Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
Toxicology ; 504: 153793, 2024 May.
Article in English | MEDLINE | ID: mdl-38574843

ABSTRACT

Photodynamic therapy (PDT) utilizes the potential of photosensitizing substances to absorb light energy and produce reactive oxygen species. Tetra-cationic porphyrins, which have organic or coordination compounds attached to their periphery, are heterocyclic derivatives with well-described antimicrobial and antitumoral properties. This is due to their ability to produce reactive oxygen species and their photobiological properties in solution. Consequently, these molecules are promising candidates as new and more effective photosensitizers with biomedical, environmental, and other biomedical applications. Prior to human exposure, it is essential to establish the toxicological profile of these molecules using in vivo models. In this study, we used Caenorhabditis elegans, a small free-living nematode, as a model for assessing toxic effects and predicting toxicity in preclinical research. We evaluated the toxic effects of porphyrins (neutral and tetra-cationic) on nematodes under dark/light conditions. Our findings demonstrate that tetra-methylated porphyrins (3TMeP and 4TMeP) at a concentration of 3.3 µg/mL (1.36 and 0.93 µM) exhibit high toxicity (as evidenced by reduced survival, development, and locomotion) under dark conditions. Moreover, photoactivated tetra-methylated porphyrins induce higher ROS levels compared to neutral (3TPyP and 4TPyP), tetra-palladated (3PdTPyP and 4PdTPyP), and tetra-platinated (3PtTPyP and 4PtTPyP) porphyrins, which may be responsible for the observed toxic effects.


Subject(s)
Caenorhabditis elegans , Light , Photosensitizing Agents , Porphyrins , Animals , Caenorhabditis elegans/drug effects , Porphyrins/toxicity , Porphyrins/chemistry , Photosensitizing Agents/toxicity , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Reactive Oxygen Species/metabolism , Photochemotherapy/methods , Cations/toxicity , Dose-Response Relationship, Drug
2.
ACS Appl Mater Interfaces ; 13(38): 45201-45213, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34525803

ABSTRACT

Overproduction of reactive oxygen species (ROS) within tumors can cause oxidative stress on tumor cells to induce death, which has motivated us to develop ROS-mediated tumor therapies, such as typical photodynamic therapy (PDT) and Fenton reaction-mediated chemodynamic therapy (CDT). However, these therapeutic modalities suffer from compromised treatment efficacy owing to their limited generation of highly reactive ROS in a tumor microenvironment (TME). In this work, a nanoscale iron-based metal-organic framework, MIL-101(Fe), is synthesized as a Fenton nanocatalyst to perform the catalytic conversion of hydroxyl radicals (·OH) from hydrogen peroxide (H2O2) under the acidic environment and as a biocompatible and biodegradable nanocarrier to deliver a 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) photosensitizer for light-activated singlet oxygen (1O2) generation. By coupling such chemodynamic/photodynamic effects, the photosensitizer-integrated nanoagents (MIL-101(Fe)@TCPP) could enable more ROS production within tumors to induce amplified oxidative damage for tumor-specific synergistic therapy. In vitro results show that MIL-101(Fe)@TCPP nanoagents achieve the acid-responsive CDT and effective PDT, and synergistic CDT/PDT provides an enhanced therapeutic effect. Ultimately, based on such synergistic therapy, MIL-101(Fe)@TCPP nanoagents cause a significant tumor growth inhibition in vivo without severe side effects, showing great potential for anti-tumor application.


Subject(s)
Antineoplastic Agents/therapeutic use , Metal-Organic Frameworks/therapeutic use , Nanostructures/therapeutic use , Neoplasms/drug therapy , Oxidative Stress/drug effects , Photosensitizing Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Catalysis , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrogen Peroxide/chemistry , Hydroxyl Radical/metabolism , Iron/chemistry , Light , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/toxicity , Nanostructures/chemistry , Nanostructures/toxicity , Photosensitizing Agents/pharmacology , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Porphyrins/pharmacology , Porphyrins/radiation effects , Porphyrins/therapeutic use , Porphyrins/toxicity , Singlet Oxygen/metabolism
3.
ACS Appl Mater Interfaces ; 13(36): 42396-42410, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34472332

ABSTRACT

Chronic wound healing, impeded by bacterial infections and drug resistance, poses a threat to global human health. Antibacterial phototherapy is an effective way to fight microbial infection without causing drug resistance. Covalent organic frameworks (COFs) are a class of highly crystalline functional porous carbon-based materials composed of light atoms (e.g., carbon, nitrogen, oxygen, and borane), showing potential applications in the biomedical field. Herein, we constructed porphyrin-based COF nanosheets (TP-Por CON) for synergizing photodynamic and photothermal therapy under red light irradiation (e.g., 635 nm). Moreover, a nitric oxide (NO) donor molecule, BNN6, was encapsulated into the pore volume of the crystalline porous framework structure to moderately release NO triggered by red light irradiation for realizing gaseous therapy. Therefore, we successfully synthesized a novel TP-Por CON@BNN6-integrated heterojunction for thoroughly killing Gram-negative bacteria Escherichia coli and Gram-positive bacteria Staphylococcus aureus in vitro. Our research identified that TP-Por CON@BNN6 has favorable biocompatibility and biodegradability, low phototoxicity, anti-inflammatory properties, and excellent mice wound healing ability in vivo. This study indicates that the TP-Por CON@BNN6-integrated heterojunction with multifunctional properties provides a potential strategy for COF-based gaseous therapy and microorganism-infected chronic wound healing.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Metal-Organic Frameworks/therapeutic use , Nitric Oxide Donors/therapeutic use , Photosensitizing Agents/therapeutic use , Staphylococcal Skin Infections/drug therapy , Wound Healing/drug effects , Animals , Anti-Inflammatory Agents/radiation effects , Anti-Inflammatory Agents/toxicity , Cell Line , Escherichia coli/drug effects , Light , Metal-Organic Frameworks/radiation effects , Metal-Organic Frameworks/toxicity , Mice, Inbred BALB C , Nitric Oxide Donors/radiation effects , Nitric Oxide Donors/toxicity , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Porphyrins/radiation effects , Porphyrins/therapeutic use , Porphyrins/toxicity , Staphylococcus aureus/drug effects
4.
ACS Appl Mater Interfaces ; 13(24): 27934-27944, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34101408

ABSTRACT

Due to conventional photodynamic therapy encountering serious problems of phototoxicity and low tissue-penetrating depth of light, other dynamic therapy-based therapeutic methods such as sonodynamic therapy (SDT) are expected to be developed. To improve the therapeutic response to SDT, more effective sonosensitizers are imperative. In this study, a novel water-soluble iridium(III)-porphyrin sonosensitizer (IrTMPPS) was synthesized and used for SDT. IrTMPPS generated ample singlet oxygen (1O2) under US irradiation and especially showed distinguished US-activatable abilities at more than 10 cm deep-tissue depths. Interestingly, under US irradiation, IrTMPPS sonocatalytically oxidized intracellular NADH, which would enhance SDT efficiency by breaking the redox balance in the tumor. Moreover, IrTMPPS displayed great sonocytotoxicity toward various cancer cells, and in vivo experiments demonstrated efficient tumor inhibition and anti-metastasis to the lungs in the presence of IrTMPPS and US irradiation. This report gives a novel idea of metal-based sonosensitizers for sonotherapy by fully taking advantage of non-invasiveness, water solubility, and deep tumor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Porphyrins/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Iridium/chemistry , Iridium/therapeutic use , Iridium/toxicity , Mice , NAD/chemistry , NAD/metabolism , Neoplasms/pathology , Oxidation-Reduction , Porphyrins/chemical synthesis , Porphyrins/toxicity , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/toxicity , Singlet Oxygen/metabolism , Ultrasonic Waves , Zebrafish
5.
ACS Appl Mater Interfaces ; 12(23): 26391-26398, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32395978

ABSTRACT

Herein, a dual-modal fluorescent/colorimetric "Signal-On" nanoprobe based on PCN-222 nanorods (NRs) toward phosphate was proposed for the first time. Due to the high affinity of the zirconium node in PCN-222 NRs for phosphate, the structure collapse of PCN-222 NRs was triggered by phosphate, resulting in the release of the tetrakis(4-carboxyphenyl)porphyrin (TCPP) ligand from PCN-222 NRs as well as the enhancement of fluorescence and absorbance signals. The PCN-222 NR-based nanoprobe could be employed for phosphate detection over a wide concentration range with a detection limit down to 23 nM. The practical application of the PCN-222 NR-based nanoprobe in real samples was evaluated. Moreover, benefitting from the good biocompatibility and water dispersibility of PCN-222 NRs, this nanoprobe was successfully employed in the intracellular imaging of phosphate, revealing its promising application in the biological science. The present work would greatly extend the potential of nanostructured MOFs in the sensing and biological fields.


Subject(s)
Fluorescent Dyes/chemistry , Metal-Organic Frameworks/chemistry , Nanotubes/chemistry , Phosphates/analysis , Porphyrins/chemistry , Colorimetry/methods , Fluorescent Dyes/toxicity , HeLa Cells , Humans , Limit of Detection , Metal-Organic Frameworks/toxicity , Microscopy, Confocal , Microscopy, Fluorescence , Nanotubes/toxicity , Phosphates/chemistry , Porphyrins/toxicity , Zirconium/chemistry , Zirconium/toxicity
6.
Molecules ; 25(8)2020 Apr 19.
Article in English | MEDLINE | ID: mdl-32325815

ABSTRACT

In this study, a family of porphyrins based on 5,10,15,20-Tetrakis(4-ethylphenyl)porphyrin (1, Ph) and six metallo-derivatives (Zn2+(2, Ph-Zn), Sn4+(3, Ph-Sn), Mn2+ (4, Ph-Mn), Ni2+ (5, Ph-Ni), Al3+ (6, Ph-Al), and V3+ (7, Ph-V)) were tested as photosensitizers for photodynamic therapy against Leishmania braziliensis and panamensis. The singlet oxygen quantum yield value (ΦΔ) for (1-7) was measured using 1,3-diphenylisobenzofuran (DPBF) as a singlet oxygen trapping agent and 5,10,15,20-(tetraphenyl)-porphyrin (H2TPP) as a reference standard; besides, parasite viability was estimated by the MTT assay. After metal insertion into the porphyrin core, the ΦΔ increased from 0.76-0.90 and cell viability changed considerably. The ΦΔ and metal type changed the cytotoxic activity. Finally, (2) showed both the highest ΦΔ (0.90) and the best photodynamic activity against the parasites studied (IC50 of 1.2 µM).


Subject(s)
Leishmania braziliensis/drug effects , Leishmania/drug effects , Metalloporphyrins/chemistry , Metalloporphyrins/toxicity , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Singlet Oxygen/chemistry , Benzofurans/chemistry , Cell Survival/drug effects , Cell Survival/radiation effects , Inhibitory Concentration 50 , Leishmania/radiation effects , Leishmania braziliensis/radiation effects , Light , Metalloporphyrins/chemical synthesis , Photochemotherapy , Porphyrins/chemical synthesis , Porphyrins/toxicity
7.
J Biol Inorg Chem ; 24(8): 1269-1278, 2019 12.
Article in English | MEDLINE | ID: mdl-31486955

ABSTRACT

The discovery of aggregation inhibitors and the elucidation of their mechanism of action are key in the quest to mitigate the toxic consequences of amyloid formation. We have previously characterized the antiamyloidogenic mechanism of action of sodium phtalocyanine tetrasulfonate ([Na4(H2PcTS)]) on α-Synuclein (αS), demonstrating that specific aromatic interactions are fundamental for the inhibition of amyloid assembly. Here we studied the influence that metal preferential affinity and peripheral substituents may have on the activity of tetrapyrrolic compounds on αS aggregation. For the first time, our laboratory has extended the studies in the field of the bioinorganic chemistry and biophysics to cellular biology, using a well-established cell-based model to study αS aggregation. The interaction scenario described in our work revealed that both N- and C-terminal regions of αS represent binding interfaces for the studied compounds, a behavior that is mainly driven by the presence of negatively or positively charged substituents located at the periphery of the macrocycle. Binding modes of the tetrapyrrole ligands to αS are determined by the planarity and hydrophobicity of the aromatic ring system in the tetrapyrrolic molecule and/or the preferential affinity of the metal ion conjugated at the center of the macrocyclic ring. The different capability of phthalocyanines and meso-tetra (N-methyl-4-pyridyl) porphine tetrachloride ([H2PrTPCl4]) to modulate αS aggregation in vitro was reproduced in cell-based models of αS aggregation, demonstrating unequivocally that the modulation exerted by these compounds on amyloid assembly is a direct consequence of their interaction with the target protein.


Subject(s)
Amyloidogenic Proteins/metabolism , Indoles/metabolism , Porphyrins/metabolism , Protein Multimerization/drug effects , Zinc/metabolism , alpha-Synuclein/metabolism , Amino Acid Sequence , Amyloidogenic Proteins/chemistry , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/chemistry , Indoles/toxicity , Porphyrins/chemistry , Porphyrins/toxicity , Protein Binding , Zinc/chemistry , alpha-Synuclein/chemistry
8.
J Mater Chem B ; 7(31): 4843-4853, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31389968

ABSTRACT

Hyaluronic acid (HA)-based nanocarriers are of great interest in the drug delivery field due to the tumor targetability via CD44-mediated recognition and endocytosis. However, sufficient tumor-specific release of encapsulated cargoes with steady controllability is necessary to optimize their outcome for cancer therapy. In this study, we constructed a light-activated nanocarrier TKHCENPDOX to enable on-demand drug release at the desired site (tumor). Particularly, TKHCENPDOX encapsulating doxorubicin (DOX) was self-assembled from a HA-photosensitizer conjugate (HA-TK-Ce6) containing reactive oxygen species (ROS)-sensitive thioketal (TK) linkers. Following i.v. injection, TKHCENPDOX was accumulated in the MDA-MB-231 breast tumor xenograft more efficiently through preventing drug leakage in the bloodstream and the HA-mediated targeting effect. Upon internalization into tumoral cells, 660 nm laser irradiation generated ROS during a photodynamic (PDT) process to cleave the TK linker next to Ce6, resulting in light-induced TKHCENPDOX dissociation and selective DOX release in the tumor area. Consequently, TKHCENPDOX showed a remarkable therapeutic effect and minimized toxicity in vivo. This strategy might provide new insight for designing cancer-selective nanoplatforms with active targeting and locoregional drug release simultaneously.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Nanoconjugates/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chlorophyllides , Doxorubicin/pharmacokinetics , Drug Carriers/radiation effects , Drug Carriers/toxicity , Drug Liberation/radiation effects , Female , Humans , Hyaluronic Acid/radiation effects , Hyaluronic Acid/toxicity , Light , Mice, Inbred BALB C , Mice, Inbred ICR , Nanoconjugates/radiation effects , Nanoconjugates/toxicity , Nanoparticles/chemistry , Nanoparticles/radiation effects , Nanoparticles/toxicity , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Porphyrins/pharmacology , Porphyrins/radiation effects , Porphyrins/toxicity , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
9.
Adv Mater ; 31(33): e1901965, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31237375

ABSTRACT

Antibacterial photocatalytic therapy has been reported as a promising alternative water disinfection technology for combating antibiotic-resistant bacteria. Numerous inorganic nanosystems have been developed as antibiotic replacements for bacterial infection treatment, but these are limited due to the toxicity risk of heavy metal species. Organic semiconductor photocatalytic materials have attracted great attention due to their good biocompatibility, chemically tunable electronic structure, diverse structural flexibility, suitable band gap, low cost, and the abundance of the resources they require. An all-organic composite photocatalytic nanomaterial C3 N4 /perylene-3,4,9,10-tetracarboxylic diimide (PDINH) heterostructure is created through recrystallization of PDINH on the surface of C3 N4 in situ, resulting in enhanced photocatalytic efficiency due to the formation of a basal heterostructure. The absorption spectrum of this composite structure can be extended from ultraviolet to near-infrared light (750 nm), enhancing the photocatalytic effect to produce more reactive oxygen species, which have an excellent inactivation effect on both Gram-negative and positive bacteria, while demonstrating negligible toxicity to normal tissue cells. An efficient promotion of infectious wound regeneration in mice with Staphylococcus aureus infected dermal wounds is demonstrated. This all-organic heterostructure shows great promise for use in wound disinfection.


Subject(s)
Anti-Bacterial Agents/chemistry , Imides/chemistry , Nitriles/chemistry , Perylene/analogs & derivatives , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Semiconductors , Animals , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/toxicity , Catalysis , Cell Survival , Escherichia coli/drug effects , Female , Light , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Nanostructures/chemistry , Nitriles/therapeutic use , Nitriles/toxicity , Perylene/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/toxicity , Porphyrins/therapeutic use , Porphyrins/toxicity , Reactive Oxygen Species/metabolism , Staphylococcus aureus/drug effects , Wound Healing , Wound Infection/drug therapy , Wound Infection/microbiology
10.
Carbohydr Polym ; 218: 289-298, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31221332

ABSTRACT

Bacterial infections are a growing global challenge for public health as antibiotic resistance could cause the failure of anti-infective treatment eventually. So, it is urgent to develop new potential antibacterial materials. Herein, a multifunctional chitosan (CS) functionalized magnetic Chlorin e6 (CS-MP-Ce6) was constructed to combat methicillin-resistant Staphylococcus aureus (MRSA) infection by integrating bacterial conjugation and enrichment, and near-infrared (NIR)-triggered photodynamic sterilization. CS-MP-Ce6 could efficiently capture bacteria due to positively charged property of CS, and Ce6 acted as an effective photodynamic killer to convert NIR light into local energy to enhance antibacterial activity. Specifically, after being trapped together with MRSA, CS-MP-Ce6 showed an excellent in vitro photodynamic sterilization ability. In vivo MRSA-induced abscess treatment studies showed faster healing when CS-MP-Ce6 was used as subcutaneous nano-localized energy sources with the assistance of external magnet to concentrate CS-MP-Ce6-bacteria conjugate. This work provides a promising framework for constructing a new system for efficiently combating MRSA.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Indoles/chemistry , Magnetite Nanoparticles/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Photosensitizing Agents/therapeutic use , Polymers/chemistry , Porphyrins/therapeutic use , Animals , Anti-Bacterial Agents/radiation effects , Anti-Bacterial Agents/toxicity , Chitosan/chemistry , Chlorophyllides , Infrared Rays , Mice , Photochemotherapy/methods , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Porphyrins/radiation effects , Porphyrins/toxicity , RAW 264.7 Cells
12.
Analyst ; 144(9): 3056-3063, 2019 May 07.
Article in English | MEDLINE | ID: mdl-30916075

ABSTRACT

Photodynamic therapy (PDT) involves a photosensitizing agent activated with light to induce cell death. Two-photon excited PDT (TPE-PDT) offers numerous benefits compared to traditional one-photon induced PDT, including an increased penetration depth and precision. However, the in vitro profiling and comparison of two-photon photosensitizers (PS) are still troublesome. Herein, we report the development of an in vitro screening platform of TPE-PS using a 3D osteosarcoma cell culture. The platform was tested using three different two-photon (2P) active compounds - a 2P sensitizer P2CK, a fluorescent dye Eosin Y, and a porphyrin derivative (TPP). Their 2P absorption cross-sections (σ2PA) were characterised using a fully automated z-scan setup. TPP exhibited a remarkably high σ2PA at 720 nm (8865 GM) and P2CK presented a high absorption at 850 nm (405 GM), while Eosin Y had the lowest 2P absorption at the studied wavelengths (<100 GM). The cellular uptake of PS visualized using confocal laser scanning microscopy showed that both TPP and P2CK were internalized by the cells, while Eosin Y stayed mainly in the surrounding media. The efficiency of the former two TPE-PS was quantified using the PrestoBlue metabolic assay, showing a significant reduction in cell viability after two-photon irradiation. The possibility of damage localization was demonstrated using a co-culture of adipose derived stem cells together with osteosarcoma spheroids showing no signs of damage to the surrounding healthy cells after TPE-PDT.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylidene Compounds/pharmacology , Eosine Yellowish-(YS)/pharmacology , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Antineoplastic Agents/radiation effects , Antineoplastic Agents/toxicity , Benzylidene Compounds/radiation effects , Benzylidene Compounds/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Eosine Yellowish-(YS)/radiation effects , Eosine Yellowish-(YS)/toxicity , Humans , Mesenchymal Stem Cells , Osteosarcoma/drug therapy , Photons , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Porphyrins/radiation effects , Porphyrins/toxicity
13.
J Photochem Photobiol B ; 194: 21-31, 2019 May.
Article in English | MEDLINE | ID: mdl-30897400

ABSTRACT

Aedes aegypti mosquitos are widespread vectors of several diseases and their control is of primary importance for biological and environmental reasons, and novel safe insecticides are highly desirable. An eco-friendly photosensitizing magnetic nanocarrier with larvicidal effects on Aedes aegypti was proposed. The innovative core-shell hybrid nanomaterial was synthesized by combining peculiar magnetic nanoparticles (called Surface Active Maghemite Nanoparticles - SAMNs, the core) and chlorin-e6 as photosensitizer (constituting the shell) via self-assembly in water. The hybrid nanomaterial (SAMN@chlorin) was extensively characterized and tested for the photocidal activity on larvae of Aedes aegypti. The SAMN@chlorin core-shell nanohybrid did not present any toxic effect in the dark, but, upon light exposure, showed a higher photocidal activity than free chlorin-e6. Moreover, the eco-toxicity of SAMN@chlorin was determined in adults and neonates of Daphnia magna, where delayed toxicity was observed only after prolonged (≥4 h) exposure to intense light, on the green alga Pseudokirchneriella subcapitata and on the duckweed Lemna minor on which no adverse effects were observed. The high colloidal stability, the physico-chemical robustness and the magnetic drivability of the core-shell SAMN@chlorin nanohybrid, accompanied by the high photocidal activity on Aedes aegypti larvae and reduced environmental concerns, can be proposed as a safe alternative to conventional insecticides.


Subject(s)
Aedes , Ferric Compounds/chemistry , Insecticides/chemistry , Larva , Nanoparticles/chemistry , Porphyrins/chemistry , Animals , Chlorophyceae/drug effects , Chlorophyceae/radiation effects , Daphnia/drug effects , Daphnia/radiation effects , Insecticides/toxicity , Light , Porphyrins/toxicity , Surface Properties , Water/chemistry
14.
Drug Deliv Transl Res ; 9(3): 615-624, 2019 06.
Article in English | MEDLINE | ID: mdl-30690675

ABSTRACT

Nanocarriers for drug delivery have made great progress in the treatment of cancer, but the dense extracellular collagen of tumors has greatly limited the efficiency of drug delivery. In this study, losartan is used to deplete tumor collagen and improve the delivery efficiency and photodynamic therapeutic efficacy of chlorine 6 (Ce6)-loaded periodic mesoporous organosilica nanoplatform (Ce6-PMO) for breast cancer. After pretreatment with losartan in vivo, the tumor collagen I fraction is significantly reduced by 53% compared to that of mice pretreated with saline. Importantly, the accumulation of the Ce6-PMO nanoplatforms in the tumor is remarkably enhanced via peritumoral and intravenous injection, respectively, after the mice are pretreated with losartan. Further, combination of losartan with the Ce6-PMO nanoplatforms shows the best therapeutic efficacy, and the suppression rate of tumor volume is measured up to 82%. Taken together, this study provides a very promising synergetic strategy to improve the tumor photodynamic treatment efficacy of nanoplatforms.


Subject(s)
Collagen Type I/metabolism , Losartan/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Photochemotherapy , Porphyrins/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Silicon Dioxide/administration & dosage , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyllides , Female , Losartan/toxicity , Male , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Inbred ICR , Porphyrins/toxicity , Radiation-Sensitizing Agents/toxicity , Silicon Dioxide/toxicity , Tumor Burden/drug effects
15.
Theranostics ; 8(16): 4332-4344, 2018.
Article in English | MEDLINE | ID: mdl-30214624

ABSTRACT

Photodynamic therapy (PDT) has been applied in clinical cancer treatment. Here we report an aptamer-functionalized nanoscale metal-organic framework for targeted PDT. Our nanosystem can be easily prepared and successfully used for targeted PDT with a significantly enhanced therapeutic efficacy in vitro and in vivo. Methods: By combining the strong binding ability between phosphate-terminated aptamers and Zr-based nanoscale metal-organic frameworks (Zr-NMOFs) and the intercalation of photosensitizer TMPyP4 within the G-quadruplex DNA structure, TMPyP4-G4-aptamer-NMOFs were prepared. The characteristics and photodynamic performance of TMPyP4-G4-aptamer-NMOFs were examined after preparation. Then, we studied their stability, specific recognition ability, and phototoxicity in vitro. For in vivo experiments, the nanosystem was intratumorally injected into a HeLa subcutaneous xenograft tumor mouse model. After irradiation on day 0, mice were further injected with the nanosystem on day 5 and were again subjected to laser irradiation for 30 min. Tumor volumes and body weights of all mice were measured by caliper every 2 days after the treatment. Results: The nanosystem induced 90% cell death of targeted cells. In contrast, the control cells maintained about 40% cell viability at the same concentration of nanosystem. For the in vivo experiments, the nanosystem-treated group maintained more than 76% inhibition within the entire experimental period. Conclusion: We have demonstrated that our smart TMPyP4-G4-sgc8-NMOFs nanosystem can be used for targeted cancer therapy with high efficiency.


Subject(s)
Aptamers, Nucleotide/administration & dosage , G-Quadruplexes , Metal-Organic Frameworks/administration & dosage , Molecular Targeted Therapy/methods , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Animals , Aptamers, Nucleotide/toxicity , Cell Survival/drug effects , Disease Models, Animal , Drug Carriers/administration & dosage , Drug Stability , HeLa Cells , Heterografts , Humans , Metal-Organic Frameworks/toxicity , Mice , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Photosensitizing Agents/toxicity , Porphyrins/toxicity , Treatment Outcome
16.
J Hazard Mater ; 360: 445-451, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30142595

ABSTRACT

A range of hydrophobic first-, second-, and third-generation manganese porphyrins (MnPs) was investigated as cytochrome P450 models for degradation of the antibiotic ciprofloxacin (CIP). The experiments were carried out under mild conditions; oxidants such as iodosylbenzene (PhIO), H2O2, and meta-chloroperbenzoic acid were employed. The PhIO system yielded the best results: CIP degradation ranged between 56% and 76%. CIP degradation was not directly related to MnP generation. The second-generation MnP afforded the best result with the advantage that it required less preparation effort as compared to the third-generation MnP. Some new degradation products in MnP-mediated ciprofloxacin degradation were proposed, and the products of the reaction are not cytotoxic under the conditions evaluated.


Subject(s)
Anti-Bacterial Agents/chemistry , Ciprofloxacin/chemistry , Manganese/chemistry , Porphyrins/chemistry , Anti-Bacterial Agents/toxicity , Catalysis , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Ciprofloxacin/toxicity , Humans , Manganese/toxicity , Oxidation-Reduction , Porphyrins/toxicity
17.
J Photochem Photobiol B ; 183: 266-274, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29747146

ABSTRACT

Ovarian Cancer is one of the deadliest gynecological cancer showing high resistance to chemotherapy. Non-overlapping and synergistic combination therapies are the best option to overcome this multi-pathological silent disease. Cationic peptides (CPs) with high targeting feature and ability to pass through cell membrane induce apoptosis via disruption of cancer cell membrane. Photodynamic Therapy (PDT) is a noninvasive clinically approved treatment modality combining light activated photosensitizer, light and oxygen. In this study we present, combination therapy composed of 9-mer +4 charge bearing CP and Benzoporphyrin derivative monoacid, (BPD-MA, Verteporfin) mediated PDT. In order to evaluate the effect of sequence on the outcome of the therapy, CP and BPD-MA mediated PDT was applied in two different sequence: 'CP first' 'BPD-MA first'. Treatment efficacy of combination therapy in SKOV-3 ovarian cancer cell line has been evaluated based on cell inhibition, cell death pathway, Combination index (CI), and Dose Reduction Index (DRI) values. When SKOV-3 ovarian cancer cell line treated with BPD-MA mediated PDT (5 J/cm2) and CP individually, IC30 values for each drug were determined as 1.1 µM and 240 µM respectively and apoptosis was the major death cell pathway for both of the drugs. In the case of combination therapy, SKOV-3 cell line treated with drugs in constant ratio yet on different sequence. Drugs were used in constant ratio so that one of them would not de-emphasize the effect of other in any concentration point. Our theoretical and experimental results were in agreement and showed that the treatment outcome significantly depends on the order of the treatment. For instance, while BPD-MA mediated PDT was applied prior to CP, cell inhibition at IC30 value of BPD-MA was roughly 28% with CI =3.3 suggesting antagonistic interaction between each therapy. When the sequence of treatment was changed to CP first, cell inhibition at IC30 concentration of CP was determined as 98% with CI = 0.3 creating substantial synergism between the drugs. Moreover, synergistic interactions were observed at all concentration points at CP first scenario. DRI value for CP first treatment option was much higher compared to BPD-MA first treatment making the former treatment sequence more attractive option for clinically relevant combination therapies. Based on our results, we strongly believe that 9-mer CP and BPD-MA-PDT based combination therapy, offering synergistic therapeutic outcome, may increase chances of treatment of ovarian cancer in comparison to 9-mer CP and/or BPD-MA alone case.


Subject(s)
Apoptosis/drug effects , Photosensitizing Agents/toxicity , Porphyrins/toxicity , Amino Acid Sequence , Apoptosis/radiation effects , Cations/chemistry , Cell Line, Tumor , Drug Synergism , Female , Humans , Light , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peptides/chemistry , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Verteporfin
18.
Environ Toxicol Chem ; 37(2): 336-344, 2018 02.
Article in English | MEDLINE | ID: mdl-28940243

ABSTRACT

Provisional molecular weights and chemical formulas were assigned to 4 significant previously unidentified contaminants present during active fish kills in the Red River region of Oklahoma. The provisional identifications of these contaminants were determined using high-resolution liquid chromatography-time-of-flight mass spectrometry (LC-TOFMS), LC-Fourier transform ion cyclotron resonance mass spectrometry (LC-FTICRMS), and LC-ion trap mass spectrometry (LC-ITMS). Environmental water samples were extracted using a solid-phase extraction (SPE) method, and sediment samples were extracted using a modified sonication liquid extraction method. During screening of the samples, 2 major unknown chromatographic peaks were detected at m/z 624.3 and m/z 639.3. The peak at m/z 639.3 was firmly identified, through the use of an authentic standard, as a porphyrin, specifically chlorin-e6-trimethyl ester, with m/z 639.31735 (M + H)+ and molecular formula C37 H43 N4 O6 . The other major peak, at m/z 624.3 (M + H)+ , was identified as an amide-containing porphyrin. It was discovered that the amide compound was an artifact created during the SPE process by reaction of ammonium hydroxide at 1 of 3 potential reaction sites on chlorin-e6-trimethyl ester. Other unique nontargeted chemicals were also detected and the importance of their identification is discussed. Environ Toxicol Chem 2018;37:336-344. Published 2017 Wiley Periodicals Inc. on behalf of SETAC. This article is a US government work and, as such, is in the public domain in the United States of America.


Subject(s)
Fishes/physiology , Rivers/chemistry , Water Pollutants, Chemical/analysis , Animals , Chlorophyllides , Chromatography, Liquid , Geography , Geologic Sediments/chemistry , Oklahoma , Porphyrins/chemistry , Porphyrins/toxicity , Tandem Mass Spectrometry
19.
Mol Neurobiol ; 55(1): 96-102, 2018 01.
Article in English | MEDLINE | ID: mdl-28844112

ABSTRACT

Photodynamic therapy (PDT) leads to production of reactive oxygen species (ROS) and cell destruction due to oxidative stress. We used photodynamic effect of photosensitizer radachlorin to unravel the effect of photo-induced oxidative stress on the calcium signal and lipid peroxidation in primary culture of cortical neurons and astrocytes using live cell imaging. We have found that irradiation in presence of 200 nM of radachlorin induces calcium signal in primary neurons and astrocytes. Photo-induced neuronal calcium signal depends on internal calcium stores as it was still observed in calcium-free medium and could be blocked by depletion of endoplasmic reticulum (ER) stores with inhibitor of sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) thapsigargin. Both inhibitors of phospholipase C activity U73122 and water-soluble analogue of vitamin E Trolox suppressed calcium response activated by PDT. We have also observed that the photodynamic effect of radachlorin induces lipid peroxidation in neurons and astrocytes. This data demonstrate that lipid peroxidation induced by PDT in neurons and astrocytes leads to activation of phospholipase C that results in production of inositol 1,4,5-trisphosphate (IP3).


Subject(s)
Astrocytes/radiation effects , Calcium Signaling/radiation effects , Neurons/radiation effects , Photosensitizing Agents/toxicity , Reactive Oxygen Species/radiation effects , Animals , Astrocytes/metabolism , Calcium Signaling/physiology , Coculture Techniques , Drug Combinations , Lasers/adverse effects , Neurons/metabolism , Porphyrins/toxicity , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
20.
Mol Neurobiol ; 55(1): 90-95, 2018 01.
Article in English | MEDLINE | ID: mdl-28840566

ABSTRACT

Photodynamic therapy is selective destruction of cells stained with a photosensitizer upon irradiation with light at a specific wavelength in the presence of oxygen. Cell death upon photodynamic treatment is known to occur mainly due to free radical production and subsequent development of oxidative stress. During photodynamic therapy of brain tumors, healthy cells are also damaged; considering this, it is important to investigate the effect of the treatment on normal neurons and glia. We employed live-cell imaging technique to investigate the cellular mechanism of photodynamic action of radachlorin (200 nM) on neurons and astrocytes in primary rat cell culture. We found that the photodynamic effect of radachlorin increases production of reactive oxygen species measured by dihydroethidium and significantly decrease mitochondrial membrane potential. Mitochondrial depolarization was independent of opening of mitochondrial permeability transition pore and was insensitive to blocker of this pore cyclosporine A. However, irradiation of cells with radachlorin dramatically decreased NADH autofluorescence and also reduced mitochondrial NADH pool suggesting inhibition of mitochondrial respiration by limitation of substrate. This effect could be prevented by inhibition of poly (ADP-ribose) polymerase (PARP) with DPQ. Thus, irradiation of neurons and astrocytes in the presence of radachlorin leads to activation of PARP and decrease in NADH that leads to mitochondrial dysfunction.


Subject(s)
Astrocytes/radiation effects , Lasers, Semiconductor/adverse effects , Mitochondria/radiation effects , Neurons/radiation effects , Oxidative Stress/radiation effects , Photosensitizing Agents/toxicity , Animals , Astrocytes/metabolism , Coculture Techniques , Drug Combinations , Mitochondria/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Porphyrins/toxicity , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...