1
|
Zarska M, Novak O, Jakubcova T, Novotny F, Urbancokova A, Havel F, Novak J, Raabova H, Musilek K, Filimonenko V, Bartek J, Proska J, Hodny Z. Photothermal induction of pyroptosis in malignant glioma spheroids using (16-mercaptohexadecyl)trimethylammonium bromide-modified cationic gold nanorods. Colloids Surf B Biointerfaces 2024; 243:114128. [PMID: 39094210 DOI: 10.1016/j.colsurfb.2024.114128] [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: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Plasmonic photothermal therapy (PPTT) employing plasmonic gold nanorods (GNRs) presents a potent strategy for eradication of tumors including aggressive brain gliomas. Despite its promise, there is a pressing need for a more comprehensive evaluation of PPTT using sophisticated in vitro models that closely resemble tumor tissues, thereby facilitating the elucidation of therapeutic mechanisms. In this study, we exposed 3D glioma spheroids (tumoroids) to (16-mercaptohexadecyl)trimethylammonium bromide-functionalized gold nanorods (MTAB-GNRs) and a near-infrared (NIR) laser. We demonstrate that the photothermal effect can be fine-tuned by adjusting the nanoparticle concentration and laser power. Depending on the selected parameters, the laser can trigger either regulated or non-regulated cell death (necrosis) in both mouse GL261 and human U-87 MG glioma cell lines, accompanied by translocation of phosphatidylserine in the membrane. Our investigation into the mechanism of regulated cell death induced by PPTT revealed an absence of markers associated with classical apoptosis pathways, such as cleaved caspase 3. Instead, we observed the presence of cleaved caspase 1, gasdermin D, and elevated levels of NLRP3 in NIR-irradiated tumoroids, indicating the activation of pyroptosis. This finding correlates with previous observations of lysosomal accumulation of MTAB-GNRs and the known lysosomal pathway of pyroptosis activation. We further confirmed the absence of toxic breakdown products of GNRs using electron microscopy, which showed no melting or fragmentation of gold nanoparticles under the conditions causing regulated cell death. In conclusion, PPTT using coated gold nanorods offers significant potential for glioma cell elimination occurring through the activation of pyroptosis rather than classical apoptosis pathways.
Collapse
Affiliation(s)
- Monika Zarska
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Ondrej Novak
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tereza Jakubcova
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Filip Novotny
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alexandra Urbancokova
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Filip Havel
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Laser Physics and Photonics, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Josef Novak
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Helena Raabova
- Electron Microscopy Core Facility, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic; Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| | - Vlada Filimonenko
- Electron Microscopy Core Facility, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Laboratory of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Bartek
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Genome Integrity Group, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark; Department of Medical Biochemistry and Biophysics, Science For Life Laboratory, Division of Genome Biology, Karolinska Institute, Stockholm, Sweden
| | - Jan Proska
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Laser Physics and Photonics, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Zdenek Hodny
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
2
|
Bregnhøj M, Thorning F, Ogilby PR. Singlet Oxygen Photophysics: From Liquid Solvents to Mammalian Cells. Chem Rev 2024; 124:9949-10051. [PMID: 39106038 DOI: 10.1021/acs.chemrev.4c00105] [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: 08/07/2024]
Abstract
Molecular oxygen, O2, has long provided a cornerstone for studies in chemistry, physics, and biology. Although the triplet ground state, O2(X3Σg-), has garnered much attention, the lowest excited electronic state, O2(a1Δg), commonly called singlet oxygen, has attracted appreciable interest, principally because of its unique chemical reactivity in systems ranging from the Earth's atmosphere to biological cells. Because O2(a1Δg) can be produced and deactivated in processes that involve light, the photophysics of O2(a1Δg) are equally important. Moreover, pathways for O2(a1Δg) deactivation that regenerate O2(X3Σg-), which address fundamental principles unto themselves, kinetically compete with the chemical reactions of O2(a1Δg) and, thus, have practical significance. Due to technological advances (e.g., lasers, optical detectors, microscopes), data acquired in the past ∼20 years have increased our understanding of O2(a1Δg) photophysics appreciably and facilitated both spatial and temporal control over the behavior of O2(a1Δg). One goal of this Review is to summarize recent developments that have broad ramifications, focusing on systems in which oxygen forms a contact complex with an organic molecule M (e.g., a liquid solvent). An important concept is the role played by the M+•O2-• charge-transfer state in both the formation and deactivation of O2(a1Δg).
Collapse
Affiliation(s)
- Mikkel Bregnhøj
- Department of Chemistry, Aarhus University, 140 Langelandsgade, Aarhus 8000, Denmark
| | - Frederik Thorning
- Department of Chemistry, Aarhus University, 140 Langelandsgade, Aarhus 8000, Denmark
| | - Peter R Ogilby
- Department of Chemistry, Aarhus University, 140 Langelandsgade, Aarhus 8000, Denmark
| |
Collapse
|
3
|
Turkmen Koc SN, Rezaei Benam S, Aral IP, Shahbazi R, Ulubayram K. Gold nanoparticles-mediated photothermal and photodynamic therapies for cancer. Int J Pharm 2024; 655:124057. [PMID: 38552752 DOI: 10.1016/j.ijpharm.2024.124057] [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: 11/17/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Cancer remains one of the major causes of death globally, with one out of every six deaths attributed to the disease. The impact of cancer is felt on psychological, physical, and financial levels, affecting individuals, communities, and healthcare institutions. Conventional cancer treatments have many challenges and inadequacies. Nanomedicine, however, presents a promising solution by not only overcoming these problems but also offering the advantage of combined therapy for treatment-resistant cancers. Nanoparticles specifically engineered for use in nanomedicine can be efficiently targeted to cancer cells through a combination of active and passive techniques, leading to superior tumor-specific accumulation, enhanced drug availability, and reduced systemic toxicity. Among various nanoparticle formulations designed for cancer treatment, gold nanoparticles have gained prominence in the field of nanomedicine due to their photothermal, photodynamic, and immunologic effects without the need for photosensitizers or immunotherapeutic agents. To date, there is no comprehensive literature review that focuses on the photothermal, photodynamic, and immunologic effects of gold nanoparticles. In this review, significant attention has been devoted to examining the parameters pertaining to the structure of gold nanoparticles and laser characteristics, which play a crucial role in influencing the efficacy of photothermal therapy (PTT) and photodynamic therapy (PDT). Moreover, this article provides insights into the success of PTT and PDT mediated by gold nanoparticles in primary cancer treatment, as well as the immunological effects of PTT and PDT on metastasis and recurrence, providing a promising strategy for cancer therapy. In summary, gold nanoparticles, with their unique properties, have the potential for clinical application in various cancer therapies, including the treatment of primary cancer, recurrence and metastasis.
Collapse
Affiliation(s)
- Seyma Nur Turkmen Koc
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye
| | - Sanam Rezaei Benam
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Ipek Pınar Aral
- Department of Radiation Oncology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara Bilkent City Hospital, Ankara, Türkiye
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA; Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, USA; Brown Center for Immunotherapy, Indiana University School of Medicine, Indianapolis, USA.
| | - Kezban Ulubayram
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye; Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye; Department of Bioengineering, Hacettepe University, Ankara, Türkiye.
| |
Collapse
|
4
|
Ruhoff V, Arastoo MR, Moreno-Pescador G, Bendix PM. Biological Applications of Thermoplasmonics. NANO LETTERS 2024; 24:777-789. [PMID: 38183300 PMCID: PMC10811673 DOI: 10.1021/acs.nanolett.3c03548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/08/2024]
Abstract
Thermoplasmonics has emerged as an extraordinarily versatile tool with profound applications across various biological domains ranging from medical science to cell biology and biophysics. The key feature of nanoscale plasmonic heating involves remote activation of heating by applying laser irradiation to plasmonic nanostructures that are designed to optimally convert light into heat. This unique capability paves the way for a diverse array of applications, facilitating the exploration of critical biological processes such as cell differentiation, repair, signaling, and protein functionality, and the advancement of biosensing techniques. Of particular significance is the rapid heat cycling that can be achieved through thermoplasmonics, which has ushered in remarkable technical innovations such as accelerated amplification of DNA through quantitative reverse transcription polymerase chain reaction. Finally, medical applications of photothermal therapy have recently completed clinical trials with remarkable results in prostate cancer, which will inevitably lead to the implementation of photothermal therapy for a number of diseases in the future. Within this review, we offer a survey of the latest advancements in the burgeoning field of thermoplasmonics, with a keen emphasis on its transformative applications within the realm of biosciences.
Collapse
Affiliation(s)
| | - Mohammad Reza Arastoo
- Niels
Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
| | - Guillermo Moreno-Pescador
- Niels
Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
- Copenhagen
Plant Science Center, Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg, Denmark
| | - Poul Martin Bendix
- Niels
Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
| |
Collapse
|
5
|
Florencia Tebele M, Paris G, Zelcer A. Plasmonic inhibition of bacterial adhesion on gold-decorated mesoporous zirconium oxide thin films. Colloids Surf B Biointerfaces 2023; 232:113576. [PMID: 37862951 DOI: 10.1016/j.colsurfb.2023.113576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/22/2023] [Accepted: 09/30/2023] [Indexed: 10/22/2023]
Abstract
Preventing bacterial development on surfaces is essential to avoid problems caused by biofouling. Surfaces decorated with gold nanoparticles have been shown to thermally kill bacteria under high-intensity NIR illumination. In this study, we evaluated the colonization by E. coli of nanostructured surfaces composed of mesoporous zirconia thin films, both with and without gold nanoparticles embedded into the pores. We studied the effect of the nanostructure and of low intensity visible light excitation of the gold nanoparticles on the colonization process. We found that neither the zirconia, nor the presence of pores, or even gold nanoparticles affect bacterial adhesion compared to the bare glass substrate. Therefore, mesoporous zirconia thin films are biologically inert scaffolds that enable the construction of robust surfaces containing functional nanoparticles that can affect bacterial growth. When the gold containing surfaces are irradiated with light, bacterial adhesion shows a remarkable 96 ± 4% reduction. Our studies revealed that these surfaces affect early colonization steps, prior to biofilm formation, preventing bacterial adhesion without affecting its viability. In contrast to related systems where plasmonic excitation induces membrane damage due to strong local heating, the membrane integrity is preserved, showing that these surfaces have a different working principle.
Collapse
Affiliation(s)
- M Florencia Tebele
- CIBION-CONICET, Godoy Cruz 2390, C1425FQD Ciudad de Buenos Aires, Argentina.
| | - Gastón Paris
- CIBION-CONICET, Godoy Cruz 2390, C1425FQD Ciudad de Buenos Aires, Argentina
| | - Andrés Zelcer
- CIBION-CONICET, Godoy Cruz 2390, C1425FQD Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
6
|
Influence of Parameters on the Death Pathway of Gastric Cells Induced by Gold Nanosphere Mediated Phototherapy. NANOMATERIALS 2022; 12:nano12040646. [PMID: 35214976 PMCID: PMC8878397 DOI: 10.3390/nano12040646] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/07/2022] [Accepted: 02/13/2022] [Indexed: 02/04/2023]
Abstract
Gold nanosphere (AuS) is a nanosized particle with inert, biocompatible, easily modified surface functionalization and adequate cell penetration ability. Photothermal, photochemical, and vapor effects of AuS could be activated by irradiating with nanosecond laser to cause cell death. Hence, AuS-mediated phototherapy irradiated with nanosecond laser is a promising and minimally-invasive treatment method for cancer therapy. However, various effects require different parameters to be activated. At present, few studies have reported on the influence of parameters of AuS inducing cell death under nanosecond laser irradiation. This makes it very challenging to optimize gold-nanoparticle-mediated specific or synergistic anti-cancer therapy. In this study, we revealed the main parameters and threshold values for AuS-mediated gastric cancer phototherapy with nanosecond pulsed laser irradiation, evaluated the pathway of induced cell death, and discussed the roles of photothermal, photochemical and vapor effects which can induce the cell death. The results showed that AuS-mediated phototherapy activated with nanosecond pulsed laser is an effective method for gastric therapy, mainly based on the photochemical effect. Prolonging the incubation time could decrease the irradiation dose, increase ROS-mediated photothermal effect and vapor effect, and then quickly induce cell death to improve security.
Collapse
|
7
|
Mechanistic insight into photoactivation of small inorganic molecules from the biomedical applications perspectives. BIOMEDICAL APPLICATIONS OF INORGANIC PHOTOCHEMISTRY 2022. [DOI: 10.1016/bs.adioch.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Xu J, Gu M, Hooi L, Toh TB, Thng DKH, Lim JJ, Chow EKH. Enhanced penetrative siRNA delivery by a nanodiamond drug delivery platform against hepatocellular carcinoma 3D models. NANOSCALE 2021; 13:16131-16145. [PMID: 34542130 DOI: 10.1039/d1nr03502a] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Small interfering RNA (siRNA) can cause specific gene silencing and is considered promising for treating a variety of cancers, including hepatocellular carcinoma (HCC). However, siRNA has many undesirable physicochemical properties that limit its application. Additionally, conventional methods for delivering siRNA are limited in their ability to penetrate solid tumors. In this study, nanodiamonds (NDs) were evaluated as a nanoparticle drug delivery platform for improved siRNA delivery into tumor cells. Our results demonstrated that ND-siRNA complexes could effectively be formed through electrostatic interactions. The ND-siRNA complexes allowed for efficient cellular uptake and endosomal escape that protects siRNA from degradation. Moreover, ND delivery of siRNA was more effective at penetrating tumor spheroids compared to liposomal formulations. This enhanced penetration capacity makes NDs ideal vehicles to deliver siRNA against solid tumor masses as efficient gene knockdown and decreased tumor cell proliferation were observed in tumor spheroids. Evaluation of ND-siRNA complexes within the context of a 3D cancer disease model demonstrates the potential of NDs as a promising gene delivery platform against solid tumors, such as HCC.
Collapse
Affiliation(s)
- Jingru Xu
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Mengjie Gu
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Lissa Hooi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, 117456, Singapore
| | - Dexter Kai Hao Thng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Jhin Jieh Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
- The N.1 Institute for Health, National University of Singapore, 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| |
Collapse
|
9
|
Nam K, Jeong CB, Kim H, Ahn M, Ahn S, Hur H, Kim DU, Jang J, Gwon H, Lim Y, Cho D, Lee K, Bae JY, Chang KS. Quantitative Photothermal Characterization with Bioprinted 3D Complex Tissue Constructs for Early-Stage Breast Cancer Therapy Using Gold Nanorods. Adv Healthc Mater 2021; 10:e2100636. [PMID: 34235891 PMCID: PMC11468621 DOI: 10.1002/adhm.202100636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/18/2021] [Indexed: 11/12/2022]
Abstract
Plasmonic photothermal therapy (PPTT) using gold nanoparticles (AuNPs) has shown great potential for use in selective tumor treatment, because the AuNPs can generate destructive heat preferentially upon irradiation. However, PPTT using AuNPs has not been added to practice, owing to insufficient heating methods and tissue temperature measurement techniques, leading to unreliable and inaccurate treatments. Because the photothermal properties of AuNPs vary with laser power, particle optical density, and tissue depth, the accurate prediction of heat generation is indispensable for clinical treatment. In this report, bioprinted 3D complex tissue constructs comprising processed gel obtained from porcine skin and human decellularized adipose tissue are presented for characterization of the photothermal properties of gold nanorods (AuNRs) having an aspect ratio of 3.7 irradiated by a near-infrared laser. Moreover, an analytical function is suggested for achieving PPTT that can cause thermal damage selectively on early-stage human breast cancer by regulating the heat generation of the AuNRs in the tissue.
Collapse
Affiliation(s)
- Ki‐Hwan Nam
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - Chan Bae Jeong
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - HyeMi Kim
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - Minjun Ahn
- Department of Mechanical EngineeringPohang University of Science and Technology (POSTECH)PohangKyungbuk37673Republic of Korea
| | - Sung‐Jun Ahn
- Research Division for Industry and EnvironmentKorea Atomic Energy Research Institute (KAERI)JeongeupJeollabuk‐do56212Republic of Korea
| | - Hwan Hur
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - Dong Uk Kim
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - Jinah Jang
- Department of Creative IT EngineeringSchool of Interdisciplinary Bioscience and BioengineeringPohang University of Science and Technology (POSTECH)PohangKyungbuk37673Republic of Korea
| | - Hui‐Jeong Gwon
- Research Division for Industry and EnvironmentKorea Atomic Energy Research Institute (KAERI)JeongeupJeollabuk‐do56212Republic of Korea
| | - Youn‐Mook Lim
- Research Division for Industry and EnvironmentKorea Atomic Energy Research Institute (KAERI)JeongeupJeollabuk‐do56212Republic of Korea
| | - Dong‐Woo Cho
- Department of Mechanical EngineeringPohang University of Science and Technology (POSTECH)PohangKyungbuk37673Republic of Korea
| | - Kye‐Sung Lee
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - Ji Yong Bae
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| | - Ki Soo Chang
- Center for Scientific InstrumentationDivision of Scientific Instrumentation and ManagementKorea Basic Science Institute (KBSI)Daejeon34133Republic of Korea
| |
Collapse
|
10
|
Xie J, Wang Y, Choi W, Jangili P, Ge Y, Xu Y, Kang J, Liu L, Zhang B, Xie Z, He J, Xie N, Nie G, Zhang H, Kim JS. Overcoming barriers in photodynamic therapy harnessing nano-formulation strategies. Chem Soc Rev 2021; 50:9152-9201. [PMID: 34223847 DOI: 10.1039/d0cs01370f] [Citation(s) in RCA: 226] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Photodynamic therapy (PDT) has been extensively investigated for decades for tumor treatment because of its non-invasiveness, spatiotemporal selectivity, lower side-effects, and immune activation ability. It can be a promising treatment modality in several medical fields, including oncology, immunology, urology, dermatology, ophthalmology, cardiology, pneumology, and dentistry. Nevertheless, the clinical application of PDT is largely restricted by the drawbacks of traditional photosensitizers, limited tissue penetrability of light, inefficient induction of tumor cell death, tumor resistance to the therapy, and the severe pain induced by the therapy. Recently, various photosensitizer formulations and therapy strategies have been developed to overcome these barriers. Significantly, the introduction of nanomaterials in PDT, as carriers or photosensitizers, may overcome the drawbacks of traditional photosensitizers. Based on this, nanocomposites excited by various light sources are applied in the PDT of deep-seated tumors. Modulation of cell death pathways with co-delivered reagents promotes PDT induced tumor cell death. Relief of tumor resistance to PDT with combined therapy strategies further promotes tumor inhibition. Also, the optimization of photosensitizer formulations and therapy procedures reduces pain in PDT. Here, a systematic summary of recent advances in the fabrication of photosensitizers and the design of therapy strategies to overcome barriers in PDT is presented. Several aspects important for the clinical application of PDT in cancer treatment are also discussed.
Collapse
Affiliation(s)
- Jianlei Xie
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Institute of Microscale Optoelectronics, and Otolaryngology Department and Biobank of the First Affiliated Hospital, Shenzhen Second People's Hospital, Health Science Center, Shenzhen University, Shenzhen 518060, P. R. China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Guerrero-Florez V, Mendez-Sanchez SC, Patrón-Soberano OA, Rodríguez-González V, Blach D, Martínez O F. Gold nanoparticle-mediated generation of reactive oxygen species during plasmonic photothermal therapy: a comparative study for different particle sizes, shapes, and surface conjugations. J Mater Chem B 2021; 8:2862-2875. [PMID: 32186317 DOI: 10.1039/d0tb00240b] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gold nanoparticle (AuNP)-mediated photothermal therapy represents an alternative to the effective ablation of cancer cells. However, the photothermal response of AuNPs must be tailored to improve the therapeutic efficacy of plasmonic photothermal therapy (PPT) and mitigate its side effects. This study presents an alternative to ease the tuning of photothermal efficiency and target selectivity. We use laser-treated spherical and anisotropic AuNPs of different sizes and biocompatible folic acid (FA)-conjugated AuNPs (FA-AuNPs) in the well-known human epithelial cervical cancer (HeLa) cell line. We show that large AuNPs produce a more significant photothermal heating effect than small ones. The thermal response of the spherical AuNPs of 9 nm was found to reach a maximum increase of 3.0 ± 1 °C, whereas with the spherical AuNPs of 14 nm, the temperature increased by over 4.4 ± 1 °C. The anisotropic AuNPs of 15 nm reached a maximum of 4.0 ± 1 °C, whereas the anisotropic AuNPs of 20 nm reached a significant increase of 5.3 ± 1 °C in the cell culture medium (MEM). Notably, the anisotropic AuNPs of 20 nm successfully demonstrate the potential for use as a photothermal agent by showing reduced viability down to 60% at a concentration of 100 μM. Besides, we reveal that high concentrations of reactive oxygen species (ROS) are formed within the irradiated cells. In combination with stress by photothermal heating, it is likely to result in significant cell death through acute necrosis by compromising the plasma membrane integrity. Cell death and ROS overproduction during PPT were characterized and quantified by transmission electron microscopy (TEM) and confocal fluorescence microscopy with different fluorescent markers. In addition, we show that FA-AuNPs induce cell death through apoptosis by internal damage, whereas diminish the ROS formation during PPT treatment. Our findings suggest the ability of plasmon-mediated ROS to sensitize cancer cells and make them vulnerable to photothermal damage, as well as the protective role of FA-AuNPs from excessive ROS formation, whereas reducing the risk of undesired side effects due to the necrotic death pathway. It allows an improvement in the efficacy of the AuNP-based photothermal therapy and a reduction in the number of exposures to high temperatures required to induce thermal stress.
Collapse
Affiliation(s)
- Valentina Guerrero-Florez
- Centro de Investigaciones en Catálisis (CICAT), Escuela de química, Universidad Industrial de Santander, Km 2 vía El Refugio, Piedecuesta, Colombia
| | - Stelia C Mendez-Sanchez
- Grupo de Investigación en Bioquímica Y Microbiología (GIBIM), Escuela de química, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Olga A Patrón-Soberano
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnología, San Luis Potosí, S. L. P., Mexico
| | - Vicente Rodríguez-González
- División de Materiales Avanzados, Instituto Potosino de Investigación Científica y Tecnología, San Luis Potosí, S. L. P., Mexico
| | - Diana Blach
- Centro de Investigaciones en Catálisis (CICAT), Escuela de química, Universidad Industrial de Santander, Km 2 vía El Refugio, Piedecuesta, Colombia
| | - Fernando Martínez O
- Centro de Investigaciones en Catálisis (CICAT), Escuela de química, Universidad Industrial de Santander, Km 2 vía El Refugio, Piedecuesta, Colombia
| |
Collapse
|
12
|
Dosumu A, Claire S, Watson LS, Girio PM, Osborne SAM, Pikramenou Z, Hodges NJ. Quantification by Luminescence Tracking of Red Emissive Gold Nanoparticles in Cells. JACS AU 2021; 1:174-186. [PMID: 33778810 PMCID: PMC7990080 DOI: 10.1021/jacsau.0c00033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 05/11/2023]
Abstract
Optical microscopy techniques are ideal for live cell imaging for real-time nanoparticle tracking of nanoparticle localization. However, the quantification of nanoparticle uptake is usually evaluated by analytical methods that require cell isolation. Luminescent labeling of gold nanoparticles with transition metal probes yields particles with attractive photophysical properties, enabling cellular tracking using confocal and time-resolved microscopies. In the current study, gold nanoparticles coated with a red-luminescent ruthenium transition metal complex are used to quantify and track particle uptake and localization. Analysis of the red-luminescence signal from particles is used as a metric of cellular uptake, which correlates to total cellular gold and ruthenium content, independently measured and correlated by inductively coupled plasma mass spectrometry. Tracking of the luminescence signal provides evidence of direct diffusion of the nanoparticles across the cytoplasmic membrane with particles observed in the cytoplasm and mitochondria as nonclustered "free" nanoparticles. Electron microscopy and inhibition studies identified macropinocytosis of clusters of particles into endosomes as the major mechanism of uptake. Nanoparticles were tracked inside GFP-tagged cells by following the red-luminescence signal of the ruthenium complex. Tracking of the particles demonstrates their initial location in early endosomes and, later, in lysosomes and autophagosomes. Colocalization was quantified by calculating the Pearson's correlation coefficient between red and green luminescence signals and confirmed by electron microscopy. Accumulation of particles in autophagosomes correlated with biochemical evidence of active autophagy, but there was no evidence of detachment of the luminescent label or breakup of the gold core. Instead, accumulation of particles in autophagosomes caused organelle swelling, breakdown of the surrounding membranes, and endosomal release of the nanoparticles into the cytoplasm. The phenomenon of endosomal release has important consequences for the toxicity, cellular targeting, and therapeutic future applications of gold nanoparticles.
Collapse
Affiliation(s)
- Abiola
N. Dosumu
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Sunil Claire
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Luke S. Watson
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Patricia M. Girio
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Shani A. M. Osborne
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Zoe Pikramenou
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Nikolas J. Hodges
- School
of Biosciences, School of Chemistry, and Doctoral Training Centre in Physical
Sciences for Health, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
13
|
Carrasco E, Stockert JC, Juarranz Á, Blázquez-Castro A. Plasmonic Hot-Electron Reactive Oxygen Species Generation: Fundamentals for Redox Biology. Front Chem 2020; 8:591325. [PMID: 33425851 PMCID: PMC7793889 DOI: 10.3389/fchem.2020.591325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
For decades, the possibility to generate Reactive Oxygen Species (ROS) in biological systems through the use of light was mainly restricted to the photodynamic effect: the photoexcitation of molecules which then engage in charge- or energy-transfer to molecular oxygen (O2) to initiate ROS production. However, the classical photodynamic approach presents drawbacks, like per se chemical reactivity of the photosensitizing agent or fast molecular photobleaching due to in situ ROS generation, to name a few. Recently, a new approach, which promises many advantages, has entered the scene: plasmon-driven hot-electron chemistry. The effect takes advantage of the photoexcitation of plasmonic resonances in metal nanoparticles to induce a new cohort of photochemical and redox reactions. These metal photo-transducers are considered chemically inert and can undergo billions of photoexcitation rounds without bleaching or suffering significant oxidative alterations. Also, their optimal absorption band can be shape- and size-tailored in order to match any of the near infrared (NIR) biological windows, where undesired absorption/scattering are minimal. In this mini review, the basic mechanisms and principal benefits of this light-driven approach to generate ROS will be discussed. Additionally, some significant experiments in vitro and in vivo will be presented, and tentative new avenues for further research will be advanced.
Collapse
Affiliation(s)
- Elisa Carrasco
- Department of Biology, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | - Juan Carlos Stockert
- Area Investigación, Instituto de Oncología “Angel H. Roffo”, Universidad de Buenos Aires, Buenos Aires, Argentina
- Cátedra de Histología y Embriología, Instituto de Investigación y Tecnología en Reproducción Animal, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ángeles Juarranz
- Department of Biology, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | | |
Collapse
|
14
|
Lee J, Dubbu S, Kumari N, Kumar A, Lim J, Kim S, Lee IS. Magnetothermia-Induced Catalytic Hollow Nanoreactor for Bioorthogonal Organic Synthesis in Living Cells. NANO LETTERS 2020; 20:6981-6988. [PMID: 32633963 DOI: 10.1021/acs.nanolett.0c01507] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanoreactors, in which the reactions are remotely controlled by magnetic fields, are potentially valuable in bioorthogonal chemistry for future applications. Here, we develop a silica-confined magnetothermia-induced nanoreactor (MAG-NER) by selectively growing Pd nanocrystals on a preinstalled iron-oxide core inside a hollow silica nanoshell. The growth is achieved by magnetic induction. The interfacial catalytic site is activated by stimulating localized magnetothermia, and nanocompartmentalization is realized by the size-selective porous silica. Therefore, MAG-NER can be conveniently used in complex biomedia and can even be internalized to living cells, realizing an on-demand, high-performance intramolecular carbocyclization reaction by remote operation without compromising the cell viability. This work opens avenues for the design of advanced nanoreactors that complement and augment the existing bioorthogonal chemical tools.
Collapse
Affiliation(s)
- Jihwan Lee
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Sateesh Dubbu
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Nitee Kumari
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Amit Kumar
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Jongwon Lim
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Seonock Kim
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - In Su Lee
- Creative Research Initiative Center for Nanospace-confined Chemical Reactions (NCCR), and Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| |
Collapse
|
15
|
Baffou G, Cichos F, Quidant R. Applications and challenges of thermoplasmonics. NATURE MATERIALS 2020; 19:946-958. [PMID: 32807918 DOI: 10.1038/s41563-020-0740-6] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/08/2020] [Indexed: 05/18/2023]
Abstract
Over the past two decades, there has been a growing interest in the use of plasmonic nanoparticles as sources of heat remotely controlled by light, giving rise to the field of thermoplasmonics. The ability to release heat on the nanoscale has already impacted a broad range of research activities, from biomedicine to imaging and catalysis. Thermoplasmonics is now entering an important phase: some applications have engaged in an industrial stage, while others, originally full of promise, experience some difficulty in reaching their potential. Meanwhile, innovative fundamental areas of research are being developed. In this Review, we scrutinize the current research landscape in thermoplasmonics, with a specific focus on its applications and main challenges in many different fields of science, including nanomedicine, cell biology, photothermal and hot-electron chemistry, solar light harvesting, soft matter and nanofluidics.
Collapse
Affiliation(s)
- Guillaume Baffou
- Institut Fresnel, CNRS, Aix Marseille University, Ecole Centrale Marseille, Marseille, France.
| | - Frank Cichos
- Molecular Nanophotonics Group, Peter Debye Institute for Soft Matter Physics, Universität Leipzig, Leipzig, Germany.
| | - Romain Quidant
- ICFO - Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Barcelona, Spain.
- ICREA - Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
- Nanophotonic Systems Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
De Matteis V, Cascione M, Toma CC, Rinaldi R. Engineered Gold Nanoshells Killing Tumor Cells: New Perspectives. Curr Pharm Des 2020; 25:1477-1489. [PMID: 31258061 DOI: 10.2174/1381612825666190618155127] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022]
Abstract
The current strategies to treat different kinds of cancer are mainly based on chemotherapy, surgery and radiation therapy. Unfortunately, these approaches are not specific and rather invasive as well. In this scenario, metal nano-shells, in particular gold-based nanoshells, offer interesting perspectives in the effort to counteract tumor cells, due to their unique ability to tune Surface Plasmon Resonance in different light-absorbing ranges. In particular, the Visible and Near Infrared Regions of the electromagnetic spectrum are able to penetrate through tissues. In this way, the light absorbed by the gold nanoshell at a specific wavelength is converted into heat, inducing photothermal ablation in treated cancer cells. Furthermore, inert gold shells can be easily functionalized with different types of molecules in order to bind cellular targets in a selective manner. This review summarizes the current state-of-art of nanosystems embodying gold shells, regarding methods of synthesis, bio-conjugations, bio-distribution, imaging and photothermal effects (in vitro and in vivo), providing new insights for the development of multifunctional antitumor drugs.
Collapse
Affiliation(s)
- Valeria De Matteis
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| | - Mariafrancesca Cascione
- Dipartimento di Scienze Biomediche e Oncologia Umana, Universita degli Studi di Bari "Aldo Moro", p.zza G. Cesare, c/o Policlinico, 70124 Bari, Italy
| | - Chiara C Toma
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| | - Rosaria Rinaldi
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
17
|
Kuncewicz J, Dąbrowski JM, Kyzioł A, Brindell M, Łabuz P, Mazuryk O, Macyk W, Stochel G. Perspectives of molecular and nanostructured systems with d- and f-block metals in photogeneration of reactive oxygen species for medical strategies. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Mosquera J, García I, Liz-Marzán LM. Cellular Uptake of Nanoparticles versus Small Molecules: A Matter of Size. Acc Chem Res 2018; 51:2305-2313. [PMID: 30156826 DOI: 10.1021/acs.accounts.8b00292] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The primary function of the cell membrane is to protect cells from their surroundings. This entails a strict regulation on controlling the exchange of matter between the cell and its environment. A key factor when considering potential biological applications of a particular chemical structure has to do with its ability to internalize into cells. Molecules that can readily cross cell membranes are frequently needed in biological research and medicine, since most therapeutic entities are designed to modulate intracellular components. However, the design of molecules that do not penetrate cells is also relevant toward, for example, extracellular contrast agents, which are most widely used in clinical diagnosis. Small molecules have occupied the forefront of biomedical research until recently, but the past few decades have seen an increasing use of larger chemical structures, such as proteins or nanoparticles, leading to unprecedented and often unexpectedly novel research. Great achievements have been made toward understanding the rules that govern cellular uptake, which show that cell internalization of molecules is largely affected by their size. For example, macromolecules such as proteins and nucleic acids are usually unable to internalize cells. Intriguingly, in the case of nanoparticles, larger sizes seem to facilitate internalization via endocytic pathways, through which the particles remain trapped in lysosomes and endosomes. In this Account, we aimed at presenting our personal view of how different chemical structures behave in terms of cell internalization due to their size, ranging from small drugs to large nanoparticles. We first introduce the properties of cell membranes and the main mechanisms involved in cellular uptake. We then discuss the cellular internalization of molecules, distinguishing between those with molecular weights below 1 kDa and biological macromolecules such as proteins and nucleic acids. In the last section, we review the biological behavior of nanoparticles, with a special emphasis on plasmonic nanoparticles, which feature a high potential in the biomedical field. For each group of chemical structures, we discuss the parameters affecting their cellular internalization but also strategies that can be applied to achieve the desired intracellular delivery. Particular attention is paid to approaches that allow conditional regulation of the cell internalization process using external triggers, such as activable cell penetrating peptides, due to the impact that these systems may have in drug delivery and sensing applications. The Account ends with a "Conclusions and Outlook" section, where general lessons and future directions toward further advancements are briefly presented.
Collapse
Affiliation(s)
- Jesús Mosquera
- CIC biomaGUNE and CIBER-BBN, Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain
| | - Isabel García
- CIC biomaGUNE and CIBER-BBN, Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain
| | - Luis M. Liz-Marzán
- CIC biomaGUNE and CIBER-BBN, Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
19
|
Durymanov M, Reineke J. Non-viral Delivery of Nucleic Acids: Insight Into Mechanisms of Overcoming Intracellular Barriers. Front Pharmacol 2018; 9:971. [PMID: 30186185 PMCID: PMC6111240 DOI: 10.3389/fphar.2018.00971] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022] Open
Abstract
Delivery of genes, including plasmid DNAs, short interfering RNAs (siRNAs), and messenger RNAs (mRNAs), using artificial non-viral nanotherapeutics is a promising approach in cancer gene therapy. However, multiple physiological barriers upon systemic administration remain a key challenge in clinical translation of anti-cancer gene therapeutics. Besides extracellular barriers including sequestration of gene delivery nanoparticles from the bloodstream by resident organ-specific macrophages, and their poor extravasation and tissue penetration in tumors, overcoming intracellular barriers is also necessary for successful delivery of nucleic acids. Whereas for RNA delivery the endosomal barrier holds a key importance, transfer of DNA cargo additionally requires translocation into the nucleus. Better understanding of crossing membrane barriers by nucleic acid nanoformulations is essential to the improvement of current non-viral carriers. This review aims to summarize relevant literature on intracellular trafficking of non-viral nanoparticles and determine key factors toward surmounting intracellular barriers. Moreover, recent data allowed us to propose new interpretations of current hypotheses of endosomal escape mechanisms of nucleic acid nanoformulations.
Collapse
Affiliation(s)
- Mikhail Durymanov
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD, United States
| | - Joshua Reineke
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
20
|
Vermeulen LMP, Fraire JC, Raes L, De Meester E, De Keulenaer S, Van Nieuwerburgh F, De Smedt S, Remaut K, Braeckmans K. Photothermally Triggered Endosomal Escape and Its Influence on Transfection Efficiency of Gold-Functionalized JetPEI/pDNA Nanoparticles. Int J Mol Sci 2018; 19:E2400. [PMID: 30110965 PMCID: PMC6121899 DOI: 10.3390/ijms19082400] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022] Open
Abstract
Plasmonic nanoparticles for drug delivery have attracted increasing interest over the last few years. Their localized surface plasmon resonance causes photothermal effects on laser irradiation, which allows for delivering drugs in a spatio-temporally controlled manner. Here, we explore the use of gold nanoparticles (AuNP) as carriers for pDNA in combination with pulsed laser irradiation to induce endosomal escape, which is currently considered to be one of the major bottlenecks in macromolecular drug delivery on the intracellular level. In particular, we evaluate nanocomplexes composed of JetPEI (polyethylenimine)pDNA and 10 nm AuNP, which do not exhibit endosomal escape by themselves. After incubating HeLa cells with these complexes, we evaluated endosomal escape and transfection efficiency using low- and high-energy laser pulses. At low laser energy heat is produced by the nanocomplexes, while, at higher laser energy, explosive vapour nanobubbles (VNB) are formed. We investigated the ability of heat transfer and VNB formation to induce endosomal escape and we examine the integrity of pDNA cargo after inducing both photothermal effects. We conclude that JetPEI/pDNA/AuNP complexes are unable to induce meaningful transfection efficiencies because laser treatment causes either dysfunctionality of the cargo when VNB are formed or forms too small pores in the endosomal membrane to allow pDNA to escape in case of heating. We conclude that laser-induced VNB is the most suitable to induce effective pDNA endosomal escape, but a different nanocomplex structure will be required to keep the pDNA intact.
Collapse
Affiliation(s)
- Lotte M P Vermeulen
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
- Centre for Nano- and Biophotonics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Juan C Fraire
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
- Centre for Nano- and Biophotonics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Laurens Raes
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
- Centre for Nano- and Biophotonics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Ellen De Meester
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Sarah De Keulenaer
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan De Smedt
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Katrien Remaut
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
- Centre for Nano- and Biophotonics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
- IEMN UMR 8520 and Laboratoire de Physique des Lasers, Atomes et Molécules. UMR 8523, Université de Lille, F-59655 Villeneuve d'Ascq CEDEX, France.
| |
Collapse
|
21
|
Chitosan nanoparticles as a biocompatible and efficient nanowagon for benzyl isothiocyanate. Int J Biol Macromol 2018; 115:18-28. [DOI: 10.1016/j.ijbiomac.2018.04.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/19/2018] [Accepted: 04/08/2018] [Indexed: 01/29/2023]
|
22
|
Lino MM, Simões S, Vilaça A, Antunes H, Zonari A, Ferreira L. Modulation of Angiogenic Activity by Light-Activatable miRNA-Loaded Nanocarriers. ACS NANO 2018; 12:5207-5220. [PMID: 29870221 DOI: 10.1021/acsnano.7b07538] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The combinatorial delivery of miRNAs holds great promise to modulate cell activity in the context of angiogenesis. Yet, the delivery of multiple miRNAs with spatiotemporal control remains elusive. Here, we report a plasmonic nanocarrier to control the release of two microRNAs. The nanocarrier consists of gold nanorods modified with single-stranded DNA for hybridization with complementary DNA-conjugated microRNAs. DNA strands with distinct melting temperatures enable the independent release of each microRNA with a near-infrared laser using the same wavelength but different powers. Tests in human outgrowth endothelial cells (OECs) indicate that this system can be used to silence different targets sequentially and, by doing so, to modulate cell activity with spatiotemporal resolution. Finally, using an in vivo acute wound healing animal model, it is demonstrated that the order by which each miRNA was released in transplanted OECs significantly impacted the wound healing kinetics.
Collapse
Affiliation(s)
- Miguel M Lino
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
- Faculty of Medicine , University of Coimbra , 3000-548 Coimbra , Portugal
| | - Susana Simões
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - Andreia Vilaça
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - Helena Antunes
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
- Faculty of Medicine , University of Coimbra , 3000-548 Coimbra , Portugal
- Crioestaminal , 3060-197 Cantanhede , Portugal
| | - Alessandra Zonari
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
- Faculty of Medicine , University of Coimbra , 3000-548 Coimbra , Portugal
| |
Collapse
|
23
|
Vankayala R, Hwang KC. Near-Infrared-Light-Activatable Nanomaterial-Mediated Phototheranostic Nanomedicines: An Emerging Paradigm for Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706320. [PMID: 29577458 DOI: 10.1002/adma.201706320] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/11/2017] [Indexed: 05/22/2023]
Abstract
Cancer is one of the most deadly diseases threatening the lives of humans. Although many treatment methods have been developed to tackle cancer, each modality of cancer treatment has its own limitations and drawbacks. The development of minimally invasive treatment modalities for cancers remains a great challenge. Near-infrared (NIR) light-activated nanomaterial-mediated phototherapies, including photothermal and photodynamic therapies, provide an alternative means for spatially and temporally controlled minimally invasive treatments of cancers. Nanomaterials can serve as nanocargoes for the delivery of chemo-drugs, diagnostic contrast reagents, and organic photosensitizers, and can be used to directly generate heat or reactive oxygen species for the treatment of tumors without the need for organic photosensitizers with NIR-light irradiation. Here, current progress in NIR-light-activated nanomaterial-mediated photothermal therapy and photodynamic therapy is summarized. Furthermore, the effects of size, shape, and surface functionalities of nanomaterials on intracellular uptake, macrophage clearance, biodistribution, cytotoxicities, and biomedical efficacies are discussed. The use of various types of nanomaterials, such as gold nanoparticles, carbon nanotubes, graphene, and many other inorganic nanostructures, in combination with diagnostic and therapeutic modalities for solid tumors, is briefly reviewed.
Collapse
Affiliation(s)
- Raviraj Vankayala
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| | - Kuo Chu Hwang
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| |
Collapse
|
24
|
Lino MM, Ferreira L. Light-triggerable formulations for the intracellular controlled release of biomolecules. Drug Discov Today 2018; 23:1062-1070. [DOI: 10.1016/j.drudis.2018.01.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/03/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
|
25
|
Lino MM, Simões S, Pinho S, Ferreira L. Intracellular delivery of more than one protein with spatio-temporal control. NANOSCALE 2017; 9:18668-18680. [PMID: 29165472 DOI: 10.1039/c7nr02414b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Transient, non-integrative modulation of cell function by intracellular delivery of proteins has high potential in cellular reprogramming, gene editing and therapeutic medicine applications. Unfortunately, the capacity to deliver multiple proteins intracellularly with temporal and spatial control has not been demonstrated. Here, we report a near infrared (NIR) laser-activatable nanomaterial that allows for precise control over the release of two proteins from a single nanomaterial. The nanomaterial is formed by gold nanorods (AuNRs) modified with single stranded DNA (ssDNA) to which complementary DNA-conjugated proteins are hybridized. Using DNA strands with distinct melting temperatures we are able to control independently the release of each protein with a laser using the same wavelength but with different powers. Studies in mammalian cells show that AuNRs conjugated with proteins are internalized by endocytosis and NIR laser irradiation promotes endosomal escape and the release of the proteins from the AuNRs simultaneously. Our results further demonstrate the feasibility of protein release from a carrier that has been accumulated within the cell up to 1 day while maintaining its activity.
Collapse
Affiliation(s)
- Miguel M Lino
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal.
| | | | | | | |
Collapse
|
26
|
Maltanava HM, Poznyak SK, Andreeva DV, Quevedo MC, Bastos AC, Tedim J, Ferreira MGS, Skorb EV. Light-Induced Proton Pumping with a Semiconductor: Vision for Photoproton Lateral Separation and Robust Manipulation. ACS APPLIED MATERIALS & INTERFACES 2017; 9:24282-24289. [PMID: 28654237 DOI: 10.1021/acsami.7b05209] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Energy-transfer reactions are the key for living open systems, biological chemical networking, and the development of life-inspired nanoscale machineries. It is a challenge to find simple reliable synthetic chemical networks providing a localization of the time-dependent flux of matter. In this paper, we look to photocatalytic reaction on TiO2 from different angles, focusing on proton generation and introducing a reliable, minimal-reagent-consuming, stable inorganic light-promoted proton pump. Localized illumination was applied to a TiO2 surface in solution for reversible spatially controlled "inorganic photoproton" isometric cycling, the lateral separation of water-splitting reactions. The proton flux is pumped during the irradiation of the surface of TiO2 and dynamically maintained at the irradiated surface area in the absence of any membrane or predetermined material structure. Moreover, we spatially predetermine a transient acidic pH value on the TiO2 surface in the irradiated area with the feedback-driven generation of a base as deactivator. Importantly we describe how to effectively monitor the spatial localization of the process by the in situ scanning ion-selective electrode technique (SIET) measurements for pH and the scanning vibrating electrode technique (SVET) for local photoelectrochemical studies without additional pH-sensitive dye markers. This work shows the great potential for time- and space-resolved water-splitting reactions for following the investigation of pH-stimulated processes in open systems with their flexible localization on a surface.
Collapse
Affiliation(s)
- Hanna M Maltanava
- The Research Institute for Physical Chemical Problems, Belarusian State University , Minsk 220030, Belarus
| | - Sergey K Poznyak
- The Research Institute for Physical Chemical Problems, Belarusian State University , Minsk 220030, Belarus
| | - Daria V Andreeva
- Center for Soft and Living Matter, Institute of Basic Science Ulsan, National Institute of Science and Technology , Ulsan 44919, Republic of Korea
| | - Marcela C Quevedo
- Department of Materials and Ceramic Engineering, CICECO, University of Aveiro , Aveiro 3810-193, Portugal
| | - Alexandre C Bastos
- Department of Materials and Ceramic Engineering, CICECO, University of Aveiro , Aveiro 3810-193, Portugal
| | - João Tedim
- Department of Materials and Ceramic Engineering, CICECO, University of Aveiro , Aveiro 3810-193, Portugal
| | - Mário G S Ferreira
- Department of Materials and Ceramic Engineering, CICECO, University of Aveiro , Aveiro 3810-193, Portugal
| | - Ekaterina V Skorb
- Laboratory of Solution Chemistry of Advanced Materials and Technologies (SCAMT), ITMO University , St. Petersburg 197101, Russian Federation
- Department of Chemistry and Chemical Biology, Harvard University , 12 Oxford Street, Cambridge 02138, Massachusetts, United States
| |
Collapse
|
27
|
Štefančíková L, Lacombe S, Salado D, Porcel E, Pagáčová E, Tillement O, Lux F, Depeš D, Kozubek S, Falk M. Effect of gadolinium-based nanoparticles on nuclear DNA damage and repair in glioblastoma tumor cells. J Nanobiotechnology 2016; 14:63. [PMID: 27464501 PMCID: PMC4964094 DOI: 10.1186/s12951-016-0215-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/18/2016] [Indexed: 12/03/2022] Open
Abstract
Background Tumor targeting of radiotherapy represents a great challenge. The addition of multimodal nanoparticles, such as 3 nm gadolinium-based nanoparticles (GdBNs), has been proposed as a promising strategy to amplify the effects of radiation in tumors and improve diagnostics using the same agents. This singular property named theranostic is a unique advantage of GdBNs. It has been established that the amplification of radiation effects by GdBNs appears due to fast electronic processes. However, the influence of these nanoparticles on cells is not yet understood. In particular, it remains dubious how nanoparticles activated by ionizing radiation interact with cells and their constituents. A crucial question remains open of whether damage to the nucleus is necessary for the radiosensitization exerted by GdBNs (and other nanoparticles). Methods We studied the effect of GdBNs on the induction and repair of DNA double-strand breaks (DSBs) in the nuclear DNA of U87 tumor cells irradiated with γ-rays. For this purpose, we used currently the most sensitive method of DSBs detection based on high-resolution confocal fluorescence microscopy coupled with immunodetection of two independent DSBs markers. Results We show that, in the conditions where GdBNs amplify radiation effects, they remain localized in the cytoplasm, i.e. do not penetrate into the nucleus. In addition, the presence of GdBNs in the cytoplasm neither increases induction of DSBs by γ-rays in the nuclear DNA nor affects their consequent repair. Conclusions Our results suggest that the radiosensitization mediated by GdBNs is a cytoplasmic event that is independent of the nuclear DNA breakage, a phenomenon commonly accepted as the explanation of biological radiation effects. Considering our earlier recognized colocalization of GdBNs with the lysosomes and endosomes, we revolutionary hypothesize here about these organelles as potential targets for (some) nanoparticles. If confirmed, this finding of cytoplasmically determined radiosensitization opens new perspectives of using nano-radioenhancers to improve radiotherapy without escalating the risk of pathologies related to genetic damage.
Collapse
Affiliation(s)
- Lenka Štefančíková
- Department of Cell Biology and Radiobiology, Institute of Biophysics of ASCR, Brno, Czech Republic. .,Institute des Sciences Moléculaires d'Orsay (ISMO), Université Paris Sud 11, CNRS, Université Paris Saclay, Bât 351, 91405, Orsay Cedex, France.
| | - Sandrine Lacombe
- Institute des Sciences Moléculaires d'Orsay (ISMO), Université Paris Sud 11, CNRS, Université Paris Saclay, Bât 351, 91405, Orsay Cedex, France
| | - Daniela Salado
- Institute des Sciences Moléculaires d'Orsay (ISMO), Université Paris Sud 11, CNRS, Université Paris Saclay, Bât 351, 91405, Orsay Cedex, France
| | - Erika Porcel
- Institute des Sciences Moléculaires d'Orsay (ISMO), Université Paris Sud 11, CNRS, Université Paris Saclay, Bât 351, 91405, Orsay Cedex, France
| | - Eva Pagáčová
- Department of Cell Biology and Radiobiology, Institute of Biophysics of ASCR, Brno, Czech Republic
| | - Olivier Tillement
- Institut Lumière Matière, Université Claude Bernard Lyon 1, CNRS, 69622, Villeurbanne Cedex, France
| | - François Lux
- Institut Lumière Matière, Université Claude Bernard Lyon 1, CNRS, 69622, Villeurbanne Cedex, France
| | - Daniel Depeš
- Department of Cell Biology and Radiobiology, Institute of Biophysics of ASCR, Brno, Czech Republic
| | - Stanislav Kozubek
- Department of Cell Biology and Radiobiology, Institute of Biophysics of ASCR, Brno, Czech Republic
| | - Martin Falk
- Department of Cell Biology and Radiobiology, Institute of Biophysics of ASCR, Brno, Czech Republic.
| |
Collapse
|
28
|
Rau LR, Huang WY, Liaw JW, Tsai SW. Photothermal effects of laser-activated surface plasmonic gold nanoparticles on the apoptosis and osteogenesis of osteoblast-like cells. Int J Nanomedicine 2016; 11:3461-73. [PMID: 27555768 PMCID: PMC4968862 DOI: 10.2147/ijn.s108152] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The specific properties of gold nanoparticles (AuNPs) make them a novel class of photothermal agents that can induce cancer cell damage and even death through the conversion of optical energy to thermal energy. Most relevant studies have focused on increasing the precision of cell targeting, improving the efficacy of energy transfer, and exploring additional functions. Nevertheless, most cells can uptake nanosized particles through nonspecific endocytosis; therefore, before hyperthermia via AuNPs can be applied for clinical use, it is important to understand the adverse optical–thermal effects of AuNPs on nontargeted cells. However, few studies have investigated the thermal effects induced by pulsed laser-activated AuNPs on nearby healthy cells due to nonspecific treatment. The aim of this study is to evaluate the photothermal effects induced by AuNPs plus a pulsed laser on MG63, an osteoblast-like cell line, specifically examining the effects on cell morphology, viability, death program, and differentiation. The cells were treated with media containing 50 nm AuNPs at a concentration of 5 ppm for 1 hour. Cultured cells were then exposed to irradiation at 60 mW/cm2 and 80 mW/cm2 by a Nd:YAG laser (532 nm wavelength). We observed that the cytoskeletons of MG63 cells treated with bare AuNPs followed by pulsed laser irradiation were damaged, and these cells had few bubbles on the cell membrane compared with those that were not treated (control) or were treated with AuNPs or the laser alone. There were no significant differences between the AuNPs plus laser treatment group and the other groups in terms of cell viability, death program analysis results, or alkaline phosphatase and calcium accumulation during culture for up to 21 days. However, the calcium deposit areas in the cells treated with AuNPs plus laser were larger than those in other groups during the early culture period.
Collapse
Affiliation(s)
- Lih-Rou Rau
- Graduate Institute of Biochemical and Biomedical Engineering
| | - Wan-Yu Huang
- Graduate Institute of Biochemical and Biomedical Engineering
| | - Jiunn-Woei Liaw
- Department of Mechanical Engineering; Center for Biomedical Engineering, Chang Gung University; Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan; Center for Advanced Molecular Imaging and Translation
| | - Shiao-Wen Tsai
- Graduate Institute of Biochemical and Biomedical Engineering; Center for Biomedical Engineering, Chang Gung University; Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| |
Collapse
|
29
|
Goldstein A, Soroka Y, Frušić-Zlotkin M, Lewis A, Kohen R. The bright side of plasmonic gold nanoparticles; activation of Nrf2, the cellular protective pathway. NANOSCALE 2016; 8:11748-11759. [PMID: 27224746 DOI: 10.1039/c6nr02113a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Plasmonic gold nanoparticles (AuNPs) are widely investigated for cancer therapy, due to their ability to strongly absorb light and convert it to heat and thus selectively destroy tumor cells. In this study we shed light on a new aspect of AuNPs and their plasmonic excitation, wherein they can provide anti-oxidant and anti-inflammatory protection by stimulating the cellular protective Nrf2 pathway. Our study was carried out on cells of the immune system, macrophages, and on skin cells, keratinocytes. A different response to AuNPs was noted in the two types of cells, explained by their distinct uptake profiles. In keratinocytes, the exposure to AuNPs, even at low concentrations, was sufficient to activate the Nrf2 pathway, without any irradiation, due to the presence of free AuNPs inside the cytosol. In contrast, in macrophages, the plasmonic excitation of the AuNPs by a low, non-lethal irradiation dose was required for their release from the constraining vesicles. The mechanism by which AuNPs activate the Nrf2 pathway was studied. Direct and indirect activation were suggested, based on the inherent ability of the AuNPs to react with thiol groups and to generate reactive oxygen species, in particular, under plasmonic excitation. The ability of AuNPs to directly activate the Nrf2 pathway renders them good candidates for treatment of disorders in which the up-regulation of Nrf2 is beneficial, specifically for topical treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Alona Goldstein
- The David and Ines Myers Skin Research Laboratory, The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112100, Israel. and Department of Applied Physics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Yoram Soroka
- The David and Ines Myers Skin Research Laboratory, The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112100, Israel.
| | - Marina Frušić-Zlotkin
- The David and Ines Myers Skin Research Laboratory, The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112100, Israel.
| | - Aaron Lewis
- Department of Applied Physics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Ron Kohen
- The David and Ines Myers Skin Research Laboratory, The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112100, Israel.
| |
Collapse
|
30
|
Krawinkel J, Torres-Mapa ML, Werelius K, Heisterkamp A, Rüttermann S, Romanos GE, Gerhardt-Szép S. Gold Nanoparticle-Mediated Delivery of Molecules into Primary Human Gingival Fibroblasts Using ns-Laser Pulses: A Pilot Study. MATERIALS 2016; 9:ma9050397. [PMID: 28773519 PMCID: PMC5503001 DOI: 10.3390/ma9050397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/04/2016] [Accepted: 05/16/2016] [Indexed: 11/18/2022]
Abstract
Interaction of gold nanoparticles (AuNPs) in the vicinity of cells’ membrane with a pulsed laser (λ = 532 nm, τ = 1 ns) leads to perforation of the cell membrane, thereby allowing extracellular molecules to diffuse into the cell. The objective of this study was to develop an experimental setting to deliver molecules into primary human gingival fibroblasts (pHFIB-G) by using ns-laser pulses interacting with AuNPs (study group). To compare the parameters required for manipulation of pHFIB-G with those needed for cell lines, a canine pleomorphic adenoma cell line (ZMTH3) was used (control group). Non-laser-treated cells incubated with AuNPs and the delivery molecules served as negative control. Laser irradiation (up to 35 mJ/cm2) resulted in a significant proportion of manipulated fibroblasts (up to 85%, compared to non-irradiated cells: p < 0.05), while cell viability (97%) was not reduced significantly. pHFIB-G were perforated as efficiently as ZMTH3. No significant decrease of metabolic cell activity was observed up to 72 h after laser treatment. The fibroblasts took up dextrans with molecular weights up to 500 kDa. Interaction of AuNPs and a pulsed laser beam yields a spatially selective technique for manipulation of even primary cells such as pHFIB-G in high throughput.
Collapse
Affiliation(s)
- Judith Krawinkel
- Institute of Applied Optics, Friedrich-Schiller-University Jena, Fröbelstieg 1, Jena 07743, Germany.
| | - Maria Leilani Torres-Mapa
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University Hannover, Welfengarten 1, Hannover 30167, Germany.
| | - Kristian Werelius
- Department of Postgraduate Education, J.W. Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University Hannover, Welfengarten 1, Hannover 30167, Germany.
| | - Stefan Rüttermann
- Department of Operative Dentistry, Carolinum Dental University-Institute GmbH, J.W. Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Georgios E Romanos
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, NY 11794, USA.
- Department of Oral Surgery and Implant Dentistry, Carolinum Dental University-Institute GmbH, J.W. Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Susanne Gerhardt-Szép
- Department of Operative Dentistry, Carolinum Dental University-Institute GmbH, J.W. Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| |
Collapse
|
31
|
Chadwick S, Salah D, Livesey PM, Brust M, Volk M. Singlet Oxygen Generation by Laser Irradiation of Gold Nanoparticles. THE JOURNAL OF PHYSICAL CHEMISTRY. C, NANOMATERIALS AND INTERFACES 2016; 120:10647-10657. [PMID: 27239247 PMCID: PMC4878812 DOI: 10.1021/acs.jpcc.6b02005] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/03/2016] [Indexed: 05/23/2023]
Abstract
The formation of singlet oxygen by irradiation of gold nanoparticles in their plasmon resonance band with continuous or pulsed laser light has been investigated. Citrate-stabilized nanoparticles were found to facilitate the photogeneration of singlet oxygen, albeit with low quantum yield. The reaction caused by pulsed laser irradiation makes use of the equilibrated hot electrons that can reach temperatures of several thousand degrees during the laser pulse. Although less efficient, continuous irradiation, which acts via the short-lived directly excited primary "hot" electrons only, can produce enough singlet oxygen for photodynamic cancer therapy and has significant advantages for practical applications. However, careful design of the nanoparticles is needed, since even a moderately thick capping layer can completely inhibit singlet oxygen formation. Moreover, the efficiency of the process also depends on the nanoparticle size.
Collapse
Affiliation(s)
- Samantha
J. Chadwick
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United
Kingdom
| | - Dina Salah
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United
Kingdom
- Biophysics
Group, Physics Department, Ain Shams University, Cairo, Egypt
| | - Penelope M. Livesey
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United
Kingdom
| | - Mathias Brust
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United
Kingdom
| | - Martin Volk
- Surface
Science Research Centre, Department of Chemistry, University of Liverpool, Abercromby Square, Liverpool L69 3BX, United Kingdom
| |
Collapse
|
32
|
Andries M, Pricop D, Oprica L, Creanga DE, Iacomi F. The effect of visible light on gold nanoparticles and some bioeffects on environmental fungi. Int J Pharm 2016; 505:255-61. [DOI: 10.1016/j.ijpharm.2016.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 12/14/2022]
|
33
|
Giner-Casares JJ, Henriksen-Lacey M, García I, Liz-Marzán LM. Plasmonic Surfaces for Cell Growth and Retrieval Triggered by Near-Infrared Light. Angew Chem Int Ed Engl 2016; 55:974-8. [PMID: 26594015 PMCID: PMC4737312 DOI: 10.1002/anie.201509025] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 10/31/2015] [Indexed: 01/19/2023]
Abstract
Methods for efficient detachment of cells avoiding damage are required in tissue engineering and regenerative medicine. We introduce a bottom-up approach to build plasmonic substrates using micellar block copolymer nanolithography to generate a 2D array of Au seeds, followed by chemical growth leading to anisotropic nanoparticles. The resulting plasmonic substrates show a broad plasmon band covering a wide part of the visible and near-infrared (NIR) spectral ranges. Both human and murine cells were successfully grown on the substrates. A simple functionalization step of the plasmonic substrates with the cyclic arginylglycylaspartic acid (c-RGD) peptide allowed us to tune the morphology of integrin-rich human umbilical vein endothelial cells (HUVEC). Subsequent irradiation with a NIR laser led to highly efficient detachment of the cells with cell viability confirmed using the MTT assay. We thus propose the use of such plasmonic substrates for cell growth and controlled detachment using remote near-IR irradiation, as a general method for cell culture in biomedical applications.
Collapse
Affiliation(s)
- Juan J Giner-Casares
- CIC biomaGUNE, Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain.
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain.
| | - Malou Henriksen-Lacey
- CIC biomaGUNE, Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain
| | - Isabel García
- CIC biomaGUNE, Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain.
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009, Donostia-San Sebastián, Spain.
- Ikerbasque, Basque Foundation for Science, 48013, Bilbao, Spain.
| |
Collapse
|
34
|
Optical and electron microscopy study of laser-based intracellular molecule delivery using peptide-conjugated photodispersible gold nanoparticle agglomerates. J Nanobiotechnology 2016; 14:2. [PMID: 26745990 PMCID: PMC4706709 DOI: 10.1186/s12951-015-0155-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/29/2015] [Indexed: 12/25/2022] Open
Abstract
Background Cell-penetrating peptides (CPPs) can act as carriers for therapeutic molecules such as drugs and genetic constructs for medical applications. The triggered release of the molecule into the cytoplasm can be crucial to its effective delivery. Hence, we implemented and characterized laser interaction with defined gold nanoparticle agglomerates conjugated to CPPs which enables efficient endosomal rupture and intracellular release of molecules transported. Results Gold nanoparticles generated by pulsed laser ablation in liquid were conjugated with CPPs forming agglomerates and the intracellular release of molecules was triggered via pulsed laser irradiation (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\lambda$$\end{document}λ = 532 nm, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\tau _{pulse}$$\end{document}τpulse = 1 ns). The CPPs enhance the uptake of the agglomerates along with the cargo which can be co-incubated with the agglomerates. The interaction of incident laser light with gold nanoparticle agglomerates leads to heat deposition and field enhancement in the vicinity of the particles. This highly precise effect deagglomerates the nanoparticles and disrupts the enclosing endosomal membrane. Transmission electron microscopy images confirmed this rupture for radiant exposures of 25 mJ/cm\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{2}$$\end{document}2 and above. Successful intracellular release was shown using the fluorescent dye calcein. For a radiant exposure of 35 mJ/cm\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{2}$$\end{document}2 we found calcein delivery in 81 % of the treated cells while maintaining a high percentage of cell viability. Furthermore, cell proliferation and metabolic activity were not reduced 72 h after the treatment. Conclusion CPPs trigger the uptake of the gold nanoparticle agglomerates via endocytosis and co-resident molecules in the endosomes are released by applying laser irradiation, preventing their intraendosomal degradation. Due to the highly localized effect, the cell membrane integrity is not affected. Therefore, this technique can be an efficient tool for spatially and temporally confined intracellular release. The utilization of specifically designed photodispersible gold nanoparticle agglomerates (65 nm) can open novel avenues in imaging and molecule delivery. Due to the induced deagglomeration the primary, small particles (~5 nm) are more likely to be removed from the body. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0155-8) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Giner-Casares JJ, Henriksen-Lacey M, García I, Liz-Marzán LM. Plasmonic Surfaces for Cell Growth and Retrieval Triggered by Near-Infrared Light. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201509025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Juan J. Giner-Casares
- CIC biomaGUNE; Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN); Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
| | - Malou Henriksen-Lacey
- CIC biomaGUNE; Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN); Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
| | - Isabel García
- CIC biomaGUNE; Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN); Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
| | - Luis M. Liz-Marzán
- CIC biomaGUNE; Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN); Paseo de Miramón 182 20009 Donostia-San Sebastián Spain
- Ikerbasque, Basque Foundation for Science; 48013 Bilbao Spain
| |
Collapse
|
36
|
Mukherjee A, Castanares M, Hedayati M, Wabler M, Trock B, Kulkarni P, Rodriguez R, Getzenberg RH, DeWeese TL, Ivkov R, Lupold SE. Monitoring nanoparticle-mediated cellular hyperthermia with a high-sensitivity biosensor. Nanomedicine (Lond) 2015; 9:2729-43. [PMID: 24547783 DOI: 10.2217/nnm.13.207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To develop and apply a heat-responsive and secreted reporter assay for comparing cellular response to nanoparticle (NP)- and macroscopic-mediated sublethal hyperthermia. MATERIALS & METHODS Reporter cells were heated by water bath (macroscopic heating) or iron oxide NPs activated by alternating magnetic fields (nanoscopic heating). Cellular responses to these thermal stresses were measured in the conditioned media by secreted luciferase assay. RESULTS & CONCLUSION Reporter activity was responsive to macroscopic and nanoparticle heating and activity correlated with measured macroscopic thermal dose. Significant cellular responses were observed with NP heating under doses that were insufficient to measurably change the temperature of the system. Under these conditions, the reporter response correlated with proximity to cells loaded with heated nanoparticles. These results suggest that NP and macroscopic hyperthermia may be distinctive under conditions of mild hyperthermia.
Collapse
Affiliation(s)
- Amarnath Mukherjee
- The James Buchanan Brady Urological Institute & Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Pérez-Hernández M, Del Pino P, Mitchell SG, Moros M, Stepien G, Pelaz B, Parak WJ, Gálvez EM, Pardo J, de la Fuente JM. Dissecting the molecular mechanism of apoptosis during photothermal therapy using gold nanoprisms. ACS NANO 2015; 9:52-61. [PMID: 25493329 DOI: 10.1021/nn505468v] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The photothermal response of plasmonic nanomaterials can be exploited for a number of biomedical applications in diagnostics (biosensing and optoacoustic imaging) and therapy (drug delivery and photothermal therapy). The most common cellular response to photothermal cancer treatment (ablation of solid tumors) using plasmonic nanomaterials is necrosis, a process that releases intracellular constituents into the extracellular milieu producing detrimental inflammatory responses. Here we report the use of laser-induced photothermal therapy employing gold nanoprisms (NPRs) to specifically induce apoptosis in mouse embryonic fibroblast cells transformed with the SV40 virus. Laser-irradiated "hot" NPRs activate the intrinsic/mitochondrial pathway of apoptosis (programmed cell death), which is mediated by the nuclear-encoded proteins Bak and Bax through the activation of the BH3-only protein Bid. We confirm that an apoptosis mechanism is responsible by showing how the NPR-mediated cell death is dependent on the presence of caspase-9 and caspase-3 proteins. The ability to selectively induce apoptotic cell death and to understand the subsequent mechanisms provides the foundations to predict and optimize NP-based photothermal therapy to treat cancer patients suffering from chemo- and radioresistance.
Collapse
Affiliation(s)
- Marta Pérez-Hernández
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza , 50018 Zaragoza, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kodiha M, Wang YM, Hutter E, Maysinger D, Stochaj U. Off to the organelles - killing cancer cells with targeted gold nanoparticles. Am J Cancer Res 2015; 5:357-70. [PMID: 25699096 PMCID: PMC4329500 DOI: 10.7150/thno.10657] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 12/16/2014] [Indexed: 12/18/2022] Open
Abstract
Gold nanoparticles (AuNPs) are excellent tools for cancer cell imaging and basic research. However, they have yet to reach their full potential in the clinic. At present, we are only beginning to understand the molecular mechanisms that underlie the biological effects of AuNPs, including the structural and functional changes of cancer cells. This knowledge is critical for two aspects of nanomedicine. First, it will define the AuNP-induced events at the subcellular and molecular level, thereby possibly identifying new targets for cancer treatment. Second, it could provide new strategies to improve AuNP-dependent cancer diagnosis and treatment. Our review summarizes the impact of AuNPs on selected subcellular organelles that are relevant to cancer therapy. We focus on the nucleus, its subcompartments, and mitochondria, because they are intimately linked to cancer cell survival, growth, proliferation and death. While non-targeted AuNPs can damage tumor cells, concentrating AuNPs in particular subcellular locations will likely improve tumor cell killing. Thus, it will increase cancer cell damage by photothermal ablation, mechanical injury or localized drug delivery. This concept is promising, but AuNPs have to overcome multiple hurdles to perform these tasks. AuNP size, morphology and surface modification are critical parameters for their delivery to organelles. Recent strategies explored all of these variables, and surface functionalization has become crucial to concentrate AuNPs in subcellular compartments. Here, we highlight the use of AuNPs to damage cancer cells and their organelles. We discuss current limitations of AuNP-based cancer research and conclude with future directions for AuNP-dependent cancer treatment.
Collapse
|
39
|
Carregal-Romero S, Guardia P, Yu X, Hartmann R, Pellegrino T, Parak WJ. Magnetically triggered release of molecular cargo from iron oxide nanoparticle loaded microcapsules. NANOSCALE 2015; 7:570-6. [PMID: 25415565 DOI: 10.1039/c4nr04055d] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Photothermal release of cargo molecules has been extensively studied for bioapplications. For instance, microcapsules decorated with plasmonic nanoparticles have been widely used in in vitro assays. However, some concerns about their suitability for some in vivo applications cannot be easily overcome, in particular the limited penetration depth of light (even infrared). Magnetic nanoparticles are alternative heat-mediators for local heating, which can be triggered by applying an alternating magnetic field (AMF). AMFs are much less absorbed by tissue than light and thus can penetrate deeper overcoming the above mentioned limitations. Here we present iron oxide nanocube-modified microcapsules as a platform for magnetically triggered molecular release. Layer-by-layer assembled polyelectrolyte microcapsules with 4.6 μm diameter, which had 18 nm diameter iron oxide nanocubes integrated in their walls, were synthesized. The microcapsules were further loaded with an organic fluorescent polymer (Cascade Blue-labelled dextran), which was used as a model of molecular cargo. Through an AMF the magnetic nanoparticles were able to heat their surroundings and destroy the microcapsule walls, leading to a final release of the embedded cargo to the surrounding solution. The cargo release was monitored in solution by measuring the increase in both absorbance and fluorescence signal after the exposure to an AMF. Our results demonstrate that magnetothermal release of the encapsulated material is possible using magnetic nanoparticles with a high heating performance.
Collapse
|
40
|
Lv J, Zhang X, Li N, Wang B, He S. Absorption-dependent generation of singlet oxygen from gold bipyramids excited under low power density. RSC Adv 2015. [DOI: 10.1039/c5ra15362j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Gold bipyramid-mediated singlet oxygen generation for photodynamic therapy of cancer cells in the optical biological window.
Collapse
Affiliation(s)
- Junliang Lv
- Centre for Optical and Electromagnetic Research
- SCNU-ZJU Joint Research Center of Photonics
- South China Academy of Advanced Optoelectronics
- South China Normal University (SCNU)
- 510006 Guangzhou
| | - Xin Zhang
- Centre for Optical and Electromagnetic Research
- SCNU-ZJU Joint Research Center of Photonics
- South China Academy of Advanced Optoelectronics
- South China Normal University (SCNU)
- 510006 Guangzhou
| | - Nana Li
- Centre for Optical and Electromagnetic Research
- SCNU-ZJU Joint Research Center of Photonics
- South China Academy of Advanced Optoelectronics
- South China Normal University (SCNU)
- 510006 Guangzhou
| | - Baoju Wang
- Centre for Optical and Electromagnetic Research
- SCNU-ZJU Joint Research Center of Photonics
- South China Academy of Advanced Optoelectronics
- South China Normal University (SCNU)
- 510006 Guangzhou
| | - Sailing He
- Centre for Optical and Electromagnetic Research
- SCNU-ZJU Joint Research Center of Photonics
- South China Academy of Advanced Optoelectronics
- South China Normal University (SCNU)
- 510006 Guangzhou
| |
Collapse
|
41
|
Efficient, dual-stimuli responsive cytosolic gene delivery using a RGD modified disulfide-linked polyethylenimine functionalized gold nanorod. J Control Release 2014; 196:37-51. [DOI: 10.1016/j.jconrel.2014.09.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/04/2014] [Accepted: 09/25/2014] [Indexed: 12/16/2022]
|
42
|
van der Pol E, Coumans FAW, Sturk A, Nieuwland R, van Leeuwen TG. Refractive index determination of nanoparticles in suspension using nanoparticle tracking analysis. NANO LETTERS 2014; 14:6195-201. [PMID: 25256919 DOI: 10.1021/nl503371p] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The refractive index (RI) dictates interaction between light and nanoparticles and therefore is important to health, environmental, and materials sciences. Using nanoparticle tracking analysis, we have determined the RI of heterogeneous particles <500 nm in suspension. We demonstrate feasibility of distinguishing silica and polystyrene beads based on their RI. The hitherto unknown RI of extracellular vesicles from human urine was determined at 1.37 (mean). This method enables differentiation of single nanoparticles based on their RI.
Collapse
Affiliation(s)
- Edwin van der Pol
- Biomedical Engineering and Physics, ‡ Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
43
|
Kumar A, Kumar S, Rhim WK, Kim GH, Nam JM. Oxidative Nanopeeling Chemistry-Based Synthesis and Photodynamic and Photothermal Therapeutic Applications of Plasmonic Core-Petal Nanostructures. J Am Chem Soc 2014; 136:16317-25. [DOI: 10.1021/ja5085699] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Amit Kumar
- Department of Chemistry, Seoul National University, Seoul 151-747, South Korea
| | - Sumit Kumar
- Department of Chemistry, Seoul National University, Seoul 151-747, South Korea
| | - Won-Kyu Rhim
- Department of Chemistry, Seoul National University, Seoul 151-747, South Korea
| | - Gyeong-Hwan Kim
- Department of Chemistry, Seoul National University, Seoul 151-747, South Korea
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, Seoul 151-747, South Korea
| |
Collapse
|
44
|
Pharmacological aspects of release from microcapsules - from polymeric multilayers to lipid membranes. Curr Opin Pharmacol 2014; 18:129-40. [PMID: 25450067 DOI: 10.1016/j.coph.2014.09.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/16/2014] [Accepted: 09/21/2014] [Indexed: 11/24/2022]
Abstract
This review is devoted to pharmacological applications of principles of release from capsules to overcome the membrane barrier. Many of these principles were developed in the context of polymeric multilayer capsule membrane modulation, but they are also pertinent to liposomes, polymersomes, capsosomes, particles, emulsion-based carriers and other carriers. We look at these methods from the physical, chemical or biological driving mechanisms point of view. In addition to applicability for carriers in drug delivery, these release methods are significant for another area directly related to pharmacology - modulation of the permeability of the membranes and thus promoting the action of drugs. Emerging technologies, including ionic current monitoring through a lipid membrane on a nanopore, are also highlighted.
Collapse
|
45
|
Cell localisation of gadolinium-based nanoparticles and related radiosensitising efficacy in glioblastoma cells. Cancer Nanotechnol 2014; 5:6. [PMID: 25328549 PMCID: PMC4192560 DOI: 10.1186/s12645-014-0006-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 09/09/2014] [Indexed: 11/10/2022] Open
Abstract
Recently, the addition of nanoparticles (NPs) has been proposed as a new strategy to enhance the effect of radiotherapy particularly in the treatment of aggressive tumors such as glioblastoma. The physical processes involved in radiosensitisation by nanoparticles have been well studied although further understanding of its biological impact is still lacking, and this includes the localisation of these NPs in the target cells. Most studies were performed with NPs tagged with fluorescent markers. However, the presence of these markers can influence the NPs uptake and localisation. In this study, a set of methods was used to unambiguously and fully characterise the uptake of label-free NPs, their co-localisation with cell organelles, and their radiosensitising efficacy. This set was applied to the case of gadolinium-based nanoparticles (GdBN) used to amplify the radiation killing of U87 glioblastoma cells extracted from highly aggressive human tumor. For the first time, Synchrotron Radiation Deep UV (SR-DUV) microscopy is proposed as a new tool to track label-free GdBN. It confirmed the localisation of the NPs in the cytoplasm of U87 cells and the absence of NPs in the nucleus. In a second step, Transmission Electron Microscopy (TEM) demonstrated that GdBN penetrate cells by endocytosis. Third, using confocal microscopy it was found that GdBN co-localise with lysosomes but not with mitochondria. Finally, clonogenic assay measurements proved that the presence of NPs in the lysosomes induces a neat amplification of the killing of glioblastoma cells irradiated by gamma rays. The set of combined experimental protocols—TEM, SR-DUV and confocal microscopy—demonstrates a new standard method to study the localisation of label-free NPs together with their radiosensitising properties. This will further the understanding of NP-induced radiosentisation and contribute to the development of nanoagents for radiotherapy.
Collapse
|
46
|
Zhao J, Wallace M, Melancon MP. Cancer theranostics with gold nanoshells. Nanomedicine (Lond) 2014; 9:2041-57. [DOI: 10.2217/nnm.14.136] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gold nanoshells (AuNSs) present a vivid example of integrating nanoscience in order to solve a biomedical problem. AuNSs exhibit tunable surface plasmon resonance, which can be tuned to the near-infrared region in order to realize optimal tissue penetration. The highly efficient light-to-heat transformation by AuNSs during laser irradiation causes thermal damage to the tumor without damaging healthy organs. Transient nanobubbles can form around AuNSs during laser treatment and induce mechanical stress specifically in tumor cells. AuNSs also serve as a versatile platform for the delivery of various diagnostic and therapeutic agents. In this article, we describe the physicochemical properties of AuNSs in the context of their design, preparation and application in cancer theranostics. Ultimately, we look beyond the current research on AuNSs and discussed future challenges to their successful translation into clinical use.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Michael Wallace
- Department of Interventional Radiology – Unit 1471, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Marites P Melancon
- Department of Interventional Radiology – Unit 1471, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Graduate School for Biomedical Science, The University of Texas at Houston, 6767 Bertner Avenue, Houston, TX 77030, USA
| |
Collapse
|
47
|
Palankar R, Pinchasik BE, Khlebtsov BN, Kolesnikova TA, Möhwald H, Winterhalter M, Skirtach AG. Nanoplasmonically-induced defects in lipid membrane monitored by ion current: transient nanopores versus membrane rupture. NANO LETTERS 2014; 14:4273-4279. [PMID: 24961609 DOI: 10.1021/nl500907k] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
We have developed a nanoplasmonic-based approach to induce nanometer-sized local defects in the phospholipid membranes. Here, gold nanorods and nanoparticles having plasmon resonances in the near-infrared (NIR) spectral range are used as optical absorption centers in the lipid membrane. Defects optically induced by NIR-laser irradiation of gold nanoparticles are continuously monitored by high-precision ion conductance measurement. Localized laser-mediated heating of nanorods and nanoparticle aggregates cause either (a) transient nanopores in lipid membranes or (b) irreversible rupture of the membrane. To monitor transient opening and closing, an electrophysiological setup is assembled wherein a giant liposome is spread over a micrometer hole in a glass slide forming a single bilayer of high Ohmic resistance (so-called gigaseal), while laser light is coupled in and focused on the membrane. The energy associated with the localized heating is discussed and compared with typical elastic parameters in the lipid membranes. The method presented here provides a novel methodology for better understanding of transport across artificial or natural biological membranes.
Collapse
Affiliation(s)
- Raghavendra Palankar
- ZIK HIKE, Nanostructure Group, Ernst-Moritz-Arndt-Universität Greifswald , 17489 Greifswald, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Gao L, Liu R, Gao F, Wang Y, Jiang X, Gao X. Plasmon-mediated generation of reactive oxygen species from near-infrared light excited gold nanocages for photodynamic therapy in vitro. ACS NANO 2014; 8:7260-71. [PMID: 24992260 DOI: 10.1021/nn502325j] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
We have performed fundamental assays of gold nanocages (AuNCs) as intrinsic inorganic photosensitizers mediating generation of reactive oxygen species (ROS) by plasmon-enabled photochemistry under near-infrared (NIR) one/two-photon irradiation. We disclosed that NIR light excited hot electrons transform into either ROS or hyperthermia. Electron spin resonance spectroscopy was applied to demonstrate the production of three main radical species, namely, singlet oxygen ((1)O2), superoxide radical anion (O2(-•)), and hydroxyl radical ((•)OH). The existence of hot electrons from irradiated AuNCs was confirmed by a well-designed photoelectrochemical experiment based on a three-electrode system. It could be speculated that surface plasmons excited in AuNCs first decay into hot electrons, and then the generated hot electrons sensitize oxygen to form ROS through energy and electron transfer modes. We also compared AuNCs' ROS generation efficiency in different surface chemical environments under one/two-photon irradiation and verified that, compared with one-photon irradiation, two-photon irradiation could bring about much more ROS. Furthermore, in vitro, under two-photon irradiation, ROS can trigger mitochondrial depolarization and caspase protein up-regulation to initiate tumor cell apoptosis. Meanwhile, hyperthermia mainly induces tumor cell necrosis. Our findings suggest that plasmon-mediated ROS and hyperthermia can be facilely regulated for optimized anticancer phototherapy.
Collapse
Affiliation(s)
- Liang Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | | | | | | | | | | |
Collapse
|
49
|
Synthesis and characterization of CREKA-conjugated iron oxide nanoparticles for hyperthermia applications. Acta Biomater 2014; 10:2622-9. [PMID: 24486913 DOI: 10.1016/j.actbio.2014.01.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 01/02/2014] [Accepted: 01/21/2014] [Indexed: 11/22/2022]
Abstract
One of the current challenges in the systemic delivery of nanoparticles in cancer therapy applications is the lack of effective tumor localization. Iron oxide nanoparticles (IONPs) coated with crosslinked dextran were functionalized with the tumor-homing peptide CREKA, which binds to fibrinogen complexes in the extracellular matrix of tumors. This allows for the homing of these nanoparticles to tumor tissue. The IONP core allows for particle heating upon exposure to an alternating magnetic field (AMF), while the dextran coating stabilizes the particles in suspension and decreases the cytotoxicity of the system. Magnetically mediated hyperthermia (MMH) allows for the heating of tumor tissue to increase the efficacy of traditional cancer treatments using IONPs. While MMH provides the opportunity for localized heating, this method is currently limited by the lack of particle penetration into tumor tissue, even after effective targeted delivery to the tumor site. The CREKA-conjugated nanoparticles presented were characterized for their size, stability, heating capabilities and biocompatibility. The particles had a hydrated diameter of 52nm, were stable in phosphate buffered saline solution and media with 10% v/v fetal bovine serum over at least 12h, and generated enough heat to raise solution temperatures well into the hyperthermia range (41-45°C) when exposed to an AMF, owing to an average specific absorption rate of 83.5Wg(-1). Cytotoxicity studies demonstrated that the particles have low cytotoxicity over long exposure times at low concentrations. A fibrinogen clotting assay was used to determine the binding affinity of CREKA-conjugated particles, which was significantly greater than the binding affinity of dextran, only coated IONPs demonstrating the potential for this particle system to effectively home to a variety of tumor locations. Finally, it was shown that in vitro MMH increased the effects of cisplatin compared with cisplatin or MMH treatments alone.
Collapse
|
50
|
Porcel E, Tillement O, Lux F, Mowat P, Usami N, Kobayashi K, Furusawa Y, Le Sech C, Li S, Lacombe S. Gadolinium-based nanoparticles to improve the hadrontherapy performances. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1601-8. [PMID: 24846523 DOI: 10.1016/j.nano.2014.05.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 04/25/2014] [Accepted: 05/12/2014] [Indexed: 01/04/2023]
Abstract
UNLABELLED Nanomedicine is proposed as a novel strategy to improve the performance of radiotherapy. High-Z nanoparticles are known to enhance the effects of ionizing radiation. Recently, multimodal nanoparticles such as gadolinium-based nanoagents were proposed to amplify the effects of x-rays and g-rays and to improve MRI diagnosis. For tumors sited in sensitive tissues, childhood cases and radioresistant cancers, hadrontherapy is considered superior to x-rays and g-rays. Hadrontherapy, based on fast ion radiation, has the advantage of avoiding damage to the tissues behind the tumor; however, the damage caused in front of the tumor is its major limitation. Here, we demonstrate that multimodal gadolinium-based nanoparticles amplify cell death with fast ions used as radiation. Molecular scale experiments give insights into the mechanisms underlying the amplification of radiation effects. This proof-of-concept opens up novel perspectives for multimodal nanomedicine in hadrontherapy, ultimately reducing negative radiation effects in healthy tissues in front of the tumor. FROM THE CLINICAL EDITOR Gadolinium-chelating polysiloxane nanoparticles were previously reported to amplify the anti-tumor effects of x-rays and g-rays and to serve as MRI contrast agents. Fast ion radiation-based hadrontherapy avoids damage to the tissues behind the tumor, with a major limitation of tissue damage in front of the tumor. This study demonstrates a potential role for the above nanoagents in optimizing hadrontherapy with preventive effects in healthy tissue and amplified cell death in the tumor.
Collapse
Affiliation(s)
- Erika Porcel
- Institut des Sciences Moléculaires d'Orsay, Université Paris Sud, CNRS, Orsay, France
| | - Olivier Tillement
- Institut Lumière Matière, Université Claude Bernard Lyon 1, CNRS, Villeurbanne, France
| | - François Lux
- Institut Lumière Matière, Université Claude Bernard Lyon 1, CNRS, Villeurbanne, France
| | - Pierre Mowat
- Institut Lumière Matière, Université Claude Bernard Lyon 1, CNRS, Villeurbanne, France
| | - Noriko Usami
- Photon Factory, Institute of Material Science, High Energy Accelerator Research Organization, Oho 1, Tsukuba, Ibaraki, Japan
| | - Katsumi Kobayashi
- Photon Factory, Institute of Material Science, High Energy Accelerator Research Organization, Oho 1, Tsukuba, Ibaraki, Japan
| | - Yoshiya Furusawa
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba, Japan
| | - Claude Le Sech
- Institut des Sciences Moléculaires d'Orsay, Université Paris Sud, CNRS, Orsay, France
| | - Sha Li
- Institut des Sciences Moléculaires d'Orsay, Université Paris Sud, CNRS, Orsay, France
| | - Sandrine Lacombe
- Institut des Sciences Moléculaires d'Orsay, Université Paris Sud, CNRS, Orsay, France.
| |
Collapse
|