1
|
Guerrero-Florez V, Barbara A, Kodjikian S, Oukacine F, Trens P, Cattoën X. Dynamic light scattering unveils stochastic degradation in large-pore mesoporous silica nanoparticles. J Colloid Interface Sci 2024; 676:1098-1108. [PMID: 39079273 DOI: 10.1016/j.jcis.2024.07.151] [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: 04/25/2024] [Revised: 07/02/2024] [Accepted: 07/19/2024] [Indexed: 09/19/2024]
Abstract
Mesoporous Silica Nanoparticles (MSNs) have been increasingly investigated as versatile drug delivery carriers. A particular challenge for the systemic use of MSNs lies in the control of their degradation, which has not been fully understood until now. We implemented standard dynamic light scattering (DLS) experiments and introduced a novel DLS technique in a confocal volume to track the dynamics of large-pore MSN degradation in situ. This unique DLS technique, which involves a small observation volume, was chosen for its ability to count particle by particle during the degradation process, a method that has not been commonly used in nanoparticle research. The experiments were performed in different media compositions at low particle concentrations, below the silica solubility limit. MSNs with large conical pores were prepared and studied as they offer the possibility to incorporate and release large-sized biomolecules. Large-pore MSNs followed a singular degradation mechanism following a stochastic-like behavior, a finding that challenges the common idea that all nanoparticles (NPs) degrade similarly and homogeneously over time. We showed that some NPs are observed intact over a prolonged period while most other NPs have already vanished or been transformed into swollen NPs. Thus, a heterogeneous degradation process occurs, while the total concentration of NPs undergoes an exponential decay. These large conical pores MSNs will be utilized as reliable biomolecule nanocarriers by predicting the factors underlying the NP hydrolytic stability.
Collapse
Affiliation(s)
| | - Aude Barbara
- Univ. Grenoble Alpes, CNRS, Grenoble INP, Institut Néel, 38000 Grenoble, France
| | - Stéphanie Kodjikian
- Univ. Grenoble Alpes, CNRS, Grenoble INP, Institut Néel, 38000 Grenoble, France
| | - Farid Oukacine
- Univ. Grenoble Alpes, DPM, CNRS UMR5063, F-38041 Grenoble, France
| | - Philippe Trens
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Xavier Cattoën
- Univ. Grenoble Alpes, CNRS, Grenoble INP, Institut Néel, 38000 Grenoble, France.
| |
Collapse
|
2
|
Heidari R, Assadollahi V, Shakib Manesh MH, Mirzaei SA, Elahian F. Recent advances in mesoporous silica nanoparticles formulations and drug delivery for wound healing. Int J Pharm 2024; 665:124654. [PMID: 39244073 DOI: 10.1016/j.ijpharm.2024.124654] [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: 06/22/2024] [Revised: 08/15/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Wound healing is a natural process that can be disrupted by disease. Nanotechnology is a promising platform for the development of new therapeutic agents to accelerate acute and chronic wound healing. Drug delivery by means of nanoparticles as well as wound dressings have emerged as suitable options to improving the healing process. The characteristics of mesoporous silica nanoparticles (MSNs) make them efficient carriers of pharmaceutical agents alone or in combination with dressings. In order to maximize the effect of a drug and minimize its adverse consequences, it may be possible to include targeted and intelligent release of the drug into the design of MSNs. Its use to facilitate closure of adjacent sides of a cut as a tissue adhesive, local wound healing, controlled drug release and induction of blood coagulation are possible applications of MSNs. This review summarizes research on MSN applications for wound healing. It includes a general overview, wound healing phases, MSN formulation, therapeutic possibilities of MSNs and MSN-based drug delivery systems for wound healing.
Collapse
Affiliation(s)
- Razieh Heidari
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vahideh Assadollahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Hossein Shakib Manesh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran; Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Ren M, Abdullah SW, Pei C, Guo H, Sun S. Use of virus-like particles and nanoparticle-based vaccines for combating picornavirus infections. Vet Res 2024; 55:128. [PMID: 39350170 PMCID: PMC11443892 DOI: 10.1186/s13567-024-01383-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
Picornaviridae are non-enveloped ssRNA viruses that cause diseases such as poliomyelitis, hand-foot-and-mouth disease (HFMD), hepatitis A, encephalitis, myocarditis, and foot-and-mouth disease (FMD). Virus-like particles (VLPs) vaccines mainly comprise particles formed through the self-assembly of viral capsid proteins (for enveloped viruses, envelope proteins are also an option). They do not contain the viral genome. On the other hand, the nanoparticles vaccine (NPs) is mainly composed of self-assembling biological proteins or nanomaterials, with viral antigens displayed on the surface. The presentation of viral antigens on these particles in a repetitive array can elicit a strong immune response in animals. VLPs and NPs can be powerful platforms for multivalent antigen presentation. This review summarises the development of virus-like particle vaccines (VLPs) and nanoparticle vaccines (NPs) against picornaviruses. By detailing the progress made in the fight against various picornaviruses such as poliovirus (PV), foot-and-mouth disease virus (FMDV), enterovirus (EV), Senecavirus A (SVA), and encephalomyocarditis virus (EMCV), we in turn highlight the significant strides made in vaccine technology. These advancements include diverse construction methods, expression systems, elicited immune responses, and the use of various adjuvants. We see promising prospects for the continued development and optimisation of VLPs and NPs vaccines. Future research should focus on enhancing these vaccines' immunogenicity, stability, and delivery methods. Moreover, expanding our understanding of the interplay between these vaccines and the immune system will be crucial. We hope these insights will inspire and guide fellow researchers in the ongoing quest to combat picornavirus infections more effectively.
Collapse
Affiliation(s)
- Mei Ren
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gembloux Agro-Biotech, University of Liege, Gembloux, Belgium
| | - Sahibzada Waheed Abdullah
- Livestock and dairy development department peshawar, Government of Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Mohan S, Thankaswamy J. Synthesis and characterization of piperine-modified mesoporous silica nanoparticles for biomedical applications. Biotechnol Appl Biochem 2024. [PMID: 39300710 DOI: 10.1002/bab.2672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
Mesoporous silica nanoparticles (MSNs) have displayed high-potential prospects in biomedical use, especially for drug delivery due to large surface area, tunable pore size and simple surface functionalization. The objective behind the present research is to synthesize and profile piperine-modified MSNs for their preparation due to antioxidative anticarcinogenic, anti-inflammatory properties of the alkaloid chosen as a modifier. In the study, silica piperine nanoparticles (SPN) were fabricated based on a modified Stöber method. Characterization techniques including SEM, TEM, AFM, FTIR, XRD, and DSC showed significant differences of incorporated piperine in the production process to plain MSN properties. Piperine was observed to inhibit nanoparticles' growth so that they became smaller, heterogeneous, with a changed morphology and surface chemistry. As a strong confirmation of covalent incorporation, spectroscopic data showed the presence of electrons in the piperine's functional group that were exchanged into some silanol groups and removed excessive surface energy. The antioxidant activity of SPNs revealed that the silica matrix, and moreover bioactive piperine combination resulted to significant increase in enhanced antioxidant potential. In general, the results of this study offer meaningful lessons about the utilization and manipulation of piperine to suit MSN in a bid to optimize them for biomedical uses such as drug delivery applications where its antioxidant characteristics may bring therapeutic benefits. This holistic characterization and standardization of piperine-modified MSNs sets the solid stage for further project practice and advance adjustment in aluminosilicate nanostructures designed for biomedical application.
Collapse
Affiliation(s)
- Shimi Mohan
- Department of Biomedical Engineering, Musaliar College of Engineering and Polytechnic, APJ Abdul Kalam Technological University, Kerala, India
| | - Jarin Thankaswamy
- Department of Electrical and Electronics Engineering, Jyothi Engineering College, APJ Abdul Kalam Technological University, Thrissur, Kerala, India
| |
Collapse
|
5
|
Bourang S, Noruzpour M, Jahanbakhsh Godekahriz S, Ebrahimi HAC, Amani A, Asghari Zakaria R, Yaghoubi H. Application of nanoparticles in breast cancer treatment: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6459-6505. [PMID: 38700795 DOI: 10.1007/s00210-024-03082-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 09/25/2024]
Abstract
It is estimated that cancer is the second leading cause of death worldwide. The primary or secondary cause of cancer-related mortality for women is breast cancer. The main treatment method for different types of cancer is chemotherapy with drugs. Because of less water solubility of chemotherapy drugs or their inability to pass through membranes, their body absorbs them inadequately, which lowers the treatment's effectiveness. Drug specificity and pharmacokinetics can be changed by nanotechnology using nanoparticles. Instead, targeted drug delivery allows medications to be delivered to the targeted sites. In this review, we focused on nanoparticles as carriers in targeted drug delivery, their characteristics, structure, and the previous studies related to breast cancer. It was shown that nanoparticles could reduce the negative effects of chemotherapy drugs while increasing their effectiveness. Lipid-based nanocarriers demonstrated notable results in this instance, and some products that are undergoing various stages of clinical trials are among the examples. Nanoparticles based on metal or polymers demonstrated a comparable level of efficacy. With the number of cancer cases rising globally, many researchers are now looking into novel treatment approaches, particularly the use of nanotechnology and nanoparticles in the treatment of cancer. In order to help clinicians, this article aimed to gather more information about various areas of nanoparticle application in breast cancer therapy, such as modifying their synthesis and physicochemical characterization. It also sought to gain a deeper understanding of the mechanisms underlying the interactions between nanoparticles and biologically normal or infected tissues.
Collapse
Affiliation(s)
- Shima Bourang
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Sodabeh Jahanbakhsh Godekahriz
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ca Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Amin Amani
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hashem Yaghoubi
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
6
|
Mohanan S, Guan X, Liang M, Karakoti A, Vinu A. Stimuli-Responsive Silica Silanol Conjugates: Strategic Nanoarchitectonics in Targeted Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301113. [PMID: 36967548 DOI: 10.1002/smll.202301113] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The design of novel drug delivery systems is exceptionally critical in disease treatments. Among the existing drug delivery systems, mesoporous silica nanoparticles (MSNs) have shown profuse promise owing to their structural stability, tunable morphologies/sizes, and ability to load different payload chemistry. Significantly, the presence of surface silanol groups enables functionalization with relevant drugs, imaging, and targeting agents, promoting their utility and popularity among researchers. Stimuli-responsive silanol conjugates have been developed as a novel, more effective way to conjugate, deliver, and release therapeutic drugs on demand and precisely to the selected location. Therefore, it is urgent to summarize the current understanding and the surface silanols' role in making MSN a versatile drug delivery platform. This review provides an analytical understanding of the surface silanols, chemistry, identification methods, and their property-performance correlation. The chemistry involved in converting surface silanols to a stimuli-responsive silica delivery system by endogenous/exogenous stimuli, including pH, redox potential, temperature, and hypoxia, is discussed in depth. Different chemistries for converting surface silanols to stimuli-responsive bonds are discussed in the context of drug delivery. The critical discussion is culminated by outlining the challenges in identifying silanols' role and overcoming the limitations in synthesizing stimuli-responsive mesoporous silica-based drug delivery systems.
Collapse
Affiliation(s)
- Shan Mohanan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Xinwei Guan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajay Karakoti
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| |
Collapse
|
7
|
Liu B, Zheng Q, Shi X, Shen J, Li R, Zhou J. Construction and synergistic anti-tumor study of a tumor microenvironment-based multifunctional nano-drug delivery system. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112977. [PMID: 38991294 DOI: 10.1016/j.jphotobiol.2024.112977] [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: 02/02/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
To solve the problems existing in the clinical application of hypericin (Hyp) and tirapazamine (TPZ), a nano-drug delivery system with synergistic anti-tumor functions was constructed using mesoporous silica nanoparticles (MSN) and sodium alginate (SA). The system exhibited excellent stability, physiological compatibility and targeted drug release performance in tumor tissues. In the in vitro and in vivo experiments, Hyp released from MSN killed tumor cells through photodynamic therapy (PDT). The degree of hypoxia in the tumor tissue site was exacerbated, enabling TPZ to fully exert its anti-tumor activity. Our studies suggested that the synergistic effects between the components of the nano-drug delivery system significantly improve the anti-tumor properties of Hyp and TPZ.
Collapse
Affiliation(s)
- Baoqing Liu
- College of Life Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Qinghua Zheng
- College of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, PR China
| | - Xiandong Shi
- College of Life Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Jian Shen
- College of Chemistry and Material Science, Nanjing Normal University, Nanjing 210023, PR China.
| | - Ruyan Li
- College of Life Science, Nanjing Normal University, Nanjing 210023, PR China.
| | - Jiahong Zhou
- College of Life Science, Nanjing Normal University, Nanjing 210023, PR China.
| |
Collapse
|
8
|
Samrout OE, Berlier G, Lambert JF. Amino Acid Polymerization on Silica Surfaces. Chempluschem 2024; 89:e202300642. [PMID: 38226922 DOI: 10.1002/cplu.202300642] [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: 11/11/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/17/2024]
Abstract
The polymerization of unactivated amino acids (AAs) is an important topic because of its applications in various fields including industrial medicinal chemistry and prebiotic chemistry. Silica as a promoter for this reaction, is of great interest owing to its large abundance and low cost. The amide/peptide bond synthesis on silica has been largely demonstrated but suffers from a lack of knowledge regarding its reaction mechanism, the key parameters, and surface features that influence AA adsorption and reactivity, the selectivity of the reaction product, the role of water in the reaction, etc. The present review addresses these problems by summarizing experimental and modeling results from the literature and attempts to rationalize some apparent divergences in published results. After briefly presenting the main types of silica surface sites and other relevant macroscopic features, we discuss the different deposition procedures of AAs, whose importance is often neglected. We address the possible AA adsorption mechanisms including covalent grafting and H-bonding and show that they are highly dependent on silanol types and density. We then consider how the adsorption mechanisms determine the occurrence and outcome of AA condensation (formation of cyclic dimers or of long linear chains), and outline some recent results that suggest significant polymerization selectivity in systems containing several AAs, as well as the formation of specific elements of secondary structure in the growing polypeptide chains.
Collapse
Affiliation(s)
- Ola El Samrout
- Department of Chemistry, University of Torino, Via P. Giuria 7, 10125, Torino, Italy
| | - Gloria Berlier
- Department of Chemistry, University of Torino, Via P. Giuria 7, 10125, Torino, Italy
| | - Jean-François Lambert
- Laboratoire de Réactivité de Surface, LRS, Sorbonne Université Place Jussieu, 75005, Paris, France
| |
Collapse
|
9
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [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: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
10
|
Ibrahim IAA, Alzahrani AR, Alanazi IM, Shahzad N, Shahid I, Falemban AH, Azlina MFN, Arulselvan P. Carbohydrate polymers-based surface modified nano delivery systems for enhanced target delivery to colon cancer - A review. Int J Biol Macromol 2023; 253:126581. [PMID: 37652322 DOI: 10.1016/j.ijbiomac.2023.126581] [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: 05/29/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Carbohydrate polymers-based surface-modified nano-delivery systems have gained significant attention in recent years for enhancing targeted delivery to colon cancer. These systems leverage carbohydrate polymers' unique properties, such as biocompatibility, biodegradability, and controlled release. These properties make them suitable candidates for drug delivery applications. Nano-delivery systems loaded with bioactive compounds are well-studied for targeted colorectal cancer delivery. However, those drugs' target reach is still limited in various nano-delivery systems. To overcome this limitation, surface modification of nanoparticles with carbohydrate polymers like chitosan, pectin, alginate, and guar gum showed enhanced target-reaching capacity along with enhanced anticancer efficacy. Recently, a chitosan-decorated PLGA nanoparticle was constructed with tannic acid and vitamin E and showed long-term release of specific targets along with higher anticancer efficacy. Similarly, Chitosan-conjugated glucuronic acid-coated silica nanoparticles loaded with capecitabine were studied against colon cancer and found to be the pH-responsive controlled release of capecitabine with higher anticancer efficacy. Surface-modified carbohydrate polymers have promising potential for improving colon cancer target delivery. By leveraging the unique properties of these polymers, such as surface modification, pH responsiveness, mucoadhesion, controlled drug release, and combination therapy, researchers are working toward developing more effective and targeted treatment strategies for colon cancer.
Collapse
Affiliation(s)
- Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ibrahim M Alanazi
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naiyer Shahzad
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Shahid
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Alaa Hisham Falemban
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohd Fahami Nur Azlina
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Malaysia
| | - Palanisamy Arulselvan
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, 602 105, India
| |
Collapse
|
11
|
Uskoković V, Abuna G, Hampton JR, Geraldeli S. Tunable Release of Calcium from Chitosan-Coated Bioglass. Pharmaceutics 2023; 16:39. [PMID: 38258050 PMCID: PMC10818729 DOI: 10.3390/pharmaceutics16010039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Bioglass presents a standard biomaterial for regeneration of hard tissues in orthopedics and dentistry. The notable osteo-inductive properties of bioglass are largely due to the release of calcium ions from it. However, this release is not easily controllable and can often be excessive, especially during the initial interaction of the biomaterial with the surrounding tissues. Consequently, this excessive release can deplete the calcium content of the bioglass, ultimately reducing its overall bioactivity. In this study, we have tested if applying biopolymer chitosan coatings of different thicknesses would be able to mitigate and regulate the calcium ion release from monodisperse bioglass nanoparticles. Calcium release was assessed for four different chitosan coating thicknesses at different time points over the period of 28 days using a fluorescence quencher. Expectedly, chitosan-coated particles released less calcium as the concentration of chitosan in the coating solution increased, presumably due to the increased thickness of the chitosan coating around the bioglass particles. The mechanism of release remained constant for each coating thickness, corresponding to anomalous, non-Fickian diffusion, but the degree of anomalousness increased with the deposition of chitosan. Zeta potential testing showed an expected increase in the positive double layer charge following the deposition of the chitosan coating due to the surface exposure of the amine groups of chitosan. Less intuitively, the zeta potential became less positive as thickness of the chitosan coating increased, attesting to the lower density of the surface charges within thicker coatings than within the thinner ones. Overall, the findings of this study demonstrate that chitosan coating efficiently prevents the early release of calcium from bioglass. This coating procedure also allows for the tuning of the calcium release kinetics by controlling the chitosan concentration in the parent solution.
Collapse
Affiliation(s)
- Vuk Uskoković
- TardigradeNano LLC, 7 Park Vista, Irvine, CA 92604, USA
- Department of Mechanical Engineering, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Gabriel Abuna
- School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Rd, Greenville, NC 27834, USA; (G.A.); (J.R.H.)
| | - Joseph Ryan Hampton
- School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Rd, Greenville, NC 27834, USA; (G.A.); (J.R.H.)
| | - Saulo Geraldeli
- School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Rd, Greenville, NC 27834, USA; (G.A.); (J.R.H.)
| |
Collapse
|
12
|
Alvarez-Lorenzo C, Zarur M, Seijo-Rabina A, Blanco-Fernandez B, Rodríguez-Moldes I, Concheiro A. Physical stimuli-emitting scaffolds: The role of piezoelectricity in tissue regeneration. Mater Today Bio 2023; 22:100740. [PMID: 37521523 PMCID: PMC10374602 DOI: 10.1016/j.mtbio.2023.100740] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023] Open
Abstract
The imbalance between life expectancy and quality of life is increasing due to the raising prevalence of chronic diseases. Musculoskeletal disorders and chronic wounds affect a growing percentage of people and demand more efficient tools for regenerative medicine. Scaffolds that can better mimic the natural physical stimuli that tissues receive under healthy conditions and during healing may significantly aid the regeneration process. Shape, mechanical properties, pore size and interconnectivity have already been demonstrated to be relevant scaffold features that can determine cell adhesion and differentiation. Much less attention has been paid to scaffolds that can deliver more dynamic physical stimuli, such as electrical signals. Recent developments in the precise measurement of electrical fields in vivo have revealed their key role in cell movement (galvanotaxis), growth, activation of secondary cascades, and differentiation to different lineages in a variety of tissues, not just neural. Piezoelectric scaffolds can mimic the natural bioelectric potentials and gradients in an autonomous way by generating the electric stimuli themselves when subjected to mechanical loads or, if the patient or the tissue lacks mobility, ultrasound irradiation. This review provides an analysis on endogenous bioelectrical signals, recent developments on piezoelectric scaffolds for bone, cartilage, tendon and nerve regeneration, and their main outcomes in vivo. Wound healing with piezoelectric dressings is addressed in the last section with relevant examples of performance in animal models. Results evidence that a fine adjustment of material composition and processing (electrospinning, corona poling, 3D printing, annealing) provides scaffolds that act as true emitters of electrical stimuli that activate endogenous signaling pathways for more efficient and long-term tissue repair.
Collapse
Affiliation(s)
- Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Mariana Zarur
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Alejandro Seijo-Rabina
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Barbara Blanco-Fernandez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Isabel Rodríguez-Moldes
- Grupo NEURODEVO, Departamento de Bioloxía Funcional, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
13
|
Noureddine A, Maestas-Olguin A, Tang L, Corman-Hijar JI, Olewine M, Krawchuck JA, Tsala Ebode J, Edeh C, Dang C, Negrete OA, Watt J, Howard T, Coker EN, Guo J, Brinker CJ. Future of Mesoporous Silica Nanoparticles in Nanomedicine: Protocol for Reproducible Synthesis, Characterization, Lipid Coating, and Loading of Therapeutics (Chemotherapeutic, Proteins, siRNA and mRNA). ACS NANO 2023; 17:16308-16325. [PMID: 37643407 DOI: 10.1021/acsnano.3c07621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Owing to their uniform and tunable particle size, pore size, and shape, along with their modular surface chemistry and biocompatibility, mesoporous silica nanoparticles (MSNs) have found extensive applications as nanocarriers to deliver therapeutic, diagnostic and combined "theranostic" cargos to cells and tissues. Although thoroughly investigated, MSN have garnered FDA approval for only one MSN system via oral administration. One possible reason is that there is no recognized, reproducible, and widely adopted MSN synthetic protocol, meaning not all MSNs are created equal in the laboratory nor in the eyes of the FDA. This manuscript provides the sol-gel and MSN research communities a reproducible, fully characterized synthetic protocol to synthesize MSNs and corresponding lipid-coated MSN delivery vehicles with predetermined particle size, pore size, and drug loading and release characteristics. By carefully articulating the step-by-step synthetic procedures and highlighting critical points and troubleshooting, augmented with videos and schematics, this Article will help researchers entering this rapidly expanding field to yield reliable results.
Collapse
Affiliation(s)
- Achraf Noureddine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Angelea Maestas-Olguin
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Lien Tang
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, United States
| | - Jim I Corman-Hijar
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- Biomedical Engineering Department, Pontifical Catholic University of Peru, San Miguel 15088, Peru
| | - Marian Olewine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Jacob A Krawchuck
- Center for Integrated Nanotechnologies, Sandia National Laboratories, Albuquerque, New Mexico 87123, United States
| | - Johanna Tsala Ebode
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Chuzube Edeh
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Caleb Dang
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Oscar A Negrete
- Systems Biology Department, Sandia National Laboratories, Livermore, California 94550 United States
| | - John Watt
- Center for Integrated Nanotechnologies, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Tamara Howard
- Department of Cell Biology & Physiology, UNM School of Medicine, Albuquerque, New Mexico 87131, United States
| | - Eric N Coker
- Electronic, Optical, and Nano Materials, Sandia National Laboratories, Albuquerque, New Mexico 87185, United States
| | - Jimin Guo
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - C Jeffrey Brinker
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
14
|
Li B, Liao Y, Su X, Chen S, Wang X, Shen B, Song H, Yue P. Powering mesoporous silica nanoparticles into bioactive nanoplatforms for antibacterial therapies: strategies and challenges. J Nanobiotechnology 2023; 21:325. [PMID: 37684605 PMCID: PMC10485977 DOI: 10.1186/s12951-023-02093-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
Bacterial infection has been a major threat to worldwide human health, in particular with the ever-increasing level of antimicrobial resistance. Given the complex microenvironment of bacterial infections, conventional use of antibiotics typically renders a low efficacy in infection control, thus calling for novel strategies for effective antibacterial therapies. As an excellent candidate for antibiotics delivery, mesoporous silica nanoparticles (MSNs) demonstrate unique physicochemical advantages in antibacterial therapies. Beyond the delivery capability, extensive efforts have been devoted in engineering MSNs to be bioactive to further synergize the therapeutic effect in infection control. In this review, we critically reviewed the essential properties of MSNs that benefit their antibacterial application, followed by a themed summary of strategies in manipulating MSNs into bioactive nanoplatforms for enhanced antibacterial therapies. The chemically functionalized platform, photo-synergized platform, physical antibacterial platform and targeting-directed platform are introduced in details, where the clinical translation challenges of these MSNs-based antibacterial nanoplatforms are briefly discussed afterwards. This review provides critical information of the emerging trend in turning bioinert MSNs into bioactive antibacterial agents, paving the way to inspire and translate novel MSNs-based nanotherapies in combating bacterial infection diseases.
Collapse
Affiliation(s)
- Biao Li
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China
| | - Yan Liao
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China
| | - Xiaoyu Su
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China
| | - Shuiyan Chen
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China
| | - Xinmin Wang
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China
| | - Baode Shen
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Pengfei Yue
- Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 MEILING Avenue, Nanchang, 330004, China.
| |
Collapse
|
15
|
Iravani S. Silica-based nanosystems against antibiotic-resistant bacteria and pathogenic viruses. Crit Rev Microbiol 2023; 49:598-610. [PMID: 35930235 DOI: 10.1080/1040841x.2022.2108309] [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: 04/08/2022] [Revised: 06/19/2022] [Accepted: 07/27/2022] [Indexed: 11/03/2022]
Abstract
Today, with the intensity of antibiotic abuse and self-medication, the need for the use of novel systems with high efficiency and biosafety for targeted drug delivery against antibiotic-resistant bacteria and their infections should be highly considered by researchers. Silica-based nanosystems with unique physicochemical properties such as large surface area, tuneable pore diameter, drug loading capacity, controlled particle size/morphology, and good biocompatibility are attractive candidates against antibiotic-resistant bacteria and pathogenic viruses. They can be loaded with antiviral and antimicrobial drugs or molecules through their exclusive internal porous structures or different surface linkers. In this context, smart nanosystems can be produced via suitable surface functionalization/modification with a variety of functional groups to act against different clinical pathogenic microbes or viruses, offering great opportunities for controlling and treating various infections. However, important criteria such as the ability to degrade, biocompatibility, biodegradability, cytotoxicity, stability, clearance from targeted organs should be systematically analysed to develop nanosystems or nanocarriers with high efficiency and multifunctionality. Herein, recent advancements pertaining to the application of silica-based nanosystems against antibiotic-resistant bacteria and pathogenic viruses are deliberated, focussing on important challenges and future perspectives.
Collapse
Affiliation(s)
- Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Ahmed HA, Elsohaby I, Elamin AM, El-Ghafar AEA, Elsaid GA, Elbarbary M, Mohsen RA, El Feky TM, El Bayomi RM. Extended-spectrum β-lactamase-producing E. coli from retail meat and workers: genetic diversity, virulotyping, pathotyping and the antimicrobial effect of silver nanoparticles. BMC Microbiol 2023; 23:212. [PMID: 37550643 PMCID: PMC10405496 DOI: 10.1186/s12866-023-02948-0] [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: 04/23/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND The spread of extended-spectrum β-lactamases (ESBL) producing E. coli from food animals and the environment to humans has become a significant public health concern. The objectives of this study were to determine the occurrence, pathotypes, virulotypes, genotypes, and antimicrobial resistance patterns of ESBL-producing E. coli in retail meat samples and workers in retail meat shops in Egypt and to evaluate the bactericidal efficacy of silver nanoparticles (AgNPs-H2O2) against multidrug resistant (MDR) ESBL-producing E. coli. RESULTS A total of 250 retail meat samples and 100 human worker samples (hand swabs and stool) were examined for the presence of ESBL- producing E. coli. Duck meat and workers' hand swabs were the highest proportion of ESBL- producing E. coli isolates (81.1%), followed by camel meat (61.5%). Pathotyping revealed that the isolates belonged to groups A and B1. Virulotyping showed that the most prevalent virulence gene was Shiga toxin 2 (stx2) associated gene (36.9%), while none of the isolates harbored stx1 gene. Genotyping of the identified isolates from human and meat sources by REP-PCR showed 100% similarity within the same cluster between human and meat isolates. All isolates were classified as MDR with an average multiple antibiotic resistance (MAR) index of 0.7. AgNPs-H2O2 at concentrations of 0.625, 1.25, 2.5 and 5 μg/mL showed complete bacterial growth inhibition. CONCLUSIONS Virulent MDR ESBL-producing E. coli were identified in retail meat products in Egypt, posing significant public health threats. Regular monitoring of ESBL-producing E. coli frequency and antimicrobial resistance profile in retail meat products is crucial to enhance their safety. AgNPs-H2O2 is a promising alternative for treating MDR ESBL-producing E. coli infections and reducing antimicrobial resistance risks.
Collapse
Affiliation(s)
- Heba A Ahmed
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig City, 44511, Sharkia Governorate, Egypt.
| | - Ibrahim Elsohaby
- Department of Infectious Diseases and Public Health, Jockey Club of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
- Centre for Applied One Health Research and Policy Advice (OHRP), City University of Hong Kong, Hong Kong SAR, China
- Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig City, 44511, Sharkia Governorate, Egypt
| | - Amina M Elamin
- Department of Food Hygiene, Zagazig Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig City, Egypt
| | - Abeer E Abd El-Ghafar
- Department of Bacteriology, Mansoura Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Mansoura City, Egypt
| | - Gamilat A Elsaid
- Department of Food Hygiene, Mansoura Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Mansoura City, Egypt
| | - Mervat Elbarbary
- Department of Food Hygiene, Zagazig Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig City, Egypt
| | - Rasha A Mohsen
- Department of Bacteriology, Mansoura Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Mansoura City, Egypt
| | - Tamer M El Feky
- Department of Bacteriology, Mansoura Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Mansoura City, Egypt
| | - Rasha M El Bayomi
- Department of Food Control, Faculty of Veterinary Medicine, Zagazig University, Zagazig City, 44511, Sharkia Governorate, Egypt
| |
Collapse
|
17
|
Omar H, Jakimoska S, Guillot J, Alsharaeh E, Charnay C, Cunin F, Bessière A, Durand JO, Raehm L, Lichon L, Onofre M, Gary-Bobo M. Dendritic Mesoporous Organosilica Nanoparticles with Photosensitizers for Cell Imaging, siRNA Delivery and Protein Loading. Molecules 2023; 28:5335. [PMID: 37513209 PMCID: PMC10385246 DOI: 10.3390/molecules28145335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Dendritic mesoporous organosilica nanoparticles (DMON) are a new class of biodegradable nanoparticles suitable for biomolecule delivery. We studied the photochemical internalization (PCI) and photodynamic therapy (PDT) of DMON to investigate new ways for DMON to escape from the endosomes-lysosomes and deliver biomolecules into the cytoplasm of cells. We added photosensitizers in the framework of DMON and found that DMON were loaded with siRNA or FVIII factor protein. We made four formulations with four different photosensitizers. The photosensitizers allowed us to perform imaging of DMON in cancer cells, but the presence of the tetrasulfide bond in the framework of DMON quenched the formation of singlet oxygen. Fortunately, one formulation allowed us to efficiently deliver proapoptotic siRNA in MCF-7 cancer cells leading to 31% of cancer cell death, without irradiation. As for FVIII protein, it was loaded in two formulations with drug-loading capacities (DLC) up to 25%. In conclusion, DMON are versatile nanoparticles capable of loading siRNA and delivering it into cancer cells, and also loading FVIII protein with good DLC. Due to the presence of tetrasulfide, it was not possible to perform PDT or PCI.
Collapse
Affiliation(s)
- Haneen Omar
- Chemistry Department, Collage of Science, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sara Jakimoska
- IBMM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
- ICGM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Julia Guillot
- IBMM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
- ICGM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Edreese Alsharaeh
- Chemistry Department, Collage of Science, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Clarence Charnay
- ICGM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Frédérique Cunin
- ICGM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Aurélie Bessière
- ICGM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | | | - Laurence Raehm
- ICGM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Laure Lichon
- IBMM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Mélanie Onofre
- IBMM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| | - Magali Gary-Bobo
- IBMM, University Montpellier, CNRS, ENSCM, 34193 Montpellier, France
| |
Collapse
|
18
|
Tyagi P, Harper G, McGeehan P, Davis SP. Current status and prospect for future advancements of long-acting antibody formulations. Expert Opin Drug Deliv 2023; 20:895-903. [PMID: 37249542 DOI: 10.1080/17425247.2023.2219445] [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: 11/09/2022] [Accepted: 05/25/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION Biologics, especially monoclonal antibodies (mAbs), have become a major class of therapeutics in recent years addressing the needs of millions of patients and becoming one of the best-selling treatments in the pharmaceutical market. A wide range of multifaceted chronic diseases have benefitted from antibody therapeutics. Long-term treatment for chronic diseases with mAb therapies can mean a lifetime of frequent injections. Technologies that can minimize the total number of injections present meaningful value to patients and the companies that develop them. AREAS COVERED This review summarizes the challenges encountered during the development of long-acting versions of mAbs. The focus will be on questions addressed during drug product development, delivery device selection, business implications, and understanding the market potential of long-acting presentations. EXPERT OPINION Long-acting drug delivery systems have reached the market for small molecules and peptides. However, these drug delivery systems, and their development lessons, cannot be extrapolated directly to antibodies. We must develop new delivery technologies suitable for biologics, identify critical attributes to capture dynamic changes in proteins during the encapsulation process, and develop analytical processes to evaluate long-term stability.
Collapse
Affiliation(s)
- Puneet Tyagi
- Dosage Form Design and Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Garrett Harper
- Insights & Analytics, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Shawn P Davis
- Dosage Form Design and Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
19
|
Aytar Çelik P, Erdogan-Gover K, Barut D, Enuh BM, Amasya G, Sengel-Türk CT, Derkus B, Çabuk A. Bacterial Membrane Vesicles as Smart Drug Delivery and Carrier Systems: A New Nanosystems Tool for Current Anticancer and Antimicrobial Therapy. Pharmaceutics 2023; 15:pharmaceutics15041052. [PMID: 37111538 PMCID: PMC10142793 DOI: 10.3390/pharmaceutics15041052] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Bacterial membrane vesicles (BMVs) are known to be critical communication tools in several pathophysiological processes between bacteria and host cells. Given this situation, BMVs for transporting and delivering exogenous therapeutic cargoes have been inspiring as promising platforms for developing smart drug delivery systems (SDDSs). In the first section of this review paper, starting with an introduction to pharmaceutical technology and nanotechnology, we delve into the design and classification of SDDSs. We discuss the characteristics of BMVs including their size, shape, charge, effective production and purification techniques, and the different methods used for cargo loading and drug encapsulation. We also shed light on the drug release mechanism, the design of BMVs as smart carriers, and recent remarkable findings on the potential of BMVs for anticancer and antimicrobial therapy. Furthermore, this review covers the safety of BMVs and the challenges that need to be overcome for clinical use. Finally, we discuss the recent advancements and prospects for BMVs as SDDSs and highlight their potential in revolutionizing the fields of nanomedicine and drug delivery. In conclusion, this review paper aims to provide a comprehensive overview of the state-of-the-art field of BMVs as SDDSs, encompassing their design, composition, fabrication, purification, and characterization, as well as the various strategies used for targeted delivery. Considering this information, the aim of this review is to provide researchers in the field with a comprehensive understanding of the current state of BMVs as SDDSs, enabling them to identify critical gaps and formulate new hypotheses to accelerate the progress of the field.
Collapse
Affiliation(s)
- Pınar Aytar Çelik
- Environmental Protection and Control Program, Eskisehir Osmangazi University, Eskisehir 26110, Turkey
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Kubra Erdogan-Gover
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Dilan Barut
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Blaise Manga Enuh
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Gülin Amasya
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara 06100, Turkey
| | - Ceyda Tuba Sengel-Türk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara 06100, Turkey
| | - Burak Derkus
- Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Ahmet Çabuk
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
- Department of Biology, Faculty of Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| |
Collapse
|
20
|
Aragoneses-Cazorla G, Vallet-Regí M, Gómez-Gómez MM, González B, Luque-Garcia JL. Integrated transcriptomics and metabolomics analysis reveals the biomolecular mechanisms associated to the antitumoral potential of a novel silver-based core@shell nanosystem. Mikrochim Acta 2023; 190:132. [PMID: 36914921 PMCID: PMC10011303 DOI: 10.1007/s00604-023-05712-3] [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: 11/22/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023]
Abstract
A combination of omics techniques (transcriptomics and metabolomics) has been used to elucidate the mechanisms responsible for the antitumor action of a nanosystem based on a Ag core coated with mesoporous silica on which transferrin has been anchored as a targeting ligand against tumor cells (Ag@MSNs-Tf). Transcriptomics analysis has been carried out by gene microarrays and RT-qPCR, while high-resolution mass spectrometry has been used for metabolomics. This multi-omics strategy has enabled the discovery of the effect of this nanosystem on different key molecular pathways including the glycolysis, the pentose phosphate pathway, the oxidative phosphorylation and the synthesis of fatty acids, among others.
Collapse
Affiliation(s)
- Guillermo Aragoneses-Cazorla
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - María Vallet-Regí
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (I+12), 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Saragossa, Spain
| | - Ma Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Blanca González
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (I+12), 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Saragossa, Spain
| | - Jose L Luque-Garcia
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
21
|
Pleiotrophin-Loaded Mesoporous Silica Nanoparticles as a Possible Treatment for Osteoporosis. Pharmaceutics 2023; 15:pharmaceutics15020658. [PMID: 36839981 PMCID: PMC9966378 DOI: 10.3390/pharmaceutics15020658] [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: 12/26/2022] [Revised: 01/19/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Osteoporosis is the most common type of bone disease. Conventional treatments are based on the use of antiresorptive drugs and/or anabolic agents. However, these treatments have certain limitations, such as a lack of bioavailability or toxicity in non-specific tissues. In this regard, pleiotrophin (PTN) is a protein with potent mitogenic, angiogenic, and chemotactic activity, with implications in tissue repair. On the other hand, mesoporous silica nanoparticles (MSNs) have proven to be an effective inorganic drug-delivery system for biomedical applications. In addition, the surface anchoring of cationic polymers, such as polyethylenimine (PEI), allows for greater cell internalization, increasing treatment efficacy. In order to load and release the PTN to improve its effectiveness, MSNs were successfully internalized in MC3T3-E1 mouse pre-osteoblastic cells and human mesenchymal stem cells. PTN-loaded MSNs significantly increased the viability, mineralization, and gene expression of alkaline phosphatase and Runx2 in comparison with the PTN alone in both cell lines, evidencing its positive effect on osteogenesis and osteoblast differentiation. This proof of concept demonstrates that MSN can take up and release PTN, developing a potent osteogenic and differentiating action in vitro in the absence of an osteogenic differentiation-promoting medium, presenting itself as a possible treatment to improve bone-regeneration and osteoporosis scenarios.
Collapse
|
22
|
Attia MS, Yahya A, Monaem NA, Sabry SA. Mesoporous silica nanoparticles: Their potential as drug delivery carriers and nanoscavengers in Alzheimer's and Parkinson's diseases. Saudi Pharm J 2023; 31:417-432. [PMID: 37026045 PMCID: PMC10071366 DOI: 10.1016/j.jsps.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Worldwide, populations face significant burdens from neurodegenerative disorders (NDDs), especially Alzheimer's and Parkinson's diseases. Although there are many proposed etiologies for neurodegenerative disorders, including genetic and environmental factors, the exact pathogenesis for these disorders is not fully understood. Most patients with NDDs are given lifelong treatment to improve their quality of life. There are myriad treatments for NDDs; however, these agents are limited by their side effects and difficulty in passing the blood-brain barrier (BBB). Furthermore, the central nervous system (CNS) active pharmaceuticals could offer symptomatic relief for the patient's condition without providing a complete cure or prevention by targeting the disease's cause. Recently, Mesoporous silica nanoparticles (MSNs) have gained interest in treating NDDs since their physicochemical properties and inherent ability to pass BBB make them possible drug carriers for several drugs for NDDs treatment. This paper provides insight into the pathogenesis and treatment of NDDs, along with the recent advances in applying MSNs as fibril scavengers. Moreover, the application of MSNs-based formulations in enhancing or sustaining drug release rate, and brain targeting via their responsive release properties, besides the neurotoxicity of MSNs, have been reviewed.
Collapse
Affiliation(s)
- Mohamed S. Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Corresponding author.
| | - Ahmed Yahya
- Egypt-Japan University of Science and Technology, New Borg El Arab, Alexandria 21934, Egypt
| | - Nada Abdel Monaem
- Department of chemistry, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Shereen A. Sabry
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
23
|
Natural Biopolymers as Smart Coating Materials of Mesoporous Silica Nanoparticles for Drug Delivery. Pharmaceutics 2023; 15:pharmaceutics15020447. [PMID: 36839771 PMCID: PMC9965229 DOI: 10.3390/pharmaceutics15020447] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
In recent years, the functionalization of mesoporous silica nanoparticles (MSNs) with different types of responsive pore gatekeepers have shown great potential for the formulation of drug delivery systems (DDS) with minimal premature leakage and site-specific controlled release. New nanotechnological approaches have been developed with the objective of utilizing natural biopolymers as smart materials in drug delivery applications. Natural biopolymers are sensitive to various physicochemical and biological stimuli and are endowed with intrinsic biodegradability, biocompatibility, and low immunogenicity. Their use as biocompatible smart coatings has extensively been investigated in the last few years. This review summarizes the MSNs coating procedures with natural polysaccharides and protein-based biopolymers, focusing on their application as responsive materials to endogenous stimuli. Biopolymer-coated MSNs, which conjugate the nanocarrier features of mesoporous silica with the biocompatibility and controlled delivery provided by natural coatings, have shown promising therapeutic outcomes and the potential to emerge as valuable candidates for the selective treatment of various diseases.
Collapse
|
24
|
Gándara Z, Rubio N, Castillo RR. Delivery of Therapeutic Biopolymers Employing Silica-Based Nanosystems. Pharmaceutics 2023; 15:pharmaceutics15020351. [PMID: 36839672 PMCID: PMC9963032 DOI: 10.3390/pharmaceutics15020351] [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: 12/01/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
The use of nanoparticles is crucial for the development of a new generation of nanodevices for clinical applications. Silica-based nanoparticles can be tailored with a wide range of functional biopolymers with unique physicochemical properties thus providing several advantages: (1) limitation of interparticle interaction, (2) preservation of cargo and particle integrity, (3) reduction of immune response, (4) additional therapeutic effects and (5) cell targeting. Therefore, the engineering of advanced functional coatings is of utmost importance to enhance the biocompatibility of existing biomaterials. Herein we will focus on the most recent advances reported on the delivery and therapeutic use of silica-based nanoparticles containing biopolymers (proteins, nucleotides, and polysaccharides) with proven biological effects.
Collapse
Affiliation(s)
- Zoila Gándara
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| | - Noelia Rubio
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| | - Rafael R. Castillo
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| |
Collapse
|
25
|
Rani R, Malik P, Dhania S, Mukherjee TK. Recent Advances in Mesoporous Silica Nanoparticle-Mediated Drug Delivery for Breast Cancer Treatment. Pharmaceutics 2023; 15:227. [PMID: 36678856 PMCID: PMC9860911 DOI: 10.3390/pharmaceutics15010227] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Breast cancer (BC) currently occupies the second rank in cancer-related global female deaths. Although consistent awareness and improved diagnosis have reduced mortality in recent years, late diagnosis and resistant response still limit the therapeutic efficacy of chemotherapeutic drugs (CDs), leading to relapse with consequent invasion and metastasis. Treatment with CDs is indeed well-versed but it is badly curtailed with accompanying side effects and inadequacies of site-specific drug delivery. As a result, drug carriers ensuring stealth delivery and sustained drug release with improved pharmacokinetics and biodistribution are urgently needed. Core-shell mesoporous silica nanoparticles (MSNPs) have recently been a cornerstone in this context, attributed to their high surface area, low density, robust functionalization, high drug loading capacity, size-shape-controlled functioning, and homogeneous shell architecture, enabling stealth drug delivery. Recent interest in using MSNPs as drug delivery vehicles has been due to their functionalization and size-shape-driven versatilities. With such insights, this article focuses on the preparation methods and drug delivery mechanisms of MSNPs, before discussing their emerging utility in BC treatment. The information compiled herein could consolidate the database for using inorganic nanoparticles (NPs) as BC drug delivery vehicles in terms of design, application and resolving post-therapy complications.
Collapse
Affiliation(s)
- Ruma Rani
- ICAR-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Sunena Dhania
- Department of Bio & Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Tapan Kumar Mukherjee
- Institute of Biotechnology (AIB), Amity University, Noida 201313, Uttar Pradesh, India
| |
Collapse
|
26
|
Modi SK, Gaur S, Sengupta M, Singh MS. Mechanistic insights into nanoparticle surface-bacterial membrane interactions in overcoming antibiotic resistance. Front Microbiol 2023; 14:1135579. [PMID: 37152753 PMCID: PMC10160668 DOI: 10.3389/fmicb.2023.1135579] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Antimicrobial Resistance (AMR) raises a serious concern as it contributes to the global mortality by 5 million deaths per year. The overall impact pertaining to significant membrane changes, through broad spectrum drugs have rendered the bacteria resistant over the years. The economic expenditure due to increasing drug resistance poses a global burden on healthcare community and must be dealt with immediate effect. Nanoparticles (NP) have demonstrated inherent therapeutic potential or can serve as nanocarriers of antibiotics against multidrug resistant (MDR) pathogens. These carriers can mask the antibiotics and help evade the resistance mechanism of the bacteria. The targeted delivery can be fine-tuned through surface functionalization of Nanocarriers using aptamers, antibodies etc. This review covers various molecular mechanisms acquired by resistant bacteria towards membrane modification. Mechanistic insight on 'NP surface-bacterial membrane' interactions are crucial in deciding the role of NP as therapeutic. Finally, we highlight the potential accessible membrane targets for designing smart surface-functionalized nanocarriers which can act as bacteria-targeted robots over the existing clinically available antibiotics. As the bacterial strains around us continue to evolve into resistant versions, nanomedicine can offer promising and alternative tools in overcoming AMR.
Collapse
Affiliation(s)
- Suraj Kumar Modi
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, India
- Centre of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Smriti Gaur
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Mrittika Sengupta
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, India
- Centre of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, India
- Mrittika Sengupta, ;
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, India
- Centre of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, India
- *Correspondence: Manu Smriti Singh, ;
| |
Collapse
|
27
|
Montiel-Centeno K, García-Villén F, Barrera D, Amaya-Roncancio S, Sánchez-Espejo R, Arroyo-Gómez JJ, Sandri G, Viseras C, Sapag K. Biocompatible nanoporous carbons as a carrier system for controlled release of cephalexin. Colloids Surf B Biointerfaces 2022. [DOI: 10.1016/j.colsurfb.2022.112937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Achievements in Mesoporous Bioactive Glasses for Biomedical Applications. Pharmaceutics 2022; 14:pharmaceutics14122636. [PMID: 36559130 PMCID: PMC9782017 DOI: 10.3390/pharmaceutics14122636] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Nowadays, mesoporous bioactive glasses (MBGs) are envisaged as promising candidates in the field of bioceramics for bone tissue regeneration. This is ascribed to their singular chemical composition, structural and textural properties and easy-to-functionalize surface, giving rise to accelerated bioactive responses and capacity for local drug delivery. Since their discovery at the beginning of the 21st century, pioneering research efforts focused on the design and fabrication of MBGs with optimal compositional, textural and structural properties to elicit superior bioactive behavior. The current trends conceive MBGs as multitherapy systems for the treatment of bone-related pathologies, emphasizing the need of fine-tuning surface functionalization. Herein, we focus on the recent developments in MBGs for biomedical applications. First, the role of MBGs in the design and fabrication of three-dimensional scaffolds that fulfil the highly demanding requirements for bone tissue engineering is outlined. The different approaches for developing multifunctional MBGs are overviewed, including the incorporation of therapeutic ions in the glass composition and the surface functionalization with zwitterionic moieties to prevent bacterial adhesion. The bourgeoning scientific literature on MBGs as local delivery systems of diverse therapeutic cargoes (osteogenic/antiosteoporotic, angiogenic, antibacterial, anti-inflammatory and antitumor agents) is addressed. Finally, the current challenges and future directions for the clinical translation of MBGs are discussed.
Collapse
|
29
|
Wu Y, Sun Z, Song J, Mo L, Wang X, Liu H, Ma Y. Preparation of multifunctional mesoporous SiO 2nanoparticles and anti-tumor action. NANOTECHNOLOGY 2022; 34:055101. [PMID: 36317264 DOI: 10.1088/1361-6528/ac9e5f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
A targeted drug delivery system was developed to accumulate specific drugs around tumor cells based on the redox, temperature, and enzyme synergistic responses of mesoporous silica nanoparticles. Mesoporous silica nanoparticles (MSN-NH2) and Doxorubicin (DOX) for tumor therapy were prepared and loaded into the pores of MSN- NH2 to obtain DOX@MSN(DM NPs). Hyaluronic acid (HA) was used as the backbone and disulfide bond was used as the linker arm to graft carboxylated poly (N-isopropylacrylamide)(PNIPAAm-COOH) to synthesize the macromolecular copolymer (HA-SS-PNIPAAm), which was modified to DM NPs with capped ends to obtain the nano-delivery system DOX@MSN@HA-SS-PNIPAAm(DMHSP NPs), and a control formulation was prepared in a similar way. DMHSP NPs specifically entered tumor cells via CD44 receptor-mediated endocytosis; the high GSH concentration (10 mM) of cells severed the disulfide bonds, the hyaluronidase sheared the capped HA to open the pores, and increased tumor microenvironment temperature due to immune response can trigger the release of encapsulated drugs in thermosensitive materials.In vitroandin vivoantitumor and hemolysis assays showed that DMHSP NPs can accurately target hepatocellular carcinoma cells with a good safety profile and have synergistic effects, which meant DMHSP NPs had great potential for tumor therapy.
Collapse
Affiliation(s)
- Yijun Wu
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, People's Republic of China
| | - Zhiqiang Sun
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, People's Republic of China
| | - Jinfeng Song
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, People's Republic of China
| | - Liufang Mo
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, People's Republic of China
| | - Xiaochen Wang
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, People's Republic of China
| | - Hanhan Liu
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, People's Republic of China
| | - Yunfeng Ma
- Institute of Microbial Engineering, Laboratory of Bioresource and Applied Microbiology, School of Life Sciences, Henan University, Kaifeng 475004, People's Republic of China
- Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng 475004, People's Republic of China
| |
Collapse
|
30
|
Escriche‐Navarro B, Escudero A, Lucena‐Sánchez E, Sancenón F, García‐Fernández A, Martínez‐Máñez R. Mesoporous Silica Materials as an Emerging Tool for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200756. [PMID: 35866466 PMCID: PMC9475525 DOI: 10.1002/advs.202200756] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/16/2022] [Indexed: 05/16/2023]
Abstract
Cancer immunotherapy has emerged in the past decade as a promising strategy for treating many forms of cancer by stimulating the patient's immune system. Although immunotherapy has achieved some promising results in clinics, more efforts are required to improve the limitations of current treatments related to lack of effective and targeted cancer antigens delivery to immune cells, dose-limiting toxicity, and immune-mediated adverse effects, among others. In recent years, the use of nanomaterials has proven promising to enhance cancer immunotherapy efficacy and reduce side effects. Among nanomaterials, attention has been recently paid to mesoporous silica nanoparticles (MSNs) as a potential multiplatform for enhancing cancer immunotherapy by considering their unique properties, such as high porosity, and good biocompatibility, facile surface modification, and self-adjuvanticity. This review explores the role of MSN and other nano/micro-materials as an emerging tool to enhance cancer immunotherapy, and it comprehensively summarizes the different immunotherapeutic strategies addressed to date by using MSN.
Collapse
Affiliation(s)
- Blanca Escriche‐Navarro
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Joint Unit of Nanomedicine and Sensors, Polytechnic University of Valencia, IIS La FeAv. Fernando Abril Martorell, 106Valencia46026Spain
| | - Andrea Escudero
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
| | - Elena Lucena‐Sánchez
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
| | - Félix Sancenón
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Joint Unit of Nanomedicine and Sensors, Polytechnic University of Valencia, IIS La FeAv. Fernando Abril Martorell, 106Valencia46026Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)Av. Monforte de Lemos, 3–5. Pabellón 11., Planta 0Madrid28029Spain
| | - Alba García‐Fernández
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)Av. Monforte de Lemos, 3–5. Pabellón 11., Planta 0Madrid28029Spain
| | - Ramón Martínez‐Máñez
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Joint Unit of Nanomedicine and Sensors, Polytechnic University of Valencia, IIS La FeAv. Fernando Abril Martorell, 106Valencia46026Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)Av. Monforte de Lemos, 3–5. Pabellón 11., Planta 0Madrid28029Spain
| |
Collapse
|
31
|
Farjadian F, Moghadam M, Monfared M, Mohammadi‐Samani S. Mesoporous Silica Nanostructure Modified with Azo Gatekeepers for Colon Targeted Delivery of
5‐Fluorouracil. AIChE J 2022. [DOI: 10.1002/aic.17900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
| | - Maryam Moghadam
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
- Department of Pharmaceutics, School of Pharmacy Shiraz University of Medical Science Shiraz Iran
| | - Mohammad Monfared
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| | - Soliman Mohammadi‐Samani
- Pharmaceutical Sciences Research Center Shiraz University of Medical Science Shiraz Iran
- Department of Pharmaceutics, School of Pharmacy Shiraz University of Medical Science Shiraz Iran
| |
Collapse
|
32
|
Vallet-Regí M, Schüth F, Lozano D, Colilla M, Manzano M. Engineering mesoporous silica nanoparticles for drug delivery: where are we after two decades? Chem Soc Rev 2022; 51:5365-5451. [PMID: 35642539 PMCID: PMC9252171 DOI: 10.1039/d1cs00659b] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Collapse
Affiliation(s)
- María Vallet-Regí
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ferdi Schüth
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Daniel Lozano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Montserrat Colilla
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
33
|
Lee ZH, Lee MF, Chen JH, Tsou MH, Wu ZY, Lee CZ, Huang YY, Lin SM, Lin HM. Fucoidan with three functions extracted from Sargassum aquifolium integrated rice-husk synthesis dual-imaging mesoporous silica nanoparticle. J Nanobiotechnology 2022; 20:298. [PMID: 35733216 PMCID: PMC9215008 DOI: 10.1186/s12951-022-01430-9] [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: 02/23/2022] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
In this study, we used the nanoparticle delivery system to reduce the side effect of conventional cancer treatment- radiation therapy and chemotherapy. We used rice husk silicon source mesoporous silica nanoparticle doped in Eu3+ and Gd3+ as the carrier in the delivery system and to enable fluorescence and MRI dual-imaging functions for follow-up therapy. In addition, we choose a popular seaweed extract-fucoidan was extracted from the same brown algae-Sargassum aquifolium collected from Taiwan-Pingtung-Kenting-Chuanfan Rock. In this research, we used acid hydrolysis to prepared two different molecular weight fucoidan, the small molecular fucoidan (Fus) as drug, and the molecular weight approximately 1 kDa fucoidan (Ful) as the nanoparticle gatekeeper, and as targeting molecule for overexpressed P-selectin on the surface of the metastatic tumors. The results of the cell cytotoxicity experiment showed that HCT116 cancer cells have a survival rate of approximately 58.12% when treated with 200 μg/mL fucoidan. Dual-imaging rice husk mesoporous silica nanoparticles (rMSN-EuGd) were modified with 1 kDa fucoidan (Ful) as the gatekeeper and target, and the small molecule fucoidan (Fus) was loaded into nanoparticles (Ful-Fus@rMSN-EuGd) at a concentration of 200 μg/mL. The HCT116 cancer cells had a survival rate of approximately 55.56%. The cell cytotoxicity experiment results show that Ful-Fus@rMSN-EuGd can improve the anticancer effect of fucoidan, and the nanoparticle drug delivery system using fucoidan as a drug, target, and gatekeeper was successfully synthesized.
Collapse
Affiliation(s)
- Zui-Harng Lee
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Meng-Feng Lee
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Jung-Huang Chen
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Min-Hsuan Tsou
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Zhi-Yuan Wu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Cheng-Zhang Lee
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Yu-Ya Huang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Showe-Mei Lin
- Institute of Marine Biology, National Taiwan Ocean University, Keelung, 202, Taiwan
| | - Hsiu-Mei Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202, Taiwan.
| |
Collapse
|
34
|
Mehta S, Suresh A, Nayak Y, Narayan R, Nayak UY. Hybrid nanostructures: Versatile systems for biomedical applications. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
35
|
Pei W, Cai L, Gong X, Zhang L, Zhang J, Zhu P, Jiang H, Wang C, Wang S, Chen J. Drug-loaded oleic-acid grafted mesoporous silica nanoparticles conjugated with α-lactalbumin resembling BAMLET-like anticancer agent with improved biocompatibility and therapeutic efficacy. Mater Today Bio 2022; 15:100272. [PMID: 35607417 PMCID: PMC9123267 DOI: 10.1016/j.mtbio.2022.100272] [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: 03/08/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Despite its prominent therapeutic efficacy, chemotherapy has raised serious concerns due to the severe adverse effects and multidrug resistance evoked, which propels the search for safe and green therapeutic agents. BAMLET (bovine α-lactalbumin made lethal against tumor cell) is a well-known protein-based anticancer agent of selective tumoricidal activity. Here, we prepared oleic acid-modified mesoporous silica nanoparticles (OA-MSNs) conjugated with bovine α-lactalbumin, a lipoprotein complex resembling BAMLET formed on the surface of MSNs (MSN-BAMLET) to load the anticancer drug of docetaxel (DTX). Compared to that of OA-MSNs/DTX, the obtained MSN-BAMLET/DTX with a sustained and pH-responsive drug release behaviors exhibited good biocompatibility and enhanced cytotoxic effect against cancer cells. Moreover, the presence of lipoprotein complex in MSN-BAMLET contributed to the improved dispersion of the composite in solution and the inhibitory effect on the migration of cancer cells. Furthermore, the adsorption profiles of protein corona on the obtained nanoparticles were analyzed. It was found that the marked low amount and abundance of plasma proteins were adsorbed on the α-lactalbumin coated siliceous composite demonstrated its long circulation property. Finally, in vivo study showed that MSN-BAMLET/DTX contributed to the effective cancer ablation and the prolonged survival. Therefore, the constructed MSN-BAMLET of the mesoregular structure and peculiar tumoricidal effect provides a manipulable nanoplatform as drug nanocarrier for therapeutic applications.
Collapse
Affiliation(s)
- Wei Pei
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Ling Cai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Xing Gong
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Li Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Jiarong Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Ping Zhu
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Huijun Jiang
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Chao Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Shoulin Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Jin Chen
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
36
|
Changes in Carbon and Nitrogen Metabolites before, at, and after Anthesis for Wheat Cultivars in Response to Reduced Soil Water and Zinc Foliar Application. PLANTS 2022; 11:plants11091261. [PMID: 35567261 PMCID: PMC9104341 DOI: 10.3390/plants11091261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 11/24/2022]
Abstract
Water deficit stress is one of the major constraints for commercial agriculture, as it disturbs the metabolic processes in plant. Identification of carbon and nitrogen receptors that act on drought resistance helps in breeding for drought resistance varieties. Zn fertilizer can regulate multiple antioxidant defense systems at the transcriptional level in response to drought. Two field experiments were conducted in 2018–2019 and 2019–2020 seasons to explore the effectiveness of foliar application of zinc oxide on soluble sugar, soluble proteins, and free amino acids under normal irrigation and drought-stressed environments. Three Egyptian wheat cultivars (Triticum aestivum L.) were used. The experimental design was split-plot in RCBD with three replications, applying zinc oxide levels to the whole plot and the split plots. Leaf samples were taken for analysis before anthesis, at anthesis, and after anthesis. Application of Zn increased soluble sugars. However, the free amino acids were higher under irrigation, reached the maximum at anthesis, and decreased sharply after 2 weeks from anthesis. The ranking of cultivars for the three metabolites differed according to plant stage, reflecting the response to Zn and years. Correlations between metabolites according to Zn were positive. The findings suggest the potential of foliar application of Zn to alleviate drought stress.
Collapse
|
37
|
MacCuaig WM, Samykutty A, Foote J, Luo W, Filatenkov A, Li M, Houchen C, Grizzle WE, McNally LR. Toxicity Assessment of Mesoporous Silica Nanoparticles upon Intravenous Injection in Mice: Implications for Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14050969. [PMID: 35631554 PMCID: PMC9148138 DOI: 10.3390/pharmaceutics14050969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Nanoparticles are popular tools utilized to selectively deliver drugs and contrast agents for identification and treatment of disease. To determine the usefulness and translational potential of mesoporous silica nanoparticles (MSNs), further evaluations of toxicity are required. MSNs are among the most utilized nano-delivery systems due to ease of synthesis, pore structure, and functionalization. This study aims to elucidate toxicity as a result of intravenous injection of 25 nm MSNs coated with chitosan (C) or polyethylene glycol (PEG) in mice. Following acute and chronic injections, blood was evaluated for standard blood chemistry and complete blood count analyses. Blood chemistry results primarily indicated that no abnormalities were present following acute or chronic injections of MSNs, or C/PEG-coated MSNs. After four weekly administered treatments, vital organs showed minor exacerbation of pre-existing lesions in the 35KPEG-MSN and moderate exacerbation of pre-existing lesions in uncoated MSN and 2KPEG-MSN treatment groups. In contrast, C-MSN treatment groups had minimal changes compared to controls. This study suggests 25 nm MSNs coated with chitosan should elicit minimal toxicity when administered as either single or multiple intravenous injections, but MSNs coated with PEG, especially 2KPEG may exacerbate pre-existing vascular conditions. Further studies should evaluate varying sizes and types of nanoparticles to provide a better overall understanding on the relation between nanoparticles and in vivo toxicity.
Collapse
Affiliation(s)
- William M. MacCuaig
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73109, USA
| | - Abhilash Samykutty
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
| | - Jeremy Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Wenyi Luo
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
- Department of Pathology, Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Alexander Filatenkov
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
- Department of Pathology, Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Min Li
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
- Department of Medicine, Oklahoma Health Science Center, Oklahoma City, OK 73049, USA
| | - Courtney Houchen
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
- Department of Medicine, Oklahoma Health Science Center, Oklahoma City, OK 73049, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA; (W.M.M.); (A.S.); (W.L.); (A.F.); (M.L.); (C.H.)
- Department of Surgery, Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
38
|
Moving beyond the Tip of the Iceberg: DJ-1 Implications in Cancer Metabolism. Cells 2022; 11:cells11091432. [PMID: 35563738 PMCID: PMC9103122 DOI: 10.3390/cells11091432] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
DJ-1, also called Parkinson’s protein 7 (PARK7), is ubiquitously expressed and plays multiple actions in different physiological and, especially, pathophysiological processes, as evidenced by its identification in neurodegenerative diseases and its high expression in different types of cancer. To date, the exact activity of DJ-1 in carcinogenesis has not been fully elucidated, however several recent studies disclosed its involvement in regulating fundamental pathways involved in cancer onset, development, and metastatization. At this purpose, we have dissected the role of DJ-1 in maintaining the transformed phenotype, survival, drug resistance, metastasis formation, and differentiation in cancer cells. Moreover, we have discussed the role of DJ-1 in controlling the redox status in cancer cells, along with the ability to attenuate reactive oxygen species (ROS)-dependent cell death, as well as to mediate ferropotosis. Finally, a mention to the development of therapeutic strategies targeting DJ-1 has been done. We have reported the most recent studies, aiming to shed light on the role played by DJ-1 in different cancer aspects and create the foundation for moving beyond the tip of the iceberg.
Collapse
|
39
|
Wang Y, Khan HM, Zhou C, Liao X, Tang P, Song P, Gui X, Li H, Chen Z, Liu S, Cen Y, Zhang Z, Li Z. Apoptotic cells derived micro/nano-sized extracellular vesicles in tissue regeneration. NANOTECHNOLOGY REVIEWS 2022. [DOI: 10.1515/ntrev-2022-0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
Extracellular vesicles (EVs), products released by cells in multiple biological activities, are currently widely accepted as functional particles and intercellular communicators. From the orthodox perspective, EVs derived from apoptotic cells (apoEVs) are responsible for cell debris clearance, while recent studies have demonstrated that apoEVs participate in tissue regeneration. However, the underlying mechanisms and particular functions in tissue regeneration promotion of apoEVs remain ambiguous. Some molecules, such as caspases, active during apoptosis also function in tissue regeneration triggered by apoptosis,. ApoEVs are generated in the process of apoptosis, carrying cell contents to manifest biological effects, and possessing biomarkers to target phagocytes. The regenerative effect of apoEVs might be due to their abilities to facilitate cell proliferation and regulate inflammation. Such regenerative effect has been observed in various tissues, including skin, bone, cardiovascular system, and kidney. Engineered apoEVs are produced to amplify the biological benefits of apoEVs, rendering them optional for drug delivery. Meanwhile, challenges exist in thorough mechanistic exploration and standardization of production. In this review, we discussed the link between apoptosis and regeneration, current comprehension of the origination and investigation strategies of apoEVs, and mechanisms in tissue regeneration by apoEVs and their applications. Challenges and prospects are also discussed here.
Collapse
Affiliation(s)
- Yixi Wang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Haider Mohammed Khan
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University , Chengdu Sichuan, 610041 , China
| | - Changchun Zhou
- College of Biomedical Engineering, Sichuan University , Chengdu 610064 , China
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064 , China
| | - Xiaoxia Liao
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Pei Tang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Ping Song
- College of Biomedical Engineering, Sichuan University , Chengdu 610064 , China
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064 , China
| | - Xingyu Gui
- College of Biomedical Engineering, Sichuan University , Chengdu 610064 , China
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064 , China
| | - Hairui Li
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University , Xi’an , Shaanxi, 710032 , China
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University , 610041 , Chengdu , China
| |
Collapse
|
40
|
Synthetic methods of lipid-coated mesoporous silica nanoparticles as drug carriers. Biointerphases 2022; 17:020801. [PMID: 35232023 DOI: 10.1116/6.0001688] [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
The lipid-coated mesoporous silica nanoparticles (LMSNs) that can synergistically harness the advantages and mitigate the disadvantages of the liposomes and MSNs are considered potential drug carriers. So far, several methods have been developed to prepare LMSNs, including vesicle fusion, thin-film hydration, and solvent exchange. Despite their wide application in LMSN preparation, these methods are short of detailed elaboration and comparison, which hinders their further development. In this review, for the first time, the three methods are systematically summarized, including their mechanisms, influence factors, advantages, and limitations. Although these methods are all based on lipid self-assembly, there is still a difference between them. In order to efficiently prepare LMSNs, we proposed that a suitable method should be selected based on the actual situation. It is conceivable that the elaboration and comparison in this review will make these methods easy to be understood and provide guidance for the design of LMSNs as drug carriers.
Collapse
|
41
|
Apoptotic cell-derived micro/nanosized extracellular vesicles in tissue regeneration. NANOTECHNOLOGY REVIEWS 2022. [DOI: 10.1515/ntrev-2022-0052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
Extracellular vesicles (EVs), products released by cells in multiple biological activities, are currently widely accepted as functional particles and intercellular communicators. From the orthodox perspective, EVs derived from apoptotic cells (apoEVs) are responsible for cell debris clearance, while recent studies have demonstrated that apoEVs participate in tissue regeneration. However, the underlying mechanisms and particular functions in tissue regeneration promotion of apoEVs remain ambiguous. Some molecules active during apoptosis also function in tissue regeneration triggered by apoptosis, such as caspases. ApoEVs are generated in the process of apoptosis, carrying cell contents to manifest biological effects and possess biomarkers to target phagocytes. The regenerative effect of apoEVs might be due to their abilities to facilitate cell proliferation and regulate inflammation. Such regenerative effect has been observed in various tissues, including skin, bone, cardiovascular system, and kidneys. Engineered apoEVs are produced to amplify the biological benefits of apoEVs, rendering them optional for drug delivery. Meanwhile, challenges exist in thorough mechanistic exploration and standardization of production. In this review, we discussed the link between apoptosis and regeneration, current comprehension of the origination and investigation strategies of apoEVs, and mechanisms in tissue regeneration of apoEVs and their applications. Challenges and prospects are also addressed here.
Collapse
|
42
|
Hasanzadeh A, Noori H, Jahandideh A, Haeri Moghaddam N, Kamrani Mousavi SM, Nourizadeh H, Saeedi S, Karimi M, Hamblin MR. Smart Strategies for Precise Delivery of CRISPR/Cas9 in Genome Editing. ACS APPLIED BIO MATERIALS 2022; 5:413-437. [PMID: 35040621 DOI: 10.1021/acsabm.1c01112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The emergence of CRISPR/Cas technology has enabled scientists to precisely edit genomic DNA sequences. This approach can be used to modulate gene expression for the treatment of genetic disorders and incurable diseases such as cancer. This potent genome-editing tool is based on a single guide RNA (sgRNA) strand that recognizes the targeted DNA, plus a Cas nuclease protein for binding and processing the target. CRISPR/Cas has great potential for editing many genes in different types of cells and organisms both in vitro and in vivo. Despite these remarkable advances, the risk of off-target effects has hindered the translation of CRISPR/Cas technology into clinical applications. To overcome this hurdle, researchers have devised gene regulatory systems that can be controlled in a spatiotemporal manner, by designing special sgRNA, Cas, and CRISPR/Cas delivery vehicles that are responsive to different stimuli, such as temperature, light, magnetic fields, ultrasound (US), pH, redox, and enzymatic activity. These systems can even respond to dual or multiple stimuli simultaneously, thereby providing superior spatial and temporal control over CRISPR/Cas gene editing. Herein, we summarize the latest advances on smart sgRNA, Cas, and CRISPR/Cas nanocarriers, categorized according to their stimulus type (physical, chemical, or biological).
Collapse
Affiliation(s)
- Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Hamid Noori
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Atefeh Jahandideh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Niloofar Haeri Moghaddam
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Seyede Mahtab Kamrani Mousavi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Helena Nourizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Sara Saeedi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 1449614535, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 141556559, Iran
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran 1584743311, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| |
Collapse
|
43
|
High Doses of Silica Nanoparticles Obtained by Microemulsion and Green Routes Compromise Human Alveolar Cells Morphology and Stiffness Differently. Bioinorg Chem Appl 2022; 2022:2343167. [PMID: 35140761 PMCID: PMC8820933 DOI: 10.1155/2022/2343167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/15/2021] [Accepted: 01/06/2022] [Indexed: 12/30/2022] Open
Abstract
Among all the inorganic nanomaterials used in commercial products, industry, and medicine, the amorphous silica nanoparticles (SiO2 NPs) appeared to be often tolerated in living organisms. However, despite several toxicity studies, some concerns about the exposure to high doses of SiO2 NPs with different sizes were raised. Then, we used the microemulsion method to obtain stable SiO2 NPs having different sizes (110 nm, 50 nm, and 25 nm). In addition, a new one-pot green synthetic route using leaves extract of Laurus nobilis was performed, obtaining monodispersed ultrasmall SiO2 NPs without the use of dangerous chemicals. The NPs achieved by microemulsion were further functionalized with amino groups making the NPs surface positively charged. Then, high doses of SiO2 NPs (1 mg/mL and 3 mg/mL) achieved from the two routes, having different sizes and surface charges, were used to assess their impact on human alveolar cells (A549), being the best cell model mimicking the inhalation route. Cell viability and caspase-3 induction were analyzed as well as the cellular uptake, obtaining that the smallest (25 nm) and positive-charged NPs were more able to induce cytotoxicity, reaching values of about 60% of cell death. Surprisingly, cells incubated with green SiO2 NPs did not show strong toxicity, and 70% of them remained vital. This result was unusual for ultrasmall nanoobjects, generally highly toxic. The actin reorganization, nuclear morphology alteration, and cell membrane elasticity analyses confirmed the trend achieved from the biological assays. The obtained data demonstrate that the increase in cellular softness, i.e., the decrease in Young’s modulus, could be associated with the smaller and positive NPs, recording values of about 3 kPa. On the contrary, green NPs triggered a slight decrease of stiffness values (c.a. 6 kPa) compared to the untreated cells (c.a. 8 kPa). As the softer cells were implicated in cancer progression and metastasization, this evidence strongly supported the idea of a link between the cell elasticity and physicochemical properties of NPs that, in turn, influenced the interaction with the cell membrane. Thus, the green SiO2 NPs compromised cells to a lesser extent than the other SiO2 NPs types. In this scenario, the elasticity evaluation could be an interesting tool to understand the toxicity of NPs with the aim of predicting some pathological phenomena associated with their exposure.
Collapse
|
44
|
Álvarez E, Estévez M, Gallo-Cordova A, González B, Castillo RR, Morales MDP, Colilla M, Izquierdo-Barba I, Vallet-Regí M. Superparamagnetic Iron Oxide Nanoparticles Decorated Mesoporous Silica Nanosystem for Combined Antibiofilm Therapy. Pharmaceutics 2022; 14:163. [PMID: 35057058 PMCID: PMC8778149 DOI: 10.3390/pharmaceutics14010163] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
A crucial challenge to face in the treatment of biofilm-associated infection is the ability of bacteria to develop resistance to traditional antimicrobial therapies based on the administration of antibiotics alone. This study aims to apply magnetic hyperthermia together with controlled antibiotic delivery from a unique magnetic-responsive nanocarrier for a combination therapy against biofilm. The design of the nanosystem is based on antibiotic-loaded mesoporous silica nanoparticles (MSNs) externally functionalized with a thermo-responsive polymer capping layer, and decorated in the outermost surface with superparamagnetic iron oxide nanoparticles (SPIONs). The SPIONs are able to generate heat upon application of an alternating magnetic field (AMF), reaching the temperature needed to induce a change in the polymer conformation from linear to globular, therefore triggering pore uncapping and the antibiotic cargo release. The microbiological assays indicated that exposure of E. coli biofilms to 200 µg/mL of the nanosystem and the application of an AMF (202 kHz, 30 mT) decreased the number of viable bacteria by 4 log10 units compared with the control. The results of the present study show that combined hyperthermia and antibiotic treatment is a promising approach for the effective management of biofilm-associated infections.
Collapse
Affiliation(s)
- Elena Álvarez
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
- CIBER de Bioingeniería Biomateriales y Nanomedicina CIBER-BBN, 28029 Madrid, Spain
| | - Manuel Estévez
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
| | - Alvaro Gallo-Cordova
- Instituto de Ciencia de Materiales de Madrid, ICMM/CSIC, Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain; (A.G.-C.); (M.d.P.M.)
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
- CIBER de Bioingeniería Biomateriales y Nanomedicina CIBER-BBN, 28029 Madrid, Spain
| | - Rafael R. Castillo
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
- CIBER de Bioingeniería Biomateriales y Nanomedicina CIBER-BBN, 28029 Madrid, Spain
| | - María del Puerto Morales
- Instituto de Ciencia de Materiales de Madrid, ICMM/CSIC, Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain; (A.G.-C.); (M.d.P.M.)
| | - Montserrat Colilla
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
- CIBER de Bioingeniería Biomateriales y Nanomedicina CIBER-BBN, 28029 Madrid, Spain
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
- CIBER de Bioingeniería Biomateriales y Nanomedicina CIBER-BBN, 28029 Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Faculdad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040 Madrid, Spain; (E.Á.); (M.E.); (B.G.); (R.R.C.)
- CIBER de Bioingeniería Biomateriales y Nanomedicina CIBER-BBN, 28029 Madrid, Spain
| |
Collapse
|
45
|
Corma A, Botella P, Rivero-Buceta E. Silica-Based Stimuli-Responsive Systems for Antitumor Drug Delivery and Controlled Release. Pharmaceutics 2022; 14:pharmaceutics14010110. [PMID: 35057006 PMCID: PMC8779356 DOI: 10.3390/pharmaceutics14010110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
The administration of cytotoxic drugs in classical chemotherapy is frequently limited by water solubility, low plasmatic stability, and a myriad of secondary effects associated with their diffusion to healthy tissue. In this sense, novel pharmaceutical forms able to deliver selectively these drugs to the malign cells, and imposing a space-time precise control of their discharge, are needed. In the last two decades, silica nanoparticles have been proposed as safe vehicles for antitumor molecules due to their stability in physiological medium, high surface area and easy functionalization, and good biocompatibility. In this review, we focus on silica-based nanomedicines provided with specific mechanisms for intracellular drug release. According to silica nature (amorphous, mesostructured, and hybrids) nanocarriers responding to a variety of stimuli endogenously (e.g., pH, redox potential, and enzyme activity) or exogenously (e.g., magnetic field, light, temperature, and ultrasound) are proposed. Furthermore, the incorporation of targeting molecules (e.g., monoclonal antibodies) that interact with specific cell membrane receptors allows a selective delivery to cancer cells to be carried out. Eventually, we present some remarks on the most important formulations in the pipeline for clinical approval, and we discuss the most difficult tasks to tackle in the near future, in order to extend the use of these nanomedicines to real patients.
Collapse
|
46
|
Omar AS. Nanoformulation Safety versus Toxicity; What do the Recent Studies Tell Us? INTERNATIONAL JOURNAL OF PHARMACEUTICAL RESEARCH AND ALLIED SCIENCES 2022. [DOI: 10.51847/spfpldpsvl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Manzano M. Chronology of Global Success: 20 Years of Prof Vallet-Regí Solving Questions. Pharmaceutics 2021; 13:pharmaceutics13122179. [PMID: 34959461 PMCID: PMC8708866 DOI: 10.3390/pharmaceutics13122179] [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: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Twenty years ago, a group of bold scientists led by Prof Vallet-Regí suggested for the first time the use of mesoporous materials as potential drug delivery systems. Without knowing it; these pioneers unleashed the beast of creativity around the world because that original idea has been the inspiration of hundreds of scientific groups for the design of many versatile delivery systems based on mesoporous materials. Because the dream is not the destination, it is the journey, the present review aims to summarise the chain of events that catapulted a small and young research team from the grassroots of academia to the elite of the Biomedical Engineering field.
Collapse
Affiliation(s)
- Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza de Ramón y Cajal s/n, E-28040 Madrid, Spain;
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-28034 Madrid, Spain
| |
Collapse
|
48
|
Combination of silver nanoparticles with ineffective antibiotics against extended spectrum beta-lactamases producing isolates at Alexandria Main University Hospital, Egypt. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021. [DOI: 10.1186/s43088-021-00147-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The context and purpose of the study: The spready dissemination of resistance mechanisms among extended spectrum beta-lactamases (ESBL) producing bacterial isolates has increasingly been reported. There is an urgent need to explore the antibacterial property of nontoxic biosafe compounds. This In-vitro experimental study aimed to evaluate the effect of silver nanoparticles (AgNPs) alone and in combination with ineffective antibiotics against ESBL producing isolates.
Results
Ciprofloxacin with AgNPs combination had the highest synergistic percentage against 91.43% of ESBL Escherichia coli isolates, and it was additive against 8.57% of them. As regards ESBL K. pneumoniae isolates, AgNPs with cefotaxime were synergistic against 75.00% of them, followed by ceftazidime and ciprofloxacin (62.50%). The least effective combination was ampicillin with AgNPs. The greatest enhancement of activity of the antibiotics was observed at silver minimum inhibitory concentration (MICAg) MICAg/2 and MICAg/4. At lower AgNPs concentrations, enhanced effects were less obvious. AgNPs inhibited the production of beta-lactamase enzymes in 91.43% of E. coli and 75% of Klebsiella pneumoniae isolates.
Conclusion
AgNPs are a valuable alternative to combat drug resistance, as they had synergistic effects when combined with different ineffective antibiotics against ESBL producing bacteria. AgNPs had lowered MIC values of antibiotics by several folds. Moreover, they inhibited the production of beta-lactamase enzymes.
Collapse
|
49
|
Demirdogen RE, Emen FM, Karaçolak AI, Kılıç D, Kutlu E, Meral O. Preparation of novel CaMoO4:Eu3+-MCM-41 nanocomposites and their applications and monitoring as drug release systems. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
50
|
Huang P, Lian D, Ma H, Gao N, Zhao L, Luan P, Zeng X. New advances in gated materials of mesoporous silica for drug controlled release. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.06.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|