1
|
Fang H, Rai A, Eslami SS, Huynh K, Liao HC, Salim A, Greening DW. Proteomics and Machine Learning-Based Approach to Decipher Subcellular Proteome of Mouse Heart. Mol Cell Proteomics 2025; 24:100952. [PMID: 40113211 PMCID: PMC12019842 DOI: 10.1016/j.mcpro.2025.100952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025] Open
Abstract
Protein compartmentalization to distinctive subcellular niches is critical for cardiac function and homeostasis. Here, we employed a rapid and robust workflow based on differential centrifugal-based fractionation with mass spectrometry-based proteomics and bioinformatic analyses for systemic mapping of the subcellular proteome of mouse heart. Using supervised machine learning of 450 hallmark protein markers from 16 subcellular niches, we further refined the subcellular information of 2083 proteins with high confidence. Our data validation focused on specific subcellular niches such as mitochondria, cell surface, cardiac dyad, myofibril, and nuclear, unfolding dominant subcellular localization of proteins in their native environment of mouse heart. We further provide targeted nuclear enrichment and co-immunoprecipitation-based proteomic validation from the heart of nuclear-localizing protein networks. This study provides novel insights into the molecular landscape of different subcellular niches of the heart and serves as a draft map for heart subcellular proteome.
Collapse
Affiliation(s)
- Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia
| | - Seyed Sadegh Eslami
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - Hsiao-Chi Liao
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
2
|
Emzhik M, Haeri A, Javidi J, Abdollahizad E, Qaribnejad A, Rezaee E, Torshabi M, Dadashzadeh S. Bile salt integrated cerasomes: A potential nanocarrier for enhancement of the oral bioavailability of idarubicin hydrochloride. Int J Pharm 2024; 662:124518. [PMID: 39074645 DOI: 10.1016/j.ijpharm.2024.124518] [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: 02/20/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Cerasomes are a modified form of liposomes containing both inorganic and organic parts and due to their strong polyorganosiloxane surface have remarkably high morphological stability and provide easier functionalization compared with conventional liposomes. To investigate the potential of these nanocarriers for oral delivery, bile salt integrated cerasomes (named bilocerasomes) encapsulating idarubicin hydrochloride (IDA) were prepared and characterized. The optimum formulation showed excellent stability in the simulated gastrointestinal fluids as well as under storage conditions. The oral pharmacokinetics of the IDA solution, empty nanocarrier + drug solution, and IDA-loaded bilocerasome were evaluated. The nanoformulation significantly increased the area under the drug concentration-time curve and the mean residence time (∼14.3- and 9-fold, respectively). The results obtained from cell uptake and chylomicron flow blocking approach revealed that bilocerasomes are absorbed into the intestinal cells via a clathrin/caveolin-independent endocytosis pathway and transported to the systemic circulation extensively via the intestinal lymphatic vessels. Considering the high stability of the prepared bilocerasome, noticeable participation of lymphatic transport in its systemic absorption and marked enhancement in the oral absorption of IDA, bilocerasomes can be introduced as a capable carrier for improving the oral bioavailability of drugs, particularly those that hepatic first-pass metabolism seriously limits their oral absorption.
Collapse
Affiliation(s)
- Marjan Emzhik
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jaber Javidi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Erfan Abdollahizad
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirsajad Qaribnejad
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Rezaee
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Torshabi
- Department of Dental Biomaterials, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Palomino-Cano C, Moreno E, Irache JM, Espuelas S. Targeting and activation of macrophages in leishmaniasis. A focus on iron oxide nanoparticles. Front Immunol 2024; 15:1437430. [PMID: 39211053 PMCID: PMC11357945 DOI: 10.3389/fimmu.2024.1437430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.
Collapse
Affiliation(s)
- Carmen Palomino-Cano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Esther Moreno
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Juan M. Irache
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| | - Socorro Espuelas
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| |
Collapse
|
4
|
Petcov TE, Straticiuc M, Iancu D, Mirea DA, Trușcă R, Mereuță PE, Savu DI, Mogoșanu GD, Mogoantă L, Popescu RC, Kopatz V, Jinga SI. Unveiling Nanoparticles: Recent Approaches in Studying the Internalization Pattern of Iron Oxide Nanoparticles in Mono- and Multicellular Biological Structures. J Funct Biomater 2024; 15:169. [PMID: 38921542 PMCID: PMC11204647 DOI: 10.3390/jfb15060169] [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: 03/30/2024] [Revised: 05/15/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Nanoparticle (NP)-based solutions for oncotherapy promise an improved efficiency of the anticancer response, as well as higher comfort for the patient. The current advancements in cancer treatment based on nanotechnology exploit the ability of these systems to pass biological barriers to target the tumor cell, as well as tumor cell organelles. In particular, iron oxide NPs are being clinically employed in oncological management due to this ability. When designing an efficient anti-cancer therapy based on NPs, it is important to know and to modulate the phenomena which take place during the interaction of the NPs with the tumor cells, as well as the normal tissues. In this regard, our review is focused on highlighting different approaches to studying the internalization patterns of iron oxide NPs in simple and complex 2D and 3D in vitro cell models, as well as in living tissues, in order to investigate the functionality of an NP-based treatment.
Collapse
Affiliation(s)
- Teodora Eliana Petcov
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
| | - Mihai Straticiuc
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Decebal Iancu
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Dragoș Alexandru Mirea
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Roxana Trușcă
- National Research Center for Micro and Nanomaterials, National University for Science and Technology Politehnica of Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania;
| | - Paul Emil Mereuță
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Diana Iulia Savu
- Department of Life and Environmental Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania
| | - George Dan Mogoșanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Laurențiu Mogoantă
- Research Center for Microscopic Morphology and Immunology, University of Medicine and Pharmacy of Craiova, 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Roxana Cristina Popescu
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
- Department of Life and Environmental Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania
| | - Verena Kopatz
- Department of Radiation Oncology, Medical University of Vienna, 18–20 Waehringer Guertel Street, 1090 Vienna, Austria;
| | - Sorin Ion Jinga
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
| |
Collapse
|
5
|
Niraula G, Wu C, Yu X, Malik S, Verma DS, Yang R, Zhao B, Ding S, Zhang W, Sharma SK. The Curie temperature: a key playmaker in self-regulated temperature hyperthermia. J Mater Chem B 2024; 12:286-331. [PMID: 37955235 DOI: 10.1039/d3tb01437a] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The Curie temperature is an important thermo-characteristic of magnetic materials, which causes a phase transition from ferromagnetic to paramagnetic by changing the spontaneous re-arrangement of their spins (intrinsic magnetic mechanism) due to an increase in temperature. The self-control-temperature (SCT) leads to the conversion of ferro/ferrimagnetic materials to paramagnetic materials, which can extend the temperature-based applications of these materials from industrial nanotechnology to the biomedical field. In this case, magnetic induction hyperthermia (MIH) with self-control-temperature has been proposed as a physical thermo-therapeutic method for killing cancer tumors in a biologically safe environment. Specifically, the thermal source of MIH is magnetic nanoparticles (MNPs), and thus their biocompatibility and Curie temperature are two important properties, where the former is required for their clinical application, while the latter acts as a switch to automatically control the temperature of MIH. In this review, we focus on the Curie temperature of magnetic materials and provide a complete overview beginning with basic magnetism and its inevitable relation with Curie's law, theoretical prediction and experimental measurement of the Curie temperature. Furthermore, we discuss the significance, evolution from different types of alloys to ferrites and impact of the shape, size, and concentration of particles on the Curie temperature considering the proposed SCT-based MIH together with their biocompatibility. Also, we highlight the thermal efficiency of MNPs in destroying tumor cells and the significance of a low Curie temperature. Finally, the challenges, concluding remarks, and future perspectives in promoting self-control-temperature based MIH to clinical application are discussed.
Collapse
Affiliation(s)
- Gopal Niraula
- Department of Physics, Federal University of Maranhão, São Luís, 65080-805, Brazil.
| | - Chengwei Wu
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116024, P. R. China.
| | - Xiaogang Yu
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116024, P. R. China.
| | - Sonia Malik
- LBLGC, University of Orléans, 1 Rue de Chartres-BP 6759, 45067 Orleans, France
| | - Dalip Singh Verma
- Department of Physics & Astronomical Science, Central University of Himachal Pradesh, Dharamshala, 176215, India
| | - Rengpeng Yang
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116024, P. R. China.
| | - Boxiong Zhao
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116024, P. R. China.
| | - Shuaiwen Ding
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116024, P. R. China.
| | - Wei Zhang
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116024, P. R. China.
| | - Surender Kumar Sharma
- Department of Physics, Federal University of Maranhão, São Luís, 65080-805, Brazil.
- Department of Physics, Central University of Punjab, Bathinda, 151401, India
| |
Collapse
|
6
|
Estévez M, Cicuéndez M, Crespo J, Serrano-López J, Colilla M, Fernández-Acevedo C, Oroz-Mateo T, Rada-Leza A, González B, Izquierdo-Barba I, Vallet-Regí M. Large-scale production of superparamagnetic iron oxide nanoparticles by flame spray pyrolysis: In vitro biological evaluation for biomedical applications. J Colloid Interface Sci 2023; 650:560-572. [PMID: 37429163 DOI: 10.1016/j.jcis.2023.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023]
Abstract
Despite the large number of synthesis methodologies described for superparamagnetic iron oxide nanoparticles (SPIONs), the search for their large-scale production for their widespread use in biomedical applications remains a mayor challenge. Flame Spray Pyrolysis (FSP) could be the solution to solve this limitation, since it allows the fabrication of metal oxide nanoparticles with high production yield and low manufacture costs. However, to our knowledge, to date such fabrication method has not been upgraded for biomedical purposes. Herein, SPIONs have been fabricated by FSP and their surface has been treated to be subsequently coated with dimercaptosuccinic acid (DMSA) to enhance their colloidal stability in aqueous media. The final material presents high quality in terms of nanoparticle size, homogeneous size distribution, long-term colloidal stability and magnetic properties. A thorough in vitro validation has been performed with peripheral blood cells and mesenchymal stem cells (hBM-MSCs). Specifically, hemocompatibility studies show that these functionalized FSP-SPIONs-DMSA nanoparticles do not cause platelet aggregation or impair basal monocyte function. Moreover, in vitro biocompatibility assays show a dose-dependent cellular uptake while maintaining high cell viability values and cell cycle progression without causing cellular oxidative stress. Taken together, the results suggest that the FSP-SPIONs-DMSA optimized in this work could be a worthy alternative with the benefit of a large-scale production aimed at industrialization for biomedical applications.
Collapse
Affiliation(s)
- Manuel Estévez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain.
| | - Mónica Cicuéndez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Julián Crespo
- Tecnología Navarra de Nanoproductos S.L. (TECNAN), área industrial PERGUITA, C/A, N° 1, 31210 Los Arcos (Navarra), Spain.
| | - Juana Serrano-López
- Experimental Hematology Lab, IIS- Fundación Jiménez Díaz, UAM, Madrid 28040, Spain.
| | - Montserrat Colilla
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Claudio Fernández-Acevedo
- Centro Tecnológico ĹUrederra, área industrial PERGUITA, C/A, N° 1, 31210 Los Arcos (Navarra), Spain.
| | - Tamara Oroz-Mateo
- Centro Tecnológico ĹUrederra, área industrial PERGUITA, C/A, N° 1, 31210 Los Arcos (Navarra), Spain.
| | - Amaia Rada-Leza
- Centro Tecnológico ĹUrederra, área industrial PERGUITA, C/A, N° 1, 31210 Los Arcos (Navarra), Spain.
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
7
|
Shestovskaya MV, Luss AL, Bezborodova OA, Makarov VV, Keskinov AA. Iron Oxide Nanoparticles in Cancer Treatment: Cell Responses and the Potency to Improve Radiosensitivity. Pharmaceutics 2023; 15:2406. [PMID: 37896166 PMCID: PMC10610190 DOI: 10.3390/pharmaceutics15102406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The main concept of radiosensitization is making the tumor tissue more responsive to ionizing radiation, which leads to an increase in the potency of radiation therapy and allows for decreasing radiation dose and the concomitant side effects. Radiosensitization by metal oxide nanoparticles is widely discussed, but the range of mechanisms studied is not sufficiently codified and often does not reflect the ability of nanocarriers to have a specific impact on cells. This review is focused on the magnetic iron oxide nanoparticles while they occupied a special niche among the prospective radiosensitizers due to unique physicochemical characteristics and reactivity. We collected data about the possible molecular mechanisms underlying the radiosensitizing effects of iron oxide nanoparticles (IONPs) and the main approaches to increase their therapeutic efficacy by variable modifications.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anna L. Luss
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
- The Department of Technology of Chemical, Pharmaceutical and Cosmetic Products Mendeleev of University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russia
| | - Olga A. Bezborodova
- P. Hertsen Moscow Oncology Research Institute of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, 2nd Botkinskiy p. 3, Moscow 125284, Russia;
| | - Valentin V. Makarov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| | - Anton A. Keskinov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia; (A.L.L.)
| |
Collapse
|
8
|
Bai Y, Tian D, Ren Z, Yue D, Ren Q, Pei L, Pan J. The dependences of mesenchymal stem cells commitments on the size, concentration, internalization and exposure time of Iron Oxide Nanoparticles through F-actin, Lamin A and ROS. J Biomed Mater Res A 2023. [PMID: 36939155 DOI: 10.1002/jbm.a.37534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 02/21/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023]
Abstract
Though magnetic iron oxide nanoparticles (IONPs) are approved for clinical use as contrast agents for MR imaging in United States and Europe, and are widely used to label cells in research, the relationship between IONPs and mesenchymal stem cells (MSCs) is not fully addressed. Here the effects of consistently appeared γ-Fe2 O3 on the lineage commitment of MSCs were studied to optimize applications of IONPs in MSCs upon verification of viability. 30 nm 10 μg/mL induced highest promotions on osteogenesis, while 30 and 50 nm of 100 μg/mL elicited most chondrogensis in 14 days, where the effects on ALP, GAG and SOX9 appeared after 7 days, while on RUNX2 came out after 10 days. γ-Fe2 O3 enhanced intracellular and extracellular Fe3+ and ROS, modulated F-actin and decreased Lamin A of MSCs at different time scale. The disturbances of F-actin, Lamin A or ROS altered the effects of γ-Fe2 O3 on MSC differentiation. Our results demonstrate that different size, concentration and modulation of γ-Fe2 O3 are needed in its MSC applications for bone and cartilage tissues. Furthermore, an undocumented phenomenon that the modulation of F-actin affected the Lamin A expression in MSCs was observed.
Collapse
Affiliation(s)
- Yuying Bai
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Dawei Tian
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zhengxin Ren
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Dangyang Yue
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Qian Ren
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Li Pei
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jun Pan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
9
|
Efficient Liposome Loading onto Surface of Mesenchymal Stem Cells via Electrostatic Interactions for Tumor-Targeted Drug Delivery. Biomedicines 2023; 11:biomedicines11020558. [PMID: 36831094 PMCID: PMC9953681 DOI: 10.3390/biomedicines11020558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have a tumor-homing capacity; therefore, MSCs are a promising drug delivery carrier for cancer therapy. To maintain the viability and activity of MSCs, anti-cancer drugs are preferably loaded on the surface of MSCs, rather than directly introduced into MSCs. In this study, we attempted to load liposomes on the surface of MSCs by using the magnetic anionic liposome/atelocollagen complexes that we previously developed and assessed the characters of liposome-loaded MSCs as drug carriers. We observed that large-sized magnetic anionic liposome/atelocollagen complexes were abundantly associated with MSCs via electrostatic interactions under a magnetic field, and its cellular internalization was lower than that of the small-sized complexes. Moreover, the complexes with higher atelocollagen concentrations showed lower cellular internalization than the complexes with lower atelocollagen concentrations. Based on these results, we succeeded in the efficient loading of liposomes on the surface of MSCs by using large-sized magnetic anionic liposomes complexed with a high concentration of atelocollagen. The constructed liposome-loaded MSCs showed a comparable proliferation rate and differentiation potential to non-loaded MSCs. Furthermore, the liposome-loaded MSCs efficiently adhered to vascular endothelial cells and migrated toward the conditioned medium from cancer cells in vitro and solid tumor tissue in vivo. These findings suggest that liposome-loaded MSCs could serve as an efficient cell-based drug carrier for tumor-targeted delivery.
Collapse
|
10
|
Park JE, Kim WC, Kim SK, Ahn Y, Ha SM, Kim G, Choi S, Yun WS, Kong TH, Lee SH, Park DJ, Choi JS, Key J, Seo YJ. Protection of Hearing Loss in Ototoxic Mouse Model Through SPIONs and Dexamethasone-Loaded PLGA Nanoparticle Delivery by Magnetic Attraction. Int J Nanomedicine 2022; 17:6317-6334. [DOI: 10.2147/ijn.s380810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/08/2022] [Indexed: 12/14/2022] Open
|
11
|
Youden B, Jiang R, Carrier AJ, Servos MR, Zhang X. A Nanomedicine Structure-Activity Framework for Research, Development, and Regulation of Future Cancer Therapies. ACS NANO 2022; 16:17497-17551. [PMID: 36322785 DOI: 10.1021/acsnano.2c06337] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Despite their clinical success in drug delivery applications, the potential of theranostic nanomedicines is hampered by mechanistic uncertainty and a lack of science-informed regulatory guidance. Both the therapeutic efficacy and the toxicity of nanoformulations are tightly controlled by the complex interplay of the nanoparticle's physicochemical properties and the individual patient/tumor biology; however, it can be difficult to correlate such information with observed outcomes. Additionally, as nanomedicine research attempts to gradually move away from large-scale animal testing, the need for computer-assisted solutions for evaluation will increase. Such models will depend on a clear understanding of structure-activity relationships. This review provides a comprehensive overview of the field of cancer nanomedicine and provides a knowledge framework and foundational interaction maps that can facilitate future research, assessments, and regulation. By forming three complementary maps profiling nanobio interactions and pathways at different levels of biological complexity, a clear picture of a nanoparticle's journey through the body and the therapeutic and adverse consequences of each potential interaction are presented.
Collapse
Affiliation(s)
- Brian Youden
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Runqing Jiang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, Ontario N2G 1G3, Canada
| | - Andrew J Carrier
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Xu Zhang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| |
Collapse
|
12
|
Abd El-Aziz YM, Hendam BM, Al-Salmi FA, Qahl SH, Althubaiti EH, Elsaid FG, Shati AA, Hosny NM, Fayad E, Abu Almaaty AH. Ameliorative Effect of Pomegranate Peel Extract (PPE) on Hepatotoxicity Prompted by Iron Oxide Nanoparticles (Fe 2O 3-NPs) in Mice. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3074. [PMID: 36080111 PMCID: PMC9457799 DOI: 10.3390/nano12173074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/11/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
An evaluation of the ameliorative effect of pomegranate peel extract (PPE) in counteracting the toxicity of iron oxide nanoparticles (Fe2O3-NPs) that cause hepatic tissue damage is focused on herein. Forty male albino mice were haphazardly grouped into four groups as follows: the first control group was orally gavage daily with physiological saline; the second group received 100 mg/kg of PPE by the oral route day after day; the third group received 30 mg/kg Fe2O3-NPs orally; and the fourth group received both PPE and Fe2O3-NPs by the oral route, the same as the second and third sets. Later, after the completion of the experiment, we collected the liver, blood, and bone marrow of bone specimens that were obtained for further laboratory tests. For instance, exposure to Fe2O3-NPs significantly altered serum antioxidant biomarkers by decreasing the levels of total antioxidant capacity (TAC), catalase (CAT), and glutathione s-transferase (GST). Additionally, it caused changes in the morphology of hepatocytes, hepatic sinusoids, and inflammatory Kupffer cells. Furthermore, they significantly elevated the number of chromosomal aberrations including gaps, breaks, deletions, fragments, polyploidies, and ring chromosomes. Moreover, they caused a significant overexpression of TIMP-1, TNF-α, and BAX mRNA levels. Finally, the use of PPE alleviates the toxicity of Fe2O3-NPs that were induced in the hepatic tissues of mice. It is concluded that PPE extract has mitigative roles against the damage induced by Fe2O3-NPs, as it serves as an antioxidant and hepatoprotective agent. The use of PPE as a modulator of Fe2O3-NPs' hepatotoxicity could be considered as a pioneering method in the use of phytochemicals against the toxicity of nanoparticles.
Collapse
Affiliation(s)
- Yasmin M. Abd El-Aziz
- Department of Zoology, Faculty of Science, Port Said University, Port Said 42526, Egypt
| | - Basma M. Hendam
- Department of Husbandry & Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Gomhoria St., Mansoura 35516, Egypt
| | - Fawziah A. Al-Salmi
- Department of Biology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Safa H. Qahl
- Department of Biology, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Eman H. Althubaiti
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Fahmy G. Elsaid
- Biology Department, Science College, King Khalid University, Abha 61421, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Ali A. Shati
- Biology Department, Science College, King Khalid University, Abha 61421, Saudi Arabia
| | - Nasser M. Hosny
- Department of Chemistry, Faculty of Science, Port Said University, Port Said 42526, Egypt
| | - Eman Fayad
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Ali H. Abu Almaaty
- Department of Zoology, Faculty of Science, Port Said University, Port Said 42526, Egypt
| |
Collapse
|
13
|
Delivery and assessment of a CRISPR/nCas9-based genome editing system on in vitro models of mucopolysaccharidoses IVA assisted by magnetite-based nanoparticles. Sci Rep 2022; 12:15045. [PMID: 36057729 PMCID: PMC9440901 DOI: 10.1038/s41598-022-19407-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/29/2022] [Indexed: 12/27/2022] Open
Abstract
Mucopolysaccharidosis IV A (MPS IVA) is a lysosomal disorder caused by mutations in the GALNS gene. Consequently, the glycosaminoglycans (GAGs) keratan sulfate and chondroitin 6-sulfate accumulate in the lysosomal lumen. Although enzyme replacement therapy has shown essential advantages for the patients, several challenges remain to overcome, such as the limited impact on the bone lesion and recovery of oxidative profile. Recently, we validated a CRISPR/nCas9-based gene therapy with promising results in an in vitro MPS IVA model. In this study, we have expanded the use of this CRISPR/nCas9 system to several MPS IVA fibroblasts carrying different GALNS mutations. Considering the latent need to develop more safety vectors for gene therapy, we co-delivered the CRISPR/nCas9 system with a novel non-viral vector based on magnetoliposomes (MLPs). We found that the CRISPR/nCas9 treatment led to an increase in enzyme activity between 5 and 88% of wild-type levels, as well as a reduction in GAGs accumulation, lysosomal mass, and mitochondrial-dependent oxidative stress, in a mutation-dependent manner. Noteworthy, MLPs allowed to obtain similar results to those observed with the conventional transfection agent lipofectamine. Overall, these results confirmed the potential of CRISPR/nCas9 as a genome editing tool for treating MPS IVA. We also demonstrated the potential use of MLPs as a novel delivery system for CRISPR/nCas9-based therapies.
Collapse
|
14
|
Meecham A, Cutmore LC, Protopapa P, Rigby LG, Marshall JF. Ligand-bound integrin αvβ6 internalisation and trafficking. Front Cell Dev Biol 2022; 10:920303. [PMID: 36092709 PMCID: PMC9448872 DOI: 10.3389/fcell.2022.920303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The integrin αvβ6 is expressed at low levels in most normal healthy tissue but is very often upregulated in a disease context including cancer and fibrosis. Integrins use endocytosis and trafficking as a means of regulating their surface expression and thus their functions, however little is known of how this process is regulated in the context of αvβ6. As αvβ6 is a major target for the development of therapeutics in cancer and fibrosis, understanding these dynamics is critical in the development of αvβ6-targeted therapies. Following development of a flow cytometry-based assay to measure ligand (A20FMDV2 or LAP)-bound αvβ6 endocytosis, an siRNA screen was performed to identify which genes were responsible for internalising αvβ6. These data identified 15 genes (DNM2, CBLB, DNM3, CBL, EEA1, CLTC, ARFGAP3, CAV1, CYTH2, CAV3, CAV2, IQSEC1, AP2M1, TSG101) which significantly decreased endocytosis, predominantly within dynamin-dependent pathways. Inhibition of these dynamin-dependent pathways significantly reduced αvβ6-dependent migration (αvβ6-specific migration was 547 ± 128 under control conditions, reduced to 225 ± 73 with clathrin inhibition, and 280 ± 51 with caveolin inhibition). Colocalization studies of αvβ6 with endosome markers revealed that up to 6 h post-internalisation of ligand, αvβ6 remains in Rab11-positive endosomes in a perinuclear location, with no evidence of αvβ6 degradation up to 48 h post exposure to A20FMDV2. Additionally, 60% of ligand-bound αvβ6 was recycled back to the surface by 6 h. With studies ongoing using conjugated A20FMDV2 to therapeutically target αvβ6 in cancer and fibrosis, these data have important implications. Binding of A20FMDV2 seemingly removes much of the αvβ6 from the cell membrane, and upon its recycling, a large fraction appears to still be in the ligand-bound state. While these results are observed with A20FMDV2, these data will be of value in the design of αvβ6-specific therapeutics and potentially the types of therapeutic load.
Collapse
Affiliation(s)
- Amelia Meecham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- University of California, San Diego, San Diego, CA, United States
| | - Lauren C. Cutmore
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Pantelitsa Protopapa
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lauren G. Rigby
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
15
|
Peserico A, Di Berardino C, Russo V, Capacchietti G, Di Giacinto O, Canciello A, Camerano Spelta Rapini C, Barboni B. Nanotechnology-Assisted Cell Tracking. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1414. [PMID: 35564123 PMCID: PMC9103829 DOI: 10.3390/nano12091414] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023]
Abstract
The usefulness of nanoparticles (NPs) in the diagnostic and/or therapeutic sector is derived from their aptitude for navigating intra- and extracellular barriers successfully and to be spatiotemporally targeted. In this context, the optimization of NP delivery platforms is technologically related to the exploitation of the mechanisms involved in the NP-cell interaction. This review provides a detailed overview of the available technologies focusing on cell-NP interaction/detection by describing their applications in the fields of cancer and regenerative medicine. Specifically, a literature survey has been performed to analyze the key nanocarrier-impacting elements, such as NP typology and functionalization, the ability to tune cell interaction mechanisms under in vitro and in vivo conditions by framing, and at the same time, the imaging devices supporting NP delivery assessment, and consideration of their specificity and sensitivity. Although the large amount of literature information on the designs and applications of cell membrane-coated NPs has reached the extent at which it could be considered a mature branch of nanomedicine ready to be translated to the clinic, the technology applied to the biomimetic functionalization strategy of the design of NPs for directing cell labelling and intracellular retention appears less advanced. These approaches, if properly scaled up, will present diverse biomedical applications and make a positive impact on human health.
Collapse
Affiliation(s)
- Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (C.D.B.); (V.R.); (G.C.); (O.D.G.); (A.C.); (C.C.S.R.); (B.B.)
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Wang F, Li S, Cheng KW, Rosencrans WM, Chou TF. The p97 Inhibitor UPCDC-30245 Blocks Endo-Lysosomal Degradation. Pharmaceuticals (Basel) 2022; 15:ph15020204. [PMID: 35215314 PMCID: PMC8880557 DOI: 10.3390/ph15020204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
The diverse modes of action of small molecule inhibitors provide versatile tools to investigate basic biology and develop therapeutics. However, it remains a challenging task to evaluate their exact mechanisms of action. We identified two classes of inhibitors for the p97 ATPase: ATP competitive and allosteric. We showed that the allosteric p97 inhibitor, UPCDC-30245, does not affect two well-known cellular functions of p97, endoplasmic-reticulum-associated protein degradation and the unfolded protein response pathway; instead, it strongly increases the lipidated form of microtubule-associated proteins 1A/1B light chain 3B (LC3-II), suggesting an alteration of autophagic pathways. To evaluate the molecular mechanism, we performed proteomic analysis of UPCDC-30245 treated cells. Our results revealed that UPCDC-30245 blocks endo-lysosomal degradation by inhibiting the formation of early endosome and reducing the acidity of the lysosome, an effect not observed with the potent p97 inhibitor CB-5083. This unique effect allows us to demonstrate UPCDC-30245 exhibits antiviral effects against coronavirus by blocking viral entry.
Collapse
Affiliation(s)
- Feng Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; (S.L.); (K.-W.C.); (W.M.R.)
- Correspondence: (F.W.); (T.-F.C.); Tel.: +1 626-395-6772 (T.-F.C.)
| | - Shan Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; (S.L.); (K.-W.C.); (W.M.R.)
| | - Kai-Wen Cheng
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; (S.L.); (K.-W.C.); (W.M.R.)
| | - William M. Rosencrans
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; (S.L.); (K.-W.C.); (W.M.R.)
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; (S.L.); (K.-W.C.); (W.M.R.)
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
- Correspondence: (F.W.); (T.-F.C.); Tel.: +1 626-395-6772 (T.-F.C.)
| |
Collapse
|
17
|
Nistorescu S, Udrea AM, Badea MA, Lungu I, Boni M, Tozar T, Dumitrache F, Maraloiu VA, Popescu RG, Fleaca C, Andronescu E, Dinischiotu A, Staicu A, Balas M. Low Blue Dose Photodynamic Therapy with Porphyrin-Iron Oxide Nanoparticles Complexes: In Vitro Study on Human Melanoma Cells. Pharmaceutics 2021; 13:2130. [PMID: 34959411 PMCID: PMC8705854 DOI: 10.3390/pharmaceutics13122130] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 01/10/2023] Open
Abstract
The purpose of this study was to investigate the effectiveness in photodynamic therapy of iron oxide nanoparticles (γ-Fe2O3 NPs), synthesized by laser pyrolysis technique, functionalized with 5,10,15,20-(Tetra-4-sulfonatophenyl) porphyrin tetraammonium (TPPS) on human cutaneous melanoma cells, after only 1 min blue light exposure. The efficiency of porphyrin loading on the iron oxide nanocarriers was estimated by using absorption and FTIR spectroscopy. The singlet oxygen yield was determined via transient characteristics of singlet oxygen phosphorescence at 1270 nm both for porphyrin functionalized nanoparticles and rose bengal used as standard. The irradiation was performed with a LED (405 nm, 1 mW/cm2) for 1 min after melanoma cells were treated with TPPS functionalized iron oxide nanoparticles (γ-Fe2O3 NPs_TPPS) and incubated for 24 h. Biological tests revealed a high anticancer effect of γ-Fe2O3 NPs_TPPS complexes indi-cated by the inhibition of tumor cell proliferation, reduction of cell adhesion, and induction of cell death through ROS generated by TPPS under light exposure. The biological assays were combined with the pharmacokinetic prediction of the porphyrin.
Collapse
Affiliation(s)
- Simona Nistorescu
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Ana-Maria Udrea
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
- Research Institute of the University of Bucharest, Earth, Environmental and Life Sciences, Section-ICUB, 050663 Bucharest, Romania
| | - Madalina Andreea Badea
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Iulia Lungu
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Str., 011061 Bucharest, Romania;
| | - Mihai Boni
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Tatiana Tozar
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Florian Dumitrache
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | | | - Roua Gabriela Popescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Claudiu Fleaca
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Ecaterina Andronescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Str., 011061 Bucharest, Romania;
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| | - Angela Staicu
- National Institute of Laser, Plasma and Radiation Physics, 409 Atomistilor Str., 077125 Magurele, Romania; (S.N.); (A.-M.U.); (I.L.); (M.B.); (T.T.); (F.D.); (C.F.)
| | - Mihaela Balas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.A.B.); (R.G.P.); (A.D.)
| |
Collapse
|
18
|
Teng C, Li B, Lin C, Xing X, Huang F, Yang Y, Li Y, Azevedo HS, He W. Targeted delivery of baicalein-p53 complex to smooth muscle cells reverses pulmonary hypertension. J Control Release 2021; 341:591-604. [PMID: 34896449 DOI: 10.1016/j.jconrel.2021.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is an uncommon and deadly cardiopulmonary disease. PAH stems essentially from pulmonary artery (PA) remodeling induced predominantly by over-proliferation of PA smooth muscle cells (PASMCs) and inflammation. However, effective treatments are still missing in the clinic because the available drugs consisting of vasodilators are aimed to attenuate PAH symptoms rather than inhibit the remodeling process. Here, we aimed to specifically co-deliver apoptotic executor gene p53 and anti-inflammatory baicalein to PASMCs to alleviate PAH. The targeted co-delivery system was prepared through a carrier-free approach, which was prepared by loading the conjugate, NLS (nuclear localization signal) peptide-p53 gene, onto the baicalein pure crystals, followed by coating with glucuronic acid (GA) for targeting the glucose transport-1 (GLUT-1). The co-delivery system developed has a 200-nm diameter with a rod shape and a drug-loading capacity of 62% (w/w). The prepared system was shown to target PASMCs in vitro and enabled effective gene transfection, efficient apoptosis, and inflammation suppression. In vivo, via targeting the axis lung-PAs-PASMCs, the co-delivery reversed monocrotaline-induced PAH by reducing pulmonary artery pressure, downregulating the proinflammatory cytokine TNF-α, and inhibiting remodeling of both PAs and right ventricular. The potent efficacy may closely correlate with the activation of the signaling axis Bax/Bcl-2/Cas-3. Overall, our results indicate that the co-delivery system holds a significant potential to target the axis of lung-PAs-PASMCs and treat PAH.
Collapse
Affiliation(s)
- Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Bingbing Li
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Chenshi Lin
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xuyang Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Feifei Huang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Yang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Helena S Azevedo
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
19
|
Tang L, He S, Yin Y, Liu H, Hu J, Cheng J, Wang W. Combination of Nanomaterials in Cell-Based Drug Delivery Systems for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13111888. [PMID: 34834304 PMCID: PMC8621332 DOI: 10.3390/pharmaceutics13111888] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-based drug delivery systems have shown tremendous advantages in cancer treatment due to their distinctive properties. For instance, delivery of therapeutics using tumor-tropic cells like neutrophils, lymphocytes and mesenchymal stem cells can achieve specific tumor targeting due to the "Trojan Horse" effect. Other circulatory cells like erythrocytes and platelets can greatly improve the circulation time of nanoparticles due to their innate long circulation property. Adipocytes, especially cancer-associated adipocytes, play key roles in tumor development and metabolism, therefore, adipocytes are regarded as promising bio-derived nanoplatforms for anticancer targeted drug delivery. Nanomaterials are important participants in cell-based drug delivery because of their unique physicochemical characteristics. Therefore, the integration of various nanomaterials with different cell types will endow the constructed delivery systems with many attractive properties due to the merits of both. In this review, a number of strategies based on nanomaterial-involved cell-mediated drug delivery systems for cancer treatment will be summarized. This review discusses how nanomaterials can be a benefit to cell-based therapies and how cell-derived carriers overcome the limitations of nanomaterials, which highlights recent advancements and specific biomedical applications based on nanomaterial-mediated, cell-based drug delivery systems.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyi Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 211166, China
- Correspondence: (J.C.); (W.W.)
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
- Correspondence: (J.C.); (W.W.)
| |
Collapse
|
20
|
Wei H, Hu Y, Wang J, Gao X, Qian X, Tang M. Superparamagnetic Iron Oxide Nanoparticles: Cytotoxicity, Metabolism, and Cellular Behavior in Biomedicine Applications. Int J Nanomedicine 2021; 16:6097-6113. [PMID: 34511908 PMCID: PMC8418330 DOI: 10.2147/ijn.s321984] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely investigated and applied in the field of biomedicine due to their excellent superparamagnetic properties and reliable traceability. However, with the optimization of core composition, shell types and transfection agents, the cytotoxicity and metabolism of different SPIONs have great differences, and the labeled cells also show different cellular behaviors. Therefore, a holistic review of the construction and application of SPIONs is desired. This review focuses the advances of SPIONs in the field of biomedicine in recent years. After summarizing the toxicity of different SPIONs, the uptake, distribution and metabolism of SPIONs in vitro were discussed. Then, the regulation of labeled-cells behavior is outlined. Furthermore, the major challenges in the optimization process of SPIONs and insights on its future developments are proposed.
Collapse
Affiliation(s)
- Hao Wei
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Yangnan Hu
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China
| | - Junguo Wang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Xia Gao
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Xiaoyun Qian
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Mingliang Tang
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China.,Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, 215000, People's Republic of China
| |
Collapse
|
21
|
Modification of chemically and physically obtained Fe3O4 magnetic nanoparticles with l-Lys for cell labeling. Russ Chem Bull 2021. [DOI: 10.1007/s11172-021-3205-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
Huang H, Du X, He Z, Yan Z, Han W. Nanoparticles for Stem Cell Tracking and the Potential Treatment of Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:662406. [PMID: 34277609 PMCID: PMC8283769 DOI: 10.3389/fcell.2021.662406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/12/2021] [Indexed: 01/15/2023] Open
Abstract
Stem cell-based therapies have been shown potential in regenerative medicine. In these cells, mesenchymal stem cells (MSCs) have the ability of self-renewal and being differentiated into different types of cells, such as cardiovascular cells. Moreover, MSCs have low immunogenicity and immunomodulatory properties, and can protect the myocardium, which are ideal qualities for cardiovascular repair. Transplanting mesenchymal stem cells has demonstrated improved outcomes for treating cardiovascular diseases in preclinical trials. However, there still are some challenges, such as their low rate of migration to the ischemic myocardium, low tissue retention, and low survival rate after the transplantation. To solve these problems, an ideal method should be developed to precisely and quantitatively monitor the viability of the transplanted cells in vivo for providing the guidance of clinical translation. Cell imaging is an ideal method, but requires a suitable contrast agent to label and track the cells. This article reviews the uses of nanoparticles as contrast agents for tracking MSCs and the challenges of clinical use of MSCs in the potential treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Huihua Huang
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Health Science Center, Shenzhen, China
| | - Xuejun Du
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Zhiguo He
- Advanced Materials Institute, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Zifeng Yan
- Advanced Materials Institute, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Wei Han
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
23
|
Wu MR, Lee CH, Hsiao JK. Bidirectional Enhancement of Cell Proliferation Between Iron Oxide Nanoparticle-Labeled Mesenchymal Stem Cells and Choroid Plexus in a Cell-Based Therapy Model of Ischemic Stroke. Int J Nanomedicine 2020; 15:9181-9195. [PMID: 33239875 PMCID: PMC7682617 DOI: 10.2147/ijn.s278687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/14/2020] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Stem cell therapy for ischemic stroke has shown success in experimental settings, but its translation into clinical practice is challenging. The choroid plexus (CP) plays a regulatory role in neural regeneration. Mesenchymal stem cells (MSCs) promote neurogenesis in the ventricular-subventricular zone. However, it is unclear whether MSCs interact with the CP in brain tissue repair. METHODS Rat (r)MSCs were labeled with iron oxide nanoparticles (IONs) and transduced with red fluorescent protein, and then injected into the brain of rats with ischemic stroke and monitored over time by magnetic resonance imaging. The functional recovery of rats was determined by the corner test score, Modified Neurological Severity score, and stroke volume. MSCs and CP were also co-cultured for 14 days, and the medium was analyzed with a cytokine array. RESULTS In vivo imaging and histologic analysis revealed that ION-labeled MSCs were mainly located at the injection site and migrated to the infarct area and to the CP. Functional recovery was greater in rats treated with MSCs as compared to those that received mock treatment. Bidirectional enhancement of proliferation in MSCs and CP was observed in the co-culture; moreover, MSCs migrated to the CP. Cytokine analysis revealed elevated levels of proliferation- and adhesion-related cytokines and chemokines in the culture medium. Wikipathway predictions indicated that insulin-like growth factor 1/Akt signaling (WP3675), chemokine signaling pathway (WP2292), and spinal cord injury (WP2432) are involved in the increased proliferation and migration of MSCs co-cultured with the CP. CONCLUSION Crosstalk with the CP enhances MSC proliferation and migration in a transwell assay. Moreover, MRI reveals MSC migration towards the CP in an ischemic stroke model. The secreted factors resulting from this interaction have therapeutic potential for promoting functional recovery in the brain after ischemic stroke.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Department of Medical Imaging, Taipei Tzuchi Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City23142, Taiwan
- Institute of Biomedical Engineering, National Taiwan University, Taipei10617, Taiwan
| | - Chia-Hsun Lee
- Department of Medical Imaging, Taipei Tzuchi Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City23142, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzuchi Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City23142, Taiwan
- School of Medicine, Tzu Chi University, Hualien97004, Taiwan
| |
Collapse
|