Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Bioconjug Chem ; 33(12): 2320-2331, 2022 12 21.
Article in English | MEDLINE | ID: mdl-35156818

ABSTRACT

Despite significant research regarding metastasis, there has been limited success in preventing it. However, gold nanoparticle (AuNP) technology has shown the potential to inhibit metastasis. Our earlier studies of gold nanorod-assisted plasmonic photothermal therapy (AuNRs-PPTT), where gold nanorods (AuNRs) were irradiated with near-infrared (NIR) light to induce heat, were utilized in slowing cancer cell migration in vitro. Herein, we have expanded the in vitro studies of the AuNRs-PPTT to xenograft mice to inhibit metastasis of mammary gland tumors. The study duration was 32 days from 4T1 cancer cell injections in four treatment groups: control (PBS), NIR Only, AuNRs, and AuNRs + NIR. Multiple AuNRs-PPTT treatment sessions with intratumoral AuNRs injections were conducted every 7 days on average on the mice. Photoacoustic spectroscopy has been utilized to study the distribution and aggregation of AuNRs within the tumors and the drainage of particles to the sentinel right subiliac lymph node. The photoacoustic results revealed that the AuNRs' shapes are still stable regardless of their heterogeneous distributions inside the mammalian tumor and lymph nodes. Bioluminescence imaging was used to monitor metastasis using luciferin labeling techniques and has shown that AuNRs-PPTT inhibited metastasis completely within the first 21 days. Moreover, proteomics was run to determine the most pivotal inhibitory pathways: NETosis, cell growth, cell proliferation, inflammation, and extracellular matrix (ECM) degradation. These five mechanisms are interdependent within related networks, which synergistically explains the molecular mechanism of metastasis inhibition by AuNRs-PPTT. The current in vivo data ensures the viability of PPTT applications in inhibiting metastasis in humans.


Subject(s)
Hyperthermia, Induced , Metal Nanoparticles , Nanotubes , Neoplasms , Humans , Animals , Mice , Gold/chemistry , Heterografts , Photothermal Therapy , Phototherapy/methods , Hyperthermia, Induced/methods , Metal Nanoparticles/therapeutic use , Neoplasms/therapy , Nanotubes/chemistry , Cell Line, Tumor , Mammals
2.
ACS Nano ; 12(9): 9279-9290, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30118603

ABSTRACT

Most cancer-related deaths come from metastasis. It was recently discovered that nanoparticles could inhibit cancer cell migration. Whereas most researchers focus on single-cell migration, the effect of nanoparticle treatment on collective cell migration has not been explored. Collective migration occurs commonly in many types of cancer metastasis, where a group of cancer cells move together, which requires the contractility of the cytoskeleton filaments and the connection of neighboring cells by the cell junction proteins. Here, we demonstrate that gold nanorods (AuNRs) and the introduction of near-infrared light could inhibit the cancer cell collective migration by altering the actin filaments and cell junctions with significantly triggered phosphorylation changes of essential proteins, using mass spectrometry-based phosphoproteomics. Further observation using super-resolution stochastic optical reconstruction microscopy (STORM) showed the actin cytoskeleton filament bundles were disturbed, which is difficult to differentiate under a normal fluorescence microscope. The decreased expression level of N-cadherin junctions and morphological changes of tight junction protein zonula occludens 2 were also observed. All of these results indicate possible functions of the AuNR treatments in regulating and remodeling the actin filaments and cell junction proteins, which contribute to decreasing cancer cell collective migration.


Subject(s)
Actins/metabolism , Gold/pharmacology , Intercellular Junctions/drug effects , Metal Nanoparticles/chemistry , Phototherapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gold/chemistry , HeLa Cells , Humans , Infrared Rays , MCF-7 Cells , Tumor Cells, Cultured
3.
Proc Natl Acad Sci U S A ; 114(28): E5655-E5663, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652358

ABSTRACT

Metastasis is responsible for most cancer-related deaths, but the current clinical treatments are not effective. Recently, gold nanoparticles (AuNPs) were discovered to inhibit cancer cell migration and prevent metastasis. Rationally designed AuNPs could greatly benefit their antimigration property, but the molecular mechanisms need to be explored. Cytoskeletons are cell structural proteins that closely relate to migration, and surface receptor integrins play critical roles in controlling the organization of cytoskeletons. Herein, we developed a strategy to inhibit cancer cell migration by targeting integrins, using Arg-Gly-Asp (RGD) peptide-functionalized gold nanorods. To enhance the effect, AuNRs were further activated with 808-nm near-infrared (NIR) light to generate heat for photothermal therapy (PPTT), where the temperature was adjusted not to affect the cell viability/proliferation. Our results demonstrate changes in cell morphology, observed as cytoskeleton protrusions-i.e., lamellipodia and filopodia-were reduced after treatment. The Western blot analysis indicates the downstream effectors of integrin were attracted toward the antimigration direction. Proteomics results indicated broad perturbations in four signaling pathways, Rho GTPases, actin, microtubule, and kinases-related pathways, which are the downstream regulators of integrins. Due to the dominant role of integrins in controlling cytoskeleton, focal adhesion, actomyosin contraction, and actin and microtubule assembly have been disrupted by targeting integrins. PPTT further enhanced the remodeling of cytoskeletal proteins and decreased migration. In summary, the ability of targeting AuNRs to cancer cell integrins and the introduction of PPTT stimulated broad regulation on the cytoskeleton, which provides the evidence for a potential medical application for controlling cancer metastasis.


Subject(s)
Cytoskeleton/metabolism , Gold/chemistry , Integrins/metabolism , Nanotubes/chemistry , Neoplasms/pathology , Neoplasms/therapy , Phototherapy/methods , Actomyosin/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Survival , Cytoskeletal Proteins , Disulfides , Humans , Hyperthermia, Induced , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/metabolism , Proteomics
4.
Proc Natl Acad Sci U S A ; 114(15): E3110-E3118, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28356516

ABSTRACT

Gold nanorods (AuNRs)-assisted plasmonic photothermal therapy (AuNRs-PPTT) is a promising strategy for combating cancer in which AuNRs absorb near-infrared light and convert it into heat, causing cell death mainly by apoptosis and/or necrosis. Developing a valid PPTT that induces cancer cell apoptosis and avoids necrosis in vivo and exploring its molecular mechanism of action is of great importance. Furthermore, assessment of the long-term fate of the AuNRs after treatment is critical for clinical use. We first optimized the size, surface modification [rifampicin (RF) conjugation], and concentration (2.5 nM) of AuNRs and the PPTT laser power (2 W/cm2) to achieve maximal induction of apoptosis. Second, we studied the potential mechanism of action of AuNRs-PPTT using quantitative proteomic analysis in mouse tumor tissues. Several death pathways were identified, mainly involving apoptosis and cell death by releasing neutrophil extracellular traps (NETs) (NETosis), which were more obvious upon PPTT using RF-conjugated AuNRs (AuNRs@RF) than with polyethylene glycol thiol-conjugated AuNRs. Cytochrome c and p53-related apoptosis mechanisms were identified as contributing to the enhanced effect of PPTT with AuNRs@RF. Furthermore, Pin1 and IL18-related signaling contributed to the observed perturbation of the NETosis pathway by PPTT with AuNRs@RF. Third, we report a 15-month toxicity study that showed no long-term toxicity of AuNRs in vivo. Together, these data demonstrate that our AuNRs-PPTT platform is effective and safe for cancer therapy in mouse models. These findings provide a strong framework for the translation of PPTT to the clinic.


Subject(s)
Carcinoma, Squamous Cell/therapy , Gold/pharmacology , Head and Neck Neoplasms/therapy , Hyperthermia, Induced , Lasers , Nanotubes/chemistry , Phototherapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Female , Gold/chemistry , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proteomics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Am Chem Soc ; 138(47): 15434-15442, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27809520

ABSTRACT

In cancer plasmonic photothermal therapy (PPTT), plasmonic nanoparticles are used to convert light into localized heat, leading to cancer cell death. Among plasmonic nanoparticles, gold nanorods (AuNRs) with specific dimensions enabling them to absorb near-infrared laser light have been widely used. The detailed mechanism of PPTT therapy, however, still remains poorly understood. Typically, surface-enhanced Raman spectroscopy (SERS) has been used to detect time-dependent changes in the intensity of the vibration frequencies of molecules that appear or disappear during different cellular processes. A complete proven assignment of the molecular identity of these vibrations and their biological importance has not yet been accomplished. Mass spectrometry (MS) is a powerful technique that is able to accurately identify molecules in chemical mixtures by observing their m/z values and fragmentation patterns. Here, we complemented the study of changes in SERS spectra with MS-based metabolomics and proteomics to identify the chemical species responsible for the observed changes in SERS band intensities during PPTT. We observed an increase in intensity of the bands at around 1000, 1207, and 1580 cm-1, which were assigned in the literature to phenylalanine, albeit with dispute. Our metabolomics results showed increased levels of phenylalanine, its derivatives, and phenylalanine-containing peptides, providing evidence for more confidence in the SERS peak assignments. To better understand the mechanism of phenylalanine increase upon PPTT, we combined metabolomics and proteomics results through network analysis, which proved that phenylalanine metabolism was perturbed. Furthermore, several apoptosis pathways were activated via key proteins (e.g., HADHA and ACAT1), consistent with the proposed role of altered phenylalanine metabolism in inducing apoptosis. Our study shows that the integration of the SERS with MS-based metabolomics and proteomics can assist the assignment of signals in SERS spectra and further characterize the related molecular mechanisms of the cellular processes involved in PPTT.


Subject(s)
Gold/pharmacology , Metabolomics , Nanotubes/chemistry , Neoplasms/pathology , Phenylalanine/metabolism , Phototherapy , Proteomics , Spectrum Analysis, Raman , Cell Death/drug effects , Cell Line, Tumor , Gold/chemistry , Humans , Particle Size , Surface Properties , Time Factors
6.
Int J Nanomedicine ; 11: 4849-4863, 2016.
Article in English | MEDLINE | ID: mdl-27703351

ABSTRACT

Plasmonic photothermal therapy (PPTT) is a cancer therapy in which gold nanorods are injected at the site of a tumor before near-infrared light is transiently applied to the tumor causing localized cell death. Previously, PPTT studies have been carried out on xenograft mice models. Herein, we report a study showing the feasibility of PPTT as applied to natural tumors in the mammary glands of dogs and cats, which more realistically represent their human equivalents at the molecular level. We optimized a regime of three low PPTT doses at 2-week intervals that ablated tumors mainly via apoptosis in 13 natural mammary gland tumors from seven animals. Histopathology, X-ray, blood profiles, and comprehensive examinations were used for both the diagnosis and the evaluation of tumor statuses before and after treatment. Histopathology results showed an obvious reduction in the cancer grade shortly after the first treatment and a complete regression after the third treatment. Blood tests showed no obvious change in liver and kidney functions. Similarly, X-ray diffraction showed no metastasis after 1 year of treatment. In conclusion, our study suggests the feasibility of applying the gold nanorods-PPTT on natural tumors in dogs and cats without any relapse or toxicity effects after 1 year of treatment.


Subject(s)
Apoptosis , Gold/therapeutic use , Hyperthermia, Induced/methods , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Nanotubes/chemistry , Surface Plasmon Resonance/methods , Animals , Cats , Dogs , Female , Gold/chemistry , Humans , MCF-7 Cells , Mice , Neoplasm Recurrence, Local/prevention & control , Phototherapy/methods , Spectroscopy, Near-Infrared
7.
J Photochem Photobiol B ; 164: 21-29, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27636008

ABSTRACT

BACKGROUND: The photothermal properties of gold nanoparticles (GNPs) are promising therapeutic modality for cancer. The study objective is to evaluate the therapeutic effect of the prepared PEGylated gold nano-semicubes (PEG-GNSCs) in skin cancer. The synthesized PEG-GNSCs were intermediate between cubic and rod shapes (low aspect ratio- rods). METHODS: In vitro toxicity was investigated in human skin melanoma Sk-Mel-28 cells, and skin squamous cell carcinoma was induced in CD1 mice by dimethylbenzanthracene (DMBA) and 12-O-tetradecanoyl-phorbol-13-acetate (TPA). RESULTS: The calculated IC50 in Sk-Mel-28 cells was 3.41µg/ml of PEG-GNSCs, in presence of laser exposure. Photothermal therapy using laser-stimulated PEG-GNSCs resulted in inhibited volume of skin tumors. Our findings indicated that the inflammatory mediators, nitric oxide and cycloxygenase-2, were inhibited in mice after being treated with low and high doses of PEG-GNSCs, accompanied with laser exposure. However, the tumor necrosis factor -α was markedly elevated, while there was no change in 5-lipoxygenase. The pro-angiogenic factor vascular endothelial growth factor was inhibited, while histone acetylation and apoptosis were induced in tumor-bearing groups, after being treated with laser-stimulated PEG-GNSCs. CONCLUSION: The present study indicated the promising photothermal therapeutic effect of laser-stimulated PEG-GNSCs as an effective modality to inhibit the tumor growth, the angiogenesis and partially the inflammation.


Subject(s)
Carcinogens/toxicity , Gold/chemistry , Metal Nanoparticles/therapeutic use , Phototherapy , Polyethylene Glycols/chemistry , Skin Neoplasms/therapy , Animals , Mice , Microscopy, Electron, Transmission , Skin Neoplasms/chemically induced
8.
Biomaterials ; 102: 1-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27318931

ABSTRACT

Plasmonic photothermal therapy (PPTT) is a promising cancer treatment where plasmonic nanoparticles are used to convert near infrared light to localized heat to cause cell death, mainly via apoptosis and necrosis. Modulating PPTT to induce cell apoptosis is more favorable than necrosis. Herein, we used a mild treatment condition using gold nanorods (AuNRs) to trigger apoptosis and tested how different cell lines responded to it. Three different cancer cell lines of epithelial origin: HSC (oral), MCF-7 (breast) and Huh7.5 (liver) had comparable AuNRs uptake and were heated to same environmental temperature (under 50 °C). However, Huh7.5 cells displayed a significant increase in cell apoptosis after PPTT as compared to the other two cell lines. As HSP70 is known to increase cellular resistance to heat, we determined relative HSP70 levels in these cells and results indicated that Huh7.5 cells had ten-fold decreased levels of HSP70 as compared with HSC and MCF-7 cells. We then down-regulated HSP70 with a siRNA and observed that all three cell lines displayed significant reduction in viability and an increase in apoptosis after PPTT. As an enhancement to PPTT, we conjugated AuNRs with Quercetin, an inhibitor of HSP70 which displayed anti-cancer effects via apoptosis.


Subject(s)
Apoptosis , Gold/therapeutic use , HSP70 Heat-Shock Proteins/metabolism , Neoplasms/therapy , Cell Line, Tumor , Gold/chemistry , Humans , Hyperthermia, Induced/methods , MCF-7 Cells , Nanotubes/chemistry , Nanotubes/ultrastructure , Neoplasms/metabolism , Phototherapy/methods
9.
J Phys Chem B ; 118(5): 1319-26, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24433049

ABSTRACT

The development of new and improved photothermal contrast agents for the successful treatment of cancer (or other diseases) via plasmonic photothermal therapy (PPTT) is a crucial part of the application of nanotechnology in medicine. Gold nanorods (AuNRs) have been found to be the most effective photothermal contrast agents, both in vitro and in vivo. Therefore, determining the optimum AuNR size needed for applications in PPTT is of great interest. In the present work, we utilized theoretical calculations as well as experimental techniques in vitro to determine this optimum AuNR size by comparing plasmonic properties and the efficacy as photothermal contrast agents of three different sizes of AuNRs. Our theoretical calculations showed that the contribution of absorbance to the total extinction, the electric field, and the distance at which this field extends away from the nanoparticle surface all govern the effectiveness of the amount of heat these particles generate upon NIR laser irradiation. Comparing between three different AuNRs (38 × 11, 28 × 8, and 17 × 5 nm), we determined that the 28 × 8 nm AuNR is the most effective in plasmonic photothermal heat generation. These results encouraged us to carry out in vitro experiments to compare the PPTT efficacy of the different sized AuNRs. The 28 × 8 nm AuNR was found to be the most effective photothermal contrast agent for PPTT of human oral squamous cell carcinoma. This size AuNR has the best compromise between the total amount of light absorbed and the fraction of which is converted to heat. In addition, the distance at which the electric field extends from the particle surface is most ideal for this size AuNR, as it is sufficient to allow for coupling between the fields of adjacent particles in solution (i.e., particle aggregates), resulting in effective heating in solution.


Subject(s)
Gold/chemistry , Nanotubes/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Contrast Media/chemistry , Contrast Media/therapeutic use , Contrast Media/toxicity , Humans , Infrared Rays , Lasers , Models, Theoretical , Mouth Neoplasms/therapy , Nanotubes/toxicity , Particle Size , Phototherapy , Polyethylene Glycols/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL