BMP-2 and dexamethasone on UCB MSCs was assessed using gas chromatography-mass spectrometry (GC-MS) metabolomics analysis, revealing metabolic discrepancies at 7, 14, and 21 days of induction. Whereas both agents when administered individually were able to induce collagen I, osteocalcin, and osteonectin expression, BMP-2 was less effective than dexamethasone in promoting alkaline phosphatase expression. The metabolomics analysis revealed that each agent induced distinct metabolic alterations, including changes in amino acid pools, glutaminolysis, one-carbon metabolism, glycolysis, and tricarboxylic acid cycle. Importantly, we showed that in vitro-differentiated UCB MSCs acquire a metabolic physiology similar to primary osteoblasts when induced with dexamethasone but not with BMP-2, highlighting the fact that metabolomics analysis is sensitive enough to reveal potential differences in the osteogenic efficiency and can be used as a quality control assay for evaluating the osteogenic process.
Collapse
Affiliation(s)
- Michail E Klontzas
- 1 Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London , London, United Kingdom
| | - Spyros I Vernardis
- 1 Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London , London, United Kingdom
| | - Manolis Heliotis
- 2 Department of Oral and Maxillofacial Surgery, London North West Healthcare NHS Trust, Northwick Park Hospital , London, United Kingdom
| | - Eleftherios Tsiridis
- 3 Academic Orthopaedic Unit, Aristotle University Medical School , Thessaloniki, Greece .,4 Department of Surgery and Cancer, Division of Surgery, Imperial College London , London, United Kingdom
| | - Athanasios Mantalaris
- 1 Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London , London, United Kingdom
| |
Collapse
|
24
|
Ginaldi L, De Martinis M. Osteoimmunology and Beyond. Curr Med Chem 2017; 23:3754-3774. [PMID: 27604089 PMCID: PMC5204071 DOI: 10.2174/0929867323666160907162546] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/27/2022]
Abstract
Abstract: Objective Osteoimmunology investigates interactions between skeleton and immune system. In the light of recent discoveries in this field, a new reading register of osteoporosis is actually emerging, in which bone and immune cells are strictly interconnected. Osteoporosis could therefore be considered a chronic immune mediated disease which shares with other age related disorders a common inflammatory background. Here, we highlight these recent discoveries and the new landscape that is emerging. Method Extensive literature search in PubMed central. Results While the inflammatory nature of osteoporosis has been clearly recognized, other interesting aspects of osteoimmunology are currently emerging. In addition, mounting evidence indicates that the immunoskeletal interface is involved in the regulation of important body functions beyond bone remodeling. Bone cells take part with cells of the immune system in various immunological functions, configuring a real expanded immune system, and are therefore variously involved not only as target but also as main actors in various pathological conditions affecting primarily the immune system, such as autoimmunity and immune deficiencies, as well as in aging, menopause and other diseases sharing an inflammatory background. Conclusion The review highlights the complexity of interwoven pathways and shared mechanisms of the crosstalk between the immune and bone systems. More interestingly, the interdisciplinary field of osteoimmunology is now expanding beyond bone and immune cells, defining new homeostatic networks in which other organs and systems are functionally interconnected. Therefore, the correct skeletal integrity maintenance may be also relevant to other functions outside its involvement in bone mineral homeostasis, hemopoiesis and immunity.
Collapse
Affiliation(s)
- Lia Ginaldi
- School and Unit of Allergy and Clinical Immunology, Department of Life, Health, & Environmental Sciences, University of L'Aquila, Italy.
| | | |
Collapse
|