1
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [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: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
2
|
Ye YT, Xia HY, Li J, Wang SB, Chen AZ, Kankala RK. Nanoarchitecting intelligently encapsulated designs for improved cancer therapy. Front Bioeng Biotechnol 2025; 13:1587178. [PMID: 40375976 PMCID: PMC12078215 DOI: 10.3389/fbioe.2025.1587178] [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: 03/04/2025] [Accepted: 04/14/2025] [Indexed: 05/18/2025] Open
Abstract
Despite the success in exploring various aspects of origination and therapeutic strategies, cancer has remained one of the most dreadful metabolic disorders due to failure to eradicate tumors comprehensively and frequent recurrence because of acquired resistance to the drugs. Recently, several advancements have been evidenced in the fabrication of various smart nanocarriers encapsulated with multiple components. Several reasons for smart nanoencapsulation include the enhancement of the bioavailability of drugs, precise targetability to reduce adverse effects on normal cells, and the ability to enable controlled drug release rates at the tumor sites. In addition, these smart nanocarriers protect encapsulated therapeutic cargo from deactivation, responsively delivering it based on the physiological or pathological characteristics of tumors. In this review, we present various smart approaches for cancer therapy, including organic materials, inorganic components, and their composites, as well as biomembrane-based nanoencapsulation strategies. These nanoencapsulation strategies, along with practical applications and their potential in cancer treatment, are discussed in depth, highlighting advantages and disadvantages, as well as aiming to reveal the ultimate prospects of nanoencapsulation in enhancing drug delivery efficiency and targeted cancer therapy.
Collapse
Affiliation(s)
- Ying-Tong Ye
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
| | - Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
| | - Jie Li
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
- College of Chemical Engineering, Huaqiao University, Xiamen, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
- College of Chemical Engineering, Huaqiao University, Xiamen, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, China
| |
Collapse
|
3
|
Zhan S, Cao Z, Li J, Chen F, Lai X, Yang W, Teng Y, Li Z, Zhang W, Xie J. Iron Oxide Nanoparticles Induce Macrophage Secretion of ATP and HMGB1 to Enhance Irradiation-Led Immunogenic Cell Death. Bioconjug Chem 2025; 36:80-91. [PMID: 39680043 PMCID: PMC11740999 DOI: 10.1021/acs.bioconjchem.4c00488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
ATP (adenosine triphosphate) and HMGB1 (high mobility group box 1 protein) are key players in treatments that induce immunogenic cell death (ICD). However, conventional therapies, including radiotherapy, are often insufficient to induce ICD. In this study, we explore a strategy using nanoparticle-loaded macrophages as a source of ATP and HMGB1 to complement radiation-induced intrinsic and adaptive immune responses. To this end, we tested three inorganic particles, namely, iron oxide nanoparticles (ION), aluminum oxide nanoparticles (AON), and zinc oxide nanoparticles (ZON), in vitro with bone marrow-derived dendritic cells (BMDCs) and then in vivo in syngeneic tumor models. Our results showed that ION was the most effective of the three nanoparticles in promoting the secretion of ATP and HMGB1 from macrophages without negatively affecting macrophage survival. Secretions from ION-loaded macrophages can activate BMDCs. Intratumoral injection of ION-loaded macrophages significantly enhanced tumor infiltration and activation of dendritic cells and cytotoxic T cells. Moreover, exogenous ION macrophages can enhance the efficacy of radiotherapy. In addition, direct injection of ION can also enhance the efficacy of radiotherapy, which is attributed to ION uptake by and stimulation of endogenous macrophages. Instead of directly targeting cancer cells, our strategy targets macrophages and uses them as a secretory source of ATP and HMGB1 to enhance radiation-induced ICD. Our research introduces a new nanoparticle-based immunomodulatory approach that may have applications in radiotherapy and beyond.
Collapse
Affiliation(s)
- Shuyue Zhan
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Zhengwei Cao
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jianwen Li
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Fanghui Chen
- Department
of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Xinning Lai
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Wei Yang
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Yong Teng
- Department
of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Zibo Li
- Department
of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive
Cancer Center, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Weizhong Zhang
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jin Xie
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
4
|
Sahoo A, Dixit T, Kumari A, Gupta S, Kothandaraman R, Rajeev PP, Rao MSR, Krishnan S. Facile control of giant green-emission in multifunctional ZnO quantum dots produced in a single-step process: femtosecond pulse ablation. NANOSCALE ADVANCES 2025; 7:524-535. [PMID: 39650615 PMCID: PMC11618588 DOI: 10.1039/d4na00793j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024]
Abstract
Controlling both UV and visible emissions in ZnO quantum dots (QDs) poses a significant challenge due to the inherent introduction of defects during the growth process. We have refined the photoluminescence (PL) emission characteristics of ZnO QDs through a single-step, reagent-free femtosecond pulsed laser ablation in liquid (fs-PLAL) technique. The ratio of the near band edge (NBE) to deep-level emission (DLE), which determines the shape of the QDs' optical emission spectrum, is precisely controlled by the ablation laser pulse parameters-namely, pulse energy and temporal duration. Having established our ability to control the optical properties, we have investigated the mechanisms and physics involved in controlling optical emission. The key highlight of the work is that ablation with a fs-pulse induces substantial defect states without altering the particle size, with the extent of the effect being dependent on the pulse energy and pulse duration. The spectroscopic techniques inducing Raman spectroscopy, excitation power dependent PL and transient PL study provided deep insight into the PL emission properties of these similarly sized QDs. The improved DLE in these laser-ablated QDs is explained by a surface-recombination-layer approximation process employing steady-state and transient PL. Moreover, we have demonstrated the applicability of green emission for pH sensing within a linear range of 7-10 and highlight the inherent antibacterial properties of these QDs.
Collapse
Affiliation(s)
- Anubhab Sahoo
- Department of Physics, Indian Institute of Technology Madras Chennai-600036 India +9144 2257 5898
| | - Tejendra Dixit
- Optoelectronics and Quantum Devices Group, Department of Electronics and Communication Engineering, Indian Institute of Information Technology Design and Manufacturing Kancheepuram Chennai-600127 India
| | - Anshu Kumari
- Department of Physiology, School of Medicine, University of Maryland Baltimore-21201 USA
| | - Sharad Gupta
- Disciple of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore Indore-453552 India
| | - R Kothandaraman
- Department of Chemistry, Indian Institute of Technology Madras Chennai-600036 India
| | - P P Rajeev
- Central Laser Facility, STFC Rutherford Appleton Laboratory Harwell Oxford Didcot Oxfordshire OX11 0QX UK
| | - M S Ramachandra Rao
- Department of Physics, Indian Institute of Technology Madras Chennai-600036 India +9144 2257 5898
- Quantum Center of Excellence for Diamond and Emergent Materials (QuCenDiEM) Group, Indian Institute of Technology Madras Chennai-600036 India
| | - Sivarama Krishnan
- Department of Physics, Indian Institute of Technology Madras Chennai-600036 India +9144 2257 5898
- Quantum Center of Excellence for Diamond and Emergent Materials (QuCenDiEM) Group, Indian Institute of Technology Madras Chennai-600036 India
| |
Collapse
|
5
|
Alhaddad R, Abualsoud BM, Al-Deeb I, Nsairat H. Green synthesized Zingiber officinale-ZnO nanoparticles: anticancer efficacy against 3D breast cancer model. Future Sci OA 2024; 10:2419806. [PMID: 39539163 PMCID: PMC11572278 DOI: 10.1080/20565623.2024.2419806] [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: 11/19/2023] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: ZnO NPs were prepared via green synthesis utilizing Zingiber Officinale.Methodology: Physical characterization and biological activity were performed against 2D, and 3D spheroids MCF-7 cell lines.Results: The NPs exhibited 188.9, 175.7 and 171.2 nm size with charge of -8.2, -11.7 and -9.7 mV for the 2%, 3% and 4% formulations. XRD confirmed a wurtzite hexagonal phase. FTIR spectra showed Zn-O stretching vibrations. The 2%, 3% and 4% formulations presented IC50 values of 14.7, 26.2 and 47 μg/ml, respectively, with complete destruction of MCF-7 spheroids. Elevated TNF-α levels suggested an inflammatory-mediated mechanism of action.Conclusion: 2% Zingiber officinale-derived ZnO NPs showed antitumor potential against deserving further mechanistic and in vivo explorations.
Collapse
Affiliation(s)
- Ruqaya Alhaddad
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Bassam M Abualsoud
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ibrahim Al-Deeb
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Department of Clinical Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa, 13110, Jordan
| | - Hamdi Nsairat
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| |
Collapse
|
6
|
Akhtarshenas B, Kowsar R, Hajian M, Vash NT, Soltani L, Mahdavi AH, Esfahani MHN. ρ-Coumaric acid-zinc oxide nanoparticles improve post-thaw quality of goat spermatozoa and developmental competence of fertilized oocytes in vitro. Sci Rep 2024; 14:31971. [PMID: 39738447 PMCID: PMC11686304 DOI: 10.1038/s41598-024-83585-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Excessive production of reactive oxygen species (ROS) during cryopreservation and post-thawing affects sperm quality and subsequent fertilizing capacity. Nanoparticles (NPs) with antioxidative properties can improve sperm function and male fertility. The aim of this study was to assess the effect of 100 µM ρ-coumaric acid (ρ-CA), 0.1 µM ρ-CA-NPs (PCNPs), 150 µg/mL zinc chloride (ZnCl2), 1 µg/mL zinc oxide-NPs (ZnO-NPs), ρ-CA + ZnCl2, PCNPs + ZnO-NPs, 0.001 µM of ρ-CA loaded on ZnO-NPs (ρ-CA-ZnONPs) on goat sperm parameters and fertilizing ability after cryopreservation. Semen samples from five Saanen goats were used. Various concentrations of treatments were incubated to determine the optimal concentrations for assessing sperm motility and viability. Subsequently, samples were filled with 0.5-mL straws, frozen, and stored in liquid nitrogen (- 196 °C). Evaluations of post-thaw spermatozoa parameters and fertilizing ability were performed. Addition of ρ-CA-ZnONPs and PCNPs + ZnO-NPs significantly increased sperm viability, motility, plasma membrane integrity, blastocyst rate, and blastocyst quality compared with the other treatments. Moreover, using ρ-CA-ZnONPs significantly decreased lipid peroxidation and DNA damage compared with the other treatments. In conclusion, spermatozoa are cryotolerant, resistant to post-thaw conditions, and have fertilizing ability that can be increased by adding ρ-CA-ZnONPs as an antioxidant to goat semen extenders.
Collapse
Affiliation(s)
- Bahareh Akhtarshenas
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Rasoul Kowsar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran.
| | - Mehdi Hajian
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Nima Tanhaei Vash
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Leila Soltani
- Department of Animal Sciences, Faculty of Agriculture and Natural Resources, Razi University, Kermanshah, Iran
| | - Amir Hossein Mahdavi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
7
|
Mai Q, Lu Y, Cai Q, Hu J, Lv Y, Yang Y, Wang L, Zhou Y, Liu J. pH and Pectinase Dual-Responsive Zinc Oxide Core-Shell Nanopesticide: Efficient Control of Sclerotinia Disease and Reduction of Environmental Risks. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2022. [PMID: 39728558 PMCID: PMC11728501 DOI: 10.3390/nano14242022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Sclerotinia sclerotiorum is one of the fungi that cause plant diseases. It damages plants by secreting large amounts of oxalic acid and cell wall-degrading enzymes. To meet this challenge, we designed a new pH/enzyme dual-responsive nanopesticide Pro@ZnO@Pectin (PZP). This nanopesticide uses zinc oxide (ZnO) as a carrier of prochloraz (Pro) and is encapsulated with pectin. When encountering oxalic acid released by Sclerotinia sclerotiorum, the acidic environment promotes the decomposition of ZnO; at the same time, the pectinase produced by Sclerotinia sclerotiorum can also decompose the outer pectin layer of PZP, thereby promoting the effective release of the active ingredient. Experimental data showed that PZP was able to achieve an efficient release rate of 57.25% and 68.46% when pectinase was added or under acidic conditions, respectively. In addition, in vitro tests showed that the antifungal effect of PZP was comparable to that of the commercial Pro (Pro SC) on the market, and its efficacy was 1.40 times and 1.32 times that of the Pro original drug (Pro TC), respectively. Crucially, the application of PZP significantly alleviated the detrimental impacts of Pro on wheat development. Soil wetting experiments have proved that PZP primarily remained in the soil, thereby decreasing its likelihood of contaminating water sources and reducing potential risks to non-target organisms. Moreover, PZP improved the foliar wettability of Pro, lowering the contact angle to 75.06°. Residue analyses indicated that PZP did not elevate prochloraz residue levels in tomato fruits compared to conventional applications, indicating that the nanopesticide formulation does not lead to excessive pesticide buildup. In summary, the nanopesticide PZP shows great promise for effectively managing Sclerotinia sclerotiorum while minimizing environmental impact.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jie Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China; (Q.M.); (Y.L.); (Q.C.); (J.H.); (Y.L.); (Y.Y.); (L.W.); (Y.Z.)
| |
Collapse
|
8
|
Feng Y, Tang Q, Wang B, Yang Q, Zhang Y, Lei L, Li S. Targeting the tumor microenvironment with biomaterials for enhanced immunotherapeutic efficacy. J Nanobiotechnology 2024; 22:737. [PMID: 39605063 PMCID: PMC11603847 DOI: 10.1186/s12951-024-03005-2] [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: 01/16/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
The tumor microenvironment (TME) is a complex system characterized by low oxygen, low pH, high pressure, and numerous growth factors and protein hydrolases that regulate a wide range of biological behaviors in the tumor and have a profound impact on cancer progression. Immunotherapy is an innovative approach to cancer treatment that activates the immune system, resulting in the spontaneous killing of tumor cells. However, the therapeutic efficacy of these clinically approved cancer immunotherapies (e.g., immune checkpoint blocker (ICB) therapies and chimeric antigen receptor (CAR) T-cell therapies) is far from satisfactory due to the presence of immunosuppressive TMEs created in part by tumor hypoxia, acidity, high levels of reactive oxygen species (ROS), and a dense extracellular matrix (ECM). With continuous advances in materials science and drug-delivery technologies, biomaterials hold considerable potential for targeting the TME. This article reviews the advances in biomaterial-based targeting of the TME to advance our current understanding on the role of biomaterials in enhancing tumor immunity. In addition, the strategies for remodeling the TME offer enticing advantages; however, the represent a double-edged sword. In the process of reshaping the TME, the risk of tumor growth, infiltration, and distant metastasis may increase.
Collapse
Affiliation(s)
- Yekai Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Bin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
9
|
Patel KD, Keskin-Erdogan Z, Sawadkar P, Nik Sharifulden NSA, Shannon MR, Patel M, Silva LB, Patel R, Chau DYS, Knowles JC, Perriman AW, Kim HW. Oxidative stress modulating nanomaterials and their biochemical roles in nanomedicine. NANOSCALE HORIZONS 2024; 9:1630-1682. [PMID: 39018043 DOI: 10.1039/d4nh00171k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Many pathological conditions are predominantly associated with oxidative stress, arising from reactive oxygen species (ROS); therefore, the modulation of redox activities has been a key strategy to restore normal tissue functions. Current approaches involve establishing a favorable cellular redox environment through the administration of therapeutic drugs and redox-active nanomaterials (RANs). In particular, RANs not only provide a stable and reliable means of therapeutic delivery but also possess the capacity to finely tune various interconnected components, including radicals, enzymes, proteins, transcription factors, and metabolites. Here, we discuss the roles that engineered RANs play in a spectrum of pathological conditions, such as cancer, neurodegenerative diseases, infections, and inflammation. We visualize the dual functions of RANs as both generator and scavenger of ROS, emphasizing their profound impact on diverse cellular functions. The focus of this review is solely on inorganic redox-active nanomaterials (inorganic RANs). Additionally, we deliberate on the challenges associated with current RANs-based approaches and propose potential research directions for their future clinical translation.
Collapse
Affiliation(s)
- Kapil D Patel
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Zalike Keskin-Erdogan
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
- Department of Chemical Engineering, Imperial College London, Exhibition Rd, South Kensington, SW7 2BX, London, UK
| | - Prasad Sawadkar
- Division of Surgery and Interventional Science, UCL, London, UK
- The Griffin Institute, Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospitals, London, HA1 3UJ, UK
| | - Nik Syahirah Aliaa Nik Sharifulden
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Mark Robert Shannon
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Women University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Lady Barrios Silva
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Rajkumar Patel
- Energy & Environment Sciences and Engineering (EESE), Integrated Sciences and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdongwahak-ro, Yeonsungu, Incheon 21938, Republic of Korea
| | - David Y S Chau
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Jonathan C Knowles
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Adam W Perriman
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
10
|
Wang A, Madden LA, Paunov VN. Enhanced anticancer effect of lysozyme-functionalized metformin-loaded shellac nanoparticles on a 3D cell model: role of the nanoparticle and payload concentrations. Biomater Sci 2024; 12:4735-4746. [PMID: 39083027 DOI: 10.1039/d4bm00692e] [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: 09/11/2024]
Abstract
Here we used a 3D human hepatic tumour cell culture model to assess the in vitro efficacy of "active" metformin-loaded nanoparticles (NPs) as anticancer therapeutics. The metformin nanocarrier design was repurposed from previous studies targeting bacterial and fungal biofilms with antimicrobials loaded in protease-coated nanoparticles. These active nanocarriers were constructed with shellac cores loaded with metformin as the anticancer agent and featured a surface coating of the cationic protease lysozyme. The lysozyme's role as a nanocarrier surface coating is to partially digest the extracellular matrix (ECM) of the 3D tumour cell culture which increases its porosity and the nanocarrier penetration. Hep-G2 hepatic 3D clusteroids were formed using a water-in-water (w/w) Pickering emulsion based on an aqueous two-phase system (ATPS). Our specific metformin nano-formulation, comprising 0.25 wt% lysozyme-coated, 0.4 wt% metformin-loaded, 0.2 wt% shellac NPs sterically stabilized with 0.25 wt% Poloxamer 407, demonstrated significantly enhanced anticancer efficiency on 3D hepatic tumour cell clusteroids. We examined the role of the lysozyme surface functionality of the metformin nanocarriers in their ability to kill both 2D and 3D hepatic tumour cell cultures. The anticancer efficiency at high metformin payloads was compared with that at a high concentration of nanocarriers with a lower metformin payload. It was discovered that the high metformin payload NPs were more efficient than the lower metformin payload NPs with a higher nanocarrier concentration. This study introduces a reliable in vitro model for potential targeting of solid tumours with smart nano-therapeutics, presenting a viable alternative to animal testing for evaluating anticancer nanotechnologies.
Collapse
Affiliation(s)
- Anheng Wang
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau SAR, China
- Zhuhai UM Science and Technology Research Institute, University of Macau, Hengqin, Guangdong, China
| | - Leigh A Madden
- Centre for Biomedicine, Hull York Medical School, University of Hull, HU67RX, UK
| | - Vesselin N Paunov
- Department of Chemistry, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana, 010000, Kazakhstan.
| |
Collapse
|
11
|
Ansari AA, Lv R, Gai S, Parchur AK, Solanki PR, Archana, Ansari Z, Dhayal M, Yang P, Nazeeruddin M, Tavakoli MM. ZnO nanostructures – Future frontiers in photocatalysis, solar cells, sensing, supercapacitor, fingerprint technologies, toxicity, and clinical diagnostics. Coord Chem Rev 2024; 515:215942. [DOI: 10.1016/j.ccr.2024.215942] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Khatua R, Bhar B, Dey S, Jaiswal C, J V, Mandal BB. Advances in engineered nanosystems: immunomodulatory interactions for therapeutic applications. NANOSCALE 2024; 16:12820-12856. [PMID: 38888201 DOI: 10.1039/d4nr00680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Advances in nanotechnology have led to significant progress in the design and fabrication of nanoparticles (NPs) with improved therapeutic properties. NPs have been explored for modulating the immune system, serving as carriers for drug delivery or vaccine adjuvants, or acting as therapeutics themselves against a wide range of deadly diseases. The combination of NPs with immune system-targeting moieties has facilitated the development of improved targeted immune therapies. Targeted delivery of therapeutic agents using NPs specifically to the disease-affected cells, distinguishing them from other host cells, offers the major advantage of concentrating the therapeutic effect and reducing systemic side effects. Furthermore, the properties of NPs, including size, shape, surface charge, and surface modifications, influence their interactions with the targeted biological components. This review aims to provide insights into these diverse emerging and innovative approaches that are being developed and utilized for modulating the immune system using NPs. We reviewed various types of NPs composed of different materials and their specific application for modulating the immune system. Furthermore, we focused on the mechanistic effects of these therapeutic NPs on primary immune components, including T cells, B cells, macrophages, dendritic cells, and complement systems. Additionally, a recent overview of clinically approved immunomodulatory nanomedicines and potential future perspectives, offering new paradigms of this field, is also highlighted.
Collapse
Affiliation(s)
- Rupam Khatua
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
| | - Bibrita Bhar
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
| | - Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India
| | - Chitra Jaiswal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
| | - Victoria J
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India
| |
Collapse
|
13
|
Kawish M, Siddiqui NN, Jahan H, Elhissi A, Zahid H, Khatoon B, Raza Shah M. Targeted pH-responsive delivery of rosmarinic acid via phenylboronic acid functionalized mesoporous silica nanoparticles for liver and lung cancer therapy. Pharm Dev Technol 2024; 29:541-550. [PMID: 38769920 DOI: 10.1080/10837450.2024.2356210] [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: 11/01/2023] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
Currently, chemotherapy is one of the most practiced approaches for the treatment of cancers. However, existing chemotherapeutic drugs have poor aqueous solubility, poor selectivity, higher systematic toxicity, and poor target accumulation. In this study, we designed and synthesized a boronic acid/ester-based pH-responsive nano-valve that specifically targets the microenvironment in cancer cells. The nano-valve comprises phenylboronic acid-coated mesoporous silica nanoparticles (B-MSN) loaded with polyphenolic compound Rosmarinic acid (ROS-B-MSN). The nano-valve was further coated with lignin (LIG) to achieve our desired LIG-ROS-BMSN nano-valve for targeted chemotherapy against Hep-G2 and NCI-H460 cell lines. The structure and properties of NPs were characterized by Fourier-transformed infrared spectroscopy (FTIR), Scanning Electron Microscopy (SEM) in combination with EDX, and Dynamic light scattering (DLS). The outcomes revealed that the designed LIG-ROS-BMSN were in the nanorange (144.1 ± 0.70 nm), had negative Zeta potential (-15.7 ± 0.46 mV) and had a nearly spherical morphology. In vitro, drug release investigations showed a controlled pH-dependent release profile under mild acidic conditions that could enhance the targeted chemotherapeutic response against cancer in mild acidic environments. The obtained LIG-ROS-BMSN nano valve achieved significantly lower IC50 values of (1.70 ± 0.01 μg/mL and 3.25 ± 0.14 μg/mL) against Hep-G2 and NCI-H460 cell lines as compared to ROS alone, which was (14.0 ± 0.7 μg/mL and 29.10 ± 0.25 μg/mL), respectively. The cellular morphology before and after treatment was further confirmed via inverted microscopy. The outcomes of the current study imply that our designed LIG-ROS-BMSN nanovalve is a potential carrier for cancer chemotherapeutics.
Collapse
Affiliation(s)
- Muhammad Kawish
- International Center for Chemical and Biological Sciences, H.E.J Research Institute of Chemistry, University of Karachi, Karachi, Pakistan
| | - Nimra Naz Siddiqui
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Abdelbari Elhissi
- College of Pharmacy, QU Health, and Office of VP for Research and Graduate Studies, Qatar University, Doha, Qatar
| | - Hina Zahid
- Department of Pharmaceutical Sciences, Dow University of Health Sciences Ojha Campus Karachi, Pakistan
| | - Bushra Khatoon
- International Center for Chemical and Biological Sciences, H.E.J Research Institute of Chemistry, University of Karachi, Karachi, Pakistan
| | - Muhammad Raza Shah
- International Center for Chemical and Biological Sciences, H.E.J Research Institute of Chemistry, University of Karachi, Karachi, Pakistan
| |
Collapse
|
14
|
Swidan MM, Marzook F, Sakr TM. pH-Sensitive doxorubicin delivery using zinc oxide nanoparticles as a rectified theranostic platform: in vitro anti-proliferative, apoptotic, cell cycle arrest and in vivo radio-distribution studies. J Mater Chem B 2024; 12:6257-6274. [PMID: 38845545 DOI: 10.1039/d4tb00615a] [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: 06/28/2024]
Abstract
Despite enormous advancements in its management, cancer is the world's primary cause of mortality. Therefore, tremendous strides were made to produce intelligent theranostics with mitigated side effects and improved specificity and efficiency. Thus, we developed a pH-sensitive theranostic platform composed of dextran immobilized zinc oxide nanoparticles, loaded with doxorubicin and radiolabeled with the technetium-99m radionuclide (99mTc-labelled DOX-loaded ZnO@dextran). The platform measured 11.5 nm in diameter with -12 mV zeta potential, 88% DOX loading efficiency and 98.5% radiolabeling efficiency. It showed DOX release in a pH-responsive manner, releasing 93.1% cumulatively at pH 5 but just 7% at pH 7.4. It showed improved intracellular uptake, which resulted in a high growth suppressive effect against MCF-7 cancer cells as compared to the free DOX. It boasted a 4 times lower IC50 than DOX, indicating its significant anti-proliferative potential (0.14 and 0.55 μg ml-1, respectively). The in vitro biological evaluation revealed that its molecular mode of anti-proliferative action included downregulating Cdk-2, which provoked G1/S cell cycle arrest, and upregulating both the intracellular ROS level and caspase-3, which induced apoptosis and necrosis. The in vivo experiments in Ehrlich-ascites carcinoma bearing mice demonstrated that DOX-loaded ZnO@dextran showed a considerable 4-fold increase in anti-tumor efficacy compared to DOX. Moreover, by utilizing the diagnostic radionuclide (99mTc), the radiolabeled platform (99mTc-labelled DOX-loaded ZnO@dextran) was in vivo monitored in tumor-bearing mice, revealing high tumor accumulation (14% ID g-1 at 1 h p.i.) and reduced uptake in non-target organs with a 17.5 T/NT ratio at 1 h p.i. Hence, 99mTc-labelled DOX-loaded ZnO@dextran could be recommended as a rectified tumor-targeted theranostic platform.
Collapse
Affiliation(s)
- Mohamed M Swidan
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, PO13759, Cairo, Egypt.
| | - Fawzy Marzook
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, PO13759, Cairo, Egypt.
| | - Tamer M Sakr
- Radioactive Isotopes and Generator Department, Hot Labs Center, Egyptian Atomic Energy Authority, PO13759, Cairo, Egypt
| |
Collapse
|
15
|
Ma S, Tian Z, Liu L, Zhu J, Wang J, Zhao S, Zhu Y, Zhu J, Wang W, Jiang R, Qu Y, Lei J, Zhao J, Jiang T. Cold to Hot: Tumor Immunotherapy by Promoting Vascular Normalization Based on PDGFB Nanocomposites. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308638. [PMID: 38018295 DOI: 10.1002/smll.202308638] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/04/2023] [Indexed: 11/30/2023]
Abstract
Immunotherapy is a promising cancer therapeutic strategy. However, the "cold" tumor immune microenvironment (TIME), characterized by insufficient immune cell infiltration and immunosuppressive status, limits the efficacy of immunotherapy. Tumor vascular abnormalities due to defective pericyte coverage are gradually recognized as a profound determinant in "cold" TIME establishment by hindering immune cell trafficking. Recently, several vascular normalization strategies by improving pericyte coverage have been reported, whereas have unsatisfactory efficacy and high rates of resistance. Herein, a combinatorial strategy to induce tumor vasculature-targeted pericyte recruitment and zinc ion-mediated immune activation with a platelet-derived growth factor B (PDGFB)-loaded, cyclo (Arg-Gly-Asp-D-Phe-Lys)-modified zeolitic imidazolate framework 8 (PDGFB@ZIF8-RGD) nanoplatform is proposed. PDGFB@ZIF8-RGD effectively induced tumor vascular normalization, which facilitated trafficking and infiltration of immune effector cells, including natural killer (NK) cells, M1-like macrophages and CD8+ T cells, into tumor microenvironment. Simultaneously, vascular normalization promoted the accumulation of zinc ions inside tumors to trigger effector cell immune activation and effector molecule production. The synergy between these two effects endowed PDGFB@ZIF8-RGD with superior capabilities in reprogramming the "cold" TIME to a "hot" TIME, thereby initiating robust antitumor immunity and suppressing tumor growth. This combinatorial strategy for improving immune effector cell infiltration and activation is a promising paradigm for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Shouzheng Ma
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Zhimin Tian
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Lei Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, China
| | - Jun Zhu
- The Southern Theater Air Force Hospital, Guangzhou, 510000, China
| | - Jing Wang
- Department of Immunology, Air Force Medical University, Xi'an, 710032, China
| | - Shoujie Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yejing Zhu
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Wenchen Wang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Runmin Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yongquan Qu
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Junlong Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Medical Genetics and Development Biology, Air Force Medical University, Xi'an, 710032, China
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, 710000, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| |
Collapse
|
16
|
Kapała A, Folwarski M, Gazi A. Cross-sectional observational study: Investigation of zinc concentration in white patients with cancer. Nutrition 2024; 117:112235. [PMID: 37924623 DOI: 10.1016/j.nut.2023.112235] [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: 06/26/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 11/06/2023]
Abstract
OBJECTIVES Numerous studies describe the role of zinc in the immune system and metabolism. Zinc may influence the pathogenesis and prognosis of cancer. The aim of this study to determine the prevalence of zinc deficiency in patients with cancer. The study's primary objective was to evaluate the frequency of zinc deficiency in White patients with cancer and characterize the clinical factors predisposing individuals to decreased zinc concentration. The study also aimed to estimate the dose of zinc supplementation that would prevent deficiency. METHODS Retrospective data for this cross-sectional study were analyzed from 300 consecutive white patients diagnosed with neoplastic disease and admitted to a major oncology hospital for treatment. Zinc plasma concentration, nutritional status, body composition, and medical history of ailments and dysphagia were recorded. Supplementation was introduced in patients with zinc deficiency according to the local protocol. Zinc plasma levels were collected at follow-up visits. RESULTS Zinc deficiency was diagnosed in 68% of the patients. Poor nutritional status was significantly associated with zinc deficiency (low body mass index, weight loss, low albumin level). Low lean body mass (P = 0.003) and adipose tissue (P = 0.045) correlated with zinc deficiency. Patients with zinc deficiency reported dysphagia more frequently than those with normal zinc levels (18 versus 8%; P = 0.03). Squamous cell carcinoma was significantly associated with zinc deficiency (P = 0.043). Oral zinc supplementation resulted in reaching laboratory norms for plasma concentration in only 27% of patients with zinc deficiency and was not dependent on lower (10-15 mg) or higher (25-30 mg) dosing (P > 0.05). CONCLUSIONS Zinc deficiency is common in cachectic, malnourished patients with cancer. Nutritional guidelines for these patients should include screening for micronutrient deficiencies. Further studies are needed to determine the role, dosage, duration, and form of nutritional supplementation recommended for specific cancer diagnoses.
Collapse
Affiliation(s)
- Aleksandra Kapała
- Department of Oncology Diagnostics, Cardio-Oncology and Palliative Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Marcin Folwarski
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, Gdańsk, Poland
| | - Aleksandra Gazi
- Department of Clinical Nutrition - Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
17
|
Kahandal A, Chaudhary S, Methe S, Nagwade P, Sivaram A, Tagad CK. Galactomannan polysaccharide as a biotemplate for the synthesis of zinc oxide nanoparticles with photocatalytic, antimicrobial and anticancer applications. Int J Biol Macromol 2023; 253:126787. [PMID: 37690639 DOI: 10.1016/j.ijbiomac.2023.126787] [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: 05/07/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
Biotemplates provide a facile, rapid, and environmentally benign route for synthesizing various nanostructured materials. Herein, Locust Bean Gum (LBG), a galactomannan polysaccharide, has been used as a biotemplate for synthesizing ZnO nanoparticles (NPs) for the first time. The composition, structure, morphology, and bandgap of ZnO were investigated by Energy Dispersive X-ray Spectroscopy (EDX), X-Ray Photoelectron Spectroscopy (XPS), X-ray powder diffraction (XRD), Scanning Electron Microscopy (SEM), Transmission Electron Microscopy (TEM) and UV-vis spectroscopy. XRD data showed single-phase crystalline hexagonal NPs. FTIR spectra confirmed the presence of M-O bonding in the sample. At a concentration of 0.5 mg/mL the NPs can degrade Rhodamine B under sunlight, displaying excellent photocatalytic activity. These NPs exhibited antimicrobial activity in both Staphylococcus aureus and Bacillus subtilis. Significant cell death was observed at 500 μg/mL, 250 μg/mL, 125 μg/mL and 62.5 μg/mL of NP in breast cancer, ovarian cancer and lung cancer cell lines. Wound healing assay showed that the NPs significantly blocked the cell migration at a concentration as low as 62.5 μg/mL in all three cell lines. Further optimization of the nanostructure properties will make it a promising candidate in the field of nano-biotechnology and bioengineering owing to its wide range of potential applications.
Collapse
Affiliation(s)
- Amol Kahandal
- MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India
| | - Sanyukta Chaudhary
- MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India
| | - Saakshi Methe
- MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India
| | - Pratik Nagwade
- Department of Chemistry, Shri Anand College, Pathardi, Ahmednagar, MH, India
| | - Aruna Sivaram
- MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India.
| | - Chandrakant K Tagad
- MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India.
| |
Collapse
|
18
|
Sungu M, Isik M, Güler Ü, Eylem CC, Eskizengin H, Nemutlu E, Salih B, Derkus B. Manipulating macrophage polarization with nanoparticles to control metastatic behavior in heterotypic breast cancer micro-tissues via exosome signaling. NANOSCALE 2023; 16:394-410. [PMID: 38073471 DOI: 10.1039/d3nr04980a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
This study aimed to investigate the effects of nanoparticles on macrophage polarization and their subsequent influence on post-tumorigenic behavior. Initially, seven different nanoparticles were applied to macrophages, and Zn-Ni-FeO (100 nm) and palladium nanoparticles (PdNPs, ∼25 nm) were found to induce M1-polarization in macrophages. A co-culture experiment was then conducted to examine the effects of macrophages on MCF-7 breast cancer micro-tissues. The M2-macrophages promoted tumor proliferation, while M1- and PdNPs-induced macrophages showed anti-tumor effects by suppressing cell proliferation. To reveal the mechanisms of effect, exosomes isolated from M1 (M1-Exo), M0 (M0-Exo), M2 (M2-Exo), and PdNPs-induced (PdNPs-Exo) macrophages were applied to the heterotypic tumor micro-tissues including MCF-7, human umbilical vein endothelial cells (HUVECs), and primary human dermal fibroblasts (phDFs). M2-Exo was seen to promote the migration of cancer cells and induce epithelial-mesenchymal transition (EMT), while M1-Exo suppressed these behaviors. PdNPs-Exo was effective in suppressing the aggressive nature of breast cancer cells similar to M1-Exo, moreover, the efficacy of 5-fluorouracil (5-FU) was increased in combination with PdNPs-Exo in both MCF-7 and heterotypic micro-tissues. In conclusion, PdNPs-Exo has potential anti-tumor effects, can be used as a combination therapy to enhance the efficacy of anti-cancer drugs, as well as innovative implants for breast cancer treatment.
Collapse
Affiliation(s)
- Mustafa Sungu
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
| | - Melis Isik
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
| | - Ülkü Güler
- Department of Chemistry, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
| | - Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Hakan Eskizengin
- Department of Biology, Faculty of Science, Ankara University, 06560 Ankara, Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Bekir Salih
- Department of Chemistry, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
- Neuroscience and Neurotechnology Excellence Joint Application and Research Center (NEUROM), 06560 Ankara, Turkey
| |
Collapse
|
19
|
Abdikakharovich SA, Rauf MA, Khattak S, Shah JA, Al-Keridis LA, Alshammari N, Saeed M, Igorevich SA. Exploring the antibacterial and dermatitis-mitigating properties of chicken egg white-synthesized zinc oxide nano whiskers. Front Cell Infect Microbiol 2023; 13:1295593. [PMID: 38099219 PMCID: PMC10719619 DOI: 10.3389/fcimb.2023.1295593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 10/16/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Zinc oxide nanoparticles (ZnO-NPs) have garnered considerable interest in biomedical research primarily owing to their prospective therapeutic implications in combatting pathogenic diseases and microbial infections. The primary objective of this study was to examine the biosynthesis of zinc oxide nanowhiskers (ZnO-NWs) using chicken egg white (albumin) as a bio-template. Furthermore, this study aimed to explore the potential biomedical applications of ZnO NWs in the context of infectious diseases. Methods The NWs synthesized through biological processes were observed using electron microscopy, which allowed for detailed examination of their characteristics. The results of these investigations indicated that the NWs exhibited a size distribution ranging from approximately 10 to 100 nm. Fourier-transform infrared spectroscopy (FTIR) and scanning electron microscopy-energy dispersive X-ray spectroscopy (SEM-EDX) mapping analyses successfully corroborated the size, dimensions, and presence of biological constituents during their formation. In this study, XTT assay and confocal imaging were employed to provide evidence of the efficacy of ZnO-NWs in the eradication of bacterial biofilms. The target bacterial strains were Staphylococcus aureus and Escherichia coli. Furthermore, we sought to address pertinent concerns regarding the biocompatibility of the ZnO-NWs. This was achieved through comprehensive evaluation of the absence of cytotoxicity in normal HEK-293T and erythrocytes. Results The findings of this investigation unequivocally confirmed the biocompatibility of the ZnO-NWs. The biosynthesized ZnO-NWs demonstrated a noteworthy capacity to mitigate the dermatitis-induced consequences induced by Staphylococcus aureus in murine models after a therapeutic intervention lasting for one week. Discussion This study presents a comprehensive examination of the biosynthesis of zinc oxide nanowhiskers (ZnO-NWs) derived from chicken egg whites. These findings highlight the considerable potential of biosynthesized ZnO-NWs as a viable option for the development of therapeutic agents targeting infectious diseases. The antibacterial efficacy of ZnO-NWs against both susceptible and antibiotic-resistant bacterial strains, as well as their ability to eradicate biofilms, suggests their promising role in combating infectious diseases. Furthermore, the confirmed biocompatibility of ZnO-NWs opens avenues for their safe use in biomedical applications. Overall, this research underscores the therapeutic promise of ZnO-NWs and their potential significance in future biomedical advancements.
Collapse
Affiliation(s)
| | - Mohd A. Rauf
- School of Life Sciences, Henan University, Kaifeng, Henan, China
- Miller School of Medicine, University of Miami, Miami, FL, United States
| | | | - Junaid Ali Shah
- Department of Dermatology, Ferghana Medical Institute of Public Health, Ferghana, Uzbekistan
- College of Life Sciences, Jilin University, Changchun, China
| | | | - Nawaf Alshammari
- Biology Department, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Sadykov Aslan Igorevich
- Department of Dermatology, Ferghana Medical Institute of Public Health, Ferghana, Uzbekistan
| |
Collapse
|
20
|
Kiouri DP, Tsoupra E, Peana M, Perlepes SP, Stefanidou ME, Chasapis CT. Multifunctional role of zinc in human health: an update. EXCLI JOURNAL 2023; 22:809-827. [PMID: 37780941 PMCID: PMC10539547 DOI: 10.17179/excli2023-6335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/26/2023] [Indexed: 10/03/2023]
Abstract
Zinc is a multipurpose trace element for the human body, as it plays a crucial part in various physiological processes, such as cell growth and development, metabolism, cognitive, reproductive, and immune system function. Its significance in human health is widely acknowledged, and this has led the scientific community towards more research that aims to uncover all of its beneficial properties, especially when compared to other essential metal ions. One notable area where zinc has shown beneficial effects is in the prevention and treatment of various diseases, including cancer. This review aims to explain the involvement of zinc in specific health conditions such as cancer, coronavirus disease 2019 (COVID-19) and neurological disorders like Alzheimer's disease, as well as its impact on the gut microbiome.
Collapse
Affiliation(s)
- Despoina P. Kiouri
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
- Department of Chemistry, Laboratory of Organic Chemistry, National Kapodistrian University of Athens, 15772 Athens, Greece
| | - Evi Tsoupra
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100 Sassari, Italy
| | | | - Maria E. Stefanidou
- Department of Forensic Medicine and Toxicology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
21
|
Suliman IH, Kim K, Chen W, Kim Y, Moon JH, Son S, Nam J. Metal-Based Nanoparticles for Cancer Metalloimmunotherapy. Pharmaceutics 2023; 15:2003. [PMID: 37514189 PMCID: PMC10385358 DOI: 10.3390/pharmaceutics15072003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Although the promise of cancer immunotherapy has been partially fulfilled with the unprecedented clinical success of several immunotherapeutic interventions, some issues, such as limited response rate and immunotoxicity, still remain. Metalloimmunotherapy offers a new form of cancer immunotherapy that utilizes the inherent immunomodulatory features of metal ions to enhance anticancer immune responses. Their versatile functionalities for a multitude of direct and indirect anticancer activities together with their inherent biocompatibility suggest that metal ions can help overcome the current issues associated with cancer immunotherapy. However, metal ions exhibit poor drug-like properties due to their intrinsic physicochemical profiles that impede in vivo pharmacological performance, thus necessitating an effective pharmaceutical formulation strategy to improve their in vivo behavior. Metal-based nanoparticles provide a promising platform technology for reshaping metal ions into more drug-like formulations with nano-enabled engineering approaches. This review provides a general overview of cancer immunotherapy, the immune system and how it works against cancer cells, and the role of metal ions in the host response and immune modulation, as well as the impact of metal ions on the process via the regulation of immune cells. The preclinical studies that have demonstrated the potential of metal-based nanoparticles for cancer metalloimmunotherapy are presented for the representative nanoparticles constructed with manganese, zinc, iron, copper, calcium, and sodium ions. Lastly, the perspectives and future directions of metal-based nanoparticles are discussed, particularly with respect to their clinical applications.
Collapse
Affiliation(s)
| | - Kidong Kim
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
| | - Weihsuan Chen
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
- Department of Biological Sciences and Bioengineering, Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon 22212, Republic of Korea
| | - Yubin Kim
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
- Department of Biological Sciences and Bioengineering, Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon 22212, Republic of Korea
| | - Jeong-Hyun Moon
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
| | - Sejin Son
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
- Department of Biological Sciences and Bioengineering, Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon 22212, Republic of Korea
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
22
|
Xiao M, Tang Q, Zeng S, Yang Q, Yang X, Tong X, Zhu G, Lei L, Li S. Emerging biomaterials for tumor immunotherapy. Biomater Res 2023; 27:47. [PMID: 37194085 PMCID: PMC10189985 DOI: 10.1186/s40824-023-00369-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The immune system interacts with cancer cells in various intricate ways that can protect the individual from overproliferation of cancer cells; however, these interactions can also lead to malignancy. There has been a dramatic increase in the application of cancer immunotherapy in the last decade. However, low immunogenicity, poor specificity, weak presentation efficiency, and off-target side effects still limit its widespread application. Fortunately, advanced biomaterials effectively contribute immunotherapy and play an important role in cancer treatment, making it a research hotspot in the biomedical field. MAIN BODY This review discusses immunotherapies and the development of related biomaterials for application in the field. The review first summarizes the various types of tumor immunotherapy applicable in clinical practice as well as their underlying mechanisms. Further, it focuses on the types of biomaterials applied in immunotherapy and related research on metal nanomaterials, silicon nanoparticles, carbon nanotubes, polymer nanoparticles, and cell membrane nanocarriers. Moreover, we introduce the preparation and processing technologies of these biomaterials (liposomes, microspheres, microneedles, and hydrogels) and summarize their mechanisms when applied to tumor immunotherapy. Finally, we discuss future advancements and shortcomings related to the application of biomaterials in tumor immunotherapy. CONCLUSION Research on biomaterial-based tumor immunotherapy is booming; however, several challenges remain to be overcome to transition from experimental research to clinical application. Biomaterials have been optimized continuously and nanotechnology has achieved continuous progression, ensuring the development of more efficient biomaterials, thereby providing a platform and opportunity for breakthroughs in tumor immunotherapy.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
23
|
Yu Q, Shi W, Li S, Liu H, Zhang J. Emerging Advancements in Piezoelectric Nanomaterials for Dynamic Tumor Therapy. Molecules 2023; 28:molecules28073170. [PMID: 37049933 PMCID: PMC10095813 DOI: 10.3390/molecules28073170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Cancer is one of the deadliest diseases, having spurred researchers to explore effective therapeutic strategies for several centuries. Although efficacious, conventional chemotherapy usually introduces various side effects, such as cytotoxicity or multi−drug resistance. In recent decades, nanomaterials, possessing unique physical and chemical properties, have been used for the treatment of a wide range of cancers. Dynamic therapies, which can kill target cells using reactive oxygen species (ROS), are promising for tumor treatment, as they overcome the drawbacks of chemotherapy methods. Piezoelectric nanomaterials, featuring a unique property to convert ultrasound vibration energy into electrical energy, have also attracted increasing attention in biomedical research, as the piezoelectric effect can drive chemical reactions to generate ROS, leading to the newly emerging technique of ultrasound−driven tumor therapy. Piezoelectric materials are expected to bring a better solution for efficient and safe cancer treatment, as well as patient pain relief. In this review article, we highlight the most recent achievements of piezoelectric biomaterials for tumor therapy, including the mechanism of piezoelectric catalysis, conventional piezoelectric materials, modified piezoelectric materials and multifunctional piezoelectric materials for tumor treatment.
Collapse
Affiliation(s)
- Qian Yu
- School of Life Science, Jiangsu University, Zhenjiang 212013, China
| | - Wenhui Shi
- School of Life Science, Jiangsu University, Zhenjiang 212013, China
| | - Shun Li
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Hong Liu
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Jianming Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
24
|
Liu T, Wang L, Jiang R, Tang Y, He Y, Sun C, Lv Y, Liu S. Fluorescence Properties of ZnOQDs-GO-g-C 3N 4 Nanocomposites. MICROMACHINES 2023; 14:711. [PMID: 37420944 PMCID: PMC10145813 DOI: 10.3390/mi14040711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 07/09/2023]
Abstract
In this paper, the fluorescence properties of ZnOQD-GO-g-C3N4 composite materials (ZCGQDs) were studied. Firstly, the addition of a silane coupling agent (APTES) in the synthesis process was explored, and it was found that the addition of 0.04 g·mL-1 APTES had the largest relative fluorescence intensity and the highest quenching efficiency. The selectivity of ZCGQDs for metal ions was also investigated, and it was found that ZCGQDs showed good selectivity for Cu2+. ZCGQDs were optimally mixed with Cu2+ for 15 min. ZCGQDs also had good anti-interference capability toward Cu2+. There was a linear relationship between the concentration of Cu2+ and the fluorescence intensity of ZCGQDs in the range of 1~100 µM. The regression equation was found to be F0/F = 0.9687 + 0.12343C. The detection limit of Cu2+ was about 1.74 μM. The quenching mechanism was also analyzed.
Collapse
Affiliation(s)
- Tianze Liu
- College of Clinical Medicine, Jiamusi University, Jiamusi 154007, China
| | - Lei Wang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Ruxue Jiang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Yashi Tang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Yuxin He
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Changze Sun
- School of Mechanical Engineering, Jiamusi University, Jiamusi 154007, China
| | - Yuguang Lv
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Shuang Liu
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, China
| |
Collapse
|
25
|
Petrikaite V, D'Avanzo N, Celia C, Fresta M. Nanocarriers overcoming biological barriers induced by multidrug resistance of chemotherapeutics in 2D and 3D cancer models. Drug Resist Updat 2023; 68:100956. [PMID: 36958083 DOI: 10.1016/j.drup.2023.100956] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Multidrug resistance (MDR) is currently a big challenge in cancer therapy and limits its success in several patients. Tumors use the MDR mechanisms to colonize the host and reduce the efficacy of chemotherapeutics that are injected as single agents or combinations. MDR mechanisms are responsible for inactivation of drugs and formbiological barriers in cancer like the drug efflux pumps, aberrant extracellular matrix, hypoxic areas, altered cell death mechanisms, etc. Nanocarriers have some potential to overcome these barriers and improve the efficacy of chemotherapeutics. In fact, they are versatile and can deliver natural and synthetic biomolecules, as well as RNAi/DNAi, thus providing a controlled release of drugs and a synergistic effect in tumor tissues. Biocompatible and safe multifunctional biopolymers, with or without specific targeting molecules, modify the surface and interface properties of nanocarriers. These modifications affect the interaction of nanocarriers with cellular models as well as the selection of suitable models for in vitro experiments. MDR cancer cells, and particularly their 2D and 3D models, in combination with anatomical and physiological structures of tumor tissues, can boost the design and preparation of nanomedicines for anticancer therapy. 2D and 3D cancer cell cultures are suitable models to study the interaction, internalization, and efficacy of nanocarriers, the mechanisms of MDR in cancer cells and tissues, and they are used to tailor a personalized medicine and improve the efficacy of anticancer treatment in patients. The description of molecular mechanisms and physio-pathological pathways of these models further allow the design of nanomedicine that can efficiently overcome biological barriers involved in MDR and test the activity of nanocarriers in 2D and 3D models of MDR cancer cells.
Collapse
Affiliation(s)
- Vilma Petrikaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 13, LT-50162 Kaunas, Lithuania; Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania.
| | - Nicola D'Avanzo
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy; Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy
| | - Christian Celia
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 13, LT-50162 Kaunas, Lithuania; Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - Massimo Fresta
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Viale "S. Venuta" s.n.c., 88100 Catanzaro, Italy
| |
Collapse
|
26
|
Husk-like Zinc Oxide Nanoparticles Induce Apoptosis through ROS Generation in Epidermoid Carcinoma Cells: Effect of Incubation Period on Sol-Gel Synthesis and Anti-Cancerous Properties. Biomedicines 2023; 11:biomedicines11020320. [PMID: 36830857 PMCID: PMC9953567 DOI: 10.3390/biomedicines11020320] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
This study effectively reports the influence of experimental incubation period on the sol-gel production of husk-like zinc oxide nanoparticles (ZNPs) and their anti-cancerous abilities. The surface morphology of ZNPs was studied with the help of SEM. With the use of TEM, the diameter range of the ZNPs was estimated to be ~86 and ~231 nm for ZNPA and ZNPB, prepared by incubating zinc oxide for 2 and 10 weeks, respectively. The X-ray diffraction (XRD) investigation showed that ZNPs had a pure wurtzite crystal structure. On prolonging the experimental incubation, a relative drop in aspect ratio was observed, displaying a distinct blue-shift in the UV-visible spectrum. Furthermore, RBC lysis assay results concluded that ZNPA and ZNPB both demonstrated innoxious nature. As indicated by MTT assay, reactive oxygen species (ROS) release, and chromatin condensation investigations against the human epidermoid carcinoma (HEC) A431 cells, ZNPB demonstrated viable relevance to chemotherapy. Compared to ZNPB, ZNPA had a slightly lower IC50 against A431 cells due to its small size. This study conclusively describes a simple, affordable method to produce ZNP nano-formulations that display significant cytotoxicity against the skin cancer cell line A431, suggesting that ZNPs may be useful in the treatment of cancer.
Collapse
|
27
|
Hannachi E, Khan FA, Slimani Y, Rehman S, Trabelsi Z, Akhtar S, Al-Suhaimi EA. In Vitro Antimicrobial and Anticancer Peculiarities of Ytterbium and Cerium Co-Doped Zinc Oxide Nanoparticles. BIOLOGY 2022; 11:1836. [PMID: 36552345 PMCID: PMC9775757 DOI: 10.3390/biology11121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Zinc oxide nanoparticles (ZnO NPs) are a promising platform for their use in biomedical research, especially given their anticancer and antimicrobial activities. This work presents the synthesis of ZnO NPs doped with different amounts of rare-earth ions of ytterbium (Yb) and cerium (Ce) and the assessment of their anticancer and antimicrobial activities. The structural investigations indicated a hexagonal wurtzite structure for all prepared NPs. The particle size was reduced by raising the amount of Ce and Yb in ZnO. The anticancer capabilities of the samples were examined by the cell viability MTT assay. Post 48-h treatment showed a reduction in the cancer cell viability, which was x = 0.00 (68%), x = 0.01 (58.70%), x = 0.03 (80.94%) and x = 0.05 (64.91%), respectively. We found that samples doped with x = 0.01 and x = 0.05 of Yb and Ce showed a better inhibitory effect on HCT-116 cancer cells than unadded ZnO (x = 0.00). The IC50 for HCT-116 cells of Ce and Yb co-doped ZnO nanoparticles was calculated and the IC50 values were x = 0.01 (3.50 µg/mL), x = 0.05 (8.25 µg/mL), x = 0.00 (11.75 µg/mL), and x = 0.03 (21.50 µg/mL). The treatment-doped ZnO NPs caused apoptotic cell death in the HCT-116 cells. The nanoparticles showed inhibitory action on both C. albicans and E. coli. It can be concluded that doping ZnO NPs with Yb and Ce improves their apoptotic effects on cancer and microbial cells.
Collapse
Affiliation(s)
- Essia Hannachi
- Department of Nuclear Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Firdos Alam Khan
- Department of Stem Cell Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Yassine Slimani
- Department of Biophysics, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Suriya Rehman
- Department of Epidemic Diseases Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Zayneb Trabelsi
- Department of Physics, Faculty of Science of Bizerte, University of Carthage, Bizerte 7021, Tunisia
| | - Sultan Akhtar
- Department of Biophysics, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Ebtesam A. Al-Suhaimi
- Biology Department & Institute for Research and Medical Consultations (IRMC), College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
28
|
Li Y, Jiang C, Zhang X, Liao Z, Chen L, Li S, Tang S, Fan Z, Zhang Q. Inhibition of ABCC9 by zinc oxide nanoparticles induces ferroptosis and inhibits progression, attenuates doxorubicin resistance in breast cancer. Cancer Nanotechnol 2022; 13:3. [DOI: 10.1186/s12645-021-00109-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 01/21/2023] Open
Abstract
Abstract
Background
Zinc oxide nanoparticles (ZONs) are a type of nanomaterial that has presented anti-cancer properties in breast cancer (BC). However, the function of ABCC9 in BC and its correlation with ZONs are still elusive.
Methods
Here, we identified the crucial role of ABCC9 in modulating ferroptosis and doxorubicin (Dox) resistance in BC and the targeted function of ZONs to ABCC9.
Results
The silencing of ABCC9 significantly repressed the viability of BC cells. The knockdown of ABCC9 decreased the numbers of Edu-positive BC cells. Conversely, BC cell apoptosis was increased by the inhibition of ABCC9. Besides, the silencing of ABCC9 reduced the capability of migration and invasion of BC cells. Significantly, tumorigenicity analysis demonstrated that the tumor growth of BC cells was suppressed by the depletion of ABCC9 in the xenograft model of nude mice. Moreover, the treatment of ferroptosis activator erastin repressed cell viability of BC cells and ABCC9 overexpression rescued the repression. Similarly, the numbers of Edu-positive BC cells were inhibited by erastin and the overexpression of ABCC9 reversed the inhibitory effect of erastin. The levels of GSH were decreased and MDA, lipid ROS, and iron levels were increased by the treatment of erastin, while the ABCC9 overexpression could reverse these results in BC cells. Consistently, erastin suppressed the expression of ferroptosis inhibitory factors, including GPX4 and SLC7A11, in BC cells and the overexpression of ABCC9 rescued the expression. The IC50 value of Dox was reduced by the knockdown of ABCC9 in Dox-resistant BC cells (BC/Dox). The numbers of Edu-positive BC/Dox cells were attenuated by the depletion of ABCC9. Meanwhile, the apoptosis of BC/Dox cells was stimulated by the silencing of ABCC9. Furthermore, the treatment of ZONs attenuated Dox resistance of BC cells. ZONs remarkably repressed the expression of ABCC9 in BC/Dox cells. ZONs inhibited the cell viability of BC/Dox cells and the overexpression of ABCC9 reversed the repression. Moreover, the treatment of ZONs reduced GSH levels and enhanced MDA, lipid ROS, and iron levels in erastin-stimulated BC/Dox cells.
Conclusions
In conclusion, we discovered that the inhibition of ABCC9 by zinc oxide nanoparticles induces ferroptosis and attenuates Dox resistance in BC.
Collapse
|
29
|
Chai H, Wang W, Yuan X, Zhu C. Bio-Activated PEEK: Promising Platforms for Improving Osteogenesis through Modulating Macrophage Polarization. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120747. [PMID: 36550953 PMCID: PMC9774947 DOI: 10.3390/bioengineering9120747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/30/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022]
Abstract
The attention on orthopedic biomaterials has shifted from their direct osteogenic properties to their osteoimmunomodulation, especially the modulation of macrophage polarization. Presently, advanced technologies endow polyetheretherketone (PEEK) with good osteoimmunomodulation by modifying PEEK surface characteristics or incorporating bioactive substances with regulating macrophage polarization. Recent studies have demonstrated that the fabrication of a hydrophilic surface and the incorporation of bioactive substances into PEEK (e.g., zinc, calcium, and phosphate) are good strategies to promote osteogenesis by enhancing the polarization of M2 macrophages. Furthermore, the modification by other osteoimmunomodulatory composites (e.g., lncRNA-MM2P, IL-4, IL-10, and chitosan) and their controlled and desired release may make PEEK an optimal bio-activated implant for regulating and balancing the osteogenic system and immune system. The purpose of this review is to comprehensively evaluate the potential of bio-activated PEEK in polarizing macrophages into M2 phenotype to improve osteogenesis. For this objective, we retrieved and discussed different kinds of bio-activated PEEK regarding improving osteogenesis through modulating macrophage polarization. Meanwhile, the relevant challenges and outlook were presented. We hope that this review can shed light on the development of bio-activated PEEK with more favorable osteoimmunomodulation.
Collapse
Affiliation(s)
- Haobu Chai
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei 230001, China
| | - Wenzhi Wang
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei 230001, China
| | - Xiangwei Yuan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
- Correspondence: (X.Y.); (C.Z.)
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei 230001, China
- Correspondence: (X.Y.); (C.Z.)
| |
Collapse
|
30
|
Zeng J, Sun Y, Sun S, Jiang M, Zhang D, Li W, Liu Z, Shang H, Guan X, Zhang W. Leveraging Nanodrug Delivery System for Simultaneously Targeting Tumor Cells and M2 Tumor-Associated Macrophages for Efficient Colon Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50475-50484. [PMID: 36327132 DOI: 10.1021/acsami.2c11534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Tumor-associated macrophages (TAMs) widely exist in the solid tumors, which participate in the entire course of tumor development and execute momentous impacts. Therefore, manipulating TAMs has been identified as an expecting strategy with immense potential for cancer therapy. Herein, a nanodrug delivery system was leveraged for simultaneously targeting tumor cells and M2-type TAMs for efficient colon cancer therapy. The broad-spectrum anticancer chemotherapeutic drug doxorubicin (DOX) was hitchhiked in a mannose-modified bovine serum albumin (MAN-BSA) carrier. The DOX@MAN-BSA nanodrug delivery system was verified to possess feasible physical performances for unhindered systemic circulation and active targeting on colon tumors. DOX@MAN-BSA nanoparticles could be preferentially swallowed by colon tumor cells and M2 TAMs through mannose receptor-mediated endocytosis. Further in vivo antitumor therapy in CT26 colon tumor-bearing mice has achieved remarkable suppression efficacy with satisfactory biosafety. Leveraging the nanodrug delivery system for simultaneously targeting tumor cells and M2 TAMs has contributed a feasible strategy to collaboratively repress the malignant tumor cells and the collusive M2 TAMs for efficient cancer therapy.
Collapse
Affiliation(s)
- Jun Zeng
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Yanju Sun
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Shuo Sun
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Mingxia Jiang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Daijuan Zhang
- Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - Wentong Li
- Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - Zhijun Liu
- Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiuwen Guan
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Weifen Zhang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
31
|
Li Z, Zhang S, Liu M, Zhong T, Li H, Wang J, Zhao H, Tian Y, Wang H, Wang J, Xu M, Wang S, Zhang X. Antitumor Activity of the Zinc Oxide Nanoparticles Coated with Low-Molecular-Weight Heparin and Doxorubicin Complex In Vitro and In Vivo. Mol Pharm 2022; 19:4179-4190. [PMID: 36223494 DOI: 10.1021/acs.molpharmaceut.2c00553] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Various metal oxide nanomaterials have been widely used as carriers to prepare pH-sensitive nanomedicines to respond to the acidic tumor microenvironment promoting antitumor efficiency. Herein, we used zinc oxide nanoparticles (ZnO NPs) as metal oxide nanomaterial coated with low-molecular-weight heparin (LMHP) and doxorubicin (DOX) complex (LMHP-DOX) to prepare ZnO-LD NPs for controllable pH-triggered DOX release on the targeted site. Our results indicated that the released DOX from ZnO-LD NPs was pH-sensitive. The oxygen produced by ZnO-LD NPs in H2O2 solution was observed in in vitro experiment. The ZnO-LD NPs entered into both PC-3M and 4T1 tumor cells via clathrin-mediated endocytosis and micropinocytosis pathway. The intracellular reactive oxygen species (ROS) generated by ZnO-LD NPs could significantly increase the caspase 3/7 level, leading to tumor cell apoptosis. The in vitro and in vivo antitumor activity was confirmed in PC-3M and 4T1 cell lines or tumor-bearing mice models. The in vivo and in vitro tumor images via second-order nonlinearity of ZnO-LD NPs indicated that ZnO-LD NPs could penetrate deep into the tumor tissues. Therefore, the ZnO-LD NPs developed in our study could provide an efficient approach for the preparation of pH-sensitive nano delivery systems suitable for tumor therapy and imaging.
Collapse
Affiliation(s)
- Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Shuang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Man Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Ting Zhong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Heng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Yubo Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Hui Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Jingwen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Meiqi Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Shumin Wang
- Department of Ultrasound, Peking University Third Hospital, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| |
Collapse
|
32
|
Deshpande SS, Veeragoni D, Rachamalla HK, Misra S. Anticancer properties of ZnO-Curcumin nanocomposite against melanoma cancer and its genotoxicity profiling. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
33
|
Liu T, Zou L, Ji X, Xiao G. Chicken skin-derived collagen peptides chelated zinc promotes zinc absorption and represses tumor growth and invasion in vivo by suppressing autophagy. Front Nutr 2022; 9:960926. [PMID: 35990359 PMCID: PMC9381994 DOI: 10.3389/fnut.2022.960926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
To improve the utilization value of chicken by-products, we utilized the method of step-by-step hydrolysis with bromelain and flavourzyme to prepare low molecular weight chicken skin collagen peptides (CCP) (<5 kDa) and characterized the amino acids composition of the CCP. Then, we prepared novel CCP-chelated zinc (CCP–Zn) by chelating the CCP with ZnSO4. We found that the bioavailability of CCP–Zn is higher than ZnSO4. Besides, CCP, ZnSO4, or CCP–Zn effectively repressed the tumor growth, invasion, and migration in a Drosophila malignant tumor model. Moreover, the anti-tumor activity of CCP–Zn is higher than CCP or ZnSO4. Furthermore, the functional mechanism studies indicated that CCP, ZnSO4, or CCP–Zn inhibits tumor progression by reducing the autonomous and non-autonomous autophagy in tumor cells and the microenvironment. Therefore, this research provides in vivo evidence for utilizing chicken skin in the development of zinc supplements and cancer treatment in the future.
Collapse
Affiliation(s)
- Tengfei Liu
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, China.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Lifang Zou
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, China.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaowen Ji
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, China.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Guiran Xiao
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, China.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
34
|
Mahmood S, Bhattarai P, Khan NR, Subhan Z, Razaque G, Albarqi HA, Alqahtani AA, Alasiri A, Zhu L. An Investigation for Skin Tissue Regeneration Enhancement/Augmentation by Curcumin-Loaded Self-Emulsifying Drug Delivery System (SEDDS). Polymers (Basel) 2022; 14:2904. [PMID: 35890680 PMCID: PMC9315559 DOI: 10.3390/polym14142904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes, one of the global metabolic disorders, is often associated with delayed wound healing due to the elevated level of free radicals at the wound site, which hampers skin regeneration. This study aimed at developing a curcumin-loaded self-emulsifying drug delivery system (SEDDS) for diabetic wound healing and skin tissue regeneration. For this purpose, various curcumin-loaded SEDDS formulations were prepared and optimized. Then, the SEDDS formulations were characterized by the emulsion droplet size, surface charge, drug content/entrapment efficiency, drug release, and stability. In vitro, the formulations were assessed for the cellular uptake, cytotoxicity, cell migration, and inhibition of the intracellular ROS production in the NIH3T3 fibroblasts. In vivo, the formulations' wound healing and skin regeneration potential were evaluated on the induced diabetic rats. The results indicated that, after being dispersed in the aqueous medium, the optimized SEDDS formulation was readily emulsified and formed a homogenous dispersion with a droplet size of 37.29 ± 3.47 nm, surface charge of -20.75 ± 0.07 mV, and PDI value of less than 0.3. The drug content in the optimized formulation was found to be 70.51% ± 2.31%, with an encapsulation efficiency of 87.36% ± 0.61%. The SEDDS showed a delayed drug release pattern compared to the pure drug solution, and the drug release rate followed the Fickian diffusion kinetically. In the cell culture, the formulations showed lower cytotoxicity, higher cellular uptake, and increased ROS production inhibition, and promoted the cell migration in the scratch assay compared to the pure drug. The in vivo data indicated that the curcumin-loaded SEDDS-treated diabetic rats had significantly faster-wound healing and re-epithelialization compared with the untreated and pure drug-treated groups. Our findings in this work suggest that the curcumin-loaded SEDDS might have great potential in facilitating diabetic wound healing and skin tissue regeneration.
Collapse
Affiliation(s)
- Saima Mahmood
- Department of Pharmaceutics, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, KPK, Pakistan;
- Gomal Centre for Skin/Regenerative Medicine and Drug Delivery Research, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, KPK, Pakistan
| | - Prapanna Bhattarai
- Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX 77843, USA;
| | - Nauman Rahim Khan
- Gomal Centre for Skin/Regenerative Medicine and Drug Delivery Research, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, KPK, Pakistan
- Department of Pharmacy, Kohat University of Science and Technology, Kohat 26000, KPK, Pakistan
| | - Zakia Subhan
- Institute of Medical Sciences, Khyber Medical University, Kohat 26000, KPK, Pakistan;
| | - Ghulam Razaque
- Faculty of Pharmacy, University of Baluchistan, Quetta 87300, Baluchistan, Pakistan;
| | - Hassan A. Albarqi
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 55461, Saudi Arabia; (H.A.A.); (A.A.A.); (A.A.)
| | - Abdulsalam A. Alqahtani
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 55461, Saudi Arabia; (H.A.A.); (A.A.A.); (A.A.)
| | - Ali Alasiri
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 55461, Saudi Arabia; (H.A.A.); (A.A.A.); (A.A.)
| | - Lin Zhu
- Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
35
|
Tang W, Liu H, Ooi TC, Rajab NF, Cao H, Sharif R. Zinc carnosine: Frontiers advances of supplement for cancer therapy. Biomed Pharmacother 2022; 151:113157. [PMID: 35605299 DOI: 10.1016/j.biopha.2022.113157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 11/02/2022] Open
Abstract
Zinc (Zn) has an existence within large quantities in the human brain, while accumulating within synaptic vesicle. There is growing evidence that Zn metabolic equilibrium breaking participates into different diseases (e.g., vascular dementia, carcinoma, Alzheimer's disease). Carnosine refers to an endogenic dipeptide abundant in skeletal muscle and brains and exerts a variety of positive influences (e.g., carcinoma resistance, crosslinking resistance, metal chelation and oxidation limitation). A complex of Zn and carnosine, called Zinc-L-carnosine (ZnC), has been extensively employed within Zn supplement therapeutic method and the treating approach for ulcers. ZnC has been shown to play a variety of roles in the body, including inhibiting intracellular reactive oxygen species(ROS) and free radical levels, inhibiting inflammation, supplementing zinc enzymes and promoting wound healing and mucosal cell repair. The present study conducting a reviewing process for the advances of ZnC in tumor adjuvant therapy.
Collapse
Affiliation(s)
- Weiwei Tang
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia; Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Hanyuan Liu
- General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Theng Choon Ooi
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Hongyong Cao
- General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Razinah Sharif
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia; Biocompatibility Laboratory, Centre for Research and Instrumentation, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
36
|
Macrophages Are a Double-Edged Sword: Molecular Crosstalk between Tumor-Associated Macrophages and Cancer Stem Cells. Biomolecules 2022; 12:biom12060850. [PMID: 35740975 PMCID: PMC9221070 DOI: 10.3390/biom12060850] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are a subset of highly tumorigenic cells in tumors. They have enhanced self-renewal properties, are usually chemo-radioresistant, and can promote tumor recurrence and metastasis. They can recruit macrophages into the tumor microenvironment and differentiate them into tumor-associated macrophages (TAMs). TAMs maintain CSC stemness and construct niches that are favorable for CSC survival. However, how CSCs and TAMs interact is not completely understood. An understanding on these mechanisms can provide additional targeting strategies for eliminating CSCs. In this review, we comprehensively summarize the reported mechanisms of crosstalk between CSCs and TAMs and update the related signaling pathways involved in tumor progression. In addition, we discuss potential therapies targeting CSC–TAM interaction, including targeting macrophage recruitment and polarization by CSCs and inhibiting the TAM-induced promotion of CSC stemness. This review also provides the perspective on the major challenge for developing potential therapeutic strategies to overcome CSC-TAM crosstalk.
Collapse
|
37
|
Dey A, Gare S, Swain S, Bhattacharya P, Dhyani V, Giri L, Neogi S. 3D
imaging and quantification of
PLL
coated fluorescent
ZnO NP
distribution and
ROS
accumulation using
LSCM. AIChE J 2022. [DOI: 10.1002/aic.17801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Aishee Dey
- Department of Chemical Engineering Indian Institute of Technology Kharagpur India
| | - Suman Gare
- Department of Chemical Engineering Indian Institute of Technology Hyderabad India
| | - Sarpras Swain
- Department of Chemical Engineering Indian Institute of Technology Hyderabad India
| | - Proma Bhattacharya
- Department of Chemical Engineering Indian Institute of Technology Kharagpur India
| | - Vaibhav Dhyani
- Department of Chemical Engineering Indian Institute of Technology Hyderabad India
| | - Lopamudra Giri
- Department of Chemical Engineering Indian Institute of Technology Hyderabad India
| | - Sudarsan Neogi
- Department of Chemical Engineering Indian Institute of Technology Kharagpur India
| |
Collapse
|
38
|
Multifunctional Nanoplatforms as a Novel Effective Approach in Photodynamic Therapy and Chemotherapy, to Overcome Multidrug Resistance in Cancer. Pharmaceutics 2022; 14:pharmaceutics14051075. [PMID: 35631660 PMCID: PMC9143284 DOI: 10.3390/pharmaceutics14051075] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/02/2022] [Accepted: 05/14/2022] [Indexed: 12/24/2022] Open
Abstract
It is more than sixty years since the era of modern photodynamic therapy (PDT) for cancer began. Enhanced selectivity for malignant cells with a reduced selectivity for non-malignant cells and good biocompatibility along with the limited occurrence of side effects are considered to be the most significant advantages of PDT in comparison with conventional therapeutic approaches, e.g., chemotherapy. The phenomenon of multidrug resistance, which is associated with drug efflux transporters, was originally identified in relation to the application of chemotherapy. Unfortunately, over the last thirty years, numerous papers have shown that many photosensitizers are the substrates of efflux transporters, significantly restricting the effectiveness of PDT. The concept of a dynamic nanoplatform offers a possible solution to minimize the multidrug resistance effect in cells affected by PDT. Indeed, recent findings have shown that the utilization of nanoparticles could significantly enhance the therapeutic efficacy of PDT. Additionally, multifunctional nanoplatforms could induce the synergistic effect of combined treatment regimens, such as PDT with chemotherapy. Moreover, the surface modifications that are associated with nanoparticle functionalization significantly improve the target potential of PDT or chemo-PDT in multidrug resistant and cancer stem cells.
Collapse
|
39
|
A mutually beneficial macrophages-mediated delivery system realizing photo/immune therapy. J Control Release 2022; 347:14-26. [PMID: 35489548 DOI: 10.1016/j.jconrel.2022.04.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 04/24/2022] [Indexed: 11/20/2022]
Abstract
The accumulation of nanomedicines in tumor tissues determines their therapeutic efficacy. We herein exploit the tropism of macrophages to improve the accumulation and retention time of nanomedicine at tumors. Interestingly, macrophages are not merely as transporters, but killers activated by nanomedicine. The system(M@C-HA/ICG) was established by decorating macrophages with hyaluronic acid-modified hollow mesoporous carbon (C) nanoparticles loading indocyanine green (ICG). Notably, C nanoparticles with superior photothermal conversion capability not merely guarantee the efficient delivery of ICG through high drug loading efficiency and inhibiting the premature leaky, but effectually activate the polarization of macrophages. The results exhibited that those activated macrophages could release pro-inflammatory cytokines (NO, TNF-α, IL-12), while M@C-HA/ICG afforded about 2-fold higher tumor accumulation compared with pure nanoparticle C-HA/ICG and produced heat and singlet oxygen (1O2) under irradiation of an 808 nm laser, realizing the combination of photodynamic therapy (PDT), photothermal therapy (PTT) and cytokines-mediated immunotherapy. Specially, we also investigated the relationship of singlet oxygen (1O2) or temperature and tumor-killing activity for understanding the specific effectual procedure of PDT/PTT synergistic therapy. Overall, we firstly established an "all active" delivery system integrating the features of nanomedicine with biological functions of macrophages, providing a novel insight for cell-mediated delivery platform and tumor targeted multimodality anti-cancer therapy.
Collapse
|
40
|
Recent advances in ZnO-based photosensitizers: Synthesis, modification, and applications in photodynamic cancer therapy. J Colloid Interface Sci 2022; 621:440-463. [PMID: 35483177 DOI: 10.1016/j.jcis.2022.04.087] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/26/2022] [Accepted: 04/14/2022] [Indexed: 01/05/2023]
Abstract
Zinc oxide nanoparticles (ZnO NPs) are important semiconductor materials with interesting photo-responsive properties. During the past, ZnO-based NPs have received considerable attention for photodynamic therapy (PDT) due to their biocompatibility and excellent potential of generating tumor-killing reactive oxygen species (ROS) through gentle photodynamic activation. This article provides a comprehensive review of the recent developments and improvements in optical properties of ZnO NPs as photosensitizers for PDT. The optical properties of ZnO-based photosensitizers are significantly dependent on their charge separation, absorption potential, band gap engineering, and surface area, which can be adjusted/tuned by doping, compositing, and morphology control. Here, we first summarize the recent progress in the charge separation capability, absorption potential, band gap engineering, and surface area of nanosized ZnO-based photosensitizers. Then, morphology control that is closely related to their synthesis method is discussed. Following on, the state-of-art for the ZnO-based NPs in the treatment of hypoxic tumors is comprehensively reviewed. Finally, we provide some outlooks on common targeted therapy methods for more effective tumor killing, including the attachment of small molecules, antibodies, ligands molecules, and receptors to NPs which further improve their selective distribution and targeting, hence improving the therapeutic effectiveness. The current review may provide useful guidance for the researchers who are interested in this promising dynamic cancer treatment technology.
Collapse
|
41
|
Aljabali AAA, Obeid MA, Bakshi HA, Alshaer W, Ennab RM, Al-Trad B, Al Khateeb W, Al-Batayneh KM, Al-Kadash A, Alsotari S, Nsairat H, Tambuwala MM. Synthesis, Characterization, and Assessment of Anti-Cancer Potential of ZnO Nanoparticles in an In Vitro Model of Breast Cancer. Molecules 2022; 27:1827. [PMID: 35335190 PMCID: PMC8952501 DOI: 10.3390/molecules27061827] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 12/10/2022] Open
Abstract
Advanced innovations for combating variants of aggressive breast cancer and overcoming drug resistance are desired. In cancer treatment, ZnO nanoparticles (NPs) have the capacity to specifically and compellingly activate apoptosis of cancer cells. There is also a pressing need to develop innovative anti-cancer therapeutics, and recent research suggests that ZnO nanoparticles hold great potential. Here, the in vitro chemical effectiveness of ZnO NPs has been tested. Zinc oxide (ZnO) nanoparticles were synthesized using Citrullus colocynthis (L.) Schrad by green methods approach. The generated ZnO was observed to have a hexagonal wurtzite crystal arrangement. The generated nanomaterials were characterized by transmission electron microscopy (TEM), scanning electron microscopy (SEM), UV-visible spectroscopy. The crystallinity of ZnO was reported to be in the range 50-60 nm. The NPs morphology showed a strong absorbance at 374 nm with an estimated gap band of 3.20 eV to 3.32 eV. Microscopy analysis proved the morphology and distribution of the generated nanoparticles to be around 50 nm, with the elemental studies showing the elemental composition of ZnO and further confirming the purity of ZnO NPs. The cytotoxic effect of ZnO NPs was evaluated against wild-type and doxorubicin-resistant MCF-7 and MDA-MB-231 breast cancer cell lines. The results showed the ability of ZnO NPs to inhibit the prefoliation of MCF-7 and MDA-MB-231 prefoliation through the induction of apoptosis without significant differences in both wild-type and resistance to doxorubicin.
Collapse
Affiliation(s)
- Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Mohammad A. Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, County Londonderry, Northern Ireland, UK;
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (W.A.); (S.A.)
| | - Raed M. Ennab
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan;
| | - Bahaa Al-Trad
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan; (B.A.-T.); (W.A.K.); (K.M.A.-B.)
| | - Wesam Al Khateeb
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan; (B.A.-T.); (W.A.K.); (K.M.A.-B.)
| | - Khalid M. Al-Batayneh
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan; (B.A.-T.); (W.A.K.); (K.M.A.-B.)
| | | | - Shrouq Alsotari
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (W.A.); (S.A.)
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, County Londonderry, Northern Ireland, UK;
| |
Collapse
|
42
|
Chelladurai M, Margavelu G, Vijayakumar S, González-Sánchez ZI, Vijayan K, Sahadevan R. Preparation and characterization of amine-functionalized mupirocin-loaded zinc oxide nanoparticles: A potent drug delivery agent in targeting human epidermoid carcinoma (A431) cells. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Leelavathi H, Muralidharan R, Abirami N, Tamizharasan S, Kumarasamy A, Arulmozhi R. Exploration of ZnO decorated g-C3N4 amphiphilic anticancer drugs for antiproliferative activity against human cervical cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Fakhroueian Z, Rajabi S, Salehi N, Tavirani MR, Noori S, Nourbakhsh M. Anticancer properties of novel zinc oxide quantum dot nanoparticles against breast cancer stem-like cells. Anticancer Drugs 2022; 33:e311-e326. [PMID: 34419959 DOI: 10.1097/cad.0000000000001207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cancer stem cells (CSCs) play an essential role in cancer development, metastasis, relapse, and resistance to treatment. In this article, the effects of three synthesized ZnO nanofluids on proliferation, apoptosis, and stemness markers of breast cancer stem-like cells are reported. The antiproliferative and apoptotic properties of ZnO nanoparticles were evaluated on breast cancer stem-like cell-enriched mammospheres by MTS assay and flowcytometry, respectively. The expression of stemness markers, including WNT1, NOTCH1, β-catenin, CXCR4, SOX2, and ALDH3A1 was assessed by real-time PCR. Western blotting was used to analyze the phosphorylation of Janus kinase 2 (JAK2) and Signal Transducer and Activator of Transcription 3 (STAT3). Markers of stemness were significantly decreased by ZnO nanofluids, especially sample (c) with code ZnO-148 with a different order of addition of polyethylene glycol solution at the end of formulation, which considerably decreased all the markers compared to the controls. All the studied ZnO nanofluids considerably reduced viability and induced apoptosis of spheroidal and parental cells, with ZnO-148 presenting the most effective activity. Using CD95L as a death ligand and ZB4 as an extrinsic apoptotic pathway blocker, it was revealed that none of the nanoparticles induced apoptosis through the extrinsic pathway. Results also showed a marked inhibition of the JAK/STAT pathway by ZnO nanoparticles; confirmed by downregulation of Mcl-1 and Bcl-XL expression. The present data demonstrated that ZnO nanofluids could combat breast CSCs via decreasing stemness markers, stimulating apoptosis, and suppressing JAK/STAT activity.
Collapse
Affiliation(s)
- Zahra Fakhroueian
- College of Engineering, School of Chemical Engineering, Institute of Petroleum Engineering, University of Tehran
| | - Sadegh Rajabi
- Traditional Medicine and Materia Medica Research Center, Shahid Beheshti University of Medical Sciences
| | | | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Science, Shahid Beheshti University of Medical Science
| | | | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Jadhav V, Bhagare A, Ali IH, Dhayagude A, Lokhande D, Aher J, Jameel M, Dutta M. Role of Moringa oleifera on Green Synthesis of Metal/Metal Oxide Nanomaterials. JOURNAL OF NANOMATERIALS 2022; 2022. [DOI: 10.1155/2022/2147393] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 10/22/2024]
Abstract
Being an environmentally benign method biosynthesis of nanomaterial paying much more attention to researchers, it has many advantages over other routes, such as one pot, facile synthesis, and cost‐effective; synthesized material can have good affinity due to surface modification and hence became a most attractive candidate for medicinal and biological applications. Moreover, biosynthesis creates a bridge of interdisciplinary research. Biosynthesis can be done by using bacteria, microbes, plant extracts, etc. In this study, we focus on the synthesis of some metal and metal oxide nanomaterials (M/MO NMs) by using an extract of parts from the Moringa oleifera plant. It is a natural source that can serve as a capping, stabilizing, and reducing/oxidizing agent due to the presence of some of the phytochemical parameters. Moreover, it is a rich source of antioxidants, including quercetin and chlorogenic acids, such as flavonoids, phenolics, astragalin, anthocyanins, cinnamates, and carotenoids, as well as a good source of carotene, iron, potassium, calcium, terpenes, quinines, saponins, alkaloids, proteins, tannins, and vitamin. These components produce smaller particles and give a compelling impact on the activities of M/MO NMs nanoparticles. Here, we discuss nanoparticles such as FeO, CuO, ZnO, NiO, MgO, Ag, and Au.
Collapse
|
46
|
Zhu Y, Sui B, Liu X, Sun J. The reversal of drug resistance by two-dimensional titanium carbide Ti 2 C (2D Ti2C) in non-small-cell lung cancer via the depletion of intracellular antioxidant reserves. Thorac Cancer 2021; 12:3340-3355. [PMID: 34741403 PMCID: PMC8671908 DOI: 10.1111/1759-7714.14208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 12/22/2022] Open
Abstract
Background Chemoresistance is a major barrier limiting the therapeutic efficacy of late stage non‐small cell lung cancer (NSCLC). In this study, we sought to use two‐dimensional titanium carbide (2D Ti2C) to reverse cisplatin resistance in NSCLC. Methods We first achieved favorable properties as a potential anti‐tumor agent. We then compared cell viability and cisplatin uptake in chemoresistant NSCLC cells before and after the use of 2D Ti2C. Afterwards, we explored the effects of 2D Ti2C on intracellular antioxidant reserves, followed by evaluating the subsequent changes in the expression of core drug resistance genes. Finally, we confirmed the tumor inhibitory effect and bio‐safety of 2D Ti2C in a drug‐resistant lung cancer model in nude mice. Results Due to the properties of thin layer, large specific surface area, and abundant reactive groups on the surface, 2D Ti2C can deplete the antioxidant reserve systems such as the glutathione redox buffer system, γ‐glutamylcysteine synthetase (γ‐GCS), glutathione peroxidase (GPx), glutathione‐S‐transferase‐Pi (GST‐π), and metallothionein (MT), thereby increasing the intracellular accumulation of cisplatin and decreasing the expression of drug resistance genes. Conclusions 2D Ti2C can reverse NSCLC chemoresistance both in vitro and in vivo, suggesting that it may potentially become a novel and effective means to treat chemoresistant NSCLC in the clinic.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Baiyan Sui
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xin Liu
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiao Sun
- Department of Dental Materials, Shanghai Biomaterials Research & Testing Center, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
47
|
|
48
|
He Y, de Araújo Júnior RF, Cruz LJ, Eich C. Functionalized Nanoparticles Targeting Tumor-Associated Macrophages as Cancer Therapy. Pharmaceutics 2021; 13:1670. [PMID: 34683963 PMCID: PMC8540805 DOI: 10.3390/pharmaceutics13101670] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays a central role in regulating antitumor immune responses. As an important part of the TME, alternatively activated type 2 (M2) macrophages drive the development of primary and secondary tumors by promoting tumor cell proliferation, tumor angiogenesis, extracellular matrix remodeling and overall immunosuppression. Immunotherapy approaches targeting tumor-associated macrophages (TAMs) in order to reduce the immunosuppressive state in the TME have received great attention. Although these methods hold great potential for the treatment of several cancers, they also face some limitations, such as the fast degradation rate of drugs and drug-induced cytotoxicity of organs and tissues. Nanomedicine formulations that prevent TAM signaling and recruitment to the TME or deplete M2 TAMs to reduce tumor growth and metastasis represent encouraging novel strategies in cancer therapy. They allow the specific delivery of antitumor drugs to the tumor area, thereby reducing side effects associated with systemic application. In this review, we give an overview of TAM biology and the current state of nanomedicines that target M2 macrophages in the course of cancer immunotherapy, with a specific focus on nanoparticles (NPs). We summarize how different types of NPs target M2 TAMs, and how the physicochemical properties of NPs (size, shape, charge and targeting ligands) influence NP uptake by TAMs in vitro and in vivo in the TME. Furthermore, we provide a comparative analysis of passive and active NP-based TAM-targeting strategies and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Yuanyuan He
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| | - Raimundo Fernandes de Araújo Júnior
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal 59064-720, Brazil
- Cancer and Inflammation Research Laboratory (LAICI), Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59064-720, Brazil
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| | - Christina Eich
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| |
Collapse
|
49
|
Wei S, Guo W, Qian Y, Xiang J, Liu K, Gao XJ, Gao X, Chen Y. Ribosome profiling reveals translatome remodeling in cancer cells in response to zinc oxide nanoparticles. Aging (Albany NY) 2021; 13:23119-23132. [PMID: 34620733 PMCID: PMC8544296 DOI: 10.18632/aging.203606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
The anticancer effect of zinc oxide nanoparticles (ZnO NPs) largely relies on cellular responses such as alteration of gene expression. Although ZnO NPs have been reported to induce transcriptional changes, the potential of ZnO NPs to affect cellular translatome remains largely unknown. Using ribosome profiling, we demonstrated that the transcription of 78 genes and the translation of 1,448 genes are affected during one hour of ZnO NPs exposure in A549 human lung cancer cells. The mitogen-activated protein kinase (MAPK) pathway is up-regulated upon ZnO NP treatment. The upstream open reading frame (uORF) plays a pervasive role in the induction of up-regulated genes, including TLNRD1 and CCNB1IP1. Knockdown of TLNRD1 or CCNB1IP1 reduces ZnO NP-induced cytotoxicity. Together, our study characterizes the landscape of translational alteration under ZnO NPs treatment and provides potential targets to augment the anticancer effect of ZnO NPs.
Collapse
Affiliation(s)
- Saisai Wei
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Endoscopic Technique Research of Zhejiang Province, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Wenhao Guo
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Urology, Shaoxing Branch of Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Shaoxing 312000, China
| | - Yu Qian
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jie Xiang
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kangli Liu
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiang-Jing Gao
- Department of Occupational Health and Radiation Protection, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, Zhejiang, China
| | - Xiangwei Gao
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yicheng Chen
- Sir Run-Run Shaw Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
50
|
Fayed B, Jayakumar MN, Soliman SSM. Caspofungin-resistance in Candida auris is cell wall-dependent phenotype and potential prevention by zinc oxide nanoparticles. Med Mycol 2021; 59:1243-1256. [PMID: 34612496 DOI: 10.1093/mmy/myab059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 01/07/2023] Open
Abstract
Candida auris is an emergent nosocomial multi-drug-resistant yeast that represents a global health threat. Recently, C. auris clinical isolates with caspofungin resistance were identified. Mutation in FKS1 gene was determined as a mechanism of resistance. However, the ability of C. auris to develop acquired and cross-resistance has never been investigated. Herein, this resistance ability due to caspofungin and associate mechanisms were investigated. C. auris clinical isolate was successively cultured for 10 generations in the presence of caspofungin compared to fluconazole-treatment and untreated controls. This was followed by the analysis of target gene expression and phenotypic changes. The obtained results showed that caspofungin-treated C. auris exhibited elevated MIC50(caspofungin), slower growth, elevated chitin content, overexpression of caspofungin target genes, and cross-resistance to fluconazole. Interestingly, caspofungin exposure induced cell-cell adhesion and biofilm formation. C. auris gradually lost caspofungin resistance after removal of antifungal pressure, while keeping the overexpression of fungal cell wall-related genes including ALS5. We propose that C. auris ageing in the presence of caspofungin caused the development of persistent phenotypic changes in the fungal cell wall, leading to acquired and physical cross-resistance mechanisms. Surprisingly, formulation of caspofungin in zinc oxide nanoparticles prevented the aforementioned behavioral changes regardless of the pathogen generations. LAY SUMMARY Candida auris developed resistance against caspofungin. Our data indicated that this resistance mechanism is unique because of changes in the genes related to cell wall adhesions. Formulation of caspofungin in ZnO nanoparticles was able to overcome these phenotypic changes.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.,Chemistry of Natural and Microbial Product Department, National Research Centre, Cairo, 12622, Egypt
| | - Manju Nidagodu Jayakumar
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Sameh S M Soliman
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.,College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| |
Collapse
|