1
|
Pereira NC, de Oliveira Silva J, De Sousa FB, Miranda SEM, Soares DCF, de Barros ALB. [99mTc]Tc-Phosphate-buffer system as a potential tracer for bone imaging. J Radioanal Nucl Chem 2021. [DOI: 10.1007/s10967-021-07869-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
2
|
Humbert J, Will O, Peñate-Medina T, Peñate-Medina O, Jansen O, Both M, Glüer CC. Comparison of photoacoustic and fluorescence tomography for the in vivo imaging of ICG-labelled liposomes in the medullary cavity in mice. PHOTOACOUSTICS 2020; 20:100210. [PMID: 33101928 PMCID: PMC7569329 DOI: 10.1016/j.pacs.2020.100210] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 05/20/2023]
Abstract
Few reports quantitatively compare the performance of photoacoustic tomography (PAT) versus fluorescence molecular tomography (FMT) in vivo. We compared both modalities for the detection of signals from injected ICG liposomes in the tibial medullary space of 10 BALB/c mice in vivo and ex vivo. Signals significantly correlated between modalities (R² = 0.69) and within each modality in vivo versus ex vivo (PAT: R² = 0.70, FMT: R² = 0.76). Phantom studies showed that signals at 4 mm depth are detected down to 3.3 ng ICG by PAT and 33 ng by FMT, with a nominal spatial resolution below 0.5 mm in PAT and limited to 1 mm in FMT. Our study demonstrates comparable in vivo sensitivity, but superior ex vivo sensitivity and in vivo resolution for our ICG liposomes of the VevoLAZR versus the FMT2500. PAT provides a useful new tool for the high-resolution imaging of bone marrow signals, for example for monitoring drug delivery.
Collapse
Key Words
- % ID, percent initial dose
- % PA signal, percent photoacoustic signal
- BMD, bone mineral density
- Bone
- DXA, dual-energy x-ray absorptiometry
- FLI, fluorescence imaging
- FMT, fluorescence molecular tomography
- Fluorescence imaging
- Hb, deoxygenated hemoglobin
- HbO2, oxygenated hemoglobin
- ICG, indocyanine green
- In vivo imaging
- LDF, laser-doppler flowmetry
- Liposomes
- M, mean
- Medullary space
- NIR, near-infrared
- PAI, photoacoustic imaging
- PAT, photoacoustic tomography
- Photoacoustic imaging
- QUS, quantitative ultrasound
- RFU, relative fluorescence units
- SD, standard deviation
- SEM, standard error of the mean
- Tibia
- US, ultrasound
Collapse
Affiliation(s)
- Jana Humbert
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
- Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Straße 3, 24105 Kiel, Germany
- Corresponding author at: Molecular Imaging North Competence Center (MOIN CC), Am Botanischen Garten 14, 24118 Kiel, Germany.
| | - Olga Will
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Tuula Peñate-Medina
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Oula Peñate-Medina
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Olav Jansen
- Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Marcus Both
- Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Claus-Christian Glüer
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| |
Collapse
|
3
|
Lin CC, Chang WHS, Cheng TM, Chiu LH, Wang YH, Lin CAJ, Ho YS, Zuo CS, Wang YM, Lai WFT. Two new, near-infrared, fluorescent probes as potential tools for imaging bone repair. Sci Rep 2020; 10:2580. [PMID: 32054952 PMCID: PMC7018698 DOI: 10.1038/s41598-020-59522-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 01/14/2020] [Indexed: 11/30/2022] Open
Abstract
A precise imaging technique to evaluate osteogenesis, osteodifferentiation, and osseointegration following peri-implant surgery is in high clinical demand. Herein, we report the generation of two new, near-infrared (NIR) fluorescent probes for use in the molecular imaging of bone repair. The first probe aims to monitor the in vitro differentiation of human mesenchymal stem cells (MSCs) into osteoblasts. A NIR fluorochrome was conjugated to a cyclic peptide that binds to integrin α5β1, a factor that promotes osteogenesis in MSCs and therefore functioned as an osteoblast-specific marker. The second probe aims to monitor osteogenesis, and was generated by conjugating the drug pamidronate to a NIR fluorescent gold nanocluster. Pamidronate specifically binds to hydroxyapatite (HA), a mineral present in bone that is produced by osteoblasts, and therefore provides a functional marker for new bone formation. Our results show that both probes bind to their specific targets in vitro-differentiated osteoblasts, and not to undifferentiated MSCs, and emit NIR fluorescence for functional detection. This in vitro work demonstrates the ability of these probes to bind to active osteoblasts and their mineral deposits and highlight their potential utility as clinical tools for the imaging of the osseointegration process at the molecular level.
Collapse
Affiliation(s)
- Chien-Chou Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Tsai-Mu Cheng
- Ph.D. Program for Translational Medicine, College of Medicine and Technology, Taipei Medical University, Taipei, Taiwan
| | - Li-Hsuan Chiu
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Department of Research and Department of Dentistry, Taipei Medical University/Shuang-Ho Hospital, New Taipei City, Taiwan
| | - Yen-Hsun Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan
| | - Cheng-An J Lin
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung-Li, Taiwan
| | - Yuan-Soon Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun S Zuo
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan.
| | - Wen-Fu Thomas Lai
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, USA.
- Institute of Graduate Clinical Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Research and Department of Dentistry, Taipei Medical University/Shuang-Ho Hospital, New Taipei City, Taiwan.
| |
Collapse
|
4
|
Cho N, Shokeen M. Changing landscape of optical imaging in skeletal metastases. J Bone Oncol 2019; 17:100249. [PMID: 31316892 PMCID: PMC6611980 DOI: 10.1016/j.jbo.2019.100249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023] Open
Abstract
Optical imaging is an emerging strategy for in vitro and in vivo visualization of the molecular mechanisms of cancer over time. An increasing number of optical imaging contrast agents and techniques have been developed in recent years specifically for bone research and skeletal metastases. Visualizing molecular processes in relation to bone remodeling in metastasized cancers provides valuable information for understanding disease mechanisms and monitoring expression of primary molecular targets and therapeutic efficacy. This review is intended to provide an overview of tumor-specific and non-specific contrast agents in the first near-infrared window (NIR-I) window from 650 nm to 950 nm that can be used to study functional and structural aspects of skeletal remodeling of cancer in preclinical animal models. Near-infrared (NIR) optical imaging techniques, specifically NIR spectroscopy and photoacoustic imaging, and their use in skeletal metastases will also be discussed. Perspectives on the promises and challenges facing this exciting field are then given.
Collapse
Affiliation(s)
- Nicholas Cho
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, MO 63110, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Monica Shokeen
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, MO 63110, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States.,Alvin J. Siteman Cancer Center at Washington University School of Medicine and Barnes Jewish Hospital, St. Louis, MO 63110, United States
| |
Collapse
|
5
|
Alldritt I, Whitham-Agut B, Sipin M, Studholme J, Trentacoste A, Tripp JA, Cappai MG, Ditchfield P, Devièse T, Hedges REM, McCullagh JSO. Metabolomics reveals diet-derived plant polyphenols accumulate in physiological bone. Sci Rep 2019; 9:8047. [PMID: 31142795 PMCID: PMC6541599 DOI: 10.1038/s41598-019-44390-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/16/2019] [Indexed: 11/09/2022] Open
Abstract
Plant-derived secondary metabolites consumed in the diet, especially polyphenolic compounds, are known to have a range of positive health effects. They are present in circulation after ingestion and absorption and can be sequestered into cells within particular organs, but have rarely been investigated systematically in osteological tissues. However, a small number of polyphenols and similar molecules are known to bind to bone. For example alizarin, a plant derived anthraquinone and tetracycline (a naturally occurring antibiotic), are both absorbed into bone from circulation during bone formation and are used to monitor mineralization in osteological studies. Both molecules have also been identified serendipitously in archaeological human bones derived from natural sources in the diet. Whether an analogous mechanism of sequestration extends to additional diet-derived plant-polyphenols has not previously been systematically studied. We investigated whether a range of diet-derived polyphenol-like compounds bind to bone using untargeted metabolomics applied to the analysis of bone extracts from pigs fed an acorn-based diet. We analysed the diet which was rich in ellagitannins, extracts from the pig bones and surrounding tissue, post-mortem. We found direct evidence of multiple polyphenolic compounds in these extracts and matched them to the diet. We also showed that these compounds were present in the bone but not surrounding tissues. We also provide data showing that a range of polyphenolic compounds bind to hydroxyapatite in vitro. The evidence for polyphenol sequestration into physiological bone, and the range and specificity of polyphenols in human and animal diets, raises intriguing questions about potential effects on bone formation and bone health. Further studies are needed to determine the stability of the sequestered molecules post-mortem but there is also potential for (palaeo)dietary reconstruction and forensic applications.
Collapse
Affiliation(s)
- Isabelle Alldritt
- Department of Chemistry, Mansfield Road, University of Oxford, Oxford, OX1 3TA, UK
| | | | - Miguel Sipin
- Department of Chemistry, Mansfield Road, University of Oxford, Oxford, OX1 3TA, UK
| | - Jacob Studholme
- Department of Chemistry, Mansfield Road, University of Oxford, Oxford, OX1 3TA, UK
| | | | - Jennifer A Tripp
- UCL Institute of Archaeology, Gordon Square, London, WC1H 0PY, UK
| | - Maria Grazia Cappai
- Research Unit of Animal Nutrition, Department of Veterinary Medicine, University of Sassari, Sassari, PO Box 07100, Italy
| | - Peter Ditchfield
- Research Laboratory for Archaeology and the History of Art, University of Oxford, Dyson Perrins Building, South Parks Road, Oxford, OX1 3QY, UK
| | - Thibaut Devièse
- Research Laboratory for Archaeology and the History of Art, University of Oxford, Dyson Perrins Building, South Parks Road, Oxford, OX1 3QY, UK
| | - Robert E M Hedges
- Department of Chemistry, Mansfield Road, University of Oxford, Oxford, OX1 3TA, UK
- Research Laboratory for Archaeology and the History of Art, University of Oxford, Dyson Perrins Building, South Parks Road, Oxford, OX1 3QY, UK
| | - James S O McCullagh
- Department of Chemistry, Mansfield Road, University of Oxford, Oxford, OX1 3TA, UK.
| |
Collapse
|
6
|
Özalp Ö, Toru HS, Altay MA, Sindel A. Evaluation of the Efficacy of EDTA Chelation on Alveolar Bone Healing After Ultrasonic and Conventional Surgery Under Bisphosphonate Medication: A Rat Model. J Oral Maxillofac Surg 2019; 77:1982-1989. [PMID: 31095928 DOI: 10.1016/j.joms.2019.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 11/18/2022]
Abstract
PURPOSE The aims of the present study were 1) to investigate the effects of local ethylenediaminetetraacetic acid (EDTA) chelation on alveolar bone healing after systemic administration of bisphosphonates; and 2) to compare the healing in alveolar defects created with ultrasonic and conventional surgery. MATERIALS AND METHODS A total of 40 male Wistar rats were divided into 4 equal groups of 10 rats each (zoledronate and piezosurgery, zoledronate and bur, placebo and piezosurgery, and placebo and bur). The first 2 groups received weekly intraperitoneal injections of zoledronic acid (0.06 mg/kg), and the second 2 groups received saline solution for 4 weeks. After the last injections, horizontal defects 4 × 2 × 1 mm in size were created on the mandibular alveolar bone using piezosurgery or conventional bur surgery. The zoledronate groups received 10 minutes of local application of EDTA on 1 side and saline solution on the other side. In the placebo groups, only 1 side was treated (one half of the group with the bur and one half of the group with piezosurgery). All the rats were euthanized at 4 postoperative weeks for comparative histomorphometric evaluation of bone healing in the created defects. RESULTS Bone formation was found to be the greatest in the placebo groups (P < .05). Although a greater amount of bone formation was observed with piezosurgery and EDTA among the bisphosphonate-treated groups, the difference between the zoledronate groups was not statistically significant (P > .05). Similarly, no statistically significant difference was found between the use of piezosurgery and conventional bur surgery within the placebo groups (P > .05). CONCLUSIONS The findings of the present study revealed improved bone healing with the use of piezosurgery and EDTA chelation, although the difference did not reach statistical significance. Further research should be performed to clearly identify the role of EDTA as a chelating agent and in prevention of medication-related osteonecrosis of the jaws development.
Collapse
Affiliation(s)
- Öznur Özalp
- Specialist, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Akdeniz University, Antalya, Turkey.
| | - Havva Serap Toru
- Associate Professor, Department of Pathology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Mehmet Ali Altay
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Akdeniz University, Antalya, Turkey
| | - Alper Sindel
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Akdeniz University, Antalya, Turkey
| |
Collapse
|
7
|
Lee KK, Lee JG, Park CS, Lee SH, Raja N, Yun HS, Lee JS, Lee CS. Bone-targeting carbon dots: effect of nitrogen-doping on binding affinity. RSC Adv 2019; 9:2708-2717. [PMID: 35520477 PMCID: PMC9059868 DOI: 10.1039/c8ra09729a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/01/2019] [Indexed: 11/23/2022] Open
Abstract
Novel fluorescent carbon dots (CDs) for bone imaging were fabricated via a facile hydrothermal method using alendronate in the absence of a nitrogen-doping precursor to enhance bone affinity. One-step synthesized alendronate-based CDs (Alen-CDs) had strong binding activity for calcium-deficient hydroxyapatite (CDHA, the mineral component of bones) scaffold, rat femur, and bone structures of live zebrafish. This was attributed to the bisphosphonate group present on the CD surface, even after carbonization. For comparison, the surface effects of nitrogen-doped CDs obtained using ethylenediamine (EDA), i.e., Alen-EDA-CDs, were also investigated, focusing on the targeting ability of distinct surface functional groups when compared with Alen-CDs. An in vivo study to assess the impact on bone affinity revealed that Alen-CDs effectively accumulated in the bone structures of live zebrafish larvae after microinjections, as well as in the bone tissues of femur extracted from rats. Moreover, Alen-CD-treated zebrafish larvae had superior toleration, retaining skeletal fluorescence for 7 days post-injection (dpi). The sustainable capability, surpassing that of Alizarin Red S, suggests that Alen-CDs have the potential for targeted drug delivery to damaged bone tissues and provides motivation for additional in vivo investigations. To our knowledge, this is the first in vitro, ex vivo, and in vivo demonstration of direct bone-targeted deliveries, supporting the use of fluorescent CDs in the treatment of various bone diseases such as osteoporosis, Paget's disease, and metastatic bone cancer. Fluorescent carbon dots selectively bind to skull tissues with high affinity, including a strong binding activity for calcium deficient hydroxyapatite, and rat femur, for bone targeted imaging.![]()
Collapse
Affiliation(s)
- Kyung Kwan Lee
- Hazards Monitoring BNT Research Center
- Korea Research Institute of Bioscience and Biotechnology (KRIBB)
- Daejeon 34141
- Republic of Korea
- Department of Chemical Engineering and Applied Chemistry
| | - Jae-Geun Lee
- Disease Target Structure Research Center
- Korea Research Institute of Bioscience and Biotechnology (KRIBB)
- Daejeon
- Republic of Korea
- Department of Biotechnology
| | - Chul Soon Park
- Department of Polymer Engineering
- Chonnam National University
- Gwangju 61186
- Republic of Korea
| | - Sun Hyeok Lee
- Hazards Monitoring BNT Research Center
- Korea Research Institute of Bioscience and Biotechnology (KRIBB)
- Daejeon 34141
- Republic of Korea
- Department of Biotechnology
| | - Naren Raja
- Department of Biotechnology
- University of Science & Technology (UST)
- Daejeon 34113
- Republic of Korea
- Powder and Ceramics Division
| | - Hui-suk Yun
- Department of Biotechnology
- University of Science & Technology (UST)
- Daejeon 34113
- Republic of Korea
- Powder and Ceramics Division
| | - Jeong-Soo Lee
- Disease Target Structure Research Center
- Korea Research Institute of Bioscience and Biotechnology (KRIBB)
- Daejeon
- Republic of Korea
- Department of Biotechnology
| | - Chang-Soo Lee
- Hazards Monitoring BNT Research Center
- Korea Research Institute of Bioscience and Biotechnology (KRIBB)
- Daejeon 34141
- Republic of Korea
- Department of Biotechnology
| |
Collapse
|
8
|
Elsayed R, Abraham P, Awad ME, Kurago Z, Baladhandayutham B, Whitford GM, Pashley DH, McKenna CE, Elsalanty ME. Removal of matrix-bound zoledronate prevents post-extraction osteonecrosis of the jaw by rescuing osteoclast function. Bone 2018; 110:141-149. [PMID: 29408511 PMCID: PMC5878730 DOI: 10.1016/j.bone.2018.01.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/06/2018] [Accepted: 01/25/2018] [Indexed: 12/25/2022]
Abstract
Unlike other antiresorptive medications, bisphosphonate molecules accumulate in the bone matrix. Previous studies of side-effects of anti-resorptive treatment focused mainly on systemic effects. We hypothesize that matrix-bound bisphosphonate molecules contribute to the pathogenesis of bisphosphonate-related osteonecrosis of the jaw (BRONJ). In this study, we examined the effect of matrix-bound bisphosphonates on osteoclast differentiation in vitro using TRAP staining and resorption assay, with and without pretreatment with EDTA. We also tested the effect of zoledronate chelation on the healing of post-extraction defect in rats. Our results confirmed that bisphosphonates bind to, and can be chelated from, mineralized matrix in vitro in a dose-dependent manner. Matrix-bound bisphosphonates impaired the differentiation of osteoclasts, evidenced by TRAP activity and resorption assay. Zoledronate-treated rats that underwent bilateral dental extraction with unilateral EDTA treatment showed significant improvement in mucosal healing and micro-CT analysis on the chelated sides. The results suggest that matrix-bound bisphosphonates are accessible to osteoclasts and chelating agents and contribute to the pathogenesis of BRONJ. The use of topical chelating agents is a promising strategy for the prevention of BRONJ following dental procedures in bisphosphonate-treated patients.
Collapse
Affiliation(s)
- Ranya Elsayed
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Pheba Abraham
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Mohamed E Awad
- Department of Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Zoya Kurago
- Department of Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Gary M Whitford
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - David H Pashley
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Charles E McKenna
- Department of Chemistry, Dana and David Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Mohammed E Elsalanty
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
9
|
Blair HC, Larrouture QC, Li Y, Lin H, Beer-Stoltz D, Liu L, Tuan RS, Robinson LJ, Schlesinger PH, Nelson DJ. Osteoblast Differentiation and Bone Matrix Formation In Vivo and In Vitro. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:268-280. [PMID: 27846781 DOI: 10.1089/ten.teb.2016.0454] [Citation(s) in RCA: 316] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We review the characteristics of osteoblast differentiation and bone matrix synthesis. Bone in air breathing vertebrates is a specialized tissue that developmentally replaces simpler solid tissues, usually cartilage. Bone is a living organ bounded by a layer of osteoblasts that, because of transport and compartmentalization requirements, produce bone matrix exclusively as an organized tight epithelium. With matrix growth, osteoblasts are reorganized and incorporated into the matrix as living cells, osteocytes, which communicate with each other and surface epithelium by cell processes within canaliculi in the matrix. The osteoblasts secrete the organic matrix, which are dense collagen layers that alternate parallel and orthogonal to the axis of stress loading. Into this matrix is deposited extremely dense hydroxyapatite-based mineral driven by both active and passive transport and pH control. As the matrix matures, hydroxyapatite microcrystals are organized into a sophisticated composite in the collagen layer by nucleation in the protein lattice. Recent studies on differentiating osteoblast precursors revealed a sophisticated proton export network driving mineralization, a gene expression program organized with the compartmentalization of the osteoblast epithelium that produces the mature bone matrix composite, despite varying serum calcium and phosphate. Key issues not well defined include how new osteoblasts are incorporated in the epithelial layer, replacing those incorporated in the accumulating matrix. Development of bone in vitro is the subject of numerous projects using various matrices and mesenchymal stem cell-derived preparations in bioreactors. These preparations reflect the structure of bone to variable extents, and include cells at many different stages of differentiation. Major challenges are production of bone matrix approaching the in vivo density and support for trabecular bone formation. In vitro differentiation is limited by the organization and density of osteoblasts and by endogenous and exogenous inhibitors.
Collapse
Affiliation(s)
- Harry C Blair
- 1 Veteran's Affairs Medical Center , Pittsburgh, Pennsylvania.,2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | | - Yanan Li
- 3 Department of Stomatology, Chinese PLA General Hospital , Beijing, China
| | - Hang Lin
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Donna Beer-Stoltz
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Li Liu
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Rocky S Tuan
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lisa J Robinson
- 5 Department of Pathology, West Virginia University School of Medicine , Morgantown, West Virginia.,6 Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Paul H Schlesinger
- 7 Department of Cell Biology, Washington University , Saint Louis, Missouri
| | - Deborah J Nelson
- 8 Department of Neurobiology, Pharmacology & Physiology, University of Chicago , Chicago, Illinois
| |
Collapse
|
10
|
Cole LE, Vargo-Gogola T, Roeder RK. Targeted delivery to bone and mineral deposits using bisphosphonate ligands. Adv Drug Deliv Rev 2016; 99:12-27. [PMID: 26482186 DOI: 10.1016/j.addr.2015.10.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/01/2015] [Accepted: 10/09/2015] [Indexed: 01/07/2023]
Abstract
The high concentration of mineral present in bone and pathological calcifications is unique compared with all other tissues and thus provides opportunity for targeted delivery of pharmaceutical drugs, including radiosensitizers and imaging probes. Targeted delivery enables accumulation of a high local dose of a therapeutic or imaging contrast agent to diseased bone or pathological calcifications. Bisphosphonates (BPs) are the most widely utilized bone-targeting ligand due to exhibiting high binding affinity to hydroxyapatite mineral. BPs can be conjugated to an agent that would otherwise have little or no affinity for the sites of interest. This article summarizes the current state of knowledge and practice for the use of BPs as ligands for targeted delivery to bone and mineral deposits. The clinical history of BPs is briefly summarized to emphasize the success of these molecules as therapeutics for metabolic bone diseases. Mechanisms of binding and the relative binding affinity of various BPs to bone mineral are introduced, including common methods for measuring binding affinity in vitro and in vivo. Current research is highlighted for the use of BP ligands for targeted delivery of BP conjugates in various applications, including (1) therapeutic drug delivery for metabolic bone diseases, bone cancer, other bone diseases, and engineered drug delivery platforms; (2) imaging probes for scintigraphy, fluorescence, positron emission tomography, magnetic resonance imaging, and computed tomography; and (3) radiotherapy. Last, and perhaps most importantly, key structure-function relationships are considered for the design of drugs with BP ligands, including the tether length between the BP and drug, the size of the drug, the number of BP ligands per drug, cleavable tethers between the BP and drug, and conjugation schemes.
Collapse
Affiliation(s)
- Lisa E Cole
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Tracy Vargo-Gogola
- Department of Biochemistry and Molecular Biology, Indiana University Simon Cancer Center, Indiana University School of Medicine-South Bend, South Bend, IN 46617, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ryan K Roeder
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
11
|
Howie RN, Bhattacharyya M, Salama ME, Refaey ME, Isales C, Borke J, Daoudi A, Medani F, Elsalanty ME. Removal of pamidronate from bone in rats using systemic and local chelation. Arch Oral Biol 2015; 60:1699-707. [PMID: 26431826 PMCID: PMC4636948 DOI: 10.1016/j.archoralbio.2015.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/14/2015] [Accepted: 09/03/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Bisphosphonates become adsorbed on hydroxyapatite crystals in the bone matrix. In case of side-effects, stopping the treatment would not affect the bisphosphonates already deposited in bone. This study tests the feasibility of in-vivo targeted removal of bisphosphonates from bone using chelating agents. DESIGN 32 Sprague Dawley rats were given an injection of fluorescent pamidronate (OsteoSense EX; 0.16nmol/g). They were treated with either systemic (cadmium) or local [ethylenediaminetetraacetic (EDTA) or citric acid (CA)] chelating agents to induce the removal of the bisphosphonate from bone. We evaluated the decrease in fluorescence in the alveolar bone, femur, tibia, and vertebrae. We also analyzed the systemic effects of treatment. RESULTS Systemic chelation reduced the pamidronate signal universally. However, the maximum reduction was observed in the alveolar bone and femur (22% and 21%, p values 0.008 and 0.028, respectively). Systemic chelation did not impair calcium homeostasis. The chelation effect was not due to a systemic toxic effect on the liver or kidney. On the other hand local chelation at the extraction site significantly (p=0.011) decreased the pamidronate signal at bony surfaces of the socket. CONCLUSIONS Systemic and local chelating agents can remove bisphosphonate from bone. This study establishes a new concept for the prevention of side effects of bisphosphonates during high-risk situations.
Collapse
Affiliation(s)
| | | | | | | | | | - James Borke
- Western University of Health Sciences, Pomona, CA, USA
| | - Asma Daoudi
- Georgia Regents University, Augusta, GA, USA
| | | | | |
Collapse
|
12
|
Ellmann S, Beck M, Kuwert T, Uder M, Bäuerle T. Multimodal imaging of bone metastases: From preclinical to clinical applications. J Orthop Translat 2015; 3:166-177. [PMID: 30035055 PMCID: PMC5986987 DOI: 10.1016/j.jot.2015.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/17/2015] [Accepted: 07/22/2015] [Indexed: 01/02/2023] Open
Abstract
Metastases to the skeletal system are commonly observed in cancer patients, highly affecting the patients' quality of life. Imaging plays a major role in detection, follow-up, and molecular characterisation of metastatic disease. Thus, imaging techniques have been optimised and combined in a multimodal and multiparametric manner for assessment of complementary aspects in osseous metastases. This review summarises both application of the most relevant imaging techniques for bone metastasis in preclinical models and the clinical setting.
Collapse
Affiliation(s)
- Stephan Ellmann
- Institute of Radiology, University Medical Centre Erlangen, Erlangen, Germany
| | - Michael Beck
- Institute of Nuclear Medicine, University Medical Centre Erlangen, Erlangen, Germany
| | - Torsten Kuwert
- Institute of Nuclear Medicine, University Medical Centre Erlangen, Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Medical Centre Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Medical Centre Erlangen, Erlangen, Germany
| |
Collapse
|
13
|
Extracellular matrix networks in bone remodeling. Int J Biochem Cell Biol 2015; 65:20-31. [DOI: 10.1016/j.biocel.2015.05.008] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 04/18/2015] [Accepted: 05/08/2015] [Indexed: 01/21/2023]
|
14
|
Tower RJ, Campbell GM, Müller M, Glüer CC, Tiwari S. Utilizing time-lapse micro-CT-correlated bisphosphonate binding kinetics and soft tissue-derived input functions to differentiate site-specific changes in bone metabolism in vivo. Bone 2015; 74:171-81. [PMID: 25613175 DOI: 10.1016/j.bone.2015.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 11/18/2022]
Abstract
The turnover of bone is a tightly regulated process between bone formation and resorption to ensure skeletal homeostasis. This process differs between bone types, with trabecular bone often associated with higher turnover than cortical bone. Analyses of bone by micro-computed tomography (micro-CT) reveal changes in structure and mineral content, but are limited in the study of metabolic activity at a single time point, while analyses of serum markers can reveal changes in bone metabolism, but cannot delineate the origin of any aberrant findings. To obtain a site-specific assessment of bone metabolic status, bisphosphonate binding kinetics were utilized. Using a fluorescently-labeled bisphosphonate, we show that early binding kinetics monitored in vivo using fluorescent molecular tomography (FMT) can monitor changes in bone metabolism in response to bone loss, stimulated by ovariectomy (OVX), or bone gain, resulting from treatment with the anabolic bone agent parathyroid hormone (PTH), and is capable of distinguishing different, metabolically distinct skeletal sites. Using time-lapse micro-CT, longitudinal bone turnover was quantified. The spine showed a significantly greater percent resorbing volume and surface in response to OVX, while mice treated with PTH showed significantly greater resorbing volume per bone surface in the spine and significantly greater forming surfaces in the knee. Correlation studies between binding kinetics and micro-CT suggest that forming surfaces, as assessed by time-lapse micro-CT, are preferentially reflected in the rate constant values while forming and resorbing bone volumes primarily affect plateau values. Additionally, we developed a blood pool correction method which now allows for quantitative multi-compartment analyses to be conducted using FMT. These results further expand our understanding of bisphosphonate binding and the use of bisphosphonate binding kinetics as a tool to monitor site-specific changes in bone metabolism in vivo.
Collapse
Affiliation(s)
- R J Tower
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - G M Campbell
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - M Müller
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - C C Glüer
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - S Tiwari
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Germany.
| |
Collapse
|
15
|
Perosky JE, Peterson JR, Eboda ON, Morris MD, Wang SC, Levi B, Kozloff KM. Early detection of heterotopic ossification using near-infrared optical imaging reveals dynamic turnover and progression of mineralization following Achilles tenotomy and burn injury. J Orthop Res 2014; 32:1416-23. [PMID: 25087685 PMCID: PMC4408934 DOI: 10.1002/jor.22697] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/24/2014] [Indexed: 02/04/2023]
Abstract
Heterotopic ossification (HO) is the abnormal formation of bone in soft tissue. Current diagnostics have low sensitivity or specificity to incremental progression of mineralization, especially at early time points. Without accurate and reliable early diagnosis and intervention, HO progression often results in incapacitating conditions of limited range of motion, nerve entrapment, and pain. We hypothesized that non-invasive near-infrared (NIR) optical imaging can detect HO at early time points and monitor heterotopic bone turnover longitudinally. C57BL6 mice received an Achilles tenotomy on their left hind limb in combination with a dorsal burn or sham procedure. A calcium-chelating tetracycline derivative (IRDye 680RD BoneTag) was injected bi-weekly and imaged via NIR to measure accumulative fluorescence for 11 wk and compared to in vivo microCT images. Percent retention of fluorescence was calculated longitudinally to assess temporal bone resorption. NIR detected HO as early as five days and revealed a temporal response in HO formation and turnover. MicroCT could not detect HO until 5 wk. Confocal microscopy confirmed fluorophore localization to areas of HO. These findings demonstrate the ability of a near-infrared optical imaging strategy to accurately and reliably detect and monitor HO in a murine model.
Collapse
Affiliation(s)
| | | | | | | | | | - Benjamin Levi
- Department of Plastic Surgery, University of Michigan
| | | |
Collapse
|
16
|
Satkunananthan PB, Anderson MJ, De Jesus NM, Haudenschild DR, Ripplinger CM, Christiansen BA. In vivo fluorescence reflectance imaging of protease activity in a mouse model of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2014; 22:1461-9. [PMID: 25278057 PMCID: PMC4185155 DOI: 10.1016/j.joca.2014.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/10/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Joint injuries initiate a surge of inflammatory cytokines and proteases that contribute to cartilage and subchondral bone degeneration. Detecting these early processes in animal models of post-traumatic osteoarthritis (PTOA) typically involves ex vivo analysis of blood serum or synovial fluid biomarkers, or histological analysis of the joint. In this study, we used in vivo fluorescence reflectance imaging (FRI) to quantify protease, matrix metalloproteinase (MMP), and Cathepsin K activity in mice following anterior cruciate ligament (ACL) rupture. We hypothesized that these processes would be elevated at early time points following joint injury, but would return to control levels at later time points. DESIGN Mice were injured via tibial compression overload, and FRI was performed at time points from 1 to 56 days after injury using commercially available activatable fluorescent tracers to quantify protease, MMP, and cathepsin K activity in injured vs uninjured knees. PTOA was assessed at 56 days post-injury using micro-computed tomography and whole-joint histology. RESULTS Protease activity, MMP activity, and cathepsin K activity were all significantly increased in injured knees relative to uninjured knees at all time points, peaking at 1-7 days post-injury, then decreasing at later time points while still remaining elevated relative to controls. CONCLUSIONS This study establishes FRI as a reliable method for in vivo quantification of early biological processes in a translatable mouse model of PTOA, and provides crucial information about the time course of inflammation and biological activity following joint injury. These data may inform future studies aimed at targeting these early processes to inhibit PTOA development.
Collapse
Affiliation(s)
- Patrick B. Satkunananthan
- University of California-Davis Medical Center, Department of Orthopaedic Surgery,University of California-Davis, Biomedical Engineering Graduate Group
| | - Matthew J. Anderson
- University of California-Davis Medical Center, Department of Orthopaedic Surgery
| | - Nicole M. De Jesus
- University of California-Davis, Biomedical Engineering Graduate Group,University of California-Davis Medical Center, Department of Pharmacology
| | - Dominik R. Haudenschild
- University of California-Davis Medical Center, Department of Orthopaedic Surgery,University of California-Davis, Biomedical Engineering Graduate Group
| | - Crystal M. Ripplinger
- University of California-Davis, Biomedical Engineering Graduate Group,University of California-Davis Medical Center, Department of Pharmacology
| | - Blaine A. Christiansen
- University of California-Davis Medical Center, Department of Orthopaedic Surgery,University of California-Davis, Biomedical Engineering Graduate Group
| |
Collapse
|
17
|
Hyun H, Wada H, Bao K, Gravier J, Yadav Y, Laramie M, Henary M, Frangioni JV, Choi HS. Phosphonated near-infrared fluorophores for biomedical imaging of bone. Angew Chem Int Ed Engl 2014; 53:10668-72. [PMID: 25139079 PMCID: PMC4221277 DOI: 10.1002/anie.201404930] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Indexed: 01/24/2023]
Abstract
The conventional method for creating targeted contrast agents is to conjugate separate targeting and fluorophore domains. A new strategy is based on the incorporation of targeting moieties into the non-delocalized structure of pentamethine and heptamethine indocyanines. Using the known affinity of phosphonates for bone minerals in a model system, two families of bifunctional molecules that target bone without requiring a traditional bisphosphonate are synthesized. With peak fluorescence emissions at approximately 700 or 800 nm, these molecules can be used for fluorescence-assisted resection and exploration (FLARE) dual-channel imaging. Longitudinal FLARE studies in mice demonstrate that phosphonated near-infrared fluorophores remain stable in bone for over five weeks, and histological analysis confirms their incorporation into the bone matrix. Taken together, a new strategy for creating ultra-compact, targeted near-infrared fluorophores for various bioimaging applications is described.
Collapse
Affiliation(s)
- Hoon Hyun
- Division of Hematology/Oncology, Department of Medicine and Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, SL436A, Boston, MA 02215 (USA), Fax: (+1) 617-667-0981
| | - Hideyuki Wada
- Division of Hematology/Oncology, Department of Medicine and Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, SL436A, Boston, MA 02215 (USA), Fax: (+1) 617-667-0981
| | - Kai Bao
- Division of Hematology/Oncology, Department of Medicine and Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, SL436A, Boston, MA 02215 (USA), Fax: (+1) 617-667-0981
| | - Julien Gravier
- Division of Hematology/Oncology, Department of Medicine and Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, SL436A, Boston, MA 02215 (USA), Fax: (+1) 617-667-0981
| | - Yogesh Yadav
- Department of Chemistry, Georgia State University, Atlanta, GA 30303 (USA)
| | - Matt Laramie
- Department of Chemistry, Georgia State University, Atlanta, GA 30303 (USA)
| | - Maged Henary
- Department of Chemistry, Georgia State University, Atlanta, GA 30303 (USA)
| | - John V. Frangioni
- Division of Hematology/Oncology, Department of Medicine and Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, SL436A, Boston, MA 02215 (USA), Fax: (+1) 617-667-0981. Curadel, LLC, 377 Plantation Street, Worcester, MA 01605 (USA)
| | - Hak Soo Choi
- Division of Hematology/Oncology, Department of Medicine and Department of Radiology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, SL436A, Boston, MA 02215 (USA), Fax: (+1) 617-667-0981. Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 609-735, South Korea
| |
Collapse
|
18
|
Tower RJ, Campbell GM, Müller M, Will O, Glüer CC, Tiwari S. Binding kinetics of a fluorescently labeled bisphosphonate as a tool for dynamic monitoring of bone mineral deposition in vivo. J Bone Miner Res 2014; 29:1993-2003. [PMID: 24644087 DOI: 10.1002/jbmr.2224] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 02/07/2014] [Accepted: 02/18/2014] [Indexed: 12/20/2022]
Abstract
Bone mineral deposition during the modeling of new bone and remodeling of old bone can be perturbed by several pathological conditions, including osteoporosis and skeletal metastases. A site-specific marker depicting the dynamics of bone mineral deposition would provide insight into skeletal disease location and severity, and prove useful in evaluating the efficacy of pharmacological interventions. Fluorescent labels may combine advantages of both radioisotope imaging and detailed microscopic analyses. The purpose of this study was to determine if the fluorescent bisphosphonate OsteoSense could detect localized changes in bone mineral deposition in established mouse models of accelerated bone loss (ovariectomy) (OVX) and anabolic bone gain resulting from parathyroid hormone (PTH) treatment. We hypothesized that the early rate of binding, as well as the total amount of bisphosphonate, which binds over long periods of time, could be useful in evaluating changes in bone metabolism. Evaluation of the kinetic uptake of bisphosphonates revealed a significant reduction in both the rate constant and plateau binding after OVX, whereas treatment with PTH resulted in a 36-fold increase in the bisphosphonate binding rate constant compared with untreated OVX controls. Localization of bisphosphonate binding revealed initial binding at sites of ossification adjacent to the growth plate and, to a lesser extent, along more distal trabecular and cortical elements. Micro-computed tomography (CT) was used to confirm that initial bisphosphonate binding is localized to sites of low tissue mineral density, associated with new bone mineral deposition. Our results suggest monitoring binding kinetics based on fluorescently labeled bisphosphonates represents a highly sensitive, site-specific method for monitoring changes in bone mineral deposition with the potential for translation into human applications in osteoporosis and bone metastatic processes and their treatment.
Collapse
Affiliation(s)
- Robert J Tower
- Section Biomedical Imaging, Department of Diagnostic Radiology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Hyun H, Wada H, Bao K, Gravier J, Yadav Y, Laramie M, Henary M, Frangioni JV, Choi HS. Phosphonated Near-Infrared Fluorophores for Biomedical Imaging of Bone. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201404930] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Kaiplavil S, Mandelis A, Wang X, Feng T. Photothermal tomography for the functional and structural evaluation, and early mineral loss monitoring in bones. BIOMEDICAL OPTICS EXPRESS 2014; 5:2488-2502. [PMID: 25136480 PMCID: PMC4132983 DOI: 10.1364/boe.5.002488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/15/2013] [Accepted: 01/07/2014] [Indexed: 05/31/2023]
Abstract
Salient features of a new non-ionizing bone diagnostics technique, truncated-correlation photothermal coherence tomography (TC-PCT), exhibiting optical-grade contrast and capable of resolving the trabecular network in three dimensions through the cortical region with and without a soft-tissue overlayer are presented. The absolute nature and early demineralization-detection capability of a marker called thermal wave occupation index, estimated using the proposed modality, have been established. Selective imaging of regions of a specific mineral density range has been demonstrated in a mouse femur. The method is maximum-permissible-exposure compatible. In a matrix of bone and soft-tissue a depth range of ~3.8 mm has been achieved, which can be increased through instrumental and modulation waveform optimization. Furthermore, photoacoustic microscopy, a comparable modality with TC-PCT, has been used to resolve the trabecular structure and for comparison with the photothermal tomography.
Collapse
Affiliation(s)
- Sreekumar Kaiplavil
- Center for Advanced Diffusion-Wave Technologies (CADIFT), Dept. of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, M5S 3G8, Canada
| | - Andreas Mandelis
- Center for Advanced Diffusion-Wave Technologies (CADIFT), Dept. of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, M5S 3G8, Canada
| | - Xueding Wang
- Department of Radiology, University of Michigan School of Medicine, Ann Arbor, MI 48109-5667, USA
| | - Ting Feng
- Department of Radiology, University of Michigan School of Medicine, Ann Arbor, MI 48109-5667, USA
| |
Collapse
|
21
|
Lan SM, Wu YN, Wu PC, Sun CK, Shieh DB, Lin RM. Advances in noninvasive functional imaging of bone. Acad Radiol 2014; 21:281-301. [PMID: 24439341 DOI: 10.1016/j.acra.2013.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/20/2013] [Accepted: 11/26/2013] [Indexed: 02/03/2023]
Abstract
The demand for functional imaging in clinical medicine is comprehensive. Although the gold standard for the functional imaging of human bones in clinical settings is still radionuclide-based imaging modalities, nonionizing noninvasive imaging technology in small animals has greatly advanced in recent decades, especially the diffuse optical imaging to which Britton Chance made tremendous contributions. The evolution of imaging probes, instruments, and computation has facilitated exploration in the complicated biomedical research field by allowing longitudinal observation of molecular events in live cells and animals. These research-imaging tools are being used for clinical applications in various specialties, such as oncology, neuroscience, and dermatology. The Bone, a deeply located mineralized tissue, presents a challenge for noninvasive functional imaging in humans. Using nanoparticles (NP) with multiple favorable properties as bioimaging probes has provided orthopedics an opportunity to benefit from these noninvasive bone-imaging techniques. This review highlights the historical evolution of radionuclide-based imaging, computed tomography, positron emission tomography, and magnetic resonance imaging, diffuse optics-enabled in vivo technologies, vibrational spectroscopic imaging, and a greater potential for using NPs for biomedical imaging.
Collapse
|
22
|
Harmatys KM, Cole EL, Smith BD. In vivo imaging of bone using a deep-red fluorescent molecular probe bearing multiple iminodiacetate groups. Mol Pharm 2013; 10:4263-71. [PMID: 24099089 DOI: 10.1021/mp400357v] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Deep-red fluorescent molecular probes are described that have a dendritic molecular architecture with a squaraine rotaxane core scaffold and multiple peripheral iminodiacetate groups as the bone targeting units. Iminodiacetates have an inherently lower bone affinity than bisphosphonates, and a major goal of the study was to determine how many appended iminodiacetate groups are required for effective deep-red fluorescence imaging of bone in living rodents. A series of in vitro and in vivo imaging studies showed that a tetra(iminodiacetate) probe stains bones much more strongly than an analogous bis(iminodiacetate) probe. In addition, a control tetra(iminodipropionate) probe exhibited no bone targeting ability. The tetra(iminodiacetate) probe targeted the same regions of high bone turnover as the near-infrared bisphosphonate probe OsteoSense750. Longitudinal studies showed that the fluorescence image signal from living mice treated with the tetra(iminodiacetate) probe was much more stable over 19 days than the signal from OsteoSense750. The narrow emission band of the tetra(iminodiacetate) probe makes it very attractive for inclusion in multiplex imaging protocols that employ a mixture of multiple fluorescent probes in preclinical studies of bone growth or in fluorescence guided surgery. The results also suggest that molecules or nanoparticles bearing multivalent iminodiacetate groups have promise as bone targeting agents with tunable properties for various pharmaceutical applications.
Collapse
Affiliation(s)
- Kara M Harmatys
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, 46556 Indiana, United States
| | | | | |
Collapse
|
23
|
Meganck JA, Begun DL, McElderry JD, Swick A, Kozloff KM, Goldstein SA, Morris MD, Marini JC, Caird MS. Fracture healing with alendronate treatment in the Brtl/+ mouse model of osteogenesis imperfecta. Bone 2013; 56:204-12. [PMID: 23774443 PMCID: PMC3999166 DOI: 10.1016/j.bone.2013.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 05/31/2013] [Accepted: 06/01/2013] [Indexed: 11/26/2022]
Abstract
Osteogenesis imperfecta (OI) is a heritable bone dysplasia characterized by increased skeletal fragility. Patients are often treated with bisphosphonates to attempt to reduce fracture risk. However, bisphosphonates reside in the skeleton for many years and long-term administration may impact bone material quality. Acutely, there is concern about risk of non-union of fractures that occur near the time of bisphosphonate administration. This study investigated the effect of alendronate, a potent aminobisphosphonate, on fracture healing. Using the Brtl/+ murine model of type IV OI, tibial fractures were generated in 8-week-old mice that were untreated, treated with alendronate before fracture, or treated before and after fracture. After 2, 3, or 5 weeks of healing, tibiae were assessed using microcomputed tomography (μCT), torsion testing, quantitative histomorphometry, and Raman microspectroscopy. There were no morphologic, biomechanical or histomorphometric differences in callus between untreated mice and mice that received alendronate before fracture. Alendronate treatment before fracture did not cause a significant increase in cartilage retention in fracture callus. Both Brtl/+ and WT mice that received alendronate before and after fracture had increases in the callus volume, bone volume fraction and torque at failure after 5 weeks of healing. Raman microspectroscopy results did not show any effects of alendronate in wild-type mice, but calluses from Brtl/+ mice treated with alendronate during healing had a decreased mineral-to-matrix ratio, decreased crystallinity and an increased carbonate-to-phosphate ratio. Treatment with alendronate altered the dynamics of healing by preventing callus volume decreases later in the healing process. Fracture healing in Brtl/+ untreated animals was not significantly different from animals in which alendronate was halted at the time of fracture.
Collapse
Affiliation(s)
- J A Meganck
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lambers FM, Stuker F, Weigt C, Kuhn G, Koch K, Schulte FA, Ripoll J, Rudin M, Müller R. Longitudinal in vivo imaging of bone formation and resorption using fluorescence molecular tomography. Bone 2013; 52:587-95. [PMID: 23142804 DOI: 10.1016/j.bone.2012.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Revised: 10/24/2012] [Accepted: 11/01/2012] [Indexed: 11/23/2022]
Abstract
Bone research often focuses on anatomical imaging of the bone microstructure, but in order to gain better understanding in how bone remodeling is modulated through interventions also bone formation and resorption processes should be investigated. With this in mind, the purpose of this study was to establish a longitudinal in vivo imaging approach of bone formation and resorption using fluorescence molecular tomography (FMT). In this study the reproducibility, accuracy and sensitivity of FMT for bone imaging were assessed by performing longitudinal measurements with FMT and comparing it to in vivo micro-computed tomography on a set of control mice, and mice in which load-adaptation was induced in the sixth caudal vertebra. The precision error for FMT measurements, expressed as coefficient of variation, was smaller than 16%, indicating acceptable reproducibility. A correlation was found between bone resorption measured with FMT and bone resorption rate measured with in vivo micro-computed tomography only over the first 14days (R=0.81, p<0.01), but not between bone formation measured with FMT and bone formation rate measured with in vivo micro-CT. Bone formation measured by FMT was 89-109% greater (p<0.05) for mice subjected to mechanical loading than control mice. Bone resorption was 5-8% lower, but did not reach a significant difference between groups, indicating moderate sensitivity for FMT. In conclusion, in vivo FMT in mouse tail bones is feasible but needs to be optimized for monitoring load adaptation in living mice.
Collapse
Affiliation(s)
- F M Lambers
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hjortnaes J, New SEP, Aikawa E. Visualizing novel concepts of cardiovascular calcification. Trends Cardiovasc Med 2013; 23:71-9. [PMID: 23290463 DOI: 10.1016/j.tcm.2012.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/29/2012] [Accepted: 08/31/2012] [Indexed: 12/19/2022]
Abstract
Cardiovascular calcification is currently viewed as an active disease process similar to embryonic bone formation. Cardiovascular calcification mainly affects the aortic valve and arteries and is associated with increased mortality risk. Aortic valve and arterial calcification share similar risk factors, including age, gender, diabetes, chronic renal disease, and smoking. However, the exact cellular and molecular mechanism of cardiovascular calcification is unknown. Late-stage cardiovascular calcification can be visualized with conventional imaging modalities such as echocardiography and computed tomography. However, these modalities are limited in their ability to detect the development of early calcification and the progression of calcification until advanced tissue mineralization is apparent. Due to the subsequent late diagnosis of cardiovascular calcification, treatment is usually comprised of invasive interventions such as surgery. The need to understand the process of calcification is therefore warranted and requires new imaging modalities which are able to visualize early cardiovascular calcification. This review focuses on the use of new imaging techniques to visualize novel concepts of cardiovascular calcification.
Collapse
Affiliation(s)
- Jesper Hjortnaes
- Cardiovascular Medicine, Brigham & Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB741J, Boston, MA 02115, USA
| | | | | |
Collapse
|
26
|
CT-based handling and analysis of preclinical multimodality imaging data of bone metastases. BONEKEY REPORTS 2012; 1:79. [PMID: 23951472 DOI: 10.1038/bonekey.2012.79] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 03/19/2012] [Indexed: 12/19/2022]
Abstract
The pathogenesis of bone metastases is a complex and multifaceted process. Often multiple imaging modalities are needed to follow both the structural and functional changes over time during metastatic bone disease. Researchers face extended data sets of one experiment acquired with multiple modalities at multiple points in time. This review gives an overview of an integrated approach for handling these kinds of complex data. It focuses on the analysis of whole-body micro-computerized tomography and optical data handling. We show how researchers can generate side-by-side visualizations of scans taken with one imaging modality at multiple time points and with multiple modalities at one point. Moreover, we highlight methods for normalized volumes of interest selection and quantification of bone volume and thickness.
Collapse
|
27
|
van der Horst G, van der Pluijm G. Preclinical imaging of the cellular and molecular events in the multistep process of bone metastasis. Future Oncol 2012; 8:415-30. [DOI: 10.2217/fon.12.33] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Bone metastasis is a complex process that ultimately leads to devastating metastatic bone disease. It is therefore of key interest to unravel the mechanisms underlying the multistep process of skeletal metastasis and cancer-induced bone disease, and to develop better treatment and management of patients with this devastating disease. Fortunately, novel technologies are rapidly emerging that allow real-time imaging of molecules, pathogenic processes, drug delivery and drug response in preclinical in vivo models. The outcome of these experimental studies will facilitate clinical cancer research by improving the detection of cancer cell invasion, metastasis and therapy response.
Collapse
Affiliation(s)
- Geertje van der Horst
- Department of Urology, Leiden University Medical Center, J3–100, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Gabri van der Pluijm
- Department of Urology, Leiden University Medical Center, J3–100, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
28
|
Roelofs AJ, Stewart CA, Sun S, Błażewska KM, Kashemirov BA, McKenna CE, Russell RGG, Rogers MJ, Lundy MW, Ebetino FH, Coxon FP. Influence of bone affinity on the skeletal distribution of fluorescently labeled bisphosphonates in vivo. J Bone Miner Res 2012; 27:835-47. [PMID: 22228189 DOI: 10.1002/jbmr.1543] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Bisphosphonates are widely used antiresorptive drugs that bind to calcium. It has become evident that these drugs have differing affinities for bone mineral; however, it is unclear whether such differences affect their distribution on mineral surfaces. In this study, fluorescent conjugates of risedronate, and its lower-affinity analogues deoxy-risedronate and 3-PEHPC, were used to compare the localization of compounds with differing mineral affinities in vivo. Binding to dentine in vitro confirmed differences in mineral binding between compounds, which was influenced predominantly by the characteristics of the parent compound but also by the choice of fluorescent tag. In growing rats, all compounds preferentially bound to forming endocortical as opposed to resorbing periosteal surfaces in cortical bone, 1 day after administration. At resorbing surfaces, lower-affinity compounds showed preferential binding to resorption lacunae, whereas the highest-affinity compound showed more uniform labeling. At forming surfaces, penetration into the mineralizing osteoid was found to inversely correlate with mineral affinity. These differences in distribution at resorbing and forming surfaces were not observed at quiescent surfaces. Lower-affinity compounds also showed a relatively higher degree of labeling of osteocyte lacunar walls and labeled lacunae deeper within cortical bone, indicating increased penetration of the osteocyte canalicular network. Similar differences in mineralizing surface and osteocyte network penetration between high- and low-affinity compounds were evident 7 days after administration, with fluorescent conjugates at forming surfaces buried under a new layer of bone. Fluorescent compounds were incorporated into these areas of newly formed bone, indicating that "recycling" had occurred, albeit at very low levels. Taken together, these findings indicate that the bone mineral affinity of bisphosphonates is likely to influence their distribution within the skeleton.
Collapse
Affiliation(s)
- Anke J Roelofs
- Musculoskeletal Research Programme, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lambers FM, Kuhn G, Müller R. Advances in multimodality molecular imaging of bone structure and function. BONEKEY REPORTS 2012; 1:37. [PMID: 27127622 PMCID: PMC4816287 DOI: 10.1038/bonekey.2012.28] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Accepted: 01/17/2012] [Indexed: 12/14/2022]
Abstract
The skeleton is important to the body as a source of minerals and blood cells and provides a structural framework for strength, mobility and the protection of organs. Bone diseases and disorders can have deteriorating effects on the skeleton, but the biological processes underlying anatomical changes in bone diseases occurring in vivo are not well understood, mostly due to the lack of appropriate analysis techniques. Therefore, there is ongoing research in the development of novel in vivo imaging techniques and molecular markers that might help to gain more knowledge of these pathological pathways in animal models and patients. This perspective provides an overview of the latest developments in molecular imaging applied to bone. It emphasizes that multimodality imaging, the combination of multiple imaging techniques encompassing different image modalities, enhances the interpretability of data, and is imperative for the understanding of the biological processes and the associated changes in bone structure and function relationships in vivo.
Collapse
Affiliation(s)
| | - Gisela Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Liu J, Jo JI, Kawai Y, Aoki I, Tanaka C, Yamamoto M, Tabata Y. Preparation of polymer-based multimodal imaging agent to visualize the process of bone regeneration. J Control Release 2012; 157:398-405. [DOI: 10.1016/j.jconrel.2011.09.090] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 09/21/2011] [Accepted: 09/23/2011] [Indexed: 10/17/2022]
|
31
|
van der Horst G, van der Pluijm G. Preclinical models that illuminate the bone metastasis cascade. Recent Results Cancer Res 2012; 192:1-31. [PMID: 22307368 DOI: 10.1007/978-3-642-21892-7_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this chapter currently available preclinical models of tumor progression and bone metastasis, including genetically engineered mice that develop primary and metastatic carcinomas and transplantable animal models, will be described. Understanding the multistep process of incurable bone metastasis is pivotal to the development of new therapeutic strategies. Novel technologies for imaging molecules or pathologic processes in cancers and their surrounding stroma have emerged rapidly and have greatly facilitated cancer research, in particular the cellular behavior of osteotropic tumors and their response to new and existing therapeutic agents. Optical imaging, in particular, has become an important tool in preclinical bone metastasis models, clinical trials and medical practice. Advances in experimental and clinical imaging will-in the long run-result in significant improvements in diagnosis, tumor localization, enhanced drug delivery and treatment.
Collapse
|
32
|
Nakayama H, Kawase T, Okuda K, Wolff LF, Yoshie H. In-vivo near-infrared optical imaging of growing osteosarcoma cell lesions xenografted in mice: dual-channel quantitative evaluation of volume and mineralization. Acta Radiol 2011; 52:978-88. [PMID: 21969703 DOI: 10.1258/ar.2011.110131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND In a previous study using a rodent osteosarcoma-grafted rat model, in which cell-dependent mineralization was previously demonstrated to proportionally increase with growth, we performed a quantitative analysis of mineral deposit formation using (99m)Tc-HMDP and found some weaknesses, such as longer acquisition time and narrower dynamic ranges (i.e. images easily saturated). The recently developed near-infrared (NIR) optical imaging technique is expected to non-invasively evaluate changes in living small animals in a quantitative manner. PURPOSE To test the feasibility of NIR imaging with a dual-channel system as a better alternative for bone scintigraphy by quantitatively evaluating mineralization along with the growth of osteosarcoma lesions in a mouse-xenograft model. MATERIAL AND METHODS The gross volume and mineralization of osteosarcoma lesions were evaluated in living mice simultaneously with dual-channels by NIR dye-labeled probes, 2-deoxyglucose (DG) and pamidronate (OS), respectively. To verify these quantitative data, retrieved osteosarcoma lesions were then subjected to ex-vivo imaging, weighing under wet conditions, microfocus-computed tomography (μCT) analysis, and histopathological examination. RESULTS Because of less scattering and no anatomical overlapping, as generally shown, specific fluorescence signals targeted to the osteosarcoma lesions could be determined clearly by ex-vivo imaging. These data were well positively correlated with the in-vivo imaging data (r > 0.8, P < 0.02). Other good to excellent correlations (r > 0.8, P < 0.02) were observed between DG accumulation and tumor gross volume and between OS accumulation and mineralization volume. CONCLUSION This in-vivo NIR imaging technique using DG and OS is sensitive to the level to simultaneously detect and quantitatively evaluate the growth and mineralization occuring in this type of osteosarcoma lesions of living mice without either invasion or sacrifice. By possible mutual complementation, this dual imaging system might be useful for accurate diagnosis even in the presence of overlapping tissues.
Collapse
Affiliation(s)
- Hitoshi Nakayama
- Division of Oral Bioengineering, Department of Tissue Regeneration and Reconstitution, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
- Japan Science and Technology Agency Innovation Satellite Niigata, Nagaoka, Japan
| | - Tomoyuki Kawase
- Division of Oral Bioengineering, Department of Tissue Regeneration and Reconstitution, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| | - Kazuhiro Okuda
- Division of Periodontology, Department of Oral Biological Science, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| | - Larry F Wolff
- Division of Periodontology, Department of Developmental and Surgical Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| | - Hiromasa Yoshie
- Division of Periodontology, Department of Oral Biological Science, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan
| |
Collapse
|
33
|
Wolf K, Friedl P. Extracellular matrix determinants of proteolytic and non-proteolytic cell migration. Trends Cell Biol 2011; 21:736-44. [PMID: 22036198 DOI: 10.1016/j.tcb.2011.09.006] [Citation(s) in RCA: 235] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 09/20/2011] [Accepted: 09/22/2011] [Indexed: 11/25/2022]
Abstract
Cell invasion into the 3D extracellular matrix (ECM) is a multistep biophysical process involved in inflammation, tissue repair, and metastatic cancer invasion. Migrating cells navigate through tissue structures of complex and often varying physicochemical properties, including molecular composition, porosity, alignment and stiffness, by adopting strategies that involve deformation of the cell and engagement of matrix-degrading proteases. We review how the ECM determines whether or not pericellular proteolysis is required for cell migration, ranging from protease-driven invasion and secondary tissue destruction, to non-proteolytic, non-destructive movement that solely depends on cell deformability and available tissue space. These concepts call for therapeutic targeting of proteases to prevent invasion-associated tissue destruction rather than the migration process per se.
Collapse
Affiliation(s)
- Katarina Wolf
- Department of Cell Biology, Nijmegen Center for Molecular Life Science, Radboud University Nijmegen, 6500 HB Nijmegen, The Netherlands.
| | | |
Collapse
|
34
|
Las Heras F, DaCosta RS, Pritzker KPH, Haroon N, Netchev G, Tsui HW, Chiu B, Erwin WM, Tsui FWL, Inman RD. Aberrant axial mineralization precedes spinal ankylosis: a molecular imaging study in ank/ank mice. Arthritis Res Ther 2011; 13:R163. [PMID: 21992149 PMCID: PMC3308096 DOI: 10.1186/ar3482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 05/11/2011] [Accepted: 10/12/2011] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION The diagnosis of ankylosing spondylitis is made from a combination of clinical features and the presence of radiographic evidence that may be detected only after many years of inflammatory back pain. It is not uncommon to have a diagnosis confirmed 5 to 10 years after the initial onset of symptoms. Development of a more-sensitive molecular imaging technology to detect structural changes in the joints would lead to earlier diagnosis and quantitative tracking of ankylosis progression. Progressive ankylosis (ank/ank) mice have a loss of function in the Ank gene, which codes for a regulator of PPi transport. In this study, we used these ank/ank mutant mice to assess a noninvasive, quantitative measure of joint ankylosis with near-infrared (NIR) molecular imaging in vivo. METHODS Three age groups (8, 12, and 18 weeks) of ank/ank (15 mice) and wild-type littermates (12 +/+ mice) were assessed histologically and radiographically. Before imaging, OsteoSense 750 (bisphosphonate pamidronate) was injected i.v. Whole-body images were analyzed by using the multispectral Maestro imaging system. RESULTS OsteoSense 750 signals in the paw joints were higher in ank/ank mice in all three age groups compared with controls. In the spine, significantly higher OsteoSense 750 signals were detected early, in 8-week-old ank/ank mice compared with controls, although minimal radiographic differences were noted at this time point. The molecular imaging changes in the ank/ank spine (8 weeks) were supported by histologic changes, including calcium apatite crystals at the edge of the vertebral bodies and new syndesmophyte formation. CONCLUSIONS Changes in joint pathology of ank/ank mice, as evaluated by histologic and radiographic means, are qualitative, but only semiquantitative. In contrast, molecular imaging provides a quantitative assessment. Ankylosis in ank/ank mice developed simultaneously in distal and axial joints, contrary to the previous notion that it is a centripetal process. NIR imaging might be feasible for early disease diagnosis and for monitoring disease progression in ankylosing spondylitis.
Collapse
Affiliation(s)
- Facundo Las Heras
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, Ontario, M5S 1A8, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Novel mineral contrast agent for magnetic resonance studies of bone implants grown on a chick chorioallantoic membrane. Magn Reson Imaging 2011; 29:1244-54. [PMID: 21920685 DOI: 10.1016/j.mri.2011.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 05/24/2011] [Accepted: 07/27/2011] [Indexed: 11/20/2022]
Abstract
Magnetic resonance imaging (MRI) studies of tissue engineered constructs prior to implantation clearly demonstrate the utility of the MRI technique for studying the bone formation process. To test the utility of our MRI protocols for explant studies, we present a novel test platform in which osteoblast-seeded scaffolds were implanted on the chorioallantoic membrane of a chick embryo. Scaffolds from the following experimental groups were examined by high-resolution MRI: (a) cell-seeded implanted scaffolds (CIM), (b) unseeded implanted scaffolds (UCIM), (c) cell-seeded scaffolds in static culture (CIV) and (d) unseeded scaffolds in static culture (UCIV). The reduction in water proton transverse relaxation times and the concomitant increase in water proton magnetization transfer ratios for CIM and CIV scaffolds, compared to UCIV scaffolds, were consistent with the formation of a bone-like tissue within the polymer scaffold, which was confirmed by immunohistochemistry and fluorescence microscopy. However, the presence of angiogenic vessels and fibrotic adhesions around UCIM scaffolds can confound MRI findings of bone deposition. Consequently, to improve the specificity of the MRI technique for detecting mineralized deposits within explanted tissue engineered bone constructs, we introduce a novel contrast agent that uses alendronate to target a Food and Drug Administration-approved MRI contrast agent (Gd-DOTA) to bone mineral. Our contrast agent termed GdALN was used to uniquely identify mineralized deposits in representative samples from our four experimental groups. After GdALN treatment, both CIM and CIV scaffolds, containing mineralized deposits, showed marked signal enhancement on longitudinal relaxation time-weighted (T1W) images compared to UCIV scaffolds. Relative to UCIV scaffolds, some enhancement was observed in T1W images of GdALN-treated UCIM scaffolds, subjacent to the dark adhesions at the scaffold surface, possibly from dystrophic mineral formed in the fibrotic adhesions. Notably, residual dark areas on T1W images of CIM and UCIM scaffolds were attributable to blood inside infiltrating vessels. In summary, we present the efficacy of GdALN for sensitizing the MRI technique to the deposition of mineralized deposits in explanted polymeric scaffolds.
Collapse
|
36
|
Gade TP, Motley MW, Beattie BJ, Bhakta R, Boskey AL, Koutcher JA, Mayer-Kuckuk P. Imaging of alkaline phosphatase activity in bone tissue. PLoS One 2011; 6:e22608. [PMID: 21799916 PMCID: PMC3143164 DOI: 10.1371/journal.pone.0022608] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 06/26/2011] [Indexed: 11/18/2022] Open
Abstract
The purpose of this study was to develop a paradigm for quantitative molecular imaging of bone cell activity. We hypothesized the feasibility of non-invasive imaging of the osteoblast enzyme alkaline phosphatase (ALP) using a small imaging molecule in combination with 19Flourine magnetic resonance spectroscopic imaging (19FMRSI). 6, 8-difluoro-4-methylumbelliferyl phosphate (DiFMUP), a fluorinated ALP substrate that is activatable to a fluorescent hydrolysis product was utilized as a prototype small imaging molecule. The molecular structure of DiFMUP includes two Fluorine atoms adjacent to a phosphate group allowing it and its hydrolysis product to be distinguished using 19Fluorine magnetic resonance spectroscopy (19FMRS) and 19FMRSI. ALP-mediated hydrolysis of DiFMUP was tested on osteoblastic cells and bone tissue, using serial measurements of fluorescence activity. Extracellular activation of DiFMUP on ALP-positive mouse bone precursor cells was observed. Concurringly, DiFMUP was also activated on bone derived from rat tibia. Marked inhibition of the cell and tissue activation of DiFMUP was detected after the addition of the ALP inhibitor levamisole. 19FMRS and 19FMRSI were applied for the non-invasive measurement of DiFMUP hydrolysis. 19FMRS revealed a two-peak spectrum representing DiFMUP with an associated chemical shift for the hydrolysis product. Activation of DiFMUP by ALP yielded a characteristic pharmacokinetic profile, which was quantifiable using non-localized 19FMRS and enabled the development of a pharmacokinetic model of ALP activity. Application of 19FMRSI facilitated anatomically accurate, non-invasive imaging of ALP concentration and activity in rat bone. Thus, 19FMRSI represents a promising approach for the quantitative imaging of bone cell activity during bone formation with potential for both preclinical and clinical applications.
Collapse
Affiliation(s)
- Terence P. Gade
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York, New York, United States of America
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Matthew W. Motley
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Bradley J. Beattie
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Roshni Bhakta
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York, New York, United States of America
| | - Adele L. Boskey
- Mineralized Tissue Laboratory, Hospital for Special Surgery, New York, New York, United States of America
| | - Jason A. Koutcher
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Philipp Mayer-Kuckuk
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
37
|
Vasquez KO, Casavant C, Peterson JD. Quantitative whole body biodistribution of fluorescent-labeled agents by non-invasive tomographic imaging. PLoS One 2011; 6:e20594. [PMID: 21731618 PMCID: PMC3120766 DOI: 10.1371/journal.pone.0020594] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 05/04/2011] [Indexed: 11/19/2022] Open
Abstract
When small molecules or proteins are injected into live animals, their physical and chemical properties will significantly affect pharmacokinetics, tissue penetration, and the ultimate routes of metabolism and clearance. Fluorescence molecular tomography (FMT) offers the ability to non-invasively image and quantify temporal changes in fluorescence throughout the major organ systems of living animals, in a manner analogous to traditional approaches with radiolabeled agents. This approach is best used with biotherapeutics (therapeutic antibodies, or other large proteins) or large-scaffold drug-delivery vectors, that are minimally affected by low-level fluorophore conjugation. Application to small molecule drugs should take into account the significant impact of fluorophore labeling on size and physicochemical properties, however, the presents studies show that this technique is readily applied to small molecule agents developed for far-red (FR) or near infrared (NIR) imaging. Quantification by non-invasive FMT correlated well with both fluorescence from tissue homogenates as well as with planar (2D) fluorescence reflectance imaging of excised intact organs (r² = 0.996 and 0.969, respectively). Dynamic FMT imaging (multiple times from 0 to 24 h) performed in live mice after the injection of four different FR/NIR-labeled agents, including immunoglobulin, 20-50 nm nanoparticles, a large vascular imaging agent, and a small molecule integrin antagonist, showed clear differences in the percentage of injected dose per gram of tissue (%ID/g) in liver, kidney, and bladder signal. Nanoparticles and IgG1 favored liver over kidney signal, the small molecule integrin-binding agent favored rapid kidney and bladder clearance, and the vascular agent, showed both liver and kidney clearance. Further assessment of the volume of distribution of these agents by fluorescent volume added information regarding their biodistribution and highlighted the relatively poor extravasation into tissue by IgG1. These studies demonstrate the ability of quantitative FMT imaging of FR/NIR agents to non-invasively visualize and quantify the biodistribution of different agents over time.
Collapse
Affiliation(s)
- Kristine O. Vasquez
- Department of Applied Biology, PerkinElmer, Inc., Boston, Massachusetts, United States of America
| | - Chelsea Casavant
- Department of Applied Biology, PerkinElmer, Inc., Boston, Massachusetts, United States of America
| | - Jeffrey D. Peterson
- Department of Applied Biology, PerkinElmer, Inc., Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
38
|
Snoeks TJA, Khmelinskii A, Lelieveldt BPF, Kaijzel EL, Löwik CWGM. Optical advances in skeletal imaging applied to bone metastases. Bone 2011; 48:106-14. [PMID: 20688203 DOI: 10.1016/j.bone.2010.07.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 07/28/2010] [Indexed: 12/21/2022]
Abstract
Optical Imaging has evolved into one of the standard molecular imaging modalities used in pre-clinical cancer research. Bone research however, strongly depends on other imaging modalities such as SPECT, PET, x-ray and μCT. Each imaging modality has its own specific strengths and weaknesses concerning spatial resolution, sensitivity and the possibility to quantify the signal. An increasing number of bone specific optical imaging models and probes have been developed over the past years. This review gives an overview of optical imaging modalities, models and probes that can be used to study skeletal complications of cancer in small laboratory animals.
Collapse
Affiliation(s)
- T J A Snoeks
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
39
|
Kozloff KM, Volakis LI, Marini JC, Caird MS. Near-infrared fluorescent probe traces bisphosphonate delivery and retention in vivo. J Bone Miner Res 2010; 25:1748-58. [PMID: 20200982 DOI: 10.1002/jbmr.66] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Bisphosphonate use has expanded beyond traditional applications to include treatment of a variety of low-bone-mass conditions. Complications associated with long-term bisphosphonate treatment have been noted, generating a critical need for information describing the local bisphosphonate-cell interactions responsible for these observations. This study demonstrates that a fluorescent bisphosphonate analogue, far-red fluorescent pamidronate (FRFP), is an accurate biomarker of bisphosphonate deposition and retention in vivo and can be used to monitor site-specific local drug concentration. In vitro, FRFP is competitively inhibited from the surface of homogenized rat cortical bone by traditional bisphosphonates. In vivo, FRFP delivery to the skeleton is rapid, with fluorescence linearly correlated with bone surface area. Limb fluorescence increases linearly with injected dose of FRFP; injected FRFP does not interfere with binding of standard bisphosphonates at the doses used in this study. Long-term FRFP retention studies demonstrated that FRFP fluorescence decreases in conditions of normal bone turnover, whereas fluorescence was retained in conditions of reduced bone turnover, demonstrating preservation of local FRFP concentration. In the mandible, FRFP localized to the alveolar bone and bone surrounding the periodontal ligament and molar roots, consistent with findings of osteonecrosis of the jaw. These findings support a role for FRFP as an effective in vivo marker for bisphosphonate site-specific deposition, turnover, and long-term retention in the skeleton.
Collapse
Affiliation(s)
- Kenneth M Kozloff
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | |
Collapse
|
40
|
Peterson JD, Labranche TP, Vasquez KO, Kossodo S, Melton M, Rader R, Listello JT, Abrams MA, Misko TP. Optical tomographic imaging discriminates between disease-modifying anti-rheumatic drug (DMARD) and non-DMARD efficacy in collagen antibody-induced arthritis. Arthritis Res Ther 2010; 12:R105. [PMID: 20509880 PMCID: PMC2911895 DOI: 10.1186/ar3038] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 04/29/2010] [Accepted: 05/28/2010] [Indexed: 02/08/2023] Open
Abstract
Introduction Standard measurements used to assess murine models of rheumatoid arthritis, notably paw thickness and clinical score, do not align well with certain aspects of disease severity as assessed by histopathology. We tested the hypothesis that non-invasive optical tomographic imaging of molecular biomarkers of inflammation and bone turnover would provide a superior quantitative readout and would discriminate between a disease-modifying anti-rheumatic drug (DMARD) and a non-DMARD treatment. Methods Using two protease-activated near-infrared fluorescence imaging agents to detect inflammation-associated cathepsin and matrix metalloprotease activity, and a third agent to detect bone turnover, we quantified fluorescence in paws of mice with collagen antibody-induced arthritis. Fluorescence molecular tomographic (FMT) imaging results, which provided deep tissue detection and quantitative readouts in absolute picomoles of agent fluorescence per paw, were compared with paw swelling, clinical scores, a panel of plasma biomarkers, and histopathology to discriminate between steroid (prednisolone), DMARD (p38 mitogen-activated protein kinase (MAPK) inhibitor) and non-DMARD (celecoxib, cyclooxygenase-2 (COX-2) inhibitor) treatments. Results Paw thickness, clinical score, and plasma biomarkers failed to discriminate well between a p38 MAPK inhibitor and a COX-2 inhibitor. In contrast, FMT quantification using near-infrared agents to detect protease activity or bone resorption yielded a clear discrimination between the different classes of therapeutics. FMT results agreed well with inflammation scores, and both imaging and histopathology provided clearer discrimination between treatments as compared with paw swelling, clinical score, and serum biomarker readouts. Conclusions Non-invasive optical tomographic imaging offers a unique approach to monitoring disease pathogenesis and correlates with histopathology assessment of joint inflammation and bone resorption. The specific use of optical tomography allowed accurate three-dimensional imaging, quantitation in picomoles rather than intensity or relative fluorescence, and, for the first time, showed that non-invasive imaging assessment can predict the pathologist's histology inflammation scoring and discriminate DMARD from non-DMARD activity.
Collapse
|
41
|
Baritaux JC, Hassler K, Unser M. An efficient numerical method for general L(p) regularization in fluorescence molecular tomography. IEEE TRANSACTIONS ON MEDICAL IMAGING 2010; 29:1075-1087. [PMID: 20236875 DOI: 10.1109/tmi.2010.2042814] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Reconstruction algorithms for fluorescence tomography have to address two crucial issues: 1) the ill-posedness of the reconstruction problem, 2) the large scale of numerical problems arising from imaging of 3-D samples. Our contribution is the design and implementation of a reconstruction algorithm that incorporates general Lp regularization (p ¿ 1). The originality of this work lies in the application of general Lp constraints to fluorescence tomography, combined with an efficient matrix-free strategy that enables the algorithm to deal with large reconstruction problems at reduced memory and computational costs. In the experimental part, we specialize the application of the algorithm to the case of sparsity promoting constraints (L (1)). We validate the adequacy of L (1) regularization for the investigation of phenomena that are well described by a sparse model, using data acquired during phantom experiments.
Collapse
|
42
|
Fluorescent risedronate analogues reveal bisphosphonate uptake by bone marrow monocytes and localization around osteocytes in vivo. J Bone Miner Res 2010; 25:606-16. [PMID: 20422624 PMCID: PMC3153397 DOI: 10.1359/jbmr.091009] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bisphosphonates are effective antiresorptive agents owing to their bone-targeting property and ability to inhibit osteoclasts. It remains unclear, however, whether any non-osteoclast cells are directly affected by these drugs in vivo. Two fluorescent risedronate analogues, carboxyfluorescein-labeled risedronate (FAM-RIS) and Alexa Fluor 647-labeled risedronate (AF647-RIS), were used to address this question. Twenty-four hours after injection into 3-month-old mice, fluorescent risedronate analogues were bound to bone surfaces. More detailed analysis revealed labeling of vascular channel walls within cortical bone. Furthermore, fluorescent risedronate analogues were present in osteocytic lacunae in close proximity to vascular channels and localized to the lacunae of newly embedded osteocytes close to the bone surface. Following injection into newborn rabbits, intracellular uptake of fluorescently labeled risedronate was detected in osteoclasts, and the active analogue FAM-RIS caused accumulation of unprenylated Rap1A in these cells. In addition, CD14(high) bone marrow monocytes showed relatively high levels of uptake of fluorescently labeled risedronate, which correlated with selective accumulation of unprenylated Rap1A in CD14(+) cells, as well as osteoclasts, following treatment with risedronate in vivo. Similar results were obtained when either rabbit or human bone marrow cells were treated with fluorescent risedronate analogues in vitro. These findings suggest that the capacity of different cell types to endocytose bisphosphonate is a major determinant for the degree of cellular drug uptake in vitro as well as in vivo. In conclusion, this study shows that in addition to bone-resorbing osteoclasts, bisphosphonates may exert direct effects on bone marrow monocytes in vivo.
Collapse
|
43
|
Abstract
In clinical practice the imaging of bone tissue is based almost exclusively on x-ray or radiochemical methods. Alternative methods, such as MRI and optical imaging, can provide not only anatomical, but also physiological information, due to their ability to reflect the properties of body fluids (temperature, pH and concentration of ions). In this article we review bone targeting probes for MRI and fluorescence imaging. As bone targeting is mainly associated with phosphonate and bisphosphonate derivatives, we also focus on their sorption behavior. Also discussed in detail is the limitation of using bone-targeting probes for MRI and optical imaging mainly due to their long-time retention in bone tissue and the low permeability of tissues for light.
Collapse
|
44
|
Reumann MK, Weiser MC, Mayer-Kuckuk P. Musculoskeletal molecular imaging: a comprehensive overview. Trends Biotechnol 2010; 28:93-101. [PMID: 20045210 DOI: 10.1016/j.tibtech.2009.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 10/26/2009] [Accepted: 11/12/2009] [Indexed: 01/01/2023]
Abstract
Molecular imaging permits non-invasive visualization and measurement of molecular and cell biology in living subjects, thereby complementing conventional anatomical imaging. Herein, we review the emerging application of molecular imaging for the study of musculoskeletal biology. Utilizing mainly bioluminescence and fluorescence techniques, molecular imaging has enabled in-vivo studies of (i) the activity of osteoblasts, osteoclasts, and hormones, (ii) the mechanisms of pathological cartilage and bone destruction, (iii) skeletal gene and cell therapy with and without biomaterial support, and (iv) the cellular processes in osteolysis and osteomyelitis. In these applications, musculoskeletal molecular imaging demonstrated feasibility for research in a myriad of musculoskeletal conditions ranging from bone fracture and arthritis to skeletal cancer. Importantly, these advances herald great potential for innovative clinical imaging in orthopedics, rheumatology, and oncology.
Collapse
Affiliation(s)
- Marie K Reumann
- Bone Cell Biology and Imaging Laboratory, Caspary Research Building, Rm. 623, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | | | |
Collapse
|
45
|
Leblond F, Davis SC, Valdés PA, Pogue BW. Pre-clinical whole-body fluorescence imaging: Review of instruments, methods and applications. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2009; 98:77-94. [PMID: 20031443 DOI: 10.1016/j.jphotobiol.2009.11.007] [Citation(s) in RCA: 375] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 11/16/2009] [Accepted: 11/20/2009] [Indexed: 01/07/2023]
Abstract
Fluorescence sampling of cellular function is widely used in all aspects of biology, allowing the visualization of cellular and sub-cellular biological processes with spatial resolutions in the range from nanometers up to centimeters. Imaging of fluorescence in vivo has become the most commonly used radiological tool in all pre-clinical work. In the last decade, full-body pre-clinical imaging systems have emerged with a wide range of utilities and niche application areas. The range of fluorescent probes that can be excited in the visible to near-infrared part of the electromagnetic spectrum continues to expand, with the most value for in vivo use being beyond the 630 nm wavelength, because the absorption of light sharply decreases. Whole-body in vivo fluorescence imaging has not yet reached a state of maturity that allows its routine use in the scope of large-scale pre-clinical studies. This is in part due to an incomplete understanding of what the actual fundamental capabilities and limitations of this imaging modality are. However, progress is continuously being made in research laboratories pushing the limits of the approach to consistently improve its performance in terms of spatial resolution, sensitivity and quantification. This paper reviews this imaging technology with a particular emphasis on its potential uses and limitations, the required instrumentation, and the possible imaging geometries and applications. A detailed account of the main commercially available systems is provided as well as some perspective relating to the future of the technology development. Although the vast majority of applications of in vivo small animal imaging are based on epi-illumination planar imaging, the future success of the method relies heavily on the design of novel imaging systems based on state-of-the-art optical technology used in conjunction with high spatial resolution structural modalities such as MRI, CT or ultrasound.
Collapse
Affiliation(s)
- Frederic Leblond
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA.
| | | | | | | |
Collapse
|
46
|
Kozloff KM, Quinti L, Patntirapong S, Hauschka PV, Tung CH, Weissleder R, Mahmood U. Non-invasive optical detection of cathepsin K-mediated fluorescence reveals osteoclast activity in vitro and in vivo. Bone 2009; 44:190-8. [PMID: 19007918 PMCID: PMC2656637 DOI: 10.1016/j.bone.2008.10.036] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 09/22/2008] [Accepted: 10/01/2008] [Indexed: 11/24/2022]
Abstract
Osteoclasts degrade bone matrix by demineralization followed by degradation of type I collagen through secretion of the cysteine protease, cathepsin K. Current imaging modalities are insufficient for sensitive observation of osteoclast activity, and in vivo live imaging of osteoclast resorption of bone has yet to be demonstrated. Here, we describe a near-infrared fluorescence reporter probe whose activation by cathepsin K is shown in live osteoclast cells and in mouse models of development and osteoclast upregulation. Cathepsin K probe activity was monitored in live osteoclast cultures and correlates with cathepsin K gene expression. In ovariectomized mice, cathepsin K probe upregulation precedes detection of bone loss by micro-computed tomography. These results are the first to demonstrate non-invasive visualization of bone degrading enzymes in models of accelerated bone loss, and may provide a means for early diagnosis of upregulated resorption and rapid feedback on efficacy of treatment protocols prior to significant loss of bone in the patient.
Collapse
Affiliation(s)
- Kenneth M. Kozloff
- Center for Molecular Imaging Research, Harvard Medical School, Massachusetts General Hospital; 149 13th Street, Room 5406, Charlestown MA 02129-2060
| | - Luisa Quinti
- Center for Molecular Imaging Research, Harvard Medical School, Massachusetts General Hospital; 149 13th Street, Room 5406, Charlestown MA 02129-2060
| | - Somying Patntirapong
- Children’s Hospital Boston, Department of Orthopaedic Surgery, 300 Longwood Avenue, Enders 1007, Boston MA 02215
| | - Peter V. Hauschka
- Children’s Hospital Boston, Department of Orthopaedic Surgery, 300 Longwood Avenue, Enders 1007, Boston MA 02215
| | - Ching-Hsuan Tung
- Center for Molecular Imaging Research, Harvard Medical School, Massachusetts General Hospital; 149 13th Street, Room 5406, Charlestown MA 02129-2060
| | - Ralph Weissleder
- Center for Molecular Imaging Research, Harvard Medical School, Massachusetts General Hospital; 149 13th Street, Room 5406, Charlestown MA 02129-2060
| | - Umar Mahmood
- Center for Molecular Imaging Research, Harvard Medical School, Massachusetts General Hospital; 149 13th Street, Room 5406, Charlestown MA 02129-2060
| |
Collapse
|
47
|
Allen MR. Skeletal accumulation of bisphosphonates: implications for osteoporosis treatment. Expert Opin Drug Metab Toxicol 2009; 4:1371-8. [PMID: 18950279 DOI: 10.1517/17425255.4.11.1371] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Bisphosphonates (BPs), the gold-standard pharmacological treatment for osteoporosis, are unique in that they become physically bound to the bone matrix and therefore accumulate over time. This skeletal accumulation has important physiological implications that are not completely understood. OBJECTIVE To review concepts related to the biological effects of BP accumulation in the skeleton. METHODS Articles concerning skeletal accumulation of BP treatment were identified. RESULTS/CONCLUSIONS Skeletal accumulation of BP, dictated by both chemical and biological factors, is dose-dependent, differs among skeletal sites and likely differs among the various BPs. Bisphosphonate embedded within the skeletal matrix has lasting biological effects, the results of which have both positive and negative implications for bone remodeling. As alternative anti-remodeling agents gain approval for treatment of osteoporosis, the property of skeletal accumulation will likely be unique to BPs and therefore may be the property that determines the future use of this drug class.
Collapse
Affiliation(s)
- Matthew R Allen
- Indiana University School of Medicine, Department of Anatomy & Cell Biology, 635 Barnhill Drive, MS-5035, IN 46202, Indianapolis, USA.
| |
Collapse
|
48
|
Kaijzel EL, Snoeks TJA, Buijs JT, van der Pluijm G, Löwik CWGM. Multimodal imaging and treatment of bone metastasis. Clin Exp Metastasis 2008; 26:371-9. [PMID: 18941911 DOI: 10.1007/s10585-008-9217-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 09/29/2008] [Indexed: 02/06/2023]
Abstract
The role of molecular imaging in pre-clinical research is continuously evolving. Particularly in small animal models in biomedical research, optical imaging technologies are frequently used to visualize normal as well as aberrant cellular processes at a molecular-genetic or cellular level of function. Also in cancer metastasis research, whole body bioluminescent and fluorescent imaging techniques have become indispensable tools that allow non-invasive and real-time imaging of gene expression, tumor progression and metastasis, and response to therapeutic intervention. In this paper, we discuss the use of optical imaging strategies--either alone or in combination with CT--to study intrabone tumor growth, tumor progression and to monitor efficacy of therapeutic agents in metastatic bone disease.
Collapse
Affiliation(s)
- Eric L Kaijzel
- Department of Endocrinology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
49
|
Manigrasso MB, O'Connor JP. Comparison of fracture healing among different inbred mouse strains. Calcif Tissue Int 2008; 82:465-74. [PMID: 18528610 DOI: 10.1007/s00223-008-9144-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Accepted: 05/03/2008] [Indexed: 11/29/2022]
Abstract
Quantitative trait locus analysis can be used to identify genes critically involved in biological processes. No such analysis has been applied to identifying genes that control bone fracture healing. To determine the feasibility of such an approach, healing of femur fractures was measured between C57BL/6, DBA/2, and C3H inbred strains of mice. Healing was assessed by radiography and histology and measured by histomorphometry and biomechanical testing. In all strains, radiographic bridging of the fracture was apparent after 3 weeks of healing. Histology showed that healing occurred through endochondral ossification in all strains. Histomorphometric measurements found more bone in the C57BL/6 fracture calluses 7 and 10 days after fracture. In contrast, more cartilage was present after 7 days in the C3H callus, which rapidly declined to levels less than those of C57BL/6 or DBA/2 mice by 14 days after fracture. An endochondral ossification index was calculated by multiplying the callus percent cartilage and bone areas as a measure of endochondral ossification. At 7 and 10 days after fracture, this value was higher in C57BL/6 mice. Using torsional mechanical testing, normalized structural and material properties of the C57BL/6 healing femurs were higher than values from the DBA/2 or C3H mice 4 weeks after fracture. The data indicate that fracture healing proceeds more rapidly in C57BL/6 mice and demonstrate that genetic variability significantly contributes to the process of bone regeneration. Large enough differences exist between C57BL/6 and DBA/2 or C3H mice to permit a quantitative trait locus analysis to identify genes controlling bone regeneration.
Collapse
Affiliation(s)
- Michaele B Manigrasso
- Department of Biochemistry and Molecular Biology, UMDNJ-New Jersey Medical School and Graduate School of Biomedical Sciences, 185 South Orange Avenue, Newark, NJ 07103, USA
| | | |
Collapse
|