1
|
Dürig J, Calcagni M, Buschmann J. Transition metals in angiogenesis - A narrative review. Mater Today Bio 2023; 22:100757. [PMID: 37593220 PMCID: PMC10430620 DOI: 10.1016/j.mtbio.2023.100757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
The aim of this paper is to offer a narrative review of the literature regarding the influence of transition metals on angiogenesis, excluding lanthanides and actinides. To our knowledge there are not any reviews up to date offering such a summary, which inclined us to write this paper. Angiogenesis describes the process of blood vessel formation, which is an essential requirement for human growth and development. When the complex interplay between pro- and antiangiogenic mediators falls out of balance, angiogenesis can quickly become harmful. As it is so fundamental, both its inhibition and enhancement take part in various diseases, making it a target for therapeutic treatments. Current methods come with limitations, therefore, novel agents are constantly being researched, with metal agents offering promising results. Various transition metals have already been investigated in-depth, with studies indicating both pro- and antiangiogenic properties, respectively. The transition metals are being applied in various formulations, such as nanoparticles, complexes, or scaffold materials. Albeit the increasing attention this field is receiving, there remain many unanswered questions, mostly regarding the molecular mechanisms behind the observed effects. Notably, approximately half of all the transition metals have not yet been investigated regarding potential angiogenic effects. Considering the promising results which have already been established, it should be of great interest to begin investigating the remaining elements whilst also further analyzing the established effects.
Collapse
Affiliation(s)
- Johannes Dürig
- University of Zürich, Faculty of Medicine, Pestalozzistrasse 3, 8032, Zurich, Switzerland
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Maurizio Calcagni
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johanna Buschmann
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
2
|
Haidara MA, Al-Ani B, Bin-Jaliah I, Shams Eldeen AM, Morsy MD. Vanadyl sulphate ameliorates biomarkers of endothelial injury and coagulation and thrombosis in a rat model of hyperglycaemia. Arch Physiol Biochem 2022; 128:447-454. [PMID: 31774317 DOI: 10.1080/13813455.2019.1691602] [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] [Indexed: 02/08/2023]
Abstract
BACKGROUND We sought to determine whether the insulin mimicking agent, vanadyl sulphate (Van) can inhibit biomarkers of endothelial injury and coagulation and thrombosis induced by a moderate level of hyperglycaemia. MATERIAL AND METHODS Hyperglycaemia was induced in rats by a single injection of streptozotocin (STZ, 50 mg/kg) two weeks after being fed on a high-fat diet (model group). The treatment group started Van (20 mg/kg/day) treatment one-week post STZ injection and continued on Van until being sacrificed at week 10. RESULTS Administration of Van to the model group significantly (p < .05) ameliorated dyslipidemia and biomarkers of inflammation (TNF-α, IL-6, and hsCRP) and endothelial injury (E-selectin, P-selectin, sICAM-1, sVCAM-1, and ET-1). Van also significantly inhibited hyperglycaemia-induced blood levels of coagulation (vWF) and thrombosis (PAI-1 and fibrinogen) biomarkers. CONCLUSIONS Vanadyl sulphate effectively suppresses hyperglycaemia-induced endothelial injury, coagulation and thrombosis, which is associated with the inhibition of inflammation and dyslipidemia.
Collapse
Affiliation(s)
- Mohamed A Haidara
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ismaeel Bin-Jaliah
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asmaa M Shams Eldeen
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - M D Morsy
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Physiology, College of Medicine, Menoufia University, Shibin el Kom, Egypt
| |
Collapse
|
3
|
Li J, Li J, Wei Y, Xu N, Li J, Pu X, Wang J, Huang Z, Liao X, Yin G. Ion release behavior of vanadium-doped mesoporous bioactive glass particles and the effect of the released ions on osteogenic differentiation of BMSCs via the FAK/MAPK signaling pathway. J Mater Chem B 2021; 9:7848-7865. [PMID: 34586154 DOI: 10.1039/d1tb01479j] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vanadium is an important trace element in bone and is involved in bone metabolism, bone formation, and bone growth, but the roles of various vanadium ions, especially of pentavalent vanadium, in bone tissue regenerative repair have been underestimated and even misinterpreted for a long time. The main purposes of this study are to investigate the release profile of Si, Ca, P, and V ions from vanadium doped mesoporous bioactive glass (V-MBG) particles and to explore the effect of pentavalent vanadium ions on proliferation and osteogenic differentiation of BMSCs as well as the corresponding osteogenic signaling pathway. On the basis of preparations of V-MBG particles with different pentavalent vanadium contents, the ion release behavior from V-MBG in distilled water and simulated body fluid was systemically investigated. Furthermore, the cytocompatibility and osteogenic effect of V-MBG extracts were studied in rBMSCs, and the related molecular mechanisms were preliminarily discussed. The results of dissolution experiments showed that the V ionic concentration exhibited a burst increase and then a sustained slow increase in the two media. The resultant V ions from 1.0V-MBG, 4.0V-MBG and 10.0V-MBG at 21 days were about 1.1, 5.8, and 12.5 mg L-1 in water, respectively, and 1.6, 4.8 and 12.8 mg L-1 in SBF, respectively. The release behaviors of Si, Ca, P, and V ions were evidently affected by high contents of incorporated vanadium. The cellular results indicated that compared to the control and MBG groups, the V(V) ions in V-MBG extracts at about 19.4 μM markedly promoted the proliferation, the gene and protein expression of BMP-2 and COL-I, and the ALP activity of rBMSCs in non-osteoinductive media, but insignificantly stimulated the OCN protein synthesis. More deeply, V(V) ions at about 19.4 μM significantly upregulated the gene and protein expressions of Itga 2b, FAK, and pERK1/2, demonstrating that V(V) ions could regulate osteogenic differentiation of rBMSCs through the activation of the Itga 2b-FAK-MAPK (pERK1/2) signaling pathway. The in vivo results further confirmed that V-MBG induced and promoted new bone formation in the defect area compared to the PGC and PGC/V-M0 groups. These results would contribute to modify the perception about the biocompatibility and osteogenic promotion of pentavalent vanadium at an appropriate concentration.
Collapse
Affiliation(s)
- Jiangfeng Li
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Junying Li
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Yuhao Wei
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Na Xu
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Jingtao Li
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Xiaoming Liao
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu, 610065, P. R. China.
| |
Collapse
|
4
|
Abraham S, Vives M, Cottrell JA, Mitchell A, Lin HN, Effiong L, Iqbal E, Jingar N, Kim B, Shah N, Munoz W, Chaudhary SB, Lin SS, Benevenia J, O'Connor JP. Local insulin application has a dose-dependent effect on lumbar fusion in a rabbit model. J Tissue Eng Regen Med 2021; 15:442-452. [PMID: 33608970 DOI: 10.1002/term.3182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 11/11/2022]
Abstract
The purpose of this study was to determine if locally applied insulin has a dose-responsive effect on posterolateral lumbar fusion. Adult male New Zealand White rabbits underwent posterolateral intertransverse spinal fusions (PLFs) at L5-L6 using suboptimal amounts of autograft. Fusion sites were treated with collagen sponge soaked in saline (control, n = 11), or with insulin at low (5 or 10 units, n = 13), mid (20 units, n = 11), and high (40 units, n = 11) doses. Rabbits were euthanized at 6 weeks. The L5-L6 spine segment underwent manual palpation and radiographic evaluation performed by two fellowship trained spine surgeons blinded to treatment. Differences between groups were evaluated by analysis of variance on ranks followed by post-hoc Dunn's tests. Forty-three rabbits were euthanized at the planned 6 weeks endpoint, while three died or were euthanized prior to the endpoint. Radiographic evaluation found bilateral solid fusion in 10%, 31%, 60%, and 60% of the rabbits from the control and low, mid, and high-dose insulin-treated groups, respectively (p < 0.05). As per manual palpation, 7 of 10 rabbits in the mid-dose insulin group were fused as compared to 1 of 10 rabbits in the control group (p < 0.05). This study demonstrates that insulin enhanced the effectiveness of autograft to increase fusion success in the rabbit PLF model. The study indicates that insulin or insulin-mimetic compounds can be used to promote bone regeneration.
Collapse
Affiliation(s)
- Sangeeta Abraham
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Michael Vives
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Jessica A Cottrell
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Ashley Mitchell
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Hsuan-Ni Lin
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Linda Effiong
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Emaad Iqbal
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Neel Jingar
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Brian Kim
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Neel Shah
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - William Munoz
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Saad B Chaudhary
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Sheldon S Lin
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Joseph Benevenia
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - J Patrick O'Connor
- Department of Orthopaedics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
5
|
Sabouni K, Ozturk Y, Kacar E, Mutlu HS, Solakoglu S, Kose GT, Kok FN, Kazmanli MK, Urgen KM, Onder S. Assessment of bone healing using (Ti,Mg)N thin film coated plates and screws: Rabbit femur model. J Biomed Mater Res B Appl Biomater 2020; 109:227-237. [PMID: 32770599 DOI: 10.1002/jbm.b.34694] [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: 01/10/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 11/06/2022]
Abstract
Magnesium (Mg) based implants such as plates and screws are often preferred to treat bone defects because of the positive effects of magnesium in bone growth and healing. Their low corrosion resistance, however, leads to fast degradation and consequently failure before healing was completed. Previously, we developed Mg doped titanium nitrate (TiN) thin film coatings to address these limitations and demonstrated that <10 at% Mg doping led to enhanced mineralization in vitro. In the present study, in vivo performance of (Ti,Mg)N coated Ti6Al4V based plates and screws were studied in the rabbit model. Bone fractures were formed on femurs of 16 rabbits and then fixed with either (Ti,Mg)N coated (n = 8) or standard TiN coated (n = 8) plates and screws. X-ray imaging and μCT analyses showed enhanced bone regeneration on fracture sites fixed with (Ti,Mg)N coated plates in comparison with the Mg free ones. Bone mineral density, bone volume, and callus volume were also found to be 11.4, 23.4, and 42.8% higher, respectively, in accordance with μCT results. Furthermore, while TiN coatings promoted only primary bone regeneration, (Ti,Mg)N led to secondary bone regeneration in 6 weeks. These results indicated that Mg presence in the coatings accelerated bone regeneration in the fracture site. (Ti,Mg)N coating can be used as a practical method to increase the efficiency of existing bone fixation devices of varying geometry.
Collapse
Affiliation(s)
- Kenda Sabouni
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Yetkin Ozturk
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Erkan Kacar
- Department of Metallurgical and Materials Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Hasan Serdar Mutlu
- Department of Histology and Embryology, Istanbul University, Istanbul, Turkey
| | - Seyhun Solakoglu
- Department of Histology and Embryology, Istanbul University, Istanbul, Turkey
| | - Gamze Torun Kose
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Fatma Nese Kok
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Muhammet Kursat Kazmanli
- Department of Metallurgical and Materials Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Kamil Mustafa Urgen
- Department of Metallurgical and Materials Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Sakip Onder
- Department of Biomedical Engineering, Yildiz Technical University, Istanbul, Turkey
| |
Collapse
|
6
|
Bis(maltolato)oxovanadium(IV) Induces Angiogenesis via Phosphorylation of VEGFR2. Int J Mol Sci 2020; 21:ijms21134643. [PMID: 32629855 PMCID: PMC7370103 DOI: 10.3390/ijms21134643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 11/17/2022] Open
Abstract
VEGFR2 and VEGF-A play a pivotal role in the process of angiogenesis. VEGFR2 activation is regulated by protein tyrosine phosphatases (PTPs), enzymes that dephosphorylate the receptor and reduce angiogenesis. We aim to study the effect of PTPs blockade using bis(maltolato)oxovanadium(IV) (BMOV) on in vivo wound healing and in vitro angiogenesis. BMOV significantly improves in vivo wound closure by 45% in C57BL/6JRj mice. We found that upon VEGFR2 phosphorylation induced by endogenously produced VEGF-A, the addition of BMOV results in increased cell migration (45%), proliferation (40%) and tube formation (27%) in HUVECs compared to control. In a mouse ex vivo, aortic ring assay BMOV increased the number of sprouts by 3 folds when compared to control. However, BMOV coadministered with exogenous VEGF-A increased ECs migration, proliferation and tube formation by only 41%, 18% and 12% respectively and aortic ring sprouting by only 1-fold. We also found that BMOV enhances VEGFR2 Y951 and p38MAPK phosphorylation, but not ERK1/2. The level of phosphorylation of these residues was the same in the groups treated with BMOV supplemented with exogenous VEGF-A and exogenous VEGF-A only. Our study demonstrates that BMOV is able to enhance wound closure in vivo. Moreover, in the presence of endogenous VEGF-A, BMOV is able to stimulate in vitro angiogenesis by increasing the phosphorylation of VEGFR2 and its downstream proangiogenic enzymes. Importantly, BMOV had a stronger proangiogenic effect compared to its effect in coadministration with exogenous VEGF-A.
Collapse
|
7
|
O’Neill E, Awale G, Daneshmandi L, Umerah O, Lo KWH. The roles of ions on bone regeneration. Drug Discov Today 2018; 23:879-890. [DOI: 10.1016/j.drudis.2018.01.049] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/04/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
|
8
|
Glenske K, Donkiewicz P, Köwitsch A, Milosevic-Oljaca N, Rider P, Rofall S, Franke J, Jung O, Smeets R, Schnettler R, Wenisch S, Barbeck M. Applications of Metals for Bone Regeneration. Int J Mol Sci 2018; 19:E826. [PMID: 29534546 PMCID: PMC5877687 DOI: 10.3390/ijms19030826] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/09/2018] [Accepted: 03/11/2018] [Indexed: 02/06/2023] Open
Abstract
The regeneration of bone tissue is the main purpose of most therapies in dental medicine. For bone regeneration, calcium phosphate (CaP)-based substitute materials based on natural (allo- and xenografts) and synthetic origins (alloplastic materials) are applied for guiding the regeneration processes. The optimal bone substitute has to act as a substrate for bone ingrowth into a defect, as well as resorb in the time frame needed for complete regeneration up to the condition of restitution ad integrum. In this context, the modes of action of CaP-based substitute materials have been frequently investigated, where it has been shown that such materials strongly influence regenerative processes such as osteoblast growth or differentiation and also osteoclastic resorption due to different physicochemical properties of the materials. However, the material characteristics needed for the required ratio between new bone tissue formation and material degradation has not been found, until now. The addition of different substances such as collagen or growth factors and also of different cell types has already been tested but did not allow for sufficient or prompt application. Moreover, metals or metal ions are used differently as a basis or as supplement for different materials in the field of bone regeneration. Moreover, it has already been shown that different metal ions are integral components of bone tissue, playing functional roles in the physiological cellular environment as well as in the course of bone healing. The present review focuses on frequently used metals as integral parts of materials designed for bone regeneration, with the aim to provide an overview of currently existing knowledge about the effects of metals in the field of bone regeneration.
Collapse
Affiliation(s)
- Kristina Glenske
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, D-35392 Giessen, Germany.
| | | | | | - Nada Milosevic-Oljaca
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, D-35392 Giessen, Germany.
| | | | - Sven Rofall
- Botiss Biomaterials, D-12109 Berlin, Germany.
| | - Jörg Franke
- Clinic for Trauma Surgery and Orthopedics, Elbe Kliniken Stade-Buxtehude, D-21682 Stade, Germany.
| | - Ole Jung
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg- Eppendorf, D-20246 Hamburg, Germany.
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg- Eppendorf, D-20246 Hamburg, Germany.
| | | | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, D-35392 Giessen, Germany.
| | - Mike Barbeck
- Botiss Biomaterials, D-12109 Berlin, Germany.
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg- Eppendorf, D-20246 Hamburg, Germany.
| |
Collapse
|
9
|
Ippolito JA, Krell ES, Cottrell J, Meyer R, Clark D, Nguyen D, Sudah S, Muñoz M, Lim E, Lin A, Lee TJH, O'Connor JP, Benevenia J, Lin SS. Effects of local vanadium delivery on diabetic fracture healing. J Orthop Res 2017; 35:2174-2180. [PMID: 28084655 DOI: 10.1002/jor.23521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 01/09/2017] [Indexed: 02/04/2023]
Abstract
This study evaluated the effect of local vanadyl acetylacetonate (VAC), an insulin mimetic agent, upon the early and late parameters of fracture healing in rats using a standard femur fracture model. Mechanical testing, and radiographic scoring were performed, as well as histomorphometry, including percent bone, percent cartilage, and osteoclast numbers. Fractures treated with local 1.5 mg/kg VAC possessed significantly increased mechanical properties compared to controls at 6 weeks post-fracture, including increased torque to failure (15%; p = 0.046), shear modulus (89%; p = 0.043), and shear stress (81%; p = 0.009). The radiographic scoring analysis showed increased cortical bridging at 4 weeks and 6 weeks (119%; p = 0.036 and 209%; p = 0.002) in 1.5 mg/kg VAC treated groups. Histomorphometry of the fracture callus at days 10 and 14 showed increased percent cartilage (121%; p = 0.009 and 45%; p = 0.035) and percent mineralized tissue (66%; p = 0.035 and 58%; p = 0.006) with local VAC treated groups compared to control. Additionally, fewer osteoclasts were observed in the local VAC treated animals as compared to controls at day 14 (0.45% ± 0.29% vs. 0.83% ± 0.36% of callus area; p = 0.032). The results suggest local administration of VAC acts to modulate osteoclast activity and increase percentage of early callus cartilage, ultimately enhancing mechanical properties comparably to non-diabetic animals treated with local VAC. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2174-2180, 2017.
Collapse
Affiliation(s)
- Joseph A Ippolito
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Ethan S Krell
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Jessica Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey
| | - Ryan Meyer
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Devin Clark
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Daniel Nguyen
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Suleiman Sudah
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Maximillian Muñoz
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Elisha Lim
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Anthony Lin
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Thomas Jae Hoon Lee
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - James Patrick O'Connor
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Joseph Benevenia
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| | - Sheldon S Lin
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07101
| |
Collapse
|
10
|
Schussler SD, Uske K, Marwah P, Kemp FW, Bogden JD, Lin SS, Livingston Arinzeh T. Controlled Release of Vanadium from a Composite Scaffold Stimulates Mesenchymal Stem Cell Osteochondrogenesis. AAPS JOURNAL 2017; 19:1017-1028. [PMID: 28332167 DOI: 10.1208/s12248-017-0073-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/06/2017] [Indexed: 01/03/2023]
Abstract
Large bone defects often require the use of autograft, allograft, or synthetic bone graft augmentation; however, these treatments can result in delayed osseous integration. A tissue engineering strategy would be the use of a scaffold that could promote the normal fracture healing process of endochondral ossification, where an intermediate cartilage phase is later transformed to bone. This study investigated vanadyl acetylacetonate (VAC), an insulin mimetic, combined with a fibrous composite scaffold, consisting of polycaprolactone with nanoparticles of hydroxyapatite and beta-tricalcium phosphate, as a potential bone tissue engineering scaffold. The differentiation of human mesenchymal stem cells (MSCs) was evaluated on 0.05 and 0.025 wt% VAC containing composite scaffolds (VAC composites) in vitro using three different induction media: osteogenic (OS), chondrogenic (CCM), and chondrogenic/osteogenic (C/O) media, which mimics endochondral ossification. The controlled release of VAC was achieved over 28 days for the VAC composites, where approximately 30% of the VAC was released over this period. MSCs cultured on the VAC composites in C/O media had increased alkaline phosphatase activity, osteocalcin production, and collagen synthesis over the composite scaffold without VAC. In addition, gene expressions for chondrogenesis (Sox9) and hypertrophic markers (VEGF, MMP-13, and collagen X) were the highest on VAC composites. Almost a 1000-fold increase in VEGF gene expression and VEGF formation, as indicated by immunostaining, was achieved for cells cultured on VAC composites in C/O media, suggesting VAC will promote angiogenesis in vivo. These results demonstrate the potential of VAC composite scaffolds in supporting endochondral ossification as a bone tissue engineering strategy.
Collapse
Affiliation(s)
- S D Schussler
- Department of Chemical, Biological and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, New Jersey, 07102, USA
| | - K Uske
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, New Jersey, 07102, USA
| | - P Marwah
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, New Jersey, 07102, USA
| | - F W Kemp
- Department of Preventive Medicine and Community Health, New Jersey Medical School, Rutgers University, Newark, New Jersey, 07103, USA
| | - J D Bogden
- Department of Preventive Medicine and Community Health, New Jersey Medical School, Rutgers University, Newark, New Jersey, 07103, USA
| | - S S Lin
- Department of Orthopaedic Surgery, New Jersey Medical School, Rutgers University, Newark, New Jersey, 07103, USA
| | - Treena Livingston Arinzeh
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, New Jersey, 07102, USA.
| |
Collapse
|
11
|
Khader A, Sherman LS, Rameshwar P, Arinzeh TL. Sodium Tungstate for Promoting Mesenchymal Stem Cell Chondrogenesis. Stem Cells Dev 2016; 25:1909-1918. [PMID: 27615276 PMCID: PMC5165671 DOI: 10.1089/scd.2016.0158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023] Open
Abstract
Articular cartilage has a limited ability to heal. Mesenchymal stem cells (MSCs) derived from the bone marrow have shown promise as a cell type for cartilage regeneration strategies. In this study, sodium tungstate (Na2WO4), which is an insulin mimetic, was evaluated for the first time as an inductive factor to enhance human MSC chondrogenesis. MSCs were seeded onto three-dimensional electrospun scaffolds in growth medium (GM), complete chondrogenic induction medium (CCM) containing insulin, and CCM without insulin. Na2WO4 was added to the media leading to final concentrations of 0, 0.01, 0.1, and 1 mM. Chondrogenic differentiation was assessed by biochemical analyses, immunostaining, and gene expression. Cytotoxicity using human peripheral blood mononuclear cells (PBMCS) was also investigated. The chondrogenic differentiation of MSCs was enhanced in the presence of low concentrations of Na2WO4 compared to control, without Na2WO4. In the induction medium containing insulin, cells in 0.01 mM Na2WO4 produced significantly higher sulfated glycosaminoglycans, collagen type II, and chondrogenic gene expression than all other groups at day 28. Cells in 0.1 mM Na2WO4 had significantly higher collagen II production and significantly higher sox-9 and aggrecan gene expression compared to control at day 28. Cells in GM and induction medium without insulin containing low concentrations of Na2WO4 also expressed chondrogenic markers. Na2WO4 did not stimulate PBMC proliferation or apoptosis. The results demonstrate that Na2WO4 enhances chondrogenic differentiation of MSCs, does not have a toxic effect, and may be useful for MSC-based approaches for cartilage repair.
Collapse
Affiliation(s)
- Ateka Khader
- 1 Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Lauren S Sherman
- 2 Department of Medicine-Hematology/Oncology, Rutgers-New Jersey Medical School , Newark, New Jersey
| | - Pranela Rameshwar
- 2 Department of Medicine-Hematology/Oncology, Rutgers-New Jersey Medical School , Newark, New Jersey
| | - Treena L Arinzeh
- 1 Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| |
Collapse
|
12
|
Suliburska J, Kocyłowski R, Komorowicz I, Grzesiak M, Bogdański P, Barałkiewicz D. Concentrations of Mineral in Amniotic Fluid and Their Relations to Selected Maternal and Fetal Parameters. Biol Trace Elem Res 2016; 172:37-45. [PMID: 26547910 PMCID: PMC4893386 DOI: 10.1007/s12011-015-0557-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/25/2015] [Indexed: 12/12/2022]
Abstract
The concentrations of various trace elements in amniotic fluid (AF) change over the course of pregnancy, with gestational age and fetus growth. The aim of the present study was to evaluate the concentrations of selected essential and toxic elements in AF and their relations to maternal and fetal parameters. The study was carried out in 39 pregnant women, aged 34.6 ± 4.7 years, between weeks 16 and 26 of gestation. Amniotic fluid samples were obtained during the standard procedure of amniocentesis in high-risk patients for chromosomal abnormalities. An inductively coupled plasma mass spectrometry (ICP-MS) technique was used to determine the levels of Al, As, Ba, Cd, Co, Cr, Cu, Mg, Mn, Ni, Sr, U, and V in AF. Body mass and blood pressure were measured in all the women. The basic parameters of fetal development were also assayed. It was found that the age of the mother, the gender of the fetus, and the week of the pregnancy may affect the concentrations of mineral in the amniotic fluid. Moreover, several significant correlations between the essential and toxic elements and maternal and fetal parameters were observed. In particular, negative and positive correlations between fetal parameters and magnesium and copper levels in AF, respectively, were seen. The present findings demonstrate the association between minerals in AF and fetal development.
Collapse
Affiliation(s)
- J Suliburska
- Department of Human Nutrition and Hygiene, Poznan University of Life Sciences, ul. Wojska Polskiego 31, 60-624, Poznan, Poland.
| | - R Kocyłowski
- PreMediCare New Med Medical Centre, ul. Drużbickiego 13, 61-693, Poznan, Poland
- Department of Perinatology and Gynecology, Polish Mother's Memorial Hospital Research Institute, ul. Rzgowska 281/289, 93-338, Łódź, Poland
| | - I Komorowicz
- Department of Trace Element Analysis by Spectroscopy Method, Faculty of Chemistry, Adam Mickiewicz University in Poznań, ul. Umultowska 89b, 61-614, Poznan, Poland
| | - M Grzesiak
- Department of Perinatology and Gynecology, Polish Mother's Memorial Hospital Research Institute, ul. Rzgowska 281/289, 93-338, Łódź, Poland
| | - P Bogdański
- Department of Education and Obesity Treatment and Metabolic Disorders, University of Medical Sciences, ul. Szamarzewskiego 84, 60-569, Poznan, Poland
| | - D Barałkiewicz
- Department of Trace Element Analysis by Spectroscopy Method, Faculty of Chemistry, Adam Mickiewicz University in Poznań, ul. Umultowska 89b, 61-614, Poznan, Poland
| |
Collapse
|
13
|
Vasconcelos DM, Santos SG, Lamghari M, Barbosa MA. The two faces of metal ions: From implants rejection to tissue repair/regeneration. Biomaterials 2016; 84:262-275. [DOI: 10.1016/j.biomaterials.2016.01.046] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
|
14
|
E. Klontzas M, I. Kenanidis E, J. MacFarlane R, Michail T, E. Potoupnis M, Heliotis M, Mantalaris A, Tsiridis E. Investigational drugs for fracture healing: preclinical & clinical data. Expert Opin Investig Drugs 2016; 25:585-96. [DOI: 10.1517/13543784.2016.1161757] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Saghiri MA, Orangi J, Asatourian A, Sorenson CM, Sheibani N. Functional role of inorganic trace elements in angiogenesis part III: (Ti, Li, Ce, As, Hg, Va, Nb and Pb). Crit Rev Oncol Hematol 2015; 98:290-301. [PMID: 26638864 DOI: 10.1016/j.critrevonc.2015.10.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/27/2015] [Accepted: 10/15/2015] [Indexed: 02/02/2023] Open
Abstract
Many essential elements exist in nature with significant influence on human health. Angiogenesis is vital in developmental, repair, and regenerative processes, and its aberrant regulation contributes to pathogenesis of many diseases including cancer. Thus, it is of great importance to explore the role of these elements in such a vital process. This is third in a series of reviews that serve as an overview of the role of inorganic elements in regulation of angiogenesis and vascular function. Here we will review the roles of titanium, lithium, cerium, arsenic, mercury, vanadium, niobium, and lead in these processes. The roles of other inorganic elements in angiogenesis were discussed in part I (N, Fe, Se, P, Au, and Ca) and part II (Cr, Si, Zn, Cu, and S) of these series. The methods of exposure, structure, mechanisms, and potential activities of these elements are briefly discussed. An electronic search was performed on the role of these elements in angiogenesis from January 2005 to April 2014. These elements can promote and/or inhibit angiogenesis through different mechanisms. The anti-angiogenic effect of titanium dioxide nanoparticles comes from the inhibition of angiogenic processes, and not from its toxicity. Lithium affects vasculogenesis but not angiogenesis. Nanoceria treatment inhibited tumor growth by inhibiting angiogenesis. Vanadium treatment inhibited cell proliferation and induced cytotoxic effects through interactions with DNA. The negative impact of mercury on endothelial cell migration and tube formation activities was dose and time dependent. Lead induced IL-8 production, which is known to promote tumor angiogenesis. Thus, understanding the impact of these elements on angiogenesis will help in development of new modalities to modulate angiogenesis under various conditions.
Collapse
Affiliation(s)
- Mohammad Ali Saghiri
- Departments of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Angiogenesis and Regenerative Group, Dr. H. Afsar Lajevardi Research Cluster, Shiraz, Iran.
| | - Jafar Orangi
- Angiogenesis and Regenerative Group, Dr. H. Afsar Lajevardi Research Cluster, Shiraz, Iran
| | - Armen Asatourian
- Angiogenesis and Regenerative Group, Dr. H. Afsar Lajevardi Research Cluster, Shiraz, Iran
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
16
|
Drissi H, Paglia DN. Surgical procedures and experimental outcomes of closed fractures in rodent models. Methods Mol Biol 2015; 1226:193-211. [PMID: 25331052 DOI: 10.1007/978-1-4939-1619-1_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The closed fracture rat model, first described by Bonnarens and Einhorn, has been widely implemented in recent years to characterize various fracture phenotypes and evaluate treatment modalities. Slight modifications in the fixation depth, to reduce surgical error associated with movement/dislocation of the k-wire fixation, were previously described. Here, we describe this method which involves the creation of a medial parapatellar incision, dislocation of the patella, boring an 18 gauge hole through the center of the femur, delivery of an adjunct (if applicable), fixation of the k-wire in the greater trochanter of the femur, suturing of muscle and skin, and finally creation of the mid-diaphyseal fracture with a three-point bending fracture device. Many laboratories routinely perform surgical procedures in which a closed fracture is induced using rat or mouse models. The benefits of such surgical models range from general orthopaedic trauma applications to the assessment of the healing process in genetically modified animals. Other important applications include the assessment of the safety and efficacy of various treatment modalities as well as the characterization of bone repair in metabolic bone diseases or skeletal dysplasia.
Collapse
Affiliation(s)
- Hicham Drissi
- Department of Orthopaedic Surgery, New England Musculoskeletal Institute, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA,
| | | |
Collapse
|
17
|
Hreha J, Wey A, Cunningham C, Krell ES, Brietbart EA, Paglia DN, Montemurro NJ, Nguyen DA, Lee YJ, Komlos D, Lim E, Benevenia J, O'Connor JP, Lin SS. Local manganese chloride treatment accelerates fracture healing in a rat model. J Orthop Res 2015; 33:122-30. [PMID: 25231276 DOI: 10.1002/jor.22733] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 08/20/2014] [Indexed: 02/04/2023]
Abstract
This study investigated the effects of local delivery of manganese chloride (MnCl2), an insulin-mimetic compound, upon fracture healing using a rat femoral fracture model. Mechanical testing, histomorphometry, and immunohistochemistry were performed to assess early and late parameters of fracture healing. At 4 weeks post-fracture, maximum torque to failure was 70% higher (P<0.05) and maximum torsional rigidity increased 133% (P<0.05) in animals treated with 0.125 mg/kg MnCl2 compared to saline controls. Histological analysis of the fracture callus revealed percent new mineralized tissue was 17% higher (P<0.05) at day 10. Immunohistochemical analysis of the 0.125 mg/kg MnCl2 treated group, compared to saline controls, showed a 379% increase in the density of VEGF-C+ cells. In addition, compared to saline controls, the 0.125 mg/kg MnCl2 treated group showed a 233% and 150% increase in blood vessel density in the subperiosteal region at day 10 post-fracture as assessed by detection of PECAM and smooth muscle α actin, respectively. The results suggest that local MnCl2 treatment accelerates fracture healing by increasing mechanical parameters via a potential mechanism of amplified early angiogenesis leading to increased osteogenesis. Therefore, local administration of MnCl2 is a potential therapeutic adjunct for fracture healing.
Collapse
Affiliation(s)
- Jeremy Hreha
- Department of Orthopaedics, Rutgers-New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey, 07103
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Drissi H, Paglia DN, Alaee F, Yoshida R. Constructing the toolbox: Patient-specific genetic factors of altered fracture healing. Genes Dis 2014; 1:140-148. [PMID: 25558470 PMCID: PMC4280851 DOI: 10.1016/j.gendis.2014.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/18/2014] [Indexed: 01/10/2023] Open
Abstract
The multifaceted sequence of events that follow fracture repair can be further complicated when considering risk factors for impaired union, present in a large and growing percentage of the population. Risk factors such as diabetes, substance abuse, and poor nutrition affect both the young and old alike, and have been shown to dramatically impair the body's natural healing processes. To this end, biotherapeudic interventions such as ultrasound, electrical simulation, growth factor treatment (BMP-2, BMP-7, PDGF-BB, FGF-2) have been evaluated in preclinical models and in some cases are used widely for patients with established non-union or risk/indication or impaired healing (ie. ultrasound, BMP-2, etc.). Despite the promise of these interventions, they have been shown to be reliant on patient compliance and can produce adverse side-effects such as heterotopic ossification. Gene and cell therapy approaches have attempted to apply controlled regimens of these factors and have produced promising results. However, there are safety and efficacy concerns that may limit the translation of these approaches. In addition, none of the above mentioned approaches consider genetic variation between individual patients. Several clinical and preclinical studies have demonstrated a genetic component to fracture repair and that SNPs and genetic background variation play major roles in the determination of healing outcomes. Despite this, there is a need for preclinical data to dissect the mechanism underlying the influence of specific gene loci on the processes of fracture healing, which will be paramount in the future of patient-centered interventions for fracture repair.
Collapse
Affiliation(s)
- Hicham Drissi
- New England Musculoskeletal Institute and Department of Orthopaedic Surgery, United States
| | | | | | | |
Collapse
|
19
|
Makinen MW, Salehitazangi M. The Structural Basis of Action of Vanadyl (VO 2+) Chelates in Cells. Coord Chem Rev 2014; 279:1-22. [PMID: 25237207 DOI: 10.1016/j.ccr.2014.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Much emphasis has been given to vanadium compounds as potential therapeutic reagents for the treatment of diabetes mellitus. Thus far, no vanadium compound has proven efficacious for long-term treatment of this disease in humans. Therefore, in review of the research literature, our goal has been to identify properties of vanadium compounds that are likely to favor physiological and biochemical compatibility for further development as therapeutic reagents. We have, therefore, limited our review to those vanadium compounds that have been used in both in vivo experiments with small, laboratory animals and in in vitro studies with primary or cultured cell systems and for which pharmacokinetic and pharmacodynamics results have been reported, including vanadium tissue content, vanadium and ligand lifetime in the bloodstream, structure in solution, and interaction with serum transport proteins. Only vanadyl (VO2+) chelates fulfill these requirements despite the large variety of vanadium compounds of different oxidation states, ligand structure, and coordination geometry synthesized as potential therapeutic agents. Extensive review of research results obtained with use of organic VO2+-chelates shows that the vanadyl chelate bis(acetylacetonato)oxidovanadium(IV) [hereafter abbreviated as VO(acac)2], exhibits the greatest capacity to enhance insulin receptor kinase activity in cells compared to other organic VO2+-chelates, is associated with a dose-dependent capacity to lower plasma glucose in diabetic laboratory animals, and exhibits a sufficiently long lifetime in the blood stream to allow correlation of its dose-dependent action with blood vanadium content. The properties underlying this behavior appear to be its high stability and capacity to remain intact upon binding to serum albumin. We relate the capacity to remain intact upon binding to serum albumin to the requirement to undergo transcytosis through the vascular endothelium to gain access to target tissues in the extravascular space. Serum albumin, as the most abundant transport protein in the blood stream, serves commonly as the carrier protein for small molecules, and transcytosis of albumin through capillary endothelium is regulated by a Src protein tyrosine kinase system. In this respect it is of interest to note that inorganic VO2+ has the capacity to enhance insulin receptor kinase activity of intact 3T3-L1 adipocytes in the presence of albumin, albeit weak; however, in the presence of transferrin no activation is observed. In addition to facilitating glucose uptake, the capacity of VO2+- chelates for insulin-like, antilipolytic action in primary adipocytes has also been reviewed. We conclude that measurement of inhibition of release of only free fatty acids from adipocytes stimulated by epinephrine is not a sufficient basis to ascribe the observations to purely insulin-mimetic, antilipolytic action. Adipocytes are known to contain both phosphodiesterase-3 and phosphodiesterase-4 (PDE3 and PDE4) isozymes, of which insulin antagonizes lipolysis only through PDE3B. It is not known whether the other isozyme in adipocytes is influenced directly by VO2+- chelates. In efforts to promote improved development of VO2+- chelates for therapeutic purposes, we propose synergism of a reagent with insulin as a criterion for evaluating physiological and biochemical specificity of action. We highlight two organic compounds that exhibit synergism with insulin in cellular assays. Interestingly, the only VO2+- chelate for which this property has been demonstrated, thus far, is VO(acac)2.
Collapse
Affiliation(s)
- Marvin W Makinen
- Department of Biochemistry & Molecular Biology, Gordon Center for Integrative Science, The University of Chicago, 929 East 57 Street, Chicago, Illinois 60637 USA
| | - Marzieh Salehitazangi
- Department of Biochemistry & Molecular Biology, Gordon Center for Integrative Science, The University of Chicago, 929 East 57 Street, Chicago, Illinois 60637 USA
| |
Collapse
|
20
|
Abstract
OBJECTIVES Wnt5a expression is upregulated during fracture repair and has previously been implicated as a potential regulator of skeletal development and bone mass accrual and maintenance. Our objective was to evaluate the function of Wnt5a in fracture healing. METHODS Femoral fracture experiments on Wnt5a and Wnt5a mice were carried out. To better understand the effect of the Wnt5a on bone repair, we evaluated radiographs using a previously validated qualitative scoring system and performed microcomputed tomography analyses. Histomorphometric analyses determined the temporal distribution of stroma, cartilage matrix, and woven bone in the fracture callus. Finally, we performed tartrate-resistant acid phosphatase (TRAP) immunohistochemical staining to visualize and quantify bone resorbing cells. RESULTS Radiographic evaluations at day 21 demonstrated significantly higher cortical remodeling and bridging parameters for the Wnt5a group compared with the Wnt5a group. The bone volume fraction by microcomputed tomography was also significantly increased in Wnt5a mice. Histological and histomorphometric analyses showed that although Wnt5a mice exhibit decreased cartilage matrix production at day 7 postfracture, they displayed increased residual cartilaginous callus at days 14 and 21 compared with the Wnt5a group. In addition, the total number of multinucleated tartrate-resistant acid phosphatase-positive cells was significantly lower in the Wnt5a group than in the Wnt5a group. CONCLUSIONS The data indicate that decreased Wnt5a signaling impaired proper fracture healing, possibly through decreased cartilaginous callus formation, and delayed cartilage matrix and mineralized tissue remodeling within the fracture callus.
Collapse
|
21
|
Wey A, Cunningham C, Hreha J, Breitbart E, Cottrell J, Ippolito J, Clark D, Lin HN, Benevenia J, O'Connor JP, Lin SS, Paglia DN. Local ZnCl2 accelerates fracture healing. J Orthop Res 2014; 32:834-41. [PMID: 24574139 DOI: 10.1002/jor.22593] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 01/13/2014] [Indexed: 02/04/2023]
Abstract
This study evaluated the effect of local zinc chloride (ZnCl2 ), an insulin mimetic agent, upon the early and late parameters of fracture healing in rats using a standard femur fracture model. Mechanical testing, radiographic scoring, histomorphometry, qualitative histological scoring, PCNA immunohistochemistry, and local growth factor analysis were performed. Fractures treated with local ZnCl2 possessed significantly increased mechanical properties compared to controls at 4 weeks post fracture. The radiographic scoring analysis showed increased cortical bridging at 4 weeks in the 1.0 (p=0.0015) and 3.0 (p<0.0001) mg/kg ZnCl2 treated groups. Histomorphometry of the fracture callus at day 7 showed 177% increase (p=0.036) in percent cartilage and 133% increase (p=0.002) in percent mineralized tissue with local ZnCl2 treatment compared to controls. Qualitative histological scoring showed a 2.1× higher value at day 7 in the ZnCl2 treated group compared to control (p = 0.004). Cell proliferation and growth factors, VEGF and IGF-I, within fracture calluses treated with local ZnCl2 were increased at day 7. The results suggest local administration of ZnCl2 increases cell proliferation, causing increased growth factor production which yields improved chondrogenesis and endochondral ossification. Ultimately, these events lead to accelerated fracture healing as early as 4 weeks post fracture.
Collapse
Affiliation(s)
- Aaron Wey
- Rutgers New Jersey Medical School, Department of Orthopaedics, 90 Bergen Street, Suite 7300, Newark, New Jersey, 07103
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Paglia DN, Wey A, Hreha J, Park AG, Cunningham C, Uko L, Benevenia J, O'Connor JP, Lin SS. Local vanadium release from a calcium sulfate carrier accelerates fracture healing. J Orthop Res 2014; 32:727-34. [PMID: 24375684 DOI: 10.1002/jor.22570] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 12/05/2013] [Indexed: 02/04/2023]
Abstract
This study evaluated the efficacy of using calcium sulfate (CaSO4 ) as a carrier for intramedullary delivery of an organic vanadium salt, vanadyl acetylacetonate (VAC) after femoral fracture. VAC can act as an insulin-mimetic and can be used to accelerate fracture healing in rats. A heterogenous mixture of VAC and CaSO4 was delivered to the fracture site of BB Wistar rats, and mechanical testing, histomorphometry, micro-computed tomography (micro-CT) were performed to measure healing. At 4 weeks after fracture, maximum torque to failure, effective shear modulus, and effective shear stress were all significantly higher (p < 0.05) in rats treated with 0.25 mg/kg VAC-CaSO4 as compared to carrier control rats. Histomorphometry found a 71% increase in percent cartilage matrix (p < 0.05) and a 64% decrease in percent mineralized tissue (p < 0.05) at 2 weeks after fracture in rats treated with 0.25 mg/kg of VAC-CaSO4 . Micro-CT analyses at 4 weeks found a more organized callus structure and higher trending maximum connected z-ray. fraction for VAC-CaSO4 groups. Evaluation of radiographs and serial histological sections at 12 weeks did not show any evidence of ectopic bone formation. As compared to previous studies, CaSO4 was an effective carrier for reducing the dose of VAC required to accelerate femoral fracture healing in rats.
Collapse
Affiliation(s)
- David N Paglia
- Department of Orthopaedics, Rutgers New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey, 07103
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Orthopedic injuries are common and a source of much misery and economic stress. Several relevant tissues, such as cartilage, meniscus, and intra-articular ligaments, do not heal. And even bone, which normally regenerates spontaneously, can fail to mend. The regeneration of orthopedic tissues requires 4 key components: cells, morphogenetic signals, scaffolds, and an appropriate mechanical environment. Although differentiated cells from the tissue in question can be used, most cellular research focuses on the use of mesenchymal stem cells. These can be retrieved from many different tissues, and one unresolved question is the degree to which the origin of the cells matters. Embryonic and induced pluripotent stem cells are also under investigation. Morphogenetic signals are most frequently supplied by individual recombinant growth factors or native mixtures provided by, for example, platelet-rich plasma; mesenchymal stem cells are also a rich source of trophic factors. Obstacles to the sustained delivery of individual growth factors can be addressed by gene transfer or smart scaffolds, but we still lack detailed, necessary information on which delivery profiles are needed. Scaffolds may be based on natural products, synthetic materials, or devitalized extracellular matrix. Strategies to combine these components to regenerate tissue can follow traditional tissue engineering practices, but these are costly, cumbersome, and not well suited to treating large numbers of individuals. More expeditious approaches make full use of intrinsic biological processes in vivo to avoid the need for ex vivo expansion of autologous cells and multiple procedures. Clinical translation remains a bottleneck.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Collaborative Research Center, AO Foundation, Davos, Switzerland.
| |
Collapse
|
24
|
Patent Highlights. Pharm Pat Anal 2013. [DOI: 10.4155/ppa.13.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Snapshot of recent key developments in the patent literature of relevance to the advancement of pharmaceutical and medical R&D
Collapse
|
25
|
Paglia DN, Wey A, Breitbart EA, Faiwiszewski J, Mehta SK, Al-Zube L, Vaidya S, Cottrell JA, Graves D, Benevenia J, O’Connor JP, Lin SS. Effects of local insulin delivery on subperiosteal angiogenesis and mineralized tissue formation during fracture healing. J Orthop Res 2013; 31:783-91. [PMID: 23238777 PMCID: PMC6446235 DOI: 10.1002/jor.22288] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/08/2012] [Indexed: 02/04/2023]
Abstract
Local insulin delivery has been shown to improve osseous healing in diabetic animals. The purpose of this study was to quantify the effects of local intramedullary delivery of saline or Ultralente insulin (UL) on various fracture healing parameters using an in vivo non-diabetic BB Wistar rat model. Quantitation of local insulin levels showed a rapid release of insulin from the fractured femora, demonstrating complete release at 2 days. RT-PCR analysis revealed that the expression of early osteogenic markers (Col1α2, osteopontin) was significantly enhanced with UL treatment when compared with saline controls (p < 0.05). Significant differences in VEGF + cells and vascularity were evident between the treatment and control groups at day 7 (p < 0.05). At day 21, histomorphometric analysis demonstrated a significant increase in percent mineralized tissue in the UL-treated animals compared with controls (p < 0.05), particularly within the subperiosteal region of the fracture callus. Mechanical testing at 4 weeks showed significantly greater mechanical strength for UL-treated animals (p < 0.05), but healing in control animals caught up at 6 weeks post-fracture. These results suggest that the primary osteogenic effect of UL during the early stages of fracture healing (1-3 weeks) is through an increase in osteogenic gene expression, subperiosteal angiogenesis, and mineralized tissue formation.
Collapse
Affiliation(s)
- David N. Paglia
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| | - Aaron Wey
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| | - Eric A. Breitbart
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| | - Jonathan Faiwiszewski
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| | - Siddhant K. Mehta
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| | - Loay Al-Zube
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, Newark, New Jersey,Department of Biomedical Engineering, The Hashemite University, Zarqa 13115, Jordan
| | - Swaroopa Vaidya
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103,Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, Newark, New Jersey
| | - Jessica A. Cottrell
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, Newark, New Jersey
| | - Dana Graves
- Department of Periodontics, University of Pennsylvania, 240 South 40 Street, Philadelphia, PA, 19104
| | - Joseph Benevenia
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| | - J. Patrick O’Connor
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, Newark, New Jersey
| | - Sheldon S. Lin
- Department of Orthopaedics, University of Medicine and Dentistry of New Jersey—New Jersey Medical School, 90 Bergen Street, Suite 7300, Newark, New Jersey 07103
| |
Collapse
|