1
|
Khan MA, Khan MA, Siddiqui S, Misra A, Yadav K, Srivastava A, Trivedi A, Husain I, Ahmad R. Phytoestrogens as potential anti-osteoporosis nutraceuticals: Major sources and mechanism(s) of action. J Steroid Biochem Mol Biol 2025; 251:106740. [PMID: 40139537 DOI: 10.1016/j.jsbmb.2025.106740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
By 2050, the global aging population is predicted to reach 1.5 billion, highlighting the need to enhance the quality of life of the elderly population. Osteoporotic fractures are projected to affect one in three women and one in five men over age 50. Initial treatments for osteoporosis in postmenopausal women include antiresorptive agents such as bisphosphonates, strontium ranelate, estrogen replacement therapy (ERT) and selective estrogen receptor modulators (SERMs). However, these do not rebuild bone, limiting their effectiveness. Denosumab, an FDA-approved antiresorptive monoclonal antibody, also has drawbacks including high costs, biannual subcutaneous injections, slow healing, impaired bone growth and side effects like eczema, flatulence, cellulitis, osteonecrosis of the jaw (ONJ) and an increased risk of spinal fractures after discontinuation of treatment. Nutraceuticals, particularly phytoestrogens, are gaining attention for their health benefits and safety in osteoporosis prevention, management and treatment. Phytoestrogens are plant metabolites similar to mammalian estrogens and include isoflavones, coumestans, lignans, stilbenes, and flavonoids. They interact with estrogen receptor isoforms ERα and ERβ, acting as agonists or antagonists based on concentration and bioavailability. Their tissue-selective activities are particularly significant: anti-estrogenic effects in reproductive tissues may lower the risk of hormone-related cancers (such as ovarian, uterine, breast and prostate), while estrogenic effects on bone could contribute to the preservation of bone mineral density.Phytoestrogens are, thus, used in managing breast and prostate cancers, cardiovascular diseases, menopause and osteoporosis. The present review focuses on the botanical origin, classification, sources and mechanism(s) of action of major phytoestrogens, their potential in prevention and management of osteoporosis and the requirement for additional clinical trials to achieve more definitive outcomes in order to confirm their efficacy and dosage safety.
Collapse
Affiliation(s)
- Mohammad Amir Khan
- Dept. of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India
| | - Mohsin Ali Khan
- Dept. of Research & Development, Era University, Lucknow, UP 226003, India
| | - Sahabjada Siddiqui
- Dept. of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India
| | - Aparna Misra
- Dept. of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India
| | - Kusum Yadav
- Dept. of Biochemistry, University of Lucknow, Lucknow, UP 226003, India
| | - Aditi Srivastava
- Dept. of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India
| | - Anchal Trivedi
- Dept. of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India
| | - Ishrat Husain
- Dept. of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India
| | - Rumana Ahmad
- Dept. of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Hardoi Road, Lucknow, UP 226003, India.
| |
Collapse
|
2
|
Abo-Elenin MHH, Kamel R, Nofal S, Ahmed AAE. The crucial role of beta-catenin in the osteoprotective effect of semaglutide in an ovariectomized rat model of osteoporosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2677-2693. [PMID: 39254876 PMCID: PMC11920005 DOI: 10.1007/s00210-024-03378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
Postmenopausal osteoporosis is a common chronic medical illness resulting from an imbalance between bone resorption and bone formation along with microarchitecture degeneration attributed to estrogen deficiency and often accompanied by other medical conditions such as weight gain, depression, and insomnia. Semaglutide (SEM) is a recently introduced GLP-1 receptor agonist (GLP-1RA) for the treatment of obesity and type 2 diabetes mellitus by mitigating insulin resistance. It has been discovered that the beneficial effects of GLP-1 are associated with alterations in lipolysis, adipogenesis, and anti-inflammatory processes. GLP-1 analogs transmit signals directly to adipose tissue. Mesenchymal stem cells (MSCs) are multidisciplinary cells that originate from bone marrow, migrate to injury sites, and promote bone regeneration. MSCs can differentiate into osteoblasts, adipose cells, and cartilage cells. Our aim is to investigate the role of semaglutide on bone formation and the Wnt signaling pathway. Osteoporosis was induced in female rats by ovariectomy, and the ovariectomized rats were treated with alendronate as standard treatment with a dose of 3 mg/kg orally and semaglutide with two doses (150 mcg/kg and 300 mcg/kg) S.C. for 10 successive weeks. Semaglutide ameliorates bone detrimental changes induced by ovariectomy. It improves bone microarchitecture and preserves bone mineral content. Semaglutide ameliorates ovariectomy-induced osteoporosis and increases the expression of β-catenin, leading to increased bone formation and halted receptor activator of nuclear factor kappa-Β ligand (RANKL's) activation. Semaglutide can be used as a potential prophylactic and therapeutic drug against osteoporosis, possibly by activating Wnt signaling and decreasing bone resorption.
Collapse
Affiliation(s)
| | - Rehab Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo City, Egypt
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo City, Egypt
| | - Amany Ali Eissa Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo City, Egypt
| |
Collapse
|
3
|
Hirata M, Tominari T, Matsumoto C, Kasuga U, Ikeda K, Miyaura C, Grundler FMW, Inada M. Polymethoxyflavones and Bone Metabolism. Nutrients 2025; 17:822. [PMID: 40077692 PMCID: PMC11901578 DOI: 10.3390/nu17050822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Phytochemicals, such as flavonoids, are bioactive compounds produced by plants, including citrus fruits, that exhibit antioxidant effects on mammalian cells and tissues. Polymethoxyflavones (PMFs) are a family of flavonoids found in the pulp and peel of citrus fruits, and have been reported to have potent antioxidant activity implicated in the prevention of human diseases. Several studies have shown that PMFs have a protective effect on bone resorption in mouse models of diseases, including osteoporosis, rheumatoid arthritis, and periodontal disease. PMFs significantly suppressed the differentiation of osteoclasts (bone resorptive cells) through indirect and direct mechanisms. The indirect effect of PMFs is the suppression of inflammatory mediator production, such as prostaglandin E2 (PGE2), and the reduction of osteoclastic inducers, such as the receptor activator of NF-κB ligand (RANKL), in osteoblasts (bone-forming cells). The direct effect of PMF suppresses osteoclast differentiation and function by inhibiting the NF-κB signaling pathway. In silico molecular docking studies indicated that PMFs target the ATP-binding pocket of IKKβ and inhibit the NF-κB signaling pathway. These findings suggest that PMFs protect against bone destruction by interfering with the NF-κB pathway in osteoblasts and osteoclasts. In this review, we summarize the latest findings regarding the effects of PMFs on various bone resorption-related diseases in mouse models.
Collapse
Affiliation(s)
- Michiko Hirata
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
| | - Tsukasa Tominari
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
| | - Chiho Matsumoto
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
| | - Urara Kasuga
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
| | - Keisuke Ikeda
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
| | - Chisato Miyaura
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
| | - Florian M. W. Grundler
- Inada Research Unit, Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan;
- Institute of Crop Science and Resource Conservation, University of Bonn, Karlrobert-Kreiten-Strasse 13, 53115 Bonn, Germany
| | - Masaki Inada
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan; (M.H.); (T.T.); (C.M.); (C.M.)
- Inada Research Unit, Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi 184-8588, Tokyo, Japan;
| |
Collapse
|
4
|
Huang H, Asghar S, Lin L, Chen S, Yuan C, Sang M, Xiao Y. Design and evaluation of a multi-responsive dual-modality bone-targeted drug delivery vehicle for the treatment of osteosarcoma. Int J Pharm 2025; 671:125191. [PMID: 39788397 DOI: 10.1016/j.ijpharm.2025.125191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The combination of chemotherapy and photothermal therapy not only improves the therapeutic effect but also limits the side effects of drugs. Herein, a multi-responsive dual-modality bone-targeted drug delivery vehicle for the treatment of osteosarcoma was designed by utilizing alendronate sodium as a bone-targeting ligand for the targeted delivery of doxorubicin (DOX) loaded polydopamine nanoparticles (PDA NPs) coated with γ-polyglutamic acid (APC@PDA/DOX NPs). The average size of spherical NPs was 140.0 nm with a zeta potential of -25.63 mV. The drug loading and encapsulation efficiency were 11.63 % and 96.44 %, respectively. The constructed NPs were responsive to acidic pH, redox conditions, and near-infrared light as the drug release rate of the system reached 70 %. Cell experiments showed that APC@PDA/DOX NPs significantly enhanced cytotoxicity in mouse K7M2 osteosarcoma cells due to PDA-induced hyperthermia and DOX-induced cytotoxicity. Compared with the free DOX solution, the area under the curve of APC@PDA/DOX NPs increased by 8.52 times, iterating the significantly prolonged circulation time of NPs in vivo that manifested in higher bioavailability. The biodistribution study showed that APC@PDA/DOX NPs enacted excellent bone targeting and tumor tissue localization. In general, APC@PDA/DOX NPs may offer a feasible and effective strategy for osteosarcoma-targeted therapy.
Collapse
Affiliation(s)
- Huilian Huang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004 China
| | - Sajid Asghar
- Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ling Lin
- School of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Su Chen
- School of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Chenjun Yuan
- School of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Muhui Sang
- Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin 214400 China.
| | - Yanyu Xiao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004 China; School of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China.
| |
Collapse
|
5
|
Wang J, Cai L, Li N, Luo Z, Ren H, Zhang B, Zhao Y. Developing mRNA Nanomedicines with Advanced Targeting Functions. NANO-MICRO LETTERS 2025; 17:155. [PMID: 39979495 PMCID: PMC11842722 DOI: 10.1007/s40820-025-01665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025]
Abstract
The emerging messenger RNA (mRNA) nanomedicines have sprung up for disease treatment. Developing targeted mRNA nanomedicines has become a thrilling research hotspot in recent years, as they can be precisely delivered to specific organs or tissues to enhance efficiency and avoid side effects. Herein, we give a comprehensive review on the latest research progress of mRNA nanomedicines with targeting functions. mRNA and its carriers are first described in detail. Then, mechanisms of passive targeting, endogenous targeting, and active targeting are outlined, with a focus on various biological barriers that mRNA may encounter during in vivo delivery. Next, emphasis is placed on summarizing mRNA-based organ-targeting strategies. Lastly, the advantages and challenges of mRNA nanomedicines in clinical translation are mentioned. This review is expected to inspire researchers in this field and drive further development of mRNA targeting technology.
Collapse
Affiliation(s)
- Ji Wang
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Lijun Cai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Ning Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Zhiqiang Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Haozhen Ren
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
| | - Yuanjin Zhao
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China.
| |
Collapse
|
6
|
Kozicka D, Krześniak M, Grymel M, Adamek J, Łasut-Szyszka B, Cichoń T, Kuźnik A. Ultrasound-assisted synthesis of new bisphosphonate-betulin conjugates and preliminary evaluation of their cytotoxic activity. RSC Adv 2025; 15:4086-4094. [PMID: 39926228 PMCID: PMC11800102 DOI: 10.1039/d4ra07782b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Bisphosphonates (BPs) are a well-established group of drugs that have been used for decades in the prevention and treatment of osteoporosis and cancer treatment-induced bone loss. Their unique properties such as high bone affinity, enzymatic stability as well as a multidirectional biological activity prompt the creation of BP conjugates. In this study, we designed and synthesized three new bisphosphonate conjugates with betulin, a natural product with a high safety profile and a broad spectrum of biological activity. The designed conjugates differed in the type of linker used and the number of bisphosphonate moieties attached (mono- or disubstituted derivatives). The proposed method for their synthesis proceeds under mild reaction conditions and gives good yields of products. In addition, as we have shown, the reaction can be assisted by ultrasound, which significantly reduced the reaction time (from 48 hours to 2 hours) and improved the overall product yield (up to 92%). The cytotoxicity of the new conjugates was evaluated against osteosarcoma (U-2 OS), lung adenocarcinoma (A549) and gastric adenocarcinoma (AGS) cell lines. The results of preliminary biological studies showed that the obtained conjugates had improved solubility compared to that of betulin and exhibited a cytotoxic effect on all three tested cell lines at the micromolar level. The betulin analog having two bisphosphonate groups 6 demonstrated the highest cytotoxic activity against tested cell lines (IC50 between 5.16 and 6.21 μM).
Collapse
Affiliation(s)
- Dominika Kozicka
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center, Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| | - Małgorzata Krześniak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch Wybrzeże Armii Krajowej 15 44-102 Gliwice Poland
| | - Mirosława Grymel
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center, Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| | - Jakub Adamek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center, Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| | - Barbara Łasut-Szyszka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch Wybrzeże Armii Krajowej 15 44-102 Gliwice Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch Wybrzeże Armii Krajowej 15 44-102 Gliwice Poland
| | - Anna Kuźnik
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center, Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| |
Collapse
|
7
|
Klawitter J, Easton M, Karpeisky A, Farrell KB, Thamm DH, Shokati T, Christians U, Zinnen SP. Novel Approaches to Monitor Pharmacokinetics and Metabolism of Gemcitabine-Ibandronate Conjugate in Mice and Dogs. Molecules 2025; 30:354. [PMID: 39860223 PMCID: PMC11767451 DOI: 10.3390/molecules30020354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The use of the bone-seeking properties of bisphosphonates (BPs) to target the delivery of therapeutic drugs is a promising approach for the treatment of bone metastases. Currently, the most advanced example of this approach is a gemcitabine-ibandronate conjugate (GEM-IB), where the bone-targeting BP ibandronate (IB) is covalently linked to the antineoplastic agent gemcitabine (GEM) via a spacer phosphate group. In the present study, we describe the development of a new analytical platform to evaluate the metabolism and pharmacokinetics of GEM-IB in mice and dogs and the results of proof-of-concept studies assessing the pharmacokinetics of GEM-IB in dogs and mice. METHODS We validated analytical platforms to analyze GEM-IB and five of its major metabolites IB, gemcitabine-5'-phosphate (GEMMP), gemcitabine (GEM), 2',2'-difluoro-2'-deoxyuridine-5'-phosphate (dFdUMP), and 2',2'-difluoro-2'-deoxyuridine (dFdU) and performed proof-of-concept pharmacokinetic studies in mice (5 mg/kg i.p.) and dogs (5 mg/kg i.v.). RESULTS Intra- and inter-run accuracy and imprecision (3 days) of the assays met the (FDA) acceptance criteria. The proof-of-concept plasma pharmacokinetic studies in mice showed AUCs of 1278, 10,652, 405, 38, 1063, 3389, and 38 h·ng/mL for GEM-IB, IB, GEMMP, dFdU-MP, GEM, and dFdU, respectively. In dog plasma, AUCs of 295, 5725, 83, 11, 1625, and 6569 h·ng/mL were observed for GEM-IB, IB, GEMMP, dFdUMP, GEM, and dFdU. CONCLUSIONS Pharmacokinetic studies in dogs and mice showed that GEM-IB is rapidly converted to IB and GEM; dFdU is formed (from GEM) with a delay. The rapid disappearance of GEM-IB from circulation could be explained by a combination of metabolism and rapid distribution to tissue/bone.
Collapse
Affiliation(s)
- Jost Klawitter
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
- Department of Psychiatry, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mckay Easton
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
| | | | - Kristen B. Farrell
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; (K.B.F.); (D.H.T.)
| | - Douglas H. Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; (K.B.F.); (D.H.T.)
| | - Touraj Shokati
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
| | - Uwe Christians
- Department of Anesthesiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (M.E.); (T.S.); (U.C.)
| | | |
Collapse
|
8
|
Lubojański A, Zakrzewski W, Samól K, Bieszczad-Czaja M, Świtała M, Wiglusz R, Watras A, Mielan B, Dobrzyński M. Application of Nanohydroxyapatite in Medicine-A Narrative Review. Molecules 2024; 29:5628. [PMID: 39683785 PMCID: PMC11643452 DOI: 10.3390/molecules29235628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
This review is an extensive collection of the latest literature describing the current knowledge about nanohydroxyapatite in a comprehensive way. These are hydroxyapatite particles with a size below 100 nm. Due to their size, the surface area to mass ratio of the particles increases. They are widely used in medicine due to their high potential in regenerative medicine, as a carrier of various substances, e.g., in targeted therapy. The aim of this article is to present the biological and physicochemical properties as well as the use of nanohydroxyapatite in modern medicine. Due to the potential of nanohydroxyapatite in medicine, further research is needed.
Collapse
Affiliation(s)
- Adam Lubojański
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland;
| | - Wojciech Zakrzewski
- Pre-Clinical Research Centre, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland (K.S.); (M.B.-C.); (M.Ś.); (B.M.)
| | - Kinga Samól
- Pre-Clinical Research Centre, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland (K.S.); (M.B.-C.); (M.Ś.); (B.M.)
| | - Martyna Bieszczad-Czaja
- Pre-Clinical Research Centre, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland (K.S.); (M.B.-C.); (M.Ś.); (B.M.)
| | - Mateusz Świtała
- Pre-Clinical Research Centre, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland (K.S.); (M.B.-C.); (M.Ś.); (B.M.)
| | - Rafał Wiglusz
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okólna 2, 50-422 Wrocław, Poland;
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland
- Meinig School of Biomedical Engineering, College of Engineering, Cornell University, Ithaca, NY 14853-1801, USA
| | - Adam Watras
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland;
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okólna 2, 50-422 Wrocław, Poland;
| | - Bartosz Mielan
- Pre-Clinical Research Centre, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland (K.S.); (M.B.-C.); (M.Ś.); (B.M.)
| | - Maciej Dobrzyński
- Department of Pediatric Dentistry and Preclinical Dentistry, Wroclaw Medical University, Krakowska 26, 50-425 Wrocław, Poland;
| |
Collapse
|
9
|
Wang X, Sato AY, Marino S, Akel N, Boysen G, Basnakian AG, Bellido TM, Li HY. Generation of BT-Amide, a Bone-Targeted Pyk2 Inhibitor, Effective via Oral Administration, for the Prevention of Glucocorticoid-Induced Bone Loss. J Med Chem 2024; 67:20708-20720. [PMID: 39540576 DOI: 10.1021/acs.jmedchem.4c02539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is the leading cause of iatrogenic osteoporosis due to the widespread clinical use of glucocorticoids (GC) as immunosuppressants. Previous research identified the proline-rich tyrosine kinase 2, Pyk2, as a critical mediator of GC-induced bone loss, and that blocking Pyk2 could protect the skeleton from adverse GC actions. However, systemic administration of current Pyk2 inhibitors causes harmful side effects, such as skin lesions. To address this, we developed bone-targeted (BT) Pyk2 inhibitors by conjugating them with bisphosphonates (BP), ensuring adherence to the bone matrix and reducing impact on noncalcified tissues. We synthesized BT-Amide by linking a derivative of TAE-226, a Pyk2 inhibitor, with alendronic acid. Oral administration (gavage) of BT-Amide prevented GC-induced bone loss in mice without causing skin lesions, or elevation of any organ toxicity markers. These findings introduce BT-Amide as the first orally effective bone-targeted Pyk2 inhibitor for preventing GC-induced bone loss while minimizing off-target effects.
Collapse
Affiliation(s)
- Xiuqi Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Amy Y Sato
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Silvia Marino
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Nisreen Akel
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Gunnar Boysen
- Department of Environmental Health Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Alexei G Basnakian
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, United States
| | - Teresita M Bellido
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, United States
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
- Department of Pharmacology, School of Medicine, The University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| |
Collapse
|
10
|
Shakori Poshteh S, Alipour S, Varamini P. Harnessing curcumin and nanotechnology for enhanced treatment of breast cancer bone metastasis. DISCOVER NANO 2024; 19:177. [PMID: 39527354 PMCID: PMC11554965 DOI: 10.1186/s11671-024-04126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Breast cancer (BC) bone metastasis poses a significant clinical challenge due to its impact on patient prognosis and quality of life. Curcumin (CUR), a natural polyphenol compound found in turmeric, has shown potential in cancer therapy due to its anti-inflammatory, antioxidant, and anticancer properties. However, its metabolic instability and hydrophobicity have hindered its clinical applications, leading to a short plasma half-life, poor absorption, and low bioavailability. To enhance the drug-like properties of CUR, nanotechnology-based delivery strategies have been employed, utilizing polymeric, lipidic, and inorganic nanoparticles (NPs). These approaches have effectively overcome CUR's inherent limitations by enhancing its stability and cellular bioavailability both in vitro and in vivo. Moreover, targeting molecules with high selectivity towards bone metastasized breast cancer cells can be used for site specific delivery of curcumin. Alendronate (ALN), a bone-seeking bisphosphonate, is one such moiety with high selectivity towards bone and thus can be effectively used for targeted delivery of curcumin loaded nanocarriers. This review will detail the process of bone metastasis in BC, elucidate the mechanism of action of CUR, and assess the efficacy of nanotechnology-based strategies for CUR delivery. Specifically, it will focus on how these strategies enhance CUR's stability and improve targeted delivery approaches in the treatment of BC bone metastasis.
Collapse
Affiliation(s)
- Shiva Shakori Poshteh
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Shohreh Alipour
- Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Drug and Food Control, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Pegah Varamini
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
11
|
Marina D, Ejersted C, Hommel K, Schwarz P. Single zoledronic acid infusion as a cause of acute kidney impairment requiring dialysis in two patients with osteoporosis. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240159. [PMID: 39529985 PMCID: PMC11554369 DOI: 10.20945/2359-4292-2024-0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/08/2024] [Indexed: 11/16/2024]
Abstract
Zoledronic acid is a widely used bisphosphonate for treating osteoporosis and hypercalcemia related to malignancy. It is also used to prevent bone loss induced by cancer treatment and bone metastases in various cancer types. Zoledronic acid is safe for most patients and is generally not associated with severe side effects. However, there have been reports of acute kidney impairment occurring after administration of intravenous zoledronic acid, mostly in patients with cancer (who received a high cumulative dose of this medication) or preexisting kidney impairment, and in patients with a history of nephrotoxic treatment. We report herein the cases of two patients without history of cancer, who developed dialysis-requiring acute kidney impairment after a single administration of intravenous zoledronic acid. None of the patients had previously used nephrotoxic medications, and one of them had a normal kidney function before zoledronic acid treatment. To the best of our knowledge, this report describes the first case of acute kidney impairment in a patient without risk factors. The findings of this report show that acute kidney impairment following intravenous zoledronic acid treatment can also occur in low-risk patients, highlighting the need for monitoring kidney function in all patients receiving this treatment.
Collapse
Affiliation(s)
- Djordje Marina
- Department of EndocrinologyCopenhagenDenmarkDepartment of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Charlotte Ejersted
- Odense University HospitalDepartment of EndocrinologyOdenseDenmarkDepartment of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Kristine Hommel
- Holbæk HospitalDepartment of NephrologyHolbækDenmarkDepartment of Nephrology, Holbæk Hospital, Holbæk, Denmark
| | - Peter Schwarz
- Department of EndocrinologyCopenhagenDenmarkDepartment of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- University of CopenhagenFaculty of Health SciencesCopenhagenDenmarkFaculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Wei PS, Thota N, John G, Chang E, Lee S, Wang Y, Ma Z, Tsai YH, Mei KC. Enhancing RNA-lipid nanoparticle delivery: Organ- and cell-specificity and barcoding strategies. J Control Release 2024; 375:366-388. [PMID: 39179112 PMCID: PMC11972657 DOI: 10.1016/j.jconrel.2024.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/25/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Recent advancements in RNA therapeutics highlight the critical need for precision gene delivery systems that target specific organs and cells. Lipid nanoparticles (LNPs) have emerged as key vectors in delivering mRNA and siRNA, offering protection against enzymatic degradation, enabling targeted delivery and cellular uptake, and facilitating RNA cargo release into the cytosol. This review discusses the development and optimization of organ- and cell-specific LNPs, focusing on their design, mechanisms of action, and therapeutic applications. We explore innovations such as DNA/RNA barcoding, which facilitates high-throughput screening and precise adjustments in formulations. We address major challenges, including improving endosomal escape, minimizing off-target effects, and enhancing delivery efficiencies. Notable clinical trials and recent FDA approvals illustrate the practical applications and future potential of LNP-based RNA therapies. Our findings suggest that while considerable progress has been made, continued research is essential to resolve existing limitations and bridge the gap between preclinical and clinical evaluation of the safety and efficacy of RNA therapeutics. This review highlights the dynamic progress in LNP research. It outlines a roadmap for future advancements in RNA-based precision medicine.
Collapse
Affiliation(s)
- Pu-Sheng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Nagasri Thota
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Greshma John
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Evelyn Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Sunjae Lee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Yuanjun Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Zitao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Yu-Hsuan Tsai
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Kuo-Ching Mei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA.
| |
Collapse
|
13
|
Egorov M, Goujon JY, Sicard M, Moal C, Pairel S, Le Bot R. Design, Synthesis, and Characterization of HBP-Vectorized Methotrexate Prodrug Molecule 1102-39: Evaluation of In Vitro Cytotoxicity Activity in Cell Culture Models, Preliminary In Vivo Safety and Efficacy Results in Rodents. ACS OMEGA 2024; 9:42433-42447. [PMID: 39431075 PMCID: PMC11483398 DOI: 10.1021/acsomega.4c06029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/22/2024]
Abstract
A novel bone-targeted prodrug, 1102-39, is discussed with the aim of enhancing the therapeutic effects of methotrexate (MTX) within bone tissues while minimizing systemic toxicity. Within the 1102-39 molecule, the central linker part forms a cleavable ester group, with MTX being also linked by a stable imine bond to the specially designed hydroxybisphosphonic (HBP) vector. Synthesized through a convergent approach starting from MTX, this prodrug advantageously modulates MTX's activity by selective esterification of its α-carboxyl group. In vitro tests revealed a 10-fold reduction in cytotoxicity compared to standard MTX, in alignment with prodrug behavior and correlated with gradual MTX release. In vivo in rodents, 1102-39 displayed preliminary encouraging antitumor effects on orthotopic osteosarcoma. Furthermore, various aspects of designing molecules for selective therapy in bone tissue based on bisphosphonate molecules as vectors for delivering active compounds to the bone are discussed. The 1102-39 molecule exhibits strong affinity for hydroxyapatite and a progressive release of MTX in aqueous environments, enhancing the safety and efficacy of bone-specific treatments and enabling sustained activity within bone and bone joints in the therapy of tumor and inflammation.
Collapse
Affiliation(s)
- Maxim Egorov
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| | | | - Marie Sicard
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| | | | - Samuel Pairel
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| | - Ronan Le Bot
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| |
Collapse
|
14
|
Wu Y, Sun B, Tang Y, Shen A, Lin Y, Zhao X, Li J, Monteiro MJ, Gu W. Bone targeted nano-drug and nano-delivery. Bone Res 2024; 12:51. [PMID: 39231955 PMCID: PMC11375042 DOI: 10.1038/s41413-024-00356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 09/06/2024] Open
Abstract
There are currently no targeted delivery systems to satisfactorily treat bone-related disorders. Many clinical drugs consisting of small organic molecules have a short circulation half-life and do not effectively reach the diseased tissue site. This coupled with repeatedly high dose usage that leads to severe side effects. With the advance in nanotechnology, drugs contained within a nano-delivery device or drugs aggregated into nanoparticles (nano-drugs) have shown promises in targeted drug delivery. The ability to design nanoparticles to target bone has attracted many researchers to develop new systems for treating bone related diseases and even repurposing current drug therapies. In this review, we shall summarise the latest progress in this area and present a perspective for future development in the field. We will focus on calcium-based nanoparticle systems that modulate calcium metabolism and consequently, the bone microenvironment to inhibit disease progression (including cancer). We shall also review the bone affinity drug family, bisphosphonates, as both a nano-drug and nano-delivery system for bone targeted therapy. The ability to target and release the drug in a controlled manner at the disease site represents a promising safe therapy to treat bone diseases in the future.
Collapse
Affiliation(s)
- Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Ying Tang
- Science and Technology Innovation Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aining Shen
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Yanlin Lin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Xiaohui Zhao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jingui Li
- School of Veterinary Medicine, Jiangsu Co-innovation Centre for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
15
|
Anish RJ, Nair A. Osteoporosis management-current and future perspectives - A systemic review. J Orthop 2024; 53:101-113. [PMID: 38495575 PMCID: PMC10940894 DOI: 10.1016/j.jor.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Osteoporosis is a geriatric metabolic ailment distinguished by low bone mineral density (BMD) and strength with enhanced micro-architectural retrogression of the extracellular matrix, further increasing bone fragility risk. Osteoporotic fractures and associated complications become common in women and men after 55 and 65 years, respectively. The loss in BMD markedly enhances the risk of fracture, non-skeletal injury, and subsequent pain, adversely affecting the quality of life. Methods Data summarised in this review were sourced and summarised, including contributions from 2008 to 2023, online from scientific search engines, based on scientific inclusion and exclusion criteria. Results Biochemical serum markers such as BALP, collagen, osteocalcin, and cathepsin-K levels can reveal the osteoporotic status. DEXA scan techniques evaluate the whole body's BMD and bone mineral content (BMC), crucial in osteoporosis management. Anabolic and anti-osteoporotic agents are commonly used to enhance bone formation, minimize bone resorption, and regulate remodelling. The challenges and side effects of drug therapy can be overcome by combining the various drug moieties. Conclusion The current review discusses the management protocol for osteoporosis, ranging from lifestyle modification, including physical exercise, pharmaceutical approaches, drug delivery applications, and advanced therapeutic possibilities of AI and machine learning techniques to reduce osteoporosis complications and fracture risk.
Collapse
Affiliation(s)
- Rajamohanan Jalaja Anish
- Department of Biochemistry, University of Kerala, Kariyavattom Campus, Trivandrum, 695581, India
| | - Aswathy Nair
- Department of Biochemistry, University of Kerala, Kariyavattom Campus, Trivandrum, 695581, India
| |
Collapse
|
16
|
Cheng Z, Li J, Chen Z, Ren W. Role of bisphosphonates in osteoporosis caused by adult growth hormone deficiency. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:839-847. [PMID: 39311779 PMCID: PMC11420967 DOI: 10.11817/j.issn.1672-7347.2024.230469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
In recent years, growth hormone and insulin-like growth factors have become key regulators of bone metabolism and remodeling, crucial for maintaining healthy bone mass throughout life. Studies have shown that adult growth hormone deficiency leads to alterations in bone remodeling, significantly affecting bone microarchitecture and increasing fracture risk. Although recombinant human growth hormone replacement therapy can mitigate these adverse effects, improving bone density, and reduce fracture risk, its effectiveness in treating osteoporosis, especially in adults with established growth hormone deficiency, seems limited. Bisphosphonates inhibit bone resorption by targeting farnesyl pyrophosphate synthase in osteoclasts, and clinical trials have confirmed their efficacy in improving osteoporosis. Therefore, for adult growth hormone deficiency patients with osteoporosis, the use of bisphosphonates alongside growth hormone replacement therapy is recommended.
Collapse
Affiliation(s)
- Zhiling Cheng
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011.
| | - Jie Li
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011
| | - Zhongpei Chen
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011
| | - Wei Ren
- Department of Endocrinology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
17
|
Abdulrehman T, Qadri S, Haik Y, Sultan A, Skariah S, Kumar S, Mendoza Z, Yadav KK, Titus A, Khader S. Advances in the targeted theragnostics of osteomyelitis caused by Staphylococcus aureus. Arch Microbiol 2024; 206:288. [PMID: 38834761 DOI: 10.1007/s00203-024-04015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
Bone infections caused by Staphylococcus aureus may lead to an inflammatory condition called osteomyelitis, which results in progressive bone loss. Biofilm formation, intracellular survival, and the ability of S. aureus to evade the immune response result in recurrent and persistent infections that present significant challenges in treating osteomyelitis. Moreover, people with diabetes are prone to osteomyelitis due to their compromised immune system, and in life-threatening cases, this may lead to amputation of the affected limbs. In most cases, bone infections are localized; thus, early detection and targeted therapy may prove fruitful in treating S. aureus-related bone infections and preventing the spread of the infection. Specific S. aureus components or overexpressed tissue biomarkers in bone infections could be targeted to deliver active therapeutics, thereby reducing drug dosage and systemic toxicity. Compounds like peptides and antibodies can specifically bind to S. aureus or overexpressed disease markers and combining these with therapeutics or imaging agents can facilitate targeted delivery to the site of infection. The effectiveness of photodynamic therapy and hyperthermia therapy can be increased by the addition of targeting molecules to these therapies enabling site-specific therapy delivery. Strategies like host-directed therapy focus on modulating the host immune mechanisms or signaling pathways utilized by S. aureus for therapeutic efficacy. Targeted therapeutic strategies in conjunction with standard surgical care could be potential treatment strategies for S. aureus-associated osteomyelitis to overcome antibiotic resistance and disease recurrence. This review paper presents information about the targeting strategies and agents for the therapy and diagnostic imaging of S. aureus bone infections.
Collapse
Affiliation(s)
- Tahir Abdulrehman
- eHealth Program, DeGroote School of Business, McMaster University, Hamilton, ON, Canada
- Health Policy, Management and Informatics, Allied Health, Credit Valley Hospital, Mississauga, ON, Canada
| | - Shahnaz Qadri
- School of Pharmacy, Texas A&M University, Kingsville, USA.
| | - Yousef Haik
- Department of Mechanical & Nuclear Engineering, University of Sharjah, Sharjah, UAE.
| | - Ali Sultan
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sini Skariah
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Shourya Kumar
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Zachary Mendoza
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Kamlesh K Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Anoop Titus
- Department of Preventive Cardiology, Houston Methodist, Houston, TX, USA
| | - Shameer Khader
- School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
18
|
Rizzi F, Panniello A, Comparelli R, Arduino I, Fanizza E, Iacobazzi RM, Perrone MG, Striccoli M, Curri ML, Scilimati A, Denora N, Depalo N. Luminescent Alendronic Acid-Conjugated Micellar Nanostructures for Potential Application in the Bone-Targeted Delivery of Cholecalciferol. Molecules 2024; 29:2367. [PMID: 38792228 PMCID: PMC11123821 DOI: 10.3390/molecules29102367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Vitamin D, an essential micronutrient crucial for skeletal integrity and various non-skeletal physiological functions, exhibits limited bioavailability and stability in vivo. This study is focused on the development of polyethylene glycol (PEG)-grafted phospholipid micellar nanostructures co-encapsulating vitamin D3 and conjugated with alendronic acid, aimed at active bone targeting. Furthermore, these nanostructures are rendered optically traceable in the UV-visible region of the electromagnetic spectrum via the simultaneous encapsulation of vitamin D3 with carbon dots, a newly emerging class of fluorescents, biocompatible nanoparticles characterized by their resistance to photobleaching and environmental friendliness, which hold promise for future in vitro bioimaging studies. A systematic investigation is conducted to optimize experimental parameters for the preparation of micellar nanostructures with an average hydrodynamic diameter below 200 nm, ensuring colloidal stability in physiological media while preserving the optical luminescent properties of the encapsulated carbon dots. Comprehensive chemical-physical characterization of these micellar nanostructures is performed employing optical and morphological techniques. Furthermore, their binding affinity for the principal inorganic constituent of bone tissue is assessed through a binding assay with hydroxyapatite nanoparticles, indicating significant potential for active bone-targeting. These formulated nanostructures hold promise for novel therapeutic interventions to address skeletal-related complications in cancer affected patients in the future.
Collapse
Affiliation(s)
- Federica Rizzi
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| | - Annamaria Panniello
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
| | - Roberto Comparelli
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| | - Ilaria Arduino
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (I.A.); (R.M.I.)
| | - Elisabetta Fanizza
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
- Department of Chemistry, University of Bari, 70125 Bari, Italy
| | - Rosa Maria Iacobazzi
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (I.A.); (R.M.I.)
| | - Maria Grazia Perrone
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (M.G.P.); (A.S.)
| | - Marinella Striccoli
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
| | - Maria Lucia Curri
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
- Department of Chemistry, University of Bari, 70125 Bari, Italy
| | - Antonio Scilimati
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (M.G.P.); (A.S.)
| | - Nunzio Denora
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (I.A.); (R.M.I.)
| | - Nicoletta Depalo
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| |
Collapse
|
19
|
Hu K, Zhong L, Lin W, Zhao G, Pu W, Feng Z, Zhou M, Ding J, Zhang J. Pathogenesis-Guided Rational Engineering of Nanotherapies for the Targeted Treatment of Abdominal Aortic Aneurysm by Inhibiting Neutrophilic Inflammation. ACS NANO 2024; 18:6650-6672. [PMID: 38369729 DOI: 10.1021/acsnano.4c00120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Abdominal aortic aneurysm (AAA) remains a fatal disease in the elderly. Currently, no drugs can be clinically used for AAA therapy. Considering the pivotal role of neutrophils in the pathogenesis of AAA, herein we propose the targeted therapy of AAA by site-specifically regulating neutrophilic inflammation. Based on a luminol-conjugated α-cyclodextrin material (LaCD), intrinsically anti-inflammatory nanoparticles (NPs) were engineered by simple nanoprecipitation, which were examined as a nanotherapy (defined as LaCD NP). After efficient accumulation in the aneurysmal aorta and localization in pathologically relevant inflammatory cells in rats with CaCl2-induced AAA, LaCD NP significantly alleviated AAA progression, as implicated by the decreased aortic expansion, suppressed elastin degradation, inhibited calcification, and improved structural integrity of the abdominal aorta. By functionalizing LaCD NP with alendronate, a calcification-targeting moiety, the in vivo aneurysmal targeting capability of LaCD NP was considerably enhanced, thereby affording significantly potentiated therapeutic outcomes in AAA rats. Mechanistically, LaCD NP can effectively inhibit neutrophil-mediated inflammatory responses in the aneurysmal aorta. Particularly, LaCD NP potently attenuated the formation of neutrophil extracellular traps (NETs), thereby suppressing NETs-mediated pro-inflammatory events and NETosis-associated negative effects responsible for AAA progression. Consequently, we demonstrated the effectiveness and underlying mechanisms of anti-NETosis nanotherapies for the targeted treatment of AAA. Our findings provide promising insights into discovering precision therapies for AAA and other inflammatory vascular diseases.
Collapse
Affiliation(s)
- Kaiyao Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Ling Zhong
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Wenjie Lin
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Guanli Zhao
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Zhiqiang Feng
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Min Zhou
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Jun Ding
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, People's Republic of China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing 400039, People's Republic of China
| |
Collapse
|
20
|
Shi BY, Sriram V, Wu SY, Huang D, Cheney A, Metzger MF, Sundberg O, Lyons KM, McKenna CE, Nishimura I, Kremen TJ. Novel bisphosphonate-based cathepsin K-triggered compound targets the enthesis without impairing soft tissue-to-bone healing. Front Bioeng Biotechnol 2024; 12:1308161. [PMID: 38433822 PMCID: PMC10905384 DOI: 10.3389/fbioe.2024.1308161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024] Open
Abstract
Background: Osteoadsorptive fluorogenic sentinel 3 (OFS-3) is a recently described compound that contains a bone-targeting bisphosphonate (BP) and cathepsin K (Ctsk)-triggered fluorescence signal. A prior study in a murine Achilles repair model demonstrated its effectiveness at targeting the site of tendon-to-bone repair, but the intrinsic effect of this novel bisphosphonate chaperone on tendon-to-bone healing has not been previously explored. We hypothesized that application of this bisphosphonate-fluorophore cargo conjugate would not affect the biomechanical properties or histologic appearance of tendon-bone repairs. Materials and Methods: Right hindlimb Achilles tendon-to-bone repair was performed on 12-week old male mice. Animals were divided into 2 groups of 18 each: 1) Achilles repair with OFS-3 applied directly to the repair site prior to closure, and 2) Achilles repair with saline applied prior to closure. Repaired hindlimbs from 12 animals per group were harvested at 6 weeks for biomechanical analysis with a custom 3D-printed jig. At 4 and 6 weeks, repaired hindlimbs from the remaining animals were assessed histologically using H&E, immunohistochemistry (IHC) staining for the presence of Ctsk, and second harmonic generation (SHG) imaging to evaluate collagen fibers. Results: At 6 weeks, there was no significant difference in failure load, stiffness, toughness, or displacement to failure between repaired hindlimbs that received OFS-3 versus saline. There was no difference in tissue healing on H&E or Ctsk staining on immunohistochemistry between animals that received OFS-3 versus saline. Finally, second harmonic generation imaging demonstrated no difference in collagen fiber parameters between the two groups. Conclusion: OFS-3 did not significantly affect the biomechanical properties or histologic appearance of murine Achilles tendon-to-bone repairs. This study demonstrates that OFS-3 can target the site of tendon-to-bone repair without causing intrinsic negative effects on healing. Further development of this drug delivery platform to target growth factors to the site of tendon-bone repair is warranted.
Collapse
Affiliation(s)
- Brendan Y. Shi
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - Varun Sriram
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - Shannon Y. Wu
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - Dave Huang
- Department of Orthopaedic Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Alexis Cheney
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - Melodie F. Metzger
- Department of Orthopaedic Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Oskar Sundberg
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Karen M. Lyons
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Charles E. McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Ichiro Nishimura
- Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California at Los Angeles, Los Angeles, CA, United States
| | - Thomas J. Kremen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
21
|
Abukhder M, Nasri Elmi S, Van Der Lith S, Hawesa N, Abukhder D, Abid H, Liu L. Bisphosphonate therapy in the management of diffuse sclerosing osteomyelitis of the mandible: a systematic review and narrative synthesis. Ann Med Surg (Lond) 2024; 86:950-957. [PMID: 38333319 PMCID: PMC10849439 DOI: 10.1097/ms9.0000000000001561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/18/2023] [Indexed: 02/10/2024] Open
Abstract
Background Diffuse sclerosing osteomyelitis (DSO) affecting the mandible is an uncommon condition characterised by recurrent pain and functional disturbances. Traditional treatments involving antibiotics, steroids, and analgesics have generally yielded unsatisfactory results. Numerous articles have proposed the utilisation of bisphosphonate therapy as an alternative approach to achieve sustained symptom relief. This study aims to consolidate the available evidence on the effectiveness of bisphosphonate therapy in managing DSO. Methods A systematic review protocol was registered with PROSPERO and reported in accordance with the Preferred Reporting for Items for Systematic Reviews and Meta-Analyses. Comprehensive electronic search strategies were devised, and studies were screened based on predefined inclusion and exclusion criteria. Results Ten articles met the eligibility criteria, encompassing a total of 135 patients diagnosed with DSO who received bisphosphonate treatment. All included studies consistently reported a reduction in pain levels and swelling, along with a decrease in the cumulative use of analgesics. The majority of patients reported long-lasting symptom improvement with bisphosphonate therapy. Notably, four studies documented improvements in maximal mouth opening, with one study reporting a mean increase of 9.6mm. Furthermore, six studies observed improvements in panoramic radiographs and cone beam computed tomography scans, with one publication describing two patients exhibiting near-normal bone architecture. Importantly, all studies reported the absence of long-term complications. Conclusions Bisphosphonate therapy emerges as a promising treatment modality for DSO, exhibiting efficacy in symptom alleviation and radiological enhancement while conferring lasting benefits. Nevertheless, further prospective studies are warranted to refine treatment protocols and substantiate these findings.
Collapse
Affiliation(s)
| | | | | | | | | | - Husnain Abid
- Sandwell General Hospital, Lyndon, West Bromwich, West Midlands, B71 4HJ, UK
| | - Linda Liu
- Northwick Park Hospita, Harrow HA1 3UJ
| |
Collapse
|
22
|
Ren Y, Weeks J, Xue T, Rainbolt J, de Mesy Bentley KL, Shu Y, Liu Y, Masters E, Cherian P, McKenna CE, Neighbors J, Ebetino FH, Schwarz EM, Sun S, Xie C. Evidence of bisphosphonate-conjugated sitafloxacin eradication of established methicillin-resistant S. aureus infection with osseointegration in murine models of implant-associated osteomyelitis. Bone Res 2023; 11:51. [PMID: 37848449 PMCID: PMC10582111 DOI: 10.1038/s41413-023-00287-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/05/2023] [Accepted: 08/21/2023] [Indexed: 10/19/2023] Open
Abstract
Eradication of MRSA osteomyelitis requires elimination of distinct biofilms. To overcome this, we developed bisphosphonate-conjugated sitafloxacin (BCS, BV600072) and hydroxybisphosphonate-conjugate sitafloxacin (HBCS, BV63072), which achieve "target-and-release" drug delivery proximal to the bone infection and have prophylactic efficacy against MRSA static biofilm in vitro and in vivo. Here we evaluated their therapeutic efficacy in a murine 1-stage exchange femoral plate model with bioluminescent MRSA (USA300LAC::lux). Osteomyelitis was confirmed by CFU on the explants and longitudinal bioluminescent imaging (BLI) after debridement and implant exchange surgery on day 7, and mice were randomized into seven groups: 1) Baseline (harvested at day 7, no treatment); 2) HPBP (bisphosphonate control for BCS) + vancomycin; 3) HPHBP (hydroxybisphosphonate control for HBCS) + vancomycin; 4) vancomycin; 5) sitafloxacin; 6) BCS + vancomycin; and 7) HBCS + vancomycin. BLI confirmed infection persisted in all groups except for mice treated with BCS or HBCS + vancomycin. Radiology revealed catastrophic femur fractures in all groups except mice treated with BCS or HBCS + vancomycin, which also displayed decreases in peri-implant bone loss, osteoclast numbers, and biofilm. To confirm this, we assessed the efficacy of vancomycin, sitafloxacin, and HBCS monotherapy in a transtibial implant model. The results showed complete lack of vancomycin efficacy while all mice treated with HBCS had evidence of infection control, and some had evidence of osseous integrated septic implants, suggestive of biofilm eradication. Taken together these studies demonstrate that HBCS adjuvant with standard of care debridement and vancomycin therapy has the potential to eradicate MRSA osteomyelitis.
Collapse
Affiliation(s)
- Youliang Ren
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jason Weeks
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Thomas Xue
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Joshua Rainbolt
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Pathology and Center for Advanced Research Technologies, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ye Shu
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yuting Liu
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Elysia Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | | | - Charles E McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - Jeffrey Neighbors
- Department of Pharmacology, Pennsylvania State University, Hershey, PA, 17033, USA
| | - Frank H Ebetino
- BioVinc, LLC, Pasadena, CA, 91107, USA
- Department of Chemistry, University of Rochester, Rochester, NY, 14642, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | | | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
23
|
Lima WJDM, Pontes JCXD, Figueiredo LSD, Araújo RDS, Paiva Sousa MCD, Aquino JDS, Castro RDD, Alves AF. Obesity influences the development of bisphosphonate-induced osteonecrosis in Wistar rats. J Appl Oral Sci 2023; 31:e20230133. [PMID: 37792808 PMCID: PMC10547385 DOI: 10.1590/1678-7757-2023-0133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/05/2023] [Accepted: 08/03/2023] [Indexed: 10/06/2023] Open
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is characterized by bone exposure for more than eight weeks in patients who have used or been treated with antiresorptive or antiangiogenic drugs, without a history of radiation therapy or metastatic diseases in the jaws. Obesity is associated with changes in periodontal tissues and oral microbiota that are linked to bone alterations. This study aimed to analyze the influence of obesity on the development of bisphosphonate-induced osteonecrosis. The experiment randomly and simply divided 24 male Wistar rats (Rattus norvegicus) into four groups: healthy, with osteonecrosis, obese, and obese with osteonecrosis (n=6 per group). Osteonecrosis was induced through weekly intraperitoneal injection for eight weeks at a dose of 250 µg/kg of zoledronic acid in a 4 mg/5 mL solution, combined with trauma (exodontia). Obesity was induced through a high glycaemic index diet. Each group was qualitatively and quantitatively evaluated regarding the development of models and pathological anatomy of the lesions. The results were expressed in mean percentage and standard deviation and statistically analyzed using one-way analysis of variance (ANOVA) followed by Tukey's post-hoc test, with a significance level of 5% (p<0.05) to establish differences found between the groups. Animals in the osteonecrosis group and the obese with osteonecrosis group presented larger necrosis areas (averages: 172.83±18,19 µm2 and 290.33±15,77 µm2, respectively) (p<0,0001). Bone sequestration, hepatic steatosis, and increased adipocyte size were observed in the obese group (average: 97.75±1.91 µm2) and in the obese with osteonecrosis group (average: 98.41±1.56 µm2), indicating greater tissue damage in these groups (p<0,0001). All parameters analyzed (through histological, morphometric, and murinometric analyses) increased for the obese and obese with osteonecrosis groups, suggesting a possible influence of obesity on the results. However, further studies are needed to confirm the role of obesity in the possible exacerbation of osteonecrosis and understand the underlying mechanisms.
Collapse
Affiliation(s)
- Wilson José de Miranda Lima
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, João Pessoa, Paraíba, Brasil
| | - Jannerson Cesar Xavier de Pontes
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, João Pessoa, Paraíba, Brasil
| | - Ludmila Silva de Figueiredo
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, João Pessoa, Paraíba, Brasil
| | - Rubens da Silva Araújo
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, João Pessoa, Paraíba, Brasil
| | | | - Jailane de Souza Aquino
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Ciências da Nutrição,João Pessoa, Paraíba, Brasil
| | - Ricardo Dias de Castro
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, João Pessoa, Paraíba, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Clínica e Odontologia Social, João Pessoa, Paraíba, Brasil
| | - Adriano Francisco Alves
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, João Pessoa, Paraíba, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Fisiologia e Patologia, João Pessoa, Paraíba, Brasil
| |
Collapse
|
24
|
Ha P, Kwak JH, Zhang Y, Shi J, Tran L, Liu TP, Pan HC, Lee S, Kim JK, Chen E, Shirazi-Fard Y, Stodieck LS, Lin A, Zheng Z, Dong SN, Zhang X, Wu BM, Ting K, Soo C. Bisphosphonate conjugation enhances the bone-specificity of NELL-1-based systemic therapy for spaceflight-induced bone loss in mice. NPJ Microgravity 2023; 9:75. [PMID: 37723136 PMCID: PMC10507033 DOI: 10.1038/s41526-023-00319-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Microgravity-induced bone loss results in a 1% bone mineral density loss monthly and can be a mission critical factor in long-duration spaceflight. Biomolecular therapies with dual osteogenic and anti-resorptive functions are promising for treating extreme osteoporosis. We previously confirmed that NELL-like molecule-1 (NELL-1) is crucial for bone density maintenance. We further PEGylated NELL-1 (NELL-polyethylene glycol, or NELL-PEG) to increase systemic delivery half-life from 5.5 to 15.5 h. In this study, we used a bio-inert bisphosphonate (BP) moiety to chemically engineer NELL-PEG into BP-NELL-PEG and specifically target bone tissues. We found conjugation with BP improved hydroxyapatite (HA) binding and protein stability of NELL-PEG while preserving NELL-1's osteogenicity in vitro. Furthermore, BP-NELL-PEG showed superior in vivo bone specificity without observable pathology in liver, spleen, lungs, brain, heart, muscles, or ovaries of mice. Finally, we tested BP-NELL-PEG through spaceflight exposure onboard the International Space Station (ISS) at maximal animal capacity (n = 40) in a long-term (9 week) osteoporosis therapeutic study and found that BP-NELL-PEG significantly increased bone formation in flight and ground control mice without obvious adverse health effects. Our results highlight BP-NELL-PEG as a promising therapeutic to mitigate extreme bone loss from long-duration microgravity exposure and musculoskeletal degeneration on Earth, especially when resistance training is not possible due to incapacity (e.g., bone fracture, stroke).
Collapse
Affiliation(s)
- Pin Ha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jin Hee Kwak
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yulong Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Forsyth Institute, Cambridge, MA, 02142, USA
| | - Jiayu Shi
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luan Tran
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy Pan Liu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin-Chuan Pan
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Samantha Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jong Kil Kim
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eric Chen
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yasaman Shirazi-Fard
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Louis S Stodieck
- BioServe Space Technologies and Aerospace Engineering Sciences, University of Colorado, Boulder, CO, 80303, USA
| | - Andy Lin
- Office of Advanced Research Computing, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Stella Nuo Dong
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin M Wu
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Kang Ting
- Forsyth Institute, Cambridge, MA, 02142, USA.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
25
|
Ma M, Zeng H, Yang P, Xu J, Zhang X, He W. Drug Delivery and Therapy Strategies for Osteoporosis Intervention. Molecules 2023; 28:6652. [PMID: 37764428 PMCID: PMC10534890 DOI: 10.3390/molecules28186652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
With the advent of the aging society, osteoporosis (OP) risk increases yearly. Currently, the clinical usage of anti-OP drugs is challenged by recurrent side effects and poor patient compliance, regardless of oral, intravenous, or subcutaneous administration. Properly using a drug delivery system or formulation strategy can achieve targeted drug delivery to the bone, diminish side effects, improve bioavailability, and prolong the in vivo residence time, thus effectively curing osteoporosis. This review expounds on the pathogenesis of OP and the clinical medicaments used for OP intervention, proposes the design approach for anti-OP drug delivery, emphatically discusses emerging novel anti-OP drug delivery systems, and enumerates anti-OP preparations under clinical investigation. Our findings may contribute to engineering anti-OP drug delivery and OP-targeting therapy.
Collapse
Affiliation(s)
- Mingyang Ma
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (M.M.); (H.Z.)
| | - Huiling Zeng
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (M.M.); (H.Z.)
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, China;
| | - Jiabing Xu
- Taizhou Institute for Drug Control, Taizhou 225316, China;
| | - Xingwang Zhang
- Department of Pharmaceutics, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
26
|
Ashique S, Faiyazuddin M, Afzal O, Gowri S, Hussain A, Mishra N, Garg A, Maqsood S, Akhtar MS, Altamimi AS. Advanced nanoparticles, the hallmark of targeted drug delivery for osteosarcoma-an updated review. J Drug Deliv Sci Technol 2023; 87:104753. [DOI: 10.1016/j.jddst.2023.104753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
27
|
Liu X, Li F, Dong Z, Gu C, Mao D, Chen J, Luo L, Huang Y, Xiao J, Li Z, Liu Z, Yang Y. Metal-polyDNA nanoparticles reconstruct osteoporotic microenvironment for enhanced osteoporosis treatment. SCIENCE ADVANCES 2023; 9:eadf3329. [PMID: 37531423 PMCID: PMC10396296 DOI: 10.1126/sciadv.adf3329] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
Current clinical approaches to osteoporosis primarily target osteoclast biology, overlooking the synergistic role of bone cells, immune cells, cytokines, and inorganic components in creating an abnormal osteoporotic microenvironment. Here, metal-polyDNA nanoparticles (Ca-polyCpG MDNs) composed of Ca2+ and ultralong single-stranded CpG sequences were developed to reconstruct the osteoporotic microenvironment and suppress osteoporosis. Ca-polyCpG MDNs can neutralize osteoclast-secreted hydrogen ions, provide calcium repletion, promote remineralization, and repair bone defects. Besides, the immune-adjuvant polyCpG in MDNs could induce the secretion of osteoclastogenesis inhibitor interleukin-12 and reduce the expression of osteoclast function effector protein to inhibit osteoclast differentiation, further reducing osteoclast-mediated bone resorption. PPi4- generated during the rolling circle amplification reaction acts as bisphosphonate analog and enhances bone targeting of Ca-polyCpG MDNs. In ovariectomized mouse and rabbit models, Ca-polyCpG MDNs prevented bone resorption and promoted bone repair by restoring the osteoporotic microenvironment, providing valuable insights into osteoporosis therapy.
Collapse
Affiliation(s)
- Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Li
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Ziliang Dong
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chao Gu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dongsheng Mao
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Luo
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuting Huang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Xiao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhanchun Li
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhuang Liu
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
28
|
Kuvshinova EA, Petrakova NV, Nikitina YO, Sviridova IK, Akhmedova SA, Kirsanova VA, Karalkin PA, Komlev VS, Sergeeva NS, Kaprin AD. Functionalization of Octacalcium Phosphate Bone Graft with Cisplatin and Zoledronic Acid: Physicochemical and Bioactive Properties. Int J Mol Sci 2023; 24:11633. [PMID: 37511391 PMCID: PMC10380611 DOI: 10.3390/ijms241411633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/02/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Bones are the fourth most frequent site of metastasis from malignant tumors, including breast cancer, prostate cancer, melanoma, etc. The bioavailability of bone tissue for chemotherapy drugs is extremely low. This requires a search for new approaches of targeted drug delivery to the tumor growth zone after surgery treatment. The aim of this work was to develop a method for octacalcium phosphate (OCP) bone graft functionalization with the cytostatic drug cisplatin to provide the local release of its therapeutic concentrations into the bone defect. OCP porous ceramic granules (OCP ceramics) were used as a platform for functionalization, and bisphosphonate zoledronic acid was used to mediate the interaction between cisplatin and OCP and enhance their binding strength. The obtained OCP materials were studied using scanning electron and light microscopy, high-performance liquid chromatography, atomic emission spectroscopy, and real-time PCR. In vitro and in vivo studies were performed on normal and tumor cell lines and small laboratory animals. The bioactivity of initial OCP ceramics was explored and the efficiency of OCP functionalization with cisplatin, zoledronic acid, and their combination was evaluated. The kinetics of drug release and changes in ceramics properties after functionalization were studied. It was established that zoledronic acid changed the physicochemical and bioactive properties of OCP ceramics and prolonged cisplatin release from the ceramics. In vitro and in vivo experiments confirmed the biocompatibility, osteoconductivity, and osteoinductivity, as well as cytostatic and antitumor properties of the obtained materials. The use of OCP ceramics functionalized with a cytostatic via the described method seems to be promising in clinics when primary or metastatic tumors of the bone tissue are removed.
Collapse
Affiliation(s)
- Ekaterina A Kuvshinova
- P.A. Herzen Moscow Research Oncology Institute, Branch of FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
| | - Nataliya V Petrakova
- A.A. Baikov Institute of Metallurgy and Materials Science RAS, Leninsky Avenue 49, 119334 Moscow, Russia
| | - Yulia O Nikitina
- A.A. Baikov Institute of Metallurgy and Materials Science RAS, Leninsky Avenue 49, 119334 Moscow, Russia
| | - Irina K Sviridova
- P.A. Herzen Moscow Research Oncology Institute, Branch of FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
| | - Suraja A Akhmedova
- P.A. Herzen Moscow Research Oncology Institute, Branch of FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
| | - Valentina A Kirsanova
- P.A. Herzen Moscow Research Oncology Institute, Branch of FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
| | - Pavel A Karalkin
- P.A. Herzen Moscow Research Oncology Institute, Branch of FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
- L.L. Levshin Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, Trubetskaya 8-2, 119991 Moscow, Russia
| | - Vladimir S Komlev
- A.A. Baikov Institute of Metallurgy and Materials Science RAS, Leninsky Avenue 49, 119334 Moscow, Russia
| | - Natalia S Sergeeva
- P.A. Herzen Moscow Research Oncology Institute, Branch of FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
| | - Andrey D Kaprin
- FSBI National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinsky Pass. 3, 125284 Moscow, Russia
- Department of Urology and Operative Nephrology, Peoples' Friendship University of Russia, Miklukho-Maklay Str., 6, 117198 Moscow, Russia
| |
Collapse
|
29
|
Frase D, Lee C, Nachiappan C, Gupta R, Akkouch A. The Inflammatory Contribution of B-Lymphocytes and Neutrophils in Progression to Osteoporosis. Cells 2023; 12:1744. [PMID: 37443778 PMCID: PMC10340451 DOI: 10.3390/cells12131744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Osteoporosis is a bone disease characterized by structural deterioration and low bone mass, leading to fractures and significant health complications. In this review, we summarize the mechanisms by which B-lymphocytes and neutrophils contribute to the development of osteoporosis and potential therapeutics targeting these immune mediators to reduce the proinflammatory milieu. B-lymphocytes-typically appreciated for their canonical role in adaptive, humoral immunity-have emerged as critical regulators of bone remodeling. B-lymphocytes communicate with osteoclasts and osteoblasts through various cytokines, including IL-7, RANK, and OPG. In inflammatory conditions, B-lymphocytes promote osteoclast activation and differentiation. However, B-lymphocytes also possess immunomodulatory properties, with regulatory B-lymphocytes (Bregs) secreting TGF-β1 to restrain pathogenic osteoclastogenesis. Neutrophils, the body's most prevalent leukocyte, also contribute to the proinflammatory environment that leads to osteoporotic bone remodeling. In aged individuals, neutrophils display reduced chemotaxis, phagocytosis, and apoptosis. Understanding the delicate interplay between B-lymphocytes and neutrophils in the context of impaired bone metabolism is crucial for targeted therapies for osteoporosis.
Collapse
Affiliation(s)
- Drew Frase
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Chi Lee
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Chidambaram Nachiappan
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Richa Gupta
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Adil Akkouch
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
- Department of Orthopaedic Surgery and Medical Engineering Program, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| |
Collapse
|
30
|
Kuźnik A, Kozicka D, Październiok-Holewa A, Dąbek A, Juszczak K, Sokołowska G, Erfurt K. A method for the synthesis of unsymmetric bisphosphoric analogs of α-amino acids. RSC Adv 2023; 13:18908-18915. [PMID: 37362601 PMCID: PMC10288832 DOI: 10.1039/d3ra02981f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Herein, we describe the first universal strategy for the synthesis of unsymmetric phosphonyl-phosphinyl and phosphonyl-phosphinoyl analogs of N-protected 1-aminobisphosphonates. The proposed user-friendly procedure, based on a one-pot reaction of the α-ethoxy derivatives of phosphorus analogs of protein and non-protein α-amino acids with triphenylphosphonium tetrafluoroborate and an appropriate phosphorus nucleophile (diethyl phenylphosphonite or methyl diphenylphosphinite), provides good to very good yields of 53-91% under mild catalyst-free conditions (temperature: rt to 40 °C, time: 1 to 6 hours). The progress of the transformation, running through the corresponding phosphonium salt as a reactive intermediate, was monitored by 31P NMR spectroscopy, which is a convenient tool for the identification of the transient species formed here. In this paper, we present the full characteristics of the spectroscopic properties of all 13 synthesized models of structurally diverse N-protected unsymmetric bisphosphoric analogs of α-amino acids. Therefore, these results contribute to increasing the practical applicability of our recently reported synthesis protocol of symmetric models of α-aminobisphosphonates derivatives and thus justify its universality.
Collapse
Affiliation(s)
- Anna Kuźnik
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center of Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| | - Dominika Kozicka
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center of Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| | - Agnieszka Październiok-Holewa
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
- Biotechnology Center of Silesian University of Technology B. Krzywoustego 8 44-100 Gliwice Poland
| | - Alicja Dąbek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
| | - Karolina Juszczak
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
| | - Gloria Sokołowska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
| | - Karol Erfurt
- Department of Chemical Organic Technology and Petrochemistry, Silesian University of Technology B. Krzywoustego 4 44-100 Gliwice Poland
| |
Collapse
|
31
|
Ciobanu GA, Camen A, Ionescu M, Vlad D, Munteanu CM, Gheorghiță MI, Lungulescu CV, Staicu IE, Sin EC, Chivu L, Mercuț R, Popescu SM. Risk Factors for Medication-Related Osteonecrosis of the Jaw-A Binomial Analysis of Data of Cancer Patients from Craiova and Constanta Treated with Zoledronic Acid. J Clin Med 2023; 12:jcm12113747. [PMID: 37297941 DOI: 10.3390/jcm12113747] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
MRONJ (Medication-Related Osteonecrosis of the Jaw) is a condition observed in a subset of cancer patients who have undergone treatment with zoledronic acid in order to either prevent or treat bone metastases. The primary aim of this research was to establish the importance of risk factors in the development of medication-related osteonecrosis of the jaw in cancer patients receiving zoledronic acid therapy for bone metastases. The present study is an observational retrospective investigation conducted at two university centers, namely, Craiova and Constanța, and included cancer patients treated with zoledronic acid. The medical records of the patients were obtained over a four-year timeframe spanning from June 2018 to June 2022. The data analysis was carried out between January 2021 and October 2022. Patients were treated for cancer, bone metastases, and MRONJ according to the international guidelines. The research investigated a cohort of 174 cancer patients (109 females and 65 males) aged between 22 and 84 years (with a mean age 64.65 ± 10.72 years) seeking treatment at oncology clinics situated in Craiova and Constanța. The study conducted a binomial logistic regression to analyze ten predictor variables, namely, gender, age, smoking status, treatment duration, chemotherapy, radiotherapy, endocrine therapy, presence of diabetes mellitus (DM), obesity, and hypertension (HT). The results of the analysis revealed that only five of the ten predictor variables were statistically significant for MRONJ occurrence: duration of treatment (p < 0.005), chemotherapy (p = 0.007), and hypertension (p = 0.002) as risk factors, and endocrine therapy (p = 0.001) and obesity (p = 0.024) as protective factors.
Collapse
Affiliation(s)
- George Adrian Ciobanu
- Department of Oral Rehabilitation, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Oral and Maxillofacial Surgery, Dental Medicine Faculty, "Ovidius" University of Constanța, 900470 Constanța, Romania
| | - Adrian Camen
- Department of Oral and Maxillofacial Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihaela Ionescu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniel Vlad
- Department of Oral and Maxillofacial Surgery, Dental Medicine Faculty, "Ovidius" University of Constanța, 900470 Constanța, Romania
| | - Cristina Maria Munteanu
- Department of Oral and Maxillofacial Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mircea Ionuț Gheorghiță
- Department of Oral and Maxillofacial Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - Ionela Elisabeta Staicu
- Department of Orthodontics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Elena Claudia Sin
- Department of Oral and Maxillofacial Surgery, Dental Medicine Faculty, "Ovidius" University of Constanța, 900470 Constanța, Romania
| | - Luminița Chivu
- Department of Oral and Maxillofacial Surgery, The County Emergency Clinical Hospital "Sf. Apostol Andrei", 900591 Constanța, Romania
| | - Răzvan Mercuț
- Department of Plastic Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Sanda Mihaela Popescu
- Department of Oral Rehabilitation, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
32
|
Xu H, Wang W, Liu X, Huang W, Zhu C, Xu Y, Yang H, Bai J, Geng D. Targeting strategies for bone diseases: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:202. [PMID: 37198232 DOI: 10.1038/s41392-023-01467-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
33
|
Xie C, Ren Y, Weeks J, Xue T, Rainbolt J, Bentley KDM, Shu Y, Liu Y, Masters E, Cherian P, McKenna C, Neighbors J, Ebetino F, Schwarz E, Sun S. Evidence of Bisphosphonate-Conjugated Sitafloxacin Eradication of Established Methicillin-Resistant S. aureus Infection with Osseointegration in Murine Models of Implant-Associated Osteomyelitis. RESEARCH SQUARE 2023:rs.3.rs-2856287. [PMID: 37214929 PMCID: PMC10197753 DOI: 10.21203/rs.3.rs-2856287/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Eradication of MRSA osteomyelitis requires elimination of distinct biofilms. To overcome this, we developed bisphosphonate-conjugated sitafloxacin (BCS, BV600072) and hydroxybisphosphonate-conjugate sitafloxacin (HBCS, BV63072), which achieve "target-and-release" drug delivery proximal to the bone infection and have prophylactic efficacy against MRSA static biofilm in vitro and in vivo. Here we evaluated their therapeutic efficacy in a murine 1-stage exchange femoral plate model with bioluminescent MRSA (USA300LAC::lux). Osteomyelitis was confirmed by CFU on the explants and longitudinal bioluminescent imaging (BLI) after debridement and implant exchange surgery on day 7, and mice were randomized into seven groups: 1) Baseline (harvested at day 7, no treatment); 2) HPBP (bisphosphonate control for BCS) + vancomycin; 3) HPHBP (bisphosphonate control for HBCS) + vancomycin; 4) vancomycin; 5) sitafloxacin; 6) BCS + vancomycin; and 7) HBCS + vancomycin. BLI confirmed infection persisted in all groups except for mice treated with BCS or HBCS + vancomycin. Radiology revealed catastrophic femur fractures in all groups except mice treated with BCS or HBCS + vancomycin, which also displayed decreases in peri-implant bone loss, osteoclast numbers, and biofilm. To confirm this, we assessed the efficacy of vancomycin, sitafloxacin, and HBCS monotherapy in a transtibial implant model. The results showed complete lack of vancomycin efficacy, while all mice treated with HBCS had evidence of infection control, and some had evidence of osseous integrated septic implants, suggestive of biofilm eradication. Taken together these studies demonstrate that HBCS adjuvant with standard of care debridement and vancomycin therapy has the potential to eradicate MRSA osteomyelitis.
Collapse
Affiliation(s)
- Chao Xie
- University of Rochester Medical Center
| | | | | | | | | | | | - Ye Shu
- University of Rochester Medical Center
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chen H, Cai G, Ruan X, Lu Y, Li G, Chen Z, Guan Z, Zhang H, Sun W, Wang H. Bone-targeted bortezomib increases bone formation within Calvarial trans-sutural distraction osteogenesis. Bone 2023; 169:116677. [PMID: 36646264 DOI: 10.1016/j.bone.2023.116677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/01/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023]
Abstract
The high rate of relapse in craniofacial disharmony treatment via trans-sutural distraction osteogenesis (TSDO) is due to the failure to form a stable bone bridge in the suture gap. Bisphosphonates (BP) have a high propensity to localize to hydroxyapatite in the bone matrix and are commonly used as targeting ligands for local delivery of therapeutics into bone microenvironment. Bone-targeted Bortezomib (BP-Btz) is chemosynthetic by linking Btz (Bortezomib) to a BP residue and could target bone tissue to promote osteoblast differentiation and inhibit osteoclastogenesis. Here, suture-derived mesenchymal stem cells (SuSCs) and osteoclasts were treated with Btz and BP-Btz. Aforesaid drugs were injected locally into the sagittal sutures to explore their effects in TSDO. Further, pharmacological properties of BP-Btz in the suture expansion model were assessed by fluorescent BP analogs and levels of total ubiquitinated (Ub)-proteins. The results showed that BP-Btz could stimulate osteogenic differentiation of SuSCs, bind to bone matrix and inhibit osteoclastogenesis. Biological effects of BP-Btz were similar with those of Btz in osteoblast differentiation and osteoclastogenesis inhibition in vitro. Activated bone metabolism were detected after 14 days in the sagittal suture expansion model. Increased osteoid area, remarkably decreased osteoclast surface and enhanced osteogenesis were detected in vivo after treatment with BP-Btz. Green fluorescence signal detection and pharmacodynamic studies revealed that BP-Btz bound to suture edge, released Btz in remodeling conditions, had a higher local concentration and sustained longer than free Btz. This study delineated the clinical potential of bone-targeted Btz conjugate as an efficacious strategy to promote trans-sutural distraction osteogenesis.
Collapse
Affiliation(s)
- Hongyu Chen
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Guanhui Cai
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiaolei Ruan
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yahui Lu
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Gen Li
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhenwei Chen
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhaolan Guan
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| | - Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
35
|
Shimina GG, Bateneva AV, Tsyplenkova ES, Gamaley SG, Esina TI, Volosnikova EA, Danilenko ED. HEMOSTIMULATING PROPERTIES OF THE CONJUGATES OF GRANULOCYTE-MACROPHAGE COLONY STIMULATING FACTOR WITH ALENDRONIC ACID. PHARMACY & PHARMACOLOGY 2022. [DOI: 10.19163/2307-9266-2022-10-5-472-482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the work is to evaluate the hemostimulating activity of recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) conjugates with alendronic acid (ALN) in the model of cytostatic myelosuppression and the dynamics of rhGM-CSF accumulation as a part of the conjugate in the bone tissue and bone marrow of mice.Materials and methods. The conjugates obtained by a solid-phase synthesis using 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide or periodate oxidation, were used. A hemostimulating activity was evaluated in a model of a cytostatic myelosuppression induced by the administration of cyclophosphamide to CBA/Calac mice. RhGM-CSF preparations were injected subcutaneously for 4-5 days at the dose of 90 µg/kg. After the injections cycle had been completed, the total leukocyte and segmented neutrophil counts were carried out in the blood samples, and the total karyocyte count was carried out in the bone marrow samples.The tissue distribution of rhGM-CSF preparations was assessed in outbred CD-1 mice after a single intravenous administration at the effective dose. The content of rhGM-CSF in blood, femoral tissue and bone marrow was determined by enzyme immunoassay.Results. RhGM-CSF conjugates with ALN have been shown to retain the ability of the original protein to increase the number of leukocytes, segmented blood neutrophils, and bone marrow karyocytes under the action of conjugates. The stimulation of the neutrophil production used to be observed at earlier times than in the case of rhGM-CSF. The increase in the total number of bone marrow cells after the introduction of all three conjugates was more pronounced compared to the original protein (by 34%). The increased hemostimulatory effect of the AEG conjugate was accompanied by a more intense accumulation of rhGM-CSF in the bone tissue and bone marrow of mice. The rhGM-CSF introduced into the conjugate was detected in the bone tissue for 24 h and it circulated in the bloodstream for a longer time compared to the original protein.Conclusion. The data obtained make it possible to conclude that further work on the development of effective hemostimulating drugs based on rhGM-CSF conjugates with ALN, is promising.
Collapse
Affiliation(s)
- G. G. Shimina
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - A. V. Bateneva
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - E. S. Tsyplenkova
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - S. G. Gamaley
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - T. I. Esina
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - E. A. Volosnikova
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - E. D. Danilenko
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| |
Collapse
|
36
|
Huang K, Zapata D, Tang Y, Teng Y, Li Y. In vivo delivery of CRISPR-Cas9 genome editing components for therapeutic applications. Biomaterials 2022; 291:121876. [PMID: 36334354 PMCID: PMC10018374 DOI: 10.1016/j.biomaterials.2022.121876] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/15/2022] [Accepted: 10/23/2022] [Indexed: 12/07/2022]
Abstract
Since its mechanism discovery in 2012 and the first application for mammalian genome editing in 2013, CRISPR-Cas9 has revolutionized the genome engineering field and created countless opportunities in both basic science and translational medicine. The first clinical trial of CRISPR therapeutics was initiated in 2016, which employed ex vivo CRISPR-Cas9 edited PD-1 knockout T cells for the treatment of non-small cell lung cancer. So far there have been dozens of clinical trials registered on ClinicalTrials.gov in regard to using the CRISPR-Cas9 genome editing as the main intervention for therapeutic applications; however, most of these studies use ex vivo genome editing approach, and only a few apply the in vivo editing strategy. Compared to ex vivo editing, in vivo genome editing bypasses tedious procedures related to cell isolation, maintenance, selection, and transplantation. It is also applicable to a wide range of diseases and disorders. The main obstacles to the successful translation of in vivo therapeutic genome editing include the lack of safe and efficient delivery system and safety concerns resulting from the off-target effects. In this review, we highlight the therapeutic applications of in vivo genome editing mediated by the CRISPR-Cas9 system. Following a brief introduction of the history, biology, and functionality of CRISPR-Cas9, we showcase a series of exemplary studies in regard to the design and implementation of in vivo genome editing systems that target the brain, inner ear, eye, heart, liver, lung, muscle, skin, immune system, and tumor. Current challenges and opportunities in the field of CRISPR-enabled therapeutic in vivo genome editing are also discussed.
Collapse
Affiliation(s)
- Kun Huang
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Daniel Zapata
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yan Tang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Yamin Li
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
37
|
Tholen P, Brown CN, Keil C, Bayir A, Zeng HH, Haase H, Thompson RB, Lengyel I, Yücesan G. A 2,7-dichlorofluorescein derivative to monitor microcalcifications. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2022; 7:1415-1421. [PMID: 37927331 PMCID: PMC10624163 DOI: 10.1039/d2me00185c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Herein, we report the crystal structure of 2,7-dichlorofluorescein methyl ester (DCF-ME) and its fluorescence response to hydroxyapatite binding. The reported fluorophore is very selective for staining the bone matrix and provides turn-on fluorescence upon hydroxyapatite binding. The reported fluorophore can readily pass the cell membrane of the C2C12 cell line, and it is non-toxic for the cell line. The reported fluorophore DCF-ME may find applications in monitoring bone remodeling and microcalcification as an early diagnosis tool for breast cancer and age-related macular degeneration.
Collapse
Affiliation(s)
- Patrik Tholen
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Connor N Brown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Claudia Keil
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Ali Bayir
- The Department of Chemistry, Yıldız Technical University, 34220, Esenler, Istanbul, Turkey
| | - Hui-Hui Zeng
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Hajo Haase
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Richard B Thompson
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Gündoğ Yücesan
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
38
|
Synthesis and Antitumor Activity of a GSH Inert Bisphosphonate Platinum (II) Complex. J CHEM-NY 2022. [DOI: 10.1155/2022/3137142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma is a highly aggressive neoplasm. Traditional platinum chemotherapeutic agents for osteosarcoma inevitably have acquired drug resistance and serious side effects, which have limited their utility. To slow down the reaction of platinum drugs with glutathione (GSH) is a strategy to overcome the resistance of platinum chemotherapeutic agents. Herein, the unique design of a GSH inert bisphosphonate platinum complex cis-{di(amino)platinum[tetraethyl 2,2-bis(2-pyridinylmethyl)methylidene-1,1-bisphosphonate]} (DBPP) is reported. MTT assay demonstrates that DBPP showed moderate inhibition towards human osteosarcoma cell line U2OS cells. The cytostatic action of DBPP is related to conformational conversion from B-DNA to A-DNA and the unwinding of pUC19 DNA. DBPP could also destroy the tertiary structure of human serum albumin (HSA). Notably, 31P NMR and 1H NMR indicate that DBPP can hardly chelate with GSH, which could overcome the GSH-induced side effects. We envision that this unique design of the platinum complex would open up new ways to overcome GSH-induced resistance.
Collapse
|
39
|
Nouraeinejad A. Osteoporosis, bisphosphonates, and ocular inflammation. Eur J Ophthalmol 2022; 33:11206721221125022. [PMID: 36062621 DOI: 10.1177/11206721221125022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Osteoporosis, as a musculoskeletal disease, is very common, especially in the era of aging society. It is described by a reduction in bone strength and increased risk of fractures, which are linked to considerable morbidity, mortality, and high healthcare burdens. Bisphosphonates are the most commonly used drugs to manage osteoporosis and they consequently reduce fracture risk. However, one of the clinical challenges is fear of side effects in patients who are using bisphosphonates as the administration of these drugs is lengthy. Ocular inflammation has been reporting as one of the potential vision-threatening side effects of bisphosphonates. PURPOSE To review previously reported ocular inflammation in patients taking bisphosphonates. METHOD A literature survey was conducted using databases in order to collect data for a narrative review of published reports regarding ocular side effects of bisphosphonates. RESULTS People taking bisphosphonates were at a relatively low risk of ocular inflammation, with the onset from a few hours after exposure up to more than 3 years. The release of inflammatory mediators mainly by activated gamma delta T cells in response to bisphosphonates was the main cause of ocular inflammation. CONCLUSION Strategies for treating patients with osteoporosis should consider the potential ocular side effects of bisphosphonates. Ocular inflammation, as one of the side effects of bisphosphonates, is a serious sight-threatening sign and should be taken seriously. Greater awareness of the association between bisphosphonate use and ocular inflammation may allow for earlier identification and timely treatment of future cases.
Collapse
Affiliation(s)
- Ali Nouraeinejad
- Department of Clinical Ophthalmology, 4919University College London, London, UK
| |
Collapse
|
40
|
Hu B, Zhang Y, Zhang G, Li Z, Jing Y, Yao J, Sun S. Research progress of bone-targeted drug delivery system on metastatic bone tumors. J Control Release 2022; 350:377-388. [PMID: 36007681 DOI: 10.1016/j.jconrel.2022.08.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Bone metastases are common in malignant tumors and the effect of conventional treatment is limited. How to effectively inhibit tumor bone metastasis and deliver the drug to the bone has become an urgent issue to be solved. While bone targeting drug delivery systems have obvious advantages in the treatment of bone tumors. The research on bone-targeted anti-tumor therapy has made significant progress in recent years. We introduced the related tumor pathways of bone metastases. The tumor microenvironment plays an important role in metastatic bone tumors. We introduce a drug-loading systems based on different environment-responsive nanocomposites for anti-tumor and anti-metastatic research. According to the process of bone metastases and the structure of bone tissue, we summarized the information on bone-targeting molecules. Bisphosphate has become the first choice of bone-targeted drug delivery carrier because of its affinity with hydroxyapatite in bone. Therefore, we sought to summarize the bone-targeting molecule of bisphosphate to identify the modification effect on bone-targeting. And this paper discusses the relationship between bisphosphate bone targeting molecular structure and drug delivery carriers, to provide some new ideas for the research and development of bone-targeting drug delivery carriers. Targeted therapy will make a more outstanding contribution to the treatment of tumors.
Collapse
Affiliation(s)
- Beibei Hu
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China; State Key Laboratory Breeding Base-Hebei Province, Key Laboratory of Molecular Chemistry for Drug, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Yongkang Zhang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Guogang Zhang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Zhongqiu Li
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Yongshuai Jing
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Jun Yao
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China.
| | - Shiguo Sun
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China.
| |
Collapse
|
41
|
PT-112 Induces Mitochondrial Stress and Immunogenic Cell Death, Targeting Tumor Cells with Mitochondrial Deficiencies. Cancers (Basel) 2022; 14:cancers14163851. [PMID: 36010843 PMCID: PMC9405950 DOI: 10.3390/cancers14163851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary PT-112 is a novel pyrophosphate–platinum conjugate under Phase 1/2 clinical development for the treatment of several tumor types. In this study, using mouse tumor cells with well-characterized mitochondrial and metabolic status, we investigated the mechanisms underlying PT-112’s cancer cell death effects. Our results showed that cells with defective mitochondria were more sensitive to PT-112 when compared to cells with normal mitochondrial function. Moreover, PT-112 induced tumor cell death in those sensitive cells through non-conventional mechanisms, including increased mitochondrial stress, free radical generation and immunogenic cell death, a form of cell death that elicits an immune response. Taken together, the present findings suggest the potential for predictors of PT-112 sensitivity in the clinical setting on the basis of metabolic function. Abstract PT-112 is a novel pyrophosphate–platinum conjugate, with clinical activity reported in advanced pretreated solid tumors. While PT-112 has been shown to induce robust immunogenic cell death (ICD) in vivo but only minimally bind DNA, the molecular mechanism underlying PT-112 target disruption in cancer cells is still under elucidation. The murine L929 in vitro system was used to test whether differential metabolic status alters PT-112’s effects, including cell cytotoxicity. The results showed that tumor cells presenting mutations in mitochondrial DNA (mtDNA) (L929dt and L929dt cybrid cells) and reliant on glycolysis for survival were more sensitive to cell death induced by PT-112 compared to the parental and cybrid cells with an intact oxidative phosphorylation (OXPHOS) pathway (L929 and dtL929 cybrid cells). The type of cell death induced by PT-112 did not follow the classical apoptotic pathway: the general caspase inhibitor Z-VAD-fmk did not inhibit PT-112-induced cell death, alone or in combination with the necroptosis inhibitor necrostatin-1. Interestingly, PT-112 initiated autophagy in all cell lines, though this process was not complete. Autophagy is known to be associated with an integrated stress response in cancer cells and with subsequent ICD. PT-112 also induced a massive accumulation of mitochondrial reactive oxygen species, as well as changes in mitochondrial polarization—only in the sensitive cells harboring mitochondrial dysfunction—along with calreticulin cell-surface exposure consistent with ICD. PT-112 substantially reduced the amount of mitochondrial CoQ10 in L929 cells, while the basal CoQ10 levels were below our detection limits in L929dt cells, suggesting a potential relationship between a low basal level of CoQ10 and PT-112 sensitivity. Finally, the expression of HIF-1α was much higher in cells sensitive to PT-112 compared to cells with an intact OXPHOS pathway, suggesting potential clinical applications.
Collapse
|
42
|
Wawrzyniak N, Gramza-Michałowska A, Pruszyńska-Oszmałek E, Sassek M, Suliburska J. Effects of Calcium Lactate-Enriched Pumpkin on Calcium Status in Ovariectomized Rats. Foods 2022; 11:foods11142084. [PMID: 35885327 PMCID: PMC9325293 DOI: 10.3390/foods11142084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022] Open
Abstract
This study aimed to evaluate the effects of enriched pumpkin on calcium status in ovariectomized rats. The study was conducted in sixty female Wistar rats, which were divided into six groups: a group fed a standard diet (C) and five ovariectomized groups fed a standard diet (OVX_C) or a diet with calcium lactate (CaL), with calcium lactate-enriched pumpkin (P_CaL), with calcium lactate and alendronate (CaL_B), or with calcium lactate-enriched pumpkin with alendronate (P_CaL_B). After 12 weeks of the intervention, the rats were sacrificed, and their blood and tissues were collected. The calcium concentrations in serum and in tissues were measured using flame atomic absorption spectrometry (AAS). Serum concentrations of procollagen type-1 amino-terminal propeptide (PINP), parathyroid hormone PTH, estrogen (ES), and osteocalcin (OC) were determined with enzyme-linked immunosorbent assay (ELISA). It was found that enriched pumpkin increased the calcium level in the kidneys (194.13 ± 41.01 mg) compared to the C (87.88 ± 12.42 mg) and OVX_C (79.29 ± 7.66 mg) groups. The addition of alendronate increased the calcium level in the femurs (267.63 ± 23.63 mg) and more than six times in the kidneys (541.33 ± 62.91 mg) compared to the OVX_C group (234.53 ± 21.67 mg and 87.88 ± 12.42 mg, respectively). We found that the CaL, P_CaL, and CaL_B groups had significantly lower PINP serum concentrations (4.45 ± 0.82 ng/mL, 4.14 ± 0.69 ng/mL, and 3.77 ± 0.33 ng/mL) and higher PTH serum levels (3.39 ± 0.54 ng/dL, 3.38 ± 0.57 ng/dL, and 3.47 ± 0.28 ng/dL) than the OVX_C group (4.69 ± 0.82 ng/mL and 2.59 ± 0.45 ng/dL, respectively). In conclusion, pumpkin enriched with calcium lactate affects calcium status and normalizes PINP and PTH serum levels in ovariectomized rats. Diet with enriched pumpkin and alendronate increase calcium concentration in the femur. Enriched pumpkin causes calcium to accumulate in the kidneys of ovariectomized rats; alendronate significantly exacerbates this effect.
Collapse
Affiliation(s)
- Natalia Wawrzyniak
- Department of Human Nutrition and Dietetics, Faculty of Food and Nutrition Science, University of Life Sciences, 60-624 Poznan, Poland;
| | - Anna Gramza-Michałowska
- Department of Gastronomy Sciences and Functional Foods, Faculty of Food and Nutrition Science, University of Life Sciences, 60-624 Poznan, Poland;
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine and Animal Science, University of Life Sciences, 60-637 Poznan, Poland; (E.P.-O.); (M.S.)
| | - Maciej Sassek
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine and Animal Science, University of Life Sciences, 60-637 Poznan, Poland; (E.P.-O.); (M.S.)
| | - Joanna Suliburska
- Department of Human Nutrition and Dietetics, Faculty of Food and Nutrition Science, University of Life Sciences, 60-624 Poznan, Poland;
- Correspondence:
| |
Collapse
|
43
|
Kim JK, Ha L, Kwon YE, Lee SG, Kim DP. Rapid Flow Synthesis of a Biomimetic Carbonate Apatite as an Effective Drug Carrier. ACS APPLIED MATERIALS & INTERFACES 2022; 14:29626-29638. [PMID: 35724663 DOI: 10.1021/acsami.2c06900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A facile synthesis of apatite nanocrystals analogous to bioapatites with increased biocompatibility and biodegradability can remedy the shortcomings of the widely applied synthetic hydroxyapatite (HAp) for bone defect treatment. Here, we propose an expeditious synthesis method to develop a biomimetic B-type carbonate apatite (CAp) with a simple capillary microfluidic device at room temperature. The process not only eliminates fluctuations with the addition of carbonate but also produces safe CAp drug carriers through simultaneous alendronate incorporation to the CAp structure. CAp displayed superior mineralization on osteoblast-like MG-63 cells when compared with HAp and HAp drug carriers that were produced using identical methods. Furthermore, alendronate-incorporated CAp drug carriers potentially displayed higher cancer cell suppression when applied to breast cancer cells attached to the bone tissue model, which signifies enhanced cancer metastasis to bone suppression due to the likelihood of increased alendronate release of CAp owing to its faster dissolution. Overall, our results may provide promising opportunities for enhanced clinical CAp application for bone defect treatment, particularly for bone loss and cancer to bone metastasis.
Collapse
Affiliation(s)
- Jung-Kyun Kim
- Center for Intelligent Microprocess of Pharmaceutical Synthesis (CIMPS), Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea
| | - Laura Ha
- Center for Intelligent Microprocess of Pharmaceutical Synthesis (CIMPS), Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea
| | - Yong-Eun Kwon
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, South Korea
| | - Sang-Gil Lee
- Center for Research Equipment, Korea Basic Science Institute (KBSI), 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, South Korea
| | - Dong-Pyo Kim
- Center for Intelligent Microprocess of Pharmaceutical Synthesis (CIMPS), Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea
| |
Collapse
|
44
|
Svarca A, Grava A, Dubnika A, Ramata-Stunda A, Narnickis R, Aunina K, Rieksta E, Boroduskis M, Jurgelane I, Locs J, Loca D. Calcium Phosphate/Hyaluronic Acid Composite Hydrogels for Local Antiosteoporotic Drug Delivery. Front Bioeng Biotechnol 2022; 10:917765. [PMID: 35866026 PMCID: PMC9294454 DOI: 10.3389/fbioe.2022.917765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the bone ability of self-regeneration, large bone defects require surgical intervention. Likewise, when it comes to osteoporotic bone fractures, new approaches should be considered a supportive mechanism for the surgery. In recent years, more and more attention has been attracted to advanced drug delivery systems for local osteoporosis treatment, combining appropriate biomaterials with antiosteoporotic drugs, allowing simultaneously to regenerate the bone and locally treat the osteoporosis. Within the current research, hyaluronic acid/strontium ranelate (HA/SrRan), HA/calcium phosphate nanoparticles (HA/CaP NPs), and HA/CaP NPs/SrRan hydrogels were prepared. The effect of CaP and SrRan presence in the composites on the swelling behavior, gel fraction, molecular structure, microstructure, and SrRan and Sr2+ release, as well as in vitro cell viability was evaluated. Obtained results revealed that the route of CaP nanoparticle incorporation into the HA matrix had a significant effect on the hydrogel gel fraction, rheological properties, swelling behavior, and microstructure. Nevertheless, it had a negligible effect on the release kinetics of SrRan and Sr2+. The highest cell (3T3) viability (>80%) was observed for HA hydrogels, with and without SrRan. Moreover, the positive effect of SrRan on 3T3 cells was also demonstrated, showing a significant increase (up to 50%) in cell viability if the used concentrations of SrRan were in the range of 0.05-0.2 μg/ml.
Collapse
Affiliation(s)
- Alise Svarca
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Andra Grava
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Arita Dubnika
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Anna Ramata-Stunda
- Department of Microbiology and Biotechnology, Faculty of Biology, University of Latvia, Riga, Latvia
| | - Raimonds Narnickis
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
| | - Kristine Aunina
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Eleonora Rieksta
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Martins Boroduskis
- Department of Microbiology and Biotechnology, Faculty of Biology, University of Latvia, Riga, Latvia
| | - Inga Jurgelane
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Janis Locs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Dagnija Loca
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
45
|
Vascular Cell Adhesion Molecule 1-Mediated Targeting of Human Hematopoietic Stem Cells to Bone Marrow Is Effective in Conferring Regeneration and Survival in Lethally Irradiated Mice. Transplant Cell Ther 2022; 28:667.e1-667.e10. [DOI: 10.1016/j.jtct.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022]
|
46
|
Ren Y, Xue T, Rainbolt J, Bentley KLDM, Galloway CA, Liu Y, Cherian P, Neighbors J, Hofstee MI, Ebetino FH, Moriarty TF, Sun S, Schwarz EM, Xie C. Efficacy of Bisphosphonate-Conjugated Sitafloxacin in a Murine Model of S. aureus Osteomyelitis: Evidence of "Target & Release" Kinetics and Killing of Bacteria Within Canaliculi. Front Cell Infect Microbiol 2022; 12:910970. [PMID: 35811672 PMCID: PMC9263620 DOI: 10.3389/fcimb.2022.910970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
S. aureus infection of bone is difficult to eradicate due to its ability to colonize the osteocyte-lacuno-canalicular network (OLCN), rendering it resistant to standard-of-care (SOC) antibiotics. To overcome this, we proposed two bone-targeted bisphosphonate-conjugated antibiotics (BCA): bisphosphonate-conjugated sitafloxacin (BCS) and hydroxybisphosphonate-conjugate sitafloxacin (HBCS). Initial studies demonstrated that the BCA kills S. aureus in vitro. Here we demonstrate the in vivo efficacy of BCS and HBCS versus bisphosphonate, sitafloxacin, and vancomycin in mice with implant-associated osteomyelitis. Longitudinal bioluminescent imaging (BLI) confirmed the hypothesized "target and release"-type kinetics of BCS and HBCS. Micro-CT of the infected tibiae demonstrated that HBCS significantly inhibited peri-implant osteolysis versus placebo and free sitafloxacin (p < 0.05), which was not seen with the corresponding non-antibiotic-conjugated bisphosphonate control. TRAP-stained histology confirmed that HBCS significantly reduced peri-implant osteoclast numbers versus placebo and free sitafloxacin controls (p < 0.05). To confirm S. aureus killing, we compared the morphology of S. aureus autolysis within in vitro biofilm and infected tibiae via transmission electron microscopy (TEM). Live bacteria in vitro and in vivo presented as dense cocci ~1 μm in diameter. In vitro evidence of autolysis presented remnant cell walls of dead bacteria or "ghosts" and degenerating (non-dense) bacteria. These features of autolyzed bacteria were also present among the colonizing S. aureus within OLCN of infected tibiae from placebo-, vancomycin-, and sitafloxacin-treated mice, similar to placebo. However, most of the bacteria within OLCN of infected tibiae from BCA-treated mice were less dense and contained small vacuoles and holes >100 nm. Histomorphometry of the bacteria within the OLCN demonstrated that BCA significantly increased their diameter versus placebo and free antibiotic controls (p < 0.05). As these abnormal features are consistent with antibiotic-induced vacuolization, bacterial swelling, and necrotic phenotype, we interpret these findings to be the initial evidence of BCA-induced killing of S. aureus within the OLCN of infected bone. Collectively, these results support the bone targeting strategy of BCA to overcome the biodistribution limits of SOC antibiotics and warrant future studies to confirm the novel TEM phenotypes of bacteria within OLCN of S. aureus-infected bone of animals treated with BCS and HBCS.
Collapse
Affiliation(s)
- Youliang Ren
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | - Thomas Xue
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Joshua Rainbolt
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Karen L. de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
- Center for Advanced Research Technologies, University of Rochester Medical Center, Rochester, NY, United States
| | - Chad A. Galloway
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
- Center for Advanced Research Technologies, University of Rochester Medical Center, Rochester, NY, United States
| | - Yuting Liu
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | | | - Jeffrey Neighbors
- BioVinc LLC, Pasadena, CA, United States
- Department of Pharmacology, Pennsylvania State University, Hershey, PA, United States
| | | | - Frank H. Ebetino
- BioVinc LLC, Pasadena, CA, United States
- Department of Chemistry, University of Rochester, Rochester, NY, United States
| | | | | | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
47
|
One-Pot and Catalyst-Free Transformation of N-Protected 1-Amino-1-Ethoxyalkylphosphonates into Bisphosphonic Analogs of Protein and Non-Protein α-Amino Acids. Molecules 2022; 27:molecules27113571. [PMID: 35684508 PMCID: PMC9182278 DOI: 10.3390/molecules27113571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Abstract
Herein, we describe the development of one-pot transformation of α-ethoxy derivatives of phosphorus analogs of protein and non-protein α-amino acids into biologically important N-protected 1-aminobisphosphonates. The proposed strategy, based on the three-component reaction of 1-(N-acylamino)-1-ethoxyphosphonates with triphenylphosphonium tetrafluoroborate and triethyl phosphite, facilitates good to excellent yields under mild reaction conditions. The course of the reaction was monitored by 31P NMR spectroscopy, allowing the identification of probable intermediate species, thus making it possible to propose a reaction mechanism. In most cases, there is no need to use a catalyst to provide transformation efficiency, which increases its attractiveness both in economic and ecological terms. Furthermore, we demonstrate that the one-pot procedure can be successfully applied for the synthesis of structurally diverse N-protected bisphosphonic analogs of α-amino acids. As shown, the indirect formation of the corresponding phosphonium salt as a reactive intermediate during the conversion of 1-(N-acylamino)-1-ethoxyphosphonate into a 1-aminobisphosphonate derivative is a crucial component of the developed methodology.
Collapse
|
48
|
Xue L, Gong N, Shepherd SJ, Xiong X, Liao X, Han X, Zhao G, Song C, Huang X, Zhang H, Padilla MS, Qin J, Shi Y, Alameh MG, Pochan DJ, Wang K, Long F, Weissman D, Mitchell MJ. Rational Design of Bisphosphonate Lipid-like Materials for mRNA Delivery to the Bone Microenvironment. J Am Chem Soc 2022; 144:9926-9937. [PMID: 35616998 DOI: 10.1021/jacs.2c02706] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of lipid nanoparticle (LNP) formulations for targeting the bone microenvironment holds significant potential for nucleic acid therapeutic applications including bone regeneration, cancer, and hematopoietic stem cell therapies. However, therapeutic delivery to bone remains a significant challenge due to several biological barriers, such as low blood flow in bone, blood-bone marrow barriers, and low affinity between drugs and bone minerals, which leads to unfavorable therapeutic dosages in the bone microenvironment. Here, we construct a series of bisphosphonate (BP) lipid-like materials possessing a high affinity for bone minerals, as a means to overcome biological barriers to deliver mRNA therapeutics efficiently to the bone microenvironment in vivo. Following in vitro screening of BP lipid-like materials formulated into LNPs, we identified a lead BP-LNP formulation, 490BP-C14, with enhanced mRNA expression and localization in the bone microenvironment of mice in vivo compared to 490-C14 LNPs in the absence of BPs. Moreover, BP-LNPs enhanced mRNA delivery and secretion of therapeutic bone morphogenetic protein-2 from the bone microenvironment upon intravenous administration. These results demonstrate the potential of BP-LNPs for delivery to the bone microenvironment, which could potentially be utilized for a range of mRNA therapeutic applications including regenerative medicine, protein replacement, and gene editing therapies.
Collapse
Affiliation(s)
- Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sarah J Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Xinhong Xiong
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, Zhejiang 313001, China
| | - Xueyang Liao
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Gan Zhao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Chao Song
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Xisha Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Hanwen Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marshall S Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jingya Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yi Shi
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Darrin J Pochan
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Fanxin Long
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States.,Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
49
|
Russo S, Scotto di Carlo F, Gianfrancesco F. The Osteoclast Traces the Route to Bone Tumors and Metastases. Front Cell Dev Biol 2022; 10:886305. [PMID: 35646939 PMCID: PMC9139841 DOI: 10.3389/fcell.2022.886305] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/24/2022] [Indexed: 12/31/2022] Open
Abstract
Osteoclasts are highly specialized cells of the bone, with a unique apparatus responsible for resorption in the process of bone remodeling. They are derived from differentiation and fusion of hematopoietic precursors, committed to form mature osteoclasts in response to finely regulated stimuli produced by bone marrow-derived cells belonging to the stromal lineage. Despite a highly specific function confined to bone degradation, emerging evidence supports their relevant implication in bone tumors and metastases. In this review, we summarize the physiological role of osteoclasts and then focus our attention on their involvement in skeletal tumors, both primary and metastatic. We highlight how osteoclast-mediated bone erosion confers increased aggressiveness to primary tumors, even those with benign features. We also outline how breast and pancreas cancer cells promote osteoclastogenesis to fuel their metastatic process to the bone. Furthermore, we emphasize the role of osteoclasts in reactivating dormant cancer cells within the bone marrow niches for manifestation of overt metastases, even decades after homing of latent disseminated cells. Finally, we point out the importance of counteracting tumor progression and dissemination through pharmacological treatments based on a better understanding of molecular mechanisms underlying osteoclast lytic activity and their recruitment from cancer cells.
Collapse
Affiliation(s)
| | | | - Fernando Gianfrancesco
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council of Italy, Naples, Italy
| |
Collapse
|
50
|
Chu Y, Xia M, Wang F, Yan X, Dai Y, Dong L, Zhang Y. The uptake performance and microscopic mechanism of inorganic-organic phosphorus hybrid amorphous hydroxyapatite for multiple heavy metal ions. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.128384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|