Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.098
Filter
1.
ACS Appl Mater Interfaces ; 16(33): 43302-43316, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39111771

ABSTRACT

Photothermal therapy (PTT) is a method for eradicating tumor tissues through the use of photothermal materials and photosensitizing agents that absorb light energy from laser sources and convert it into heat, which selectively targets and destroys cancer cells while sparing healthy tissue. MXenes have been intensively investigated as photosensitizing agents for PTT. However, achieving the selectivity of MXenes to the tumor cells remains a challenge. Specific antibodies (Ab) against tumor antigens can achieve homing of the photosensitizing agents toward tumor cells, but their immobilization on MXene received little attention. Here, we offer a strategy for the selective ablation of melanoma cells using MXene-polydopamine-antiCEACAM1 Ab complexes. We coated Ti3C2Tx MXene with polydopamine (PDA), a natural compound that attaches Ab to the MXene surface, followed by conjugation with an anti-CEACAM1 Ab. Our experiments confirm the biocompatibility of the Ti3C2Tx-PDA and Ti3C2Tx-PDA-antiCEACAM1 Ab complexes across various cell types. We also established a protocol for the selective ablation of CEACAM1-positive melanoma cells using near-infrared irradiation. The obtained complexes exhibit high selectivity and efficiency in targeting and eliminating CEACAM1-positive melanoma cells while sparing CEACAM1-negative cells. These results demonstrate the potential of MXene-PDA-Ab complexes for cancer therapy. They underline the critical role of targeted therapies in oncology, offering a promising avenue for the precise and safe treatment of melanoma and possibly other cancers characterized by specific biomarkers. Future research will aim to refine these complexes for clinical use, paving the way for new strategies for cancer treatment.


Subject(s)
Indoles , Melanoma , Polymers , Polymers/chemistry , Indoles/chemistry , Indoles/pharmacology , Humans , Melanoma/pathology , Melanoma/drug therapy , Melanoma/therapy , Cell Line, Tumor , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Titanium/chemistry , Animals , Mice , Carcinoembryonic Antigen/immunology , Photothermal Therapy
2.
ACS Appl Mater Interfaces ; 16(33): 43416-43429, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39121233

ABSTRACT

The lipopolyplex, a multicomponent nonviral gene carrier, generally demonstrates superior colloidal stability, reduced cytotoxicity, and high transfection efficiency. In this study, a new concept, photochemical reaction-induced transfection, using photosensitizer (PS)-loaded lipopolyplexes was applied, which led to enhanced transfection and cytotoxic effects by photoexcitation of the photosensitizer. Hypericin, a hydrophobic photosensitizer, was encapsulated in the lipid bilayer of liposomes. The preformed nanosized hypericin liposomes enclosed the linear polyethylenimine (lPEI)/pDNA polyplexes, resulting in the formation of hypericin lipopolyplexes (Hy-LPP). The diameters of Hy-LPP containing 50 nM hypericin and 0.25 µg of pDNA were 185.6 ± 7.74 nm and 230.2 ± 4.60 nm, respectively, measured by dynamic light scattering (DLS) and atomic force microscopy (AFM). Gel electrophoresis confirmed the encapsulation of hypericin and pDNA in lipopolyplexes. Furthermore, in vitro irradiation of intracellular Hy-LPP at radiant exposures of 200, 600, and 1000 mJ/cm2 was evaluated. It demonstrated 60- to 75-fold higher in vitro luciferase expression than that in nonirradiated cells. The lactate dehydrogenase (LDH) assay supported that reduced transfection was a consequence of photocytotoxicity. The developed photosensitizer-loaded lipopolyplexes improved the transfection efficiency of an exogenous gene or induced photocytotoxicity; however, the frontier lies in the applied photochemical dose. The light-triggered photoexcitation of intracellular hypericin resulted in the generation of reactive oxygen species (ROS), leading to photoselective transfection in HepG2 cells. It was concluded that the two codelivered therapeutics resulted in enhanced transfection and a photodynamic effect by tuning the applied photochemical dose.


Subject(s)
Anthracenes , Carcinoma, Hepatocellular , Liposomes , Liver Neoplasms , Perylene , Photosensitizing Agents , Transfection , Perylene/chemistry , Perylene/analogs & derivatives , Perylene/pharmacology , Anthracenes/chemistry , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Transfection/methods , Liposomes/chemistry , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/pathology , Hep G2 Cells , DNA/chemistry , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Plasmids/chemistry , Cell Survival/drug effects
3.
ACS Nano ; 18(32): 21009-21023, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39087239

ABSTRACT

Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), induce high morbidity and mortality rates, which challenge the present approaches for the treatment of ALI/ARDS. The clinically used photosensitizer verteporfin (VER) exhibits great potential in the treatment of acute lung injury and acute respiratory distress syndrome (ALI/ARDS) by regulating macrophage polarization and reducing inflammation. Nevertheless, its hydrophobic characteristics, nonspecificity, and constrained bioavailability hinder its therapeutic efficacy. In this work, we developed a type of VER-cored artificial exosome (EVM), which was produced by using mesoporous silica nanoparticles (MSNs) to load VER, followed by the exocytosis of internalized VER-MSNs from mouse bone marrow-derived mesenchymal stem cells (mBMSCs) without further modification. Both in vitro and in vivo assessments confirmed the powerful anti-inflammation induced by EVM. EVM also showed significant higher accumulation to inflammatory lungs compared with healthy ones, which was beneficial to the treatment of ALI/ARDS. EVM improved pulmonary function, attenuated lung injury, and reduced mortality in ALI mice with high levels of biocompatibility, exhibiting a 5-fold higher survival rate than the control. This type of artificial exosome emitted near-infrared light in the presence of laser activation, which endowed EVM with trackable ability both in vitro and in vivo. Our work developed a type of clinically used photosensitizer-loaded artificial exosome with membrane integrity and traceability. To the best of our knowledge, this kind of intracellularly synthesized artificial exosome was developed and showed great potential in ALI/ARDS therapy.


Subject(s)
Acute Lung Injury , Exosomes , Silicon Dioxide , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Mice , Exosomes/metabolism , Exosomes/chemistry , Silicon Dioxide/chemistry , Verteporfin/pharmacology , Verteporfin/chemistry , Verteporfin/therapeutic use , Nanoparticles/chemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Male , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Porosity
4.
Anal Chim Acta ; 1320: 343035, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39142775

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) is a pioneering and effective anticancer modality with low adverse effects and high selectivity. Hypochlorous acid or hypochlorite (HClO/ClO-) is a type of inflammatory cytokine. The abnormal increase of ClO- in tumor cells is related to tumor pathogenesis and may be a "friend" for the design and synthesis of responsive phototherapy agents. However, preparing responsive phototherapy agents for all-in-one noninvasive diagnosis and simultaneous in situ therapy in a complex tumor environment is highly desirable but still remains an enormously demanding task. RESULTS: An acceptor-π bridge-donor-π bridge-acceptor (A-π-D-π-A) type photosensitizer TPTPy was designed and synthesized based on the phenothiazine structure which was used as the donor moiety as well as a ClO- responsive group. TPTPy was a multifunctional mitochondria targeted aggregation-induced emission (AIE) photosensitizer which could quickly and sensitively respond to ClO- with fluorescence "turn on" performance (19-fold fluorescence enhancement) and enhanced type I reactive oxygen species (ROS) generation to effectively ablate hypoxic tumor cells. The detection limit of TPTPy to ClO- was calculated to be 185.38 nM. The well-tailored TPTPy anchoring to mitochondria and producing ROS in situ could disrupt mitochondria and promote cell apoptosis. TPTPy was able to image inflammatory cells and tumor cells through ClO- response. In vivo results revealed that TPTPy was successfully utilized for PDT in tumor bearing nude mice and exhibited excellent biological safety for major organs. SIGNIFICANCE AND NOVELTY: A win-win integration strategy was proposed to design a tumor intracellular ClO- responsive photosensitizer TPTPy capable of both type I and type II ROS production to achieve photodynamic therapy of tumor. This work sheds light on the win-win integration design by taking full advantage of the characteristics of tumor microenvironment to build up responsive photosensitizer for in situ PDT of tumor.


Subject(s)
Hypochlorous Acid , Mitochondria , Photochemotherapy , Photosensitizing Agents , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/therapeutic use , Hypochlorous Acid/analysis , Hypochlorous Acid/metabolism , Animals , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/analysis , Mice, Inbred BALB C , Phenothiazines/chemistry , Phenothiazines/pharmacology , Mice, Nude , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Optical Imaging , Cell Survival/drug effects
5.
ACS Appl Bio Mater ; 7(8): 5771-5779, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39110771

ABSTRACT

Nanomaterials with photoresponsivity have garnered attention due to their fluorescence imaging, photodynamic, and photothermal therapeutic properties. In this study, a photoresponsivity nanoassembly was developed by using photosensitizers and carbon dots (CDs). Due to their multiple excitation peaks and multicolor fluorescence emission, especially their membrane-permeating properties, these nanoassemblies can label cells with multiple colors and track cell imaging in real time. Additionally, the incorporation of photosensitizers and CDs provides the nanoassemblies with the potential for photodynamic therapy (PDT) and photothermal therapy (PTT). The nanoassemblies effectively suppressed the activity of Escherichia coli and Staphylococcus aureus through PDT and PTT. Moreover, the nanoassemblies exhibited a high affinity for E. coli and S. aureus. These distinct features confer broad-spectrum antibacterial properties to the nanoassemblies. As a photoresponsivity nanoplatform, these nanoassemblies have demonstrated potential applications in the fields of bioimaging and antimicrobial.


Subject(s)
Anti-Bacterial Agents , Biocompatible Materials , Escherichia coli , Materials Testing , Particle Size , Photosensitizing Agents , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Microbial Sensitivity Tests , Humans , Quantum Dots/chemistry , Nanostructures/chemistry , Carbon/chemistry , Carbon/pharmacology , Optical Imaging , Photochemotherapy , Cell Survival/drug effects
6.
J Phys Chem B ; 128(32): 7803-7812, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39106822

ABSTRACT

The DNA binding and cellular uptake of the lambda enantiomer of two bis-tetraazaphenanthrene (TAP) Ru(II) polypyridyl complexes containing either a linear dppn (1) or a hooked bdppz (2) benzodipyridophenazine ligand are reported, and the role of different charge-transfer states of the structural isomers in the photo-oxidation of guanine is explored. Both complexes possess characteristic metal-to-ligand charge-transfer (MLCT) bands between 400 and 500 nm and emission at ca. 630 nm in an aerated aqueous solution. Transient visible absorption (TrA) spectroscopy reveals that 400 nm excitation of 1 yields a dppn-based metal-to-ligand charge-transfer (MLCT) state, which in turn populates a dppn intraligand (3IL) state. In contrast, photoexcitation of 2 results in an MLCT state on the TAP ligand and not the intercalating bdppz ligand. Both 1 and 2 bind strongly to double-stranded guanine-rich DNA with a loss of emission. Combined TrA and time-resolved infrared (TRIR) spectroscopy confirms formation of the guanine radical cation when 2 is bound to the d(G5C5)2 duplex, which is not the case when 1 is bound to the same duplex and indicates a different mechanism of action in DNA. Utilizing the long-lived triplet excited lifetime, we show good uptake and localization of 2 in live cells as well as isolated chromosomes. The observed shortening of the excited-state lifetime of 2 when internalized in cell chromosomes is consistent with DNA binding and luminescent quenching due to guanine photo-oxidation.


Subject(s)
DNA , Guanine , Intercalating Agents , Ruthenium , DNA/chemistry , DNA/metabolism , Guanine/chemistry , Ruthenium/chemistry , Ligands , Intercalating Agents/chemistry , Humans , Isomerism , Photochemical Processes , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Pyridines/chemistry , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Molecular Structure , HeLa Cells
7.
Bioconjug Chem ; 35(8): 1269-1282, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39120495

ABSTRACT

The penetration ability of visible light (<2 mm) and near-infrared (NIR) light (∼1 cm) remarkably impairs the therapeutic efficacy and clinical applications of photodynamic therapy (PDT). To address the limitation of light penetration depth, a novel self-luminescent bacterium, teLuc.FP-EcN, has been engineered through transfection of a fusion expression plasmid containing the luciferase gene teLuc and bright red fluorescent protein mScarlet-I into Escherichia coli Nissle 1917 (EcN). The engineered teLuc.FP-EcN can specifically target and colonize tumors without significant toxicity to the host. Acting as a continuous internal light source, teLuc.FP-EcN can activate the photosensitizer chlorin e6 (Ce6) to generate reactive oxygen species (ROS) and then effectively destroy tumor tissue from the inside. As a result, a significant reduction in tumor proliferation and extension of the overall survival in mouse tumor models has been observed. Furthermore, teLuc.FP-EcN-boosted PDT amplified its therapeutic effect by activating antitumor immune response, including the conversion of M2 macrophages into pro-inflammatory M1 macrophages, as well as an increase in the proportion of CD3+ T cells and a decrease in T-cell exhaustion. In conclusion, teLuc.FP-EcN can be used as an implantable light source for tumor phototherapy, which simultaneously possesses ROS generation and immune regulation.


Subject(s)
Breast Neoplasms , Photochemotherapy , Photosensitizing Agents , Photochemotherapy/methods , Animals , Female , Mice , Breast Neoplasms/drug therapy , Breast Neoplasms/therapy , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Humans , Reactive Oxygen Species/metabolism , Chlorophyllides , Mice, Inbred BALB C , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/therapeutic use , Cell Line, Tumor , Escherichia coli/drug effects
8.
Chem Commun (Camb) ; 60(68): 9082-9084, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39105653

ABSTRACT

Naphthalocyanine-based agents exhibit huge potential in photodynamic therapy, yet their photodynamic performance is restricted by the penetration depth of the external laser. Herein, we employed 18F-FDG as an internal light source to excite silicon naphthalocyanine nanoparticles to simultaneously circumvent radiative transition and boost 1O2 generation for tumor suppression.


Subject(s)
Nanoparticles , Photochemotherapy , Photosensitizing Agents , Nanoparticles/chemistry , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Animals , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Mice , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Silicon/chemistry
9.
J Photochem Photobiol B ; 258: 112999, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39126752

ABSTRACT

5-Aminolevulinic acid (5-ALA) is a prodrug of porphyrin IX (PpIX). Disadvantages of 5-ALA include poor stability, rapid elimination, poor bioavailability, and weak cell penetration, which greatly reduce the clinical effect of 5-ALA based photodynamic therapy (PDT). Presently, a novel targeting nanosystem was constructed using gold nanoparticles (AuNPs) as carriers loaded with a CSNIDARAC (CC9)-targeting peptide and 5-ALA via Au-sulphur and ionic bonds, respectively, and then wrapped in polylactic glycolic acid (PLGA) NPs via self-assembly to improve the antitumor effects and reduce the side effect. The successful preparation of ALA/CC9@ AuNPs-PLGA NPs was verified using ultraviolet-visible, Fourier transform infrared spectroscopy, and X-ray photoelectron spectroscopy. The analyses revealed good sphericity with a particle size of approximately140 nm, Zeta potential of 10.11 mV, and slow-controlled release characteristic in a weak acid environment. Confocal microscopy revealed targeting of NCL-H460 cells by NPs by actively internalising CC9 and avoiding the phagocytic action of RAW264.7 cells, and live fluorescence imaging revealed targeting of tumours in tumour-bearing mice. Compared to free 5-ALA, the nanosystem displayed amplified anticancer activity by increasing production of PpIX and reactive oxygen species to induce mitochondrial pathway apoptosis. Antitumor efficacy was consistently observed in three-dimensionally cultured cells as the loss of integrity of tumour balls. More potent anti-tumour efficacy was demonstrated in xenograft tumour models by decreased growth rate and increased tumour apoptosis. Histological analysis showed that this system was not toxic, with lowered liver toxicity of 5-ALA. Thus, ALA/CC9@AuNPs-PLGA NPs deliver 5-ALA via a carrier cascade, with excellent effects on tumour accumulation and PDT through passive enhanced permeability and retention action and active targeting. This innovative strategy for cancer therapy requires more clinical trials before being implemented.


Subject(s)
Aminolevulinic Acid , Gold , Lung Neoplasms , Metal Nanoparticles , Photochemotherapy , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Animals , Gold/chemistry , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Mice , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Cell Line, Tumor , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Drug Carriers/chemistry , Apoptosis/drug effects , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
10.
ACS Appl Mater Interfaces ; 16(32): 41843-41854, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39092532

ABSTRACT

Oxidative degradation of the pathogenic amyloid-ß-peptide (Aß) aggregation is an effective and promising method to treat Alzheimer's disease under light irradiation. However, the limited penetration of external light sources into deep tissues has hindered the development of this treatment. Therefore, we have designed an unprecedented chemiluminescence-initiated photodynamic therapy system to replace external laser irradiation, primarily composed of d-glucose-based polyoxalate (G-poly(oxalate)), the novel photosensitizer (BD-Se-QM), and bis [2,4,5-trichloro-6-(pentoxy-carbonyl) phenyl] ester. BD-Se-QM possesses excellent singlet oxygen (1O2) generation efficiency and the ability to photooxidize Aß1-42 aggregates under white light. G-poly(oxalate) not only helps the nanosystem to cross the blood-brain barrier but also has sufficient oxalate ester groups to significantly enhance the efficiency of chemiluminescence resonance energy transfer. The oxalate ester groups in BD-Se-QM/NPs can chemically react with H2O2 to produce high-energy intermediates that activate BD-Se-QM, which can generate 1O2 to inhibit Aß1-42 aggregates and also promote microglial uptake of Aß1-42, reducing the Aß1-42-induced neurotoxicity. The chemically stimulated nanoplatform not only solves the drug delivery problem but also eliminates the need for external light sources. We anticipate that this chemically excited nanosystem could also be used for targeted delivery of other small molecule drugs.


Subject(s)
Amyloid beta-Peptides , Oxidation-Reduction , Peptide Fragments , Photosensitizing Agents , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Photochemotherapy , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Humans , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Light , Hydrogen Peroxide/chemistry , Protein Aggregates/drug effects , Mice
11.
Molecules ; 29(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39125107

ABSTRACT

Photothermal, photodynamic and sonodynamic cancer therapies offer opportunities for precise tumor ablation and reduce side effects. The cyclic guanylate adenylate synthase-stimulator of interferon genes (cGAS-STING) pathway has been considered a potential target to stimulate the immune system in patients and achieve a sustained immune response. Combining photothermal, photodynamic and sonodynamic therapies with cGAS-STING agonists represents a newly developed cancer treatment demonstrating noticeable innovation in its impact on the immune system. Recent reviews have concentrated on diverse materials and their function in cancer therapy. In this review, we focus on the molecular mechanism of photothermal, photodynamic and sonodynamic cancer therapies and the connected role of cGAS-STING agonists in treating cancer.


Subject(s)
Membrane Proteins , Neoplasms , Nucleotidyltransferases , Photochemotherapy , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Photochemotherapy/methods , Signal Transduction/drug effects , Animals , Ultrasonic Therapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry
12.
Spectrochim Acta A Mol Biomol Spectrosc ; 323: 124910, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39128309

ABSTRACT

An ultrasensitive strategy for in-situ visual monitoring of ATP in a single living tumor cell during mitochondria-targeted photodynamic therapy (PDT) process with high spatiotemporal resolution was proposed using surface-enhanced Raman scattering (SERS) 3D imaging technique. The nanostructures consisting of Au-Ag2S Janus nanoparticles functionalized with both Au nanoparticles linked by a DNA chain and a mitochondrial-targeting peptide (JMDA NPs) were deliberately employed to target mitochondria. The JMDA NPs exhibit excellent SERS activity and remarkable antitumor activity. The quantization of ATP relies on the intensity of the SERS probes bonded to the DNA, which shows a strong correlation with the generated hot spot between the Janus and the Au. Consequently, spatiotemporally controlled monitoring of ATP in the mitochondria of single living cells during the PDT process was achieved. Additionally, the JMDA NPs demonstrated remarkable capability for mitochondria-targeted PDT, providing significant antitumor effects and superior therapeutic safety both in vitro and in vivo. Our work presents an effective JMDA NPs-based SERS imaging strategy for in-situ and real-time 3D visualization of intracellular ATP in living tumor cells during the mitochondria-targeted PDT process, which enables significant information on the time point of PDT treatment and is beneficial to precious PDT applications in tumor therapy.


Subject(s)
Adenosine Triphosphate , Imaging, Three-Dimensional , Metal Nanoparticles , Mitochondria , Photochemotherapy , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Photochemotherapy/methods , Mitochondria/metabolism , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/analysis , Humans , Animals , Metal Nanoparticles/chemistry , Gold/chemistry , Cell Line, Tumor , Single-Cell Analysis , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry
13.
Nat Commun ; 15(1): 7150, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39168967

ABSTRACT

Despite the prevalent of hexagonal, tetragonal, and triangular pore structures in two-dimensional covalent organic frameworks (2D COFs), the pentagonal pores remain conspicuously absent. We herein present the Cairo pentagonal tessellated COFs, achieved through precisely chosen geometry and metrics of the linkers, resulting in unprecedented mcm topology. In each pentagonal structure, porphyrin units create four uniform sides around 15.5 Å with 90° angles, while tetrabiphenyl unit establish a bottom edge about 11.6 Å with 120° angles, aligning precisely with the criteria of Cairo Pentagon. According to the narrow bandgap and strong near-infrared (NIR) absorbance, as-synthesized COFs exhibit the efficient singlet oxygen (1O2) generation and photothermal conversion, resulting in NIR photothermal combined photodynamic therapy to guide cancer cell apoptosis. Mechanistic studies reveal that the good 1O2 production capability upregulates intracellular lipid peroxidation, leading to glutathione depletion, low expression of glutathione peroxidase 4, and induction of ferroptosis. The implementation of pentagonal Cairo tessellations in this work provides a promising strategy for diversifying COFs with new topologies, along with multimodal NIR phototherapy.


Subject(s)
Apoptosis , Infrared Rays , Photochemotherapy , Singlet Oxygen , Humans , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Photochemotherapy/methods , Metal-Organic Frameworks/chemistry , Porphyrins/chemistry , Animals , Lipid Peroxidation , Cell Line, Tumor , Ferroptosis , Phototherapy/methods , Mice , Glutathione/chemistry , Glutathione/metabolism , Photosensitizing Agents/chemistry , Neoplasms/therapy , Neoplasms/metabolism
14.
J Mater Sci Mater Med ; 35(1): 51, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39172269

ABSTRACT

The use of TiO2 as a photosensitizer in photodynamic therapy is limited due to TiO2 generates reactive oxygen species only under UV irradiation. The TiO2 surface has been modified with different functional groups to achieve activation at longer wavelengths (visible light). This work reports the synthesis, characterization, and biological toxicity assay of TiO2 nanoparticles functionalized with folic acid and combined with a zinc phthalocyanine to obtain a nano-photosensitizer for its application in photodynamic therapy for glioblastoma cancer treatment. The nano-photosensitizer was prepared using the sol-gel method. Folic acid and zinc phthalocyanine were added during the hydrolysis and condensation of titanium butoxide, which was the TiO2 precursor. The samples obtained were characterized by several microscopy and spectroscopy techniques. An in vitro toxicity test was performed using the MTT assay and the C6 cellular line. The results of the characterization showed that the structure of the nanoparticles corresponds mainly to the anatase phase. Successful functionalization with folic acid and an excellent combination with phthalocyanine was also achieved. Both folic acid-functionalized TiO2 and phthalocyanine-functionalized TiO2 had no cytotoxic effect on C6 cells (even at high concentrations) in comparison to Cis-Pt, which was very toxic to C6 cells. The materials behaved similarly to the control (untreated cells). The cell viability and light microscopy images suggest that both materials could be considered biocompatible and mildly phototoxic in these cells when activated by light.


Subject(s)
Cell Survival , Folic Acid , Glioblastoma , Indoles , Isoindoles , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Titanium , Zinc Compounds , Titanium/chemistry , Folic Acid/chemistry , Glioblastoma/drug therapy , Glioblastoma/pathology , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Indoles/chemistry , Indoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Nanoparticles/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Humans , Animals , Rats
15.
ACS Appl Mater Interfaces ; 16(33): 43257-43271, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39119624

ABSTRACT

Prostate cancer presents as a challenging disease, as it is often characterized as an immunologically "cold" tumor, leading to suboptimal outcomes with current immunotherapeutic approaches in clinical settings. Photodynamic therapy (PDT) harnesses reactive oxygen species generated by photosensitizers (PSs) to disrupt the intracellular redox equilibrium. This process induces DNA damage in both the mitochondria and nucleus, activating the process of immunogenic cell death (ICD) and the cGAS-STING pathway. Ultimately, this cascade of events leads to the initiation of antitumor immune responses. Nevertheless, existing PSs face challenges, including suboptimal tumor targeting, aggregation-induced quenching, and insufficient oxygen levels in the tumor regions. To this end, a versatile bionic nanoplatform has been designed for the simultaneous delivery of the aggregation-induced emission PS TPAQ-Py-PF6 and paclitaxel (PTX). The cell membrane camouflage of the nanoplatform leads to its remarkable abilities in tumor targeting and cellular internalization. Upon laser irradiation, the utilization of TPAQ-Py-PF6 in conjunction with PTX showcases a notable and enhanced synergistic antitumor impact. Additionally, the nanoplatform has the capability of initiating the cGAS-STING pathway, leading to the generation of cytokines. The presence of damage-associated molecular patterns induced by ICD collaborates with these aforementioned cytokines lead to the recruitment and facilitation of dendritic cell maturation. Consequently, this elicits a systemic immune response against tumors. In summary, this promising strategy highlights the use of a multifunctional biomimetic nanoplatform, combining chemotherapy, PDT, and immunotherapy to enhance the effectiveness of antitumor treatment.


Subject(s)
Immunogenic Cell Death , Immunotherapy , Membrane Proteins , Nucleotidyltransferases , Photochemotherapy , Photosensitizing Agents , Prostatic Neoplasms , Humans , Nucleotidyltransferases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Immunogenic Cell Death/drug effects , Immunogenic Cell Death/radiation effects , Membrane Proteins/metabolism , Male , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Animals , Mice , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Cell Line, Tumor , Drug Delivery Systems , Porphyrins/chemistry , Porphyrins/pharmacology
16.
ACS Appl Mater Interfaces ; 16(33): 43387-43399, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39136145

ABSTRACT

A wide variety of methods are being developed to ultimately defeat cancer; while some of these strategies have shown highly positive results, there are serious obstacles to overcome to completely eradicate this disease. So, it is crucial to construct multifunctional nanostructures possessing intelligent capabilities that can be utilized to treat cancer. A possible strategy for producing these multifunctional nanostructures is to combine various cancer treatment techniques. Based on this point of view, we successfully synthesized multifunctional HCuS@Cu2S@Au-P(NIPAM-co-AAm)-PpIX nanohybrids. The peculiarities of these thermosensitive polymer-modified and protoporphyrin IX (PpIX)-loaded hollow nanohybrids are that they combine photodynamic therapy (PDT), sonodynamic therapy (SDT), and photothermal therapy (PTT) with an intelligent design. As an all-in-one nanohybrids, HCuS@Cu2S@Au-P(NIPAM-co-AAm)-PpIX nanohybrids were employed in the SDT-PDT-PTT combination therapy, which proved to have a synergistic therapeutic effect for in vitro tumor treatments against breast tumors.


Subject(s)
Copper , Photochemotherapy , Photothermal Therapy , Protoporphyrins , Humans , Copper/chemistry , Copper/pharmacology , Protoporphyrins/chemistry , Protoporphyrins/pharmacology , Protoporphyrins/therapeutic use , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemical synthesis , Gold/chemistry , Female , Ultrasonic Therapy/methods , Nanostructures/chemistry , Nanostructures/therapeutic use , Breast Neoplasms/therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Sulfides/chemistry , Sulfides/pharmacology , Animals
17.
Gen Physiol Biophys ; 43(5): 469-484, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140687

ABSTRACT

Ruthenium nitrosyl (Ru-NO) complexes are of interest as photoactive nitric oxide (NO) donor candidates for local therapeutic applications. NO plays a crucial regulatory role in skin homeostasis, concentration-dependently affecting processes like the proliferation, apoptosis, autophagy and redox balance. In this context, we investigated HE-10, a ruthenium-based photoinducible NO donor, for its pro-oxidant and cytotoxic effects under light and dark conditions in VH10 human foreskin fibroblast cells. We also tested its intracellular and extracellular NO-releasing function. Our study reveals a significant dose-dependent cytotoxic effect of HE-10, an increase in intracellular reactive oxygen and nitrogen species, and the occurrence of apoptosis in skin fibroblast cells. Furthermore, exposure to both increasing doses of HE-10 and white LED light led to substantial cellular events, including a significant induction of autophagy and G2/M phase cell cycle arrest. Paradoxically, these effects were not solely attributable to NO release based on DAF2-DA NO probe results, suggesting that intracellular photochemical reactions additional to NO photolysis contribute to HE-10's biological activity. This study shows that HE-10 exhibits both cytotoxic and potential therapeutic effects, depending on concentration and light exposure. These findings are crucial for developing targeted Ru-NO complex treatments for skin diseases and potentially certain types of skin cancer, where controlled NO release could be beneficial.


Subject(s)
Fibroblasts , Nitric Oxide , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Nitric Oxide/metabolism , Cell Line , Cell Survival/drug effects , Ruthenium/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Dose-Response Relationship, Drug , Light
18.
Artif Cells Nanomed Biotechnol ; 52(1): 384-398, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39101753

ABSTRACT

Nanotechnology-based cancer treatment has received considerable attention, and these treatments generally use drug-loaded nanoparticles (NPs) to target and destroy cancer cells. Nanotechnology combined with photodynamic therapy (PDT) has demonstrated positive outcomes in cancer therapy. Combining nanotechnology and PDT is effective in targeting metastatic cancer cells. Nanotechnology can also increase the effectiveness of PDT by targeting cells at a molecular level. Dendrimer-based nanoconjugates (DBNs) are highly stable and biocompatible, making them suitable for drug delivery applications. Moreover, the hyperbranched structures in DBNs have the capacity to load hydrophobic compounds, such as photosensitizers (PSs) and chemotherapy drugs, and deliver them efficiently to tumour cells. This review primarily focuses on DBNs and their potential applications in cancer treatment. We discuss the chemical design, mechanism of action, and targeting efficiency of DBNs in tumour metastasis, intracellular trafficking in cancer treatment, and DBNs' biocompatibility, biodegradability and clearance properties. Overall, this study will provide the most recent insights into the application of DBNs and PDT in cancer therapy.


DBNs' intracellular journey in cancer-PDT refines targeted therapy, boosting efficacy.DBN in PDT for tumour metastasis: targeting and drug release mechanisms.DBNs' biocompatibility, biodegradability and clearance were explored thoroughly.


Subject(s)
Dendrimers , Nanoconjugates , Neoplasms , Photochemotherapy , Humans , Dendrimers/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Nanoconjugates/chemistry , Nanoconjugates/therapeutic use , Animals , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Biological Transport , Intracellular Space/metabolism , Intracellular Space/drug effects , Drug Carriers/chemistry
19.
Int J Mol Sci ; 25(15)2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39126073

ABSTRACT

The spread of multidrug-resistant mycobacterium strains requires the development of new approaches to combat diseases caused by these pathogens. For that, photodynamic inactivation (PDI) is a promising approach. In this study, a tricarbocyanine (TCC) is used for the first time as a near-infrared (740 nm) activatable PDI photosensitizer to kill mycobacteria with deep light penetration. For better targeting, a novel tricarbocyanine dye functionalized with two trehalose units (TCC2Tre) is developed. The photodynamic effect of the conjugates against mycobacteria, including Mycobacterium tuberculosis, is evaluated. Under irradiation, TCC2Tre causes more effective killing of mycobacteria compared to the photosensitizer without trehalose conjugation, with 99.99% dead vegetative cells of M. tuberculosis and M. smegmatis. In addition, effective photoinactivation of dormant forms of M. smegmatis is observed after incubation with TCC2Tre. Mycobacteria treated with TCC2Tre are more sensitive to 740 nm light than the Gram-positive Micrococcus luteus and the Gram-negative Escherichia coli. For the first time, this study demonstrates the proof of principle of in vitro PDI of mycobacteria including the fast-growing M. smegmatis and the slow-growing M. tuberculosis using near-infrared activatable photosensitizers conjugated with trehalose. These findings are useful for the development of new efficient alternatives to antibiotic therapy.


Subject(s)
Infrared Rays , Mycobacterium smegmatis , Mycobacterium tuberculosis , Photosensitizing Agents , Trehalose , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/radiation effects , Trehalose/pharmacology , Trehalose/chemistry , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/radiation effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Carbocyanines/chemistry , Carbocyanines/pharmacology , Photochemotherapy/methods
20.
Nat Commun ; 15(1): 6935, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138197

ABSTRACT

Nanobodies (Nbs), the smallest antigen-binding fragments with high stability and affinity derived from the variable domain of naturally occurring heavy-chain-only antibodies in camelids, have been shown as an efficient way to improve the specificity to tumors for photodynamic therapy (PDT). Nonetheless, the rapid clearance of Nbs in vivo restricts the accumulation and retention of the photosensitizer at the tumor site causing insufficient therapeutic outcome, especially in large-volume tumors. Herein, we develop photodynamic conjugates, MNB-Pyra Nbs, through site-specific conjugation between 7D12 Nbs and type I photosensitizer MNB-Pyra (morpholine-modified nile blue structure connected to pyrazolinone) in a 1:2 ratio. The photosensitizers with long-term retention can be released at the tumor site by reactive oxygen species cleavage after illumination, accompanied with fluorescence recovery for self-reporting the occurrence of PDT. Ultimately, a single dose of MNB-Pyra Nbs demonstrate highly effective tumor suppression with high biosafety in the large-volume tumor models after three rounds of PDT. This nanobody conjugate provides a paradigm for the design of precise long-time retention photosensitizers and is expected to promote the development of PDT.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Animals , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry , Mice , Humans , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Mice, Inbred BALB C , Female , Neoplasms/drug therapy , Neoplasms/immunology , Mice, Nude , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL