Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Photochem Photobiol B ; 234: 112505, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35839543

ABSTRACT

Melanosomes have been considered crucial targets in melanoma treatments. In this study we explored the role of melanosomes in photodynamic therapy (PDT), employing the synthetic Zn(II) phthalocyanine Pc13, a potent photosensitizer that promotes melanoma cell death after irradiation. Phototoxic action is mediated by reactive oxygen species increase. The internalization mechanism of Pc13 and its consequent subcellular localization were evaluated in melanotic B16-F0 cells. Pharmacological inhibitors of dynamin or caveolae, but not of clathrin, decreased Pc13 cellular uptake and phototoxicity. Similar results were obtained when cells over-expressed dominant negative mutants of dynamin-2 and caveolin-1, indicating that Pc13 is internalized by caveolae-mediated endocytosis. Confocal microscopy analysis revealed that Pc13 targets melanosomes and damage of these structures after irradiation was demonstrated by transmission electron microscopy. Treatment of pigmented B16-F0 and WM35 melanoma cells with the melanin synthesis inhibitor phenylthiourea for 48 h led to cell depigmentation and enhanced cell death after irradiation, whereas a 3-h period of inhibition did not modify melanin content but produced a marked reduction of Pc13 phototoxicity, together with a decrease of oxidative melanin synthesis intermediates. In contrast, the effect of Pc13 in amelanotic A375 cells was not altered by phenylthiourea treatment. These results provide evidence that melanosomes have a dual role in the efficacy of PDT. While melanin antagonizes the phototoxic action of Pc13, the release of cytotoxic synthetic intermediates to cytosol after irradiation and melanosome damage is conducive to the phototoxic response. Based on these findings, we demonstrate that melanosome-targeted PDT could be an effective approach for melanoma treatment.


Subject(s)
Dermatitis, Phototoxic , Melanoma , Caveolin 1/metabolism , Caveolin 1/pharmacology , Caveolin 1/therapeutic use , Endocytosis , Humans , Indoles/chemistry , Isoindoles , Melanins/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Melanosomes/metabolism , Melanosomes/ultrastructure , Phenylthiourea/metabolism , Phenylthiourea/pharmacology , Phenylthiourea/therapeutic use
2.
Lasers Surg Med ; 53(3): 344-358, 2021 03.
Article in English | MEDLINE | ID: mdl-32525252

ABSTRACT

BACKGROUND AND OBJECTIVES: Photodynamic therapy (PDT) is an antitumor procedure clinically approved for the treatment of different cancer types. Despite strong efforts and promising results in this field, PDT has not yet been approved by any regulatory authority for the treatment of colorectal cancer, one of the most prevalent gastrointestinal tumors. In the search of novel therapeutic strategies, we examined the in vivo effect of PDT with a lipophilic phthalocyanine (Pc9) encapsulated into polymeric poloxamine micelles (T1107) in a murine colon carcinoma model. STUDY DESIGN/MATERIALS AND METHODS: In vivo assays were performed with BALB/c mice challenged with CT26 cells. Pc9 tumor uptake was evaluated with an in vivo imaging system. Immunofluorescence, western blot, and flow cytometry assays were carried out to characterize the activation of apoptosis and an antitumor immune response. RESULTS: Pc9-T1107 effectively delayed tumor growth and prolonged mice survival, without generating systemic or tissue-specific toxicity. The induction of an apoptotic response was characterized by a decrease in the expression levels of Bcl-XL , Bcl-2, procaspase 3, full length Bid, a significant increment in the amount of active caspase-3 and the detection of PARP-1 cleavage. Infiltration of CD8+ CD107a+ T cells and higher levels of interferon-γ and tumor necrosis factor-α were also found in PDT-treated tumors. CONCLUSIONS: Pc9-T1107 PDT treatment reduced tumor growth, inducing an apoptotic cell death and activating an immune response. Lasers Surg. Med. © 2020 Wiley Periodicals LLC.


Subject(s)
Colonic Neoplasms , Photochemotherapy , Animals , Apoptosis , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Immunity , Isoindoles , Mice , Mice, Inbred BALB C , Organometallic Compounds , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Zinc/pharmacology , Zinc/therapeutic use , Zinc Compounds
3.
Heliyon ; 6(6): e04182, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32566788

ABSTRACT

Natural and synthetic phenazines are widely used in biomedical sciences. In dehydrogenase histochemistry, phenazine methosulfate (PMS) is applied as a redox reagent for coupling reduced coenzymes to the reduction of tetrazolium salts into colored formazans. PMS is also currently used for cytotoxicity and viability assays of cell cultures using sulfonated tetrazoliums. Under UV (340 nm) excitation, aqueous solutions of the cationic PMS show green fluorescence (λem: 526 nm), whereas the reduced hydrophobic derivative (methyl-phenazine, MPH) shows blue fluorescence (λem: 465 nm). Under UV (365 nm) excitation, cultured cells (LM2, IGROV-1, BGC-1, and 3T3-L1 adipocytes) treated with PMS (5 µg/mL, 30 min) showed cytoplasmic granules with bright blue fluorescence, which correspond to lipid droplets labeled by the lipophilic methyl-phenazine. After formaldehyde fixation blue-fluorescing droplets could be stained with oil red O. Interestingly, PMS-treated 3T3-L1 adipocytes observed under UV excitation 24 h after labeling showed large lipid droplets with a weak green emission within a diffuse pale blue-fluorescing cytoplasm, whereas a strong green emission was observed in small lipid droplets. This fluorescence change from blue to green indicates that reoxidation of methyl-phenazine to PMS can occur. Regarding cell uptake and labeling mechanisms, QSAR models predict that the hydrophilic PMS is not significantly membrane-permeant, so most PMS reduction is expected to be extracellular and associated with a plasma membrane NAD(P)H reductase. Once formed, the lipophilic and blue-fluorescing methyl-phenazine enters live cells and mainly accumulates in lipid droplets. Overall, the results reported here indicate that PMS is an excellent fluorescent probe to investigate labeling and redox dynamics of lipid droplets in cultured cells.

4.
Free Radic Biol Med ; 152: 743-754, 2020 05 20.
Article in English | MEDLINE | ID: mdl-31962157

ABSTRACT

Melanoma is the most aggressive type of skin cancer, highly resistant to conventional therapies. Photodynamic therapy (PDT) is a minimally invasive treatment modality that combines the use of a photosensitizer, visible light and molecular oxygen, leading to oxidative stress in the specific site of irradiation. The cationic zinc(II) phthalocyanine Pc13 has shown to be a potent photosensitizer in different melanoma cell lines. In this study, we explored the intracellular signaling pathways triggered by Pc13 PDT and the role of these cascades in the phototoxic action of Pc13 in human melanoma A375 cells. Activation of MAPKs p38, ERK, JNK and PI3K-I/AKT was observed after treatment and prevented by using the antioxidant trolox. Inhibition of p38 reduced Pc13 phototoxicity, whereas blockage of JNK potentiated cell death. Results obtained indicate that p38 is involved in the cleavage of PARP-1, an important mediator of apoptosis. On the other hand, Pc13 irradiation induced the activation of an autophagic program, as evidenced by enhanced levels of Beclin-1, LC3-II and GFP-LC3 punctate staining. We also demonstrated that this autophagic response is promoted by JNK and negatively regulated by PI3K-I/AKT pathway. The blockage of autophagy increased Pc13 phototoxicity and enhanced PARP-1 cleavage, revealing a protective role of this mechanism, which tends to prevent apoptotic cell death. Furthermore, reduced susceptibility to treatment and increased activation of autophagy were detected in A375 cells submitted to repeated cycles of Pc13 PDT, indicating that autophagy could represent a mechanism of resistance to PDT. The efficacy of Pc13 PDT and an improved phototoxic action in combination with chloroquine were also demonstrated in tumor spheroids. In conclusion, we showed the interplay between apoptotic and autophagic signaling pathways triggered by Pc13 PDT-induced oxidative stress. Thus, autophagy modulation represents a promising therapeutic strategy to potentiate the efficacy of PDT in melanoma.


Subject(s)
Melanoma , Photochemotherapy , Apoptosis , Autophagy , Cell Line, Tumor , Humans , Indoles , Isoindoles , Melanoma/drug therapy , Oxidative Stress , Reactive Oxygen Species , Zinc
5.
Int J Biochem Cell Biol ; 114: 105575, 2019 09.
Article in English | MEDLINE | ID: mdl-31362060

ABSTRACT

Photodynamic therapy (PDT) is a highly specific and clinically approved method for cancer treatment in which a nontoxic drug known as photosensitizer (PS) is administered to a patient. After selective tumor irradiation, an almost complete eradication of the tumor can be reached as a consequence of reactive oxygen species (ROS) generation, which not only damage tumor cells, but also lead to tumor-associated vasculature occlusion and the induction of an immune response. Despite exhaustive investigation and encouraging results, zinc(II) phthalocyanines (ZnPcs) have not been approved as PSs for clinical use yet. This review presents an overview on the physicochemical properties of ZnPcs and biological results obtained both in vitro and in more complex models, such as 3D cell cultures, chicken chorioallantoic membranes and tumor-bearing mice. Cell death pathways induced after PDT treatment with ZnPcs are discussed in each case. Finally, combined therapeutic strategies including ZnPcs and the currently available clinical trials are mentioned.


Subject(s)
Indoles , Neoplasms, Experimental/drug therapy , Organometallic Compounds , Photochemotherapy , Photosensitizing Agents , Animals , Cell Death/drug effects , Chickens , Humans , Indoles/chemistry , Indoles/therapeutic use , Isoindoles , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Zinc Compounds
6.
Apoptosis ; 24(1-2): 119-134, 2019 02.
Article in English | MEDLINE | ID: mdl-30603830

ABSTRACT

Melanoma is an aggressive form of skin carcinoma, highly resistant to traditional therapies. Photodynamic therapy (PDT) is a non-invasive therapeutic procedure that can exert a selective cytotoxic activity toward malignant cells. In this work we evaluated the effect of a cationic zinc(II) phthalocyanine (Pc13) as photosensitizer on a panel of melanoma cells. Incubation with Pc13 and irradiation induced a concentration and light dose-dependent phototoxicity. In order to study the mechanism underlying Pc13-related cell death and to compare the effect of different doses of PDT, the most sensitive melanoma B16F0 cells were employed. By confocal imaging we showed that Pc13 targeted lysosomes and mitochondria. After irradiation, a marked increase in intracellular reactive oxygen species was observed and a complete protection from Pc13 phototoxicity was reached in the presence of the antioxidant trolox. Acridine orange/ethidium bromide staining showed morphological changes indicative of both apoptosis and necrosis. Biochemical hallmarks of apoptosis, including a significant decrease in the expression levels of Bcl-2, Bcl-xL and Bid and mitochondrial membrane permeabilization, were observed at short times post irradiation. The consequent release of cytochrome c to cytosol and caspase-3 activation led to PARP-1 cleavage and DNA fragmentation. Simultaneously, a dose dependent increase of lactate dehydrogenase in the extracellular compartment of treated cells revealed plasma membrane damage characteristic of necrosis. Taken together, these results indicate that a dual apoptotic and necrotic response is triggered by Pc13 PDT-induced oxidative stress, suggesting that combined mechanisms of cell death could result in a potent alternative for melanoma treatment.


Subject(s)
Apoptosis , Indoles/pharmacology , Melanoma/pathology , Organometallic Compounds/pharmacology , Oxidative Stress , Photochemotherapy , Photosensitizing Agents/pharmacology , Skin Neoplasms/pathology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Humans , Indoles/radiation effects , Infrared Rays/therapeutic use , Isoindoles , Melanoma/metabolism , Melanoma/therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Necrosis , Organometallic Compounds/radiation effects , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Photosensitizing Agents/radiation effects , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/radiation effects , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , Zinc Compounds
7.
Int J Biochem Cell Biol ; 103: 89-98, 2018 10.
Article in English | MEDLINE | ID: mdl-30130653

ABSTRACT

We have previously reported that the phototoxic action of the lipophilic phthalocyanine Pc9 (2,9(10),16(17),23(24) tetrakis[(2-dimethylamino)ethylsulfanyl]phthalocyaninatozinc(II)) encapsulated into poloxamine micelles is related to the induction of an apoptotic response in murine colon CT26 carcinoma cells. In the present study, we explored the intracellular signals contributing to the resulting apoptotic death. We found that Pc9-T1107 arrests cell cycle progression immediately after irradiation promoting then an apoptotic response. Thus, 3 h after irradiation the percentage of hypodiploid cells increased from 5.9 ±â€¯0.6% to 23.1 ±â€¯0.1%; activation of caspases 8 and 9 was evident; the population of cells with loss of mitochondrial membrane potential increased from 1.1 ±â€¯0.4% to 44.0 ±â€¯9.3%; the full-length forms of Bid and PARP-1 were cleaved; and a 50% decrease of the expression levels of the anti-apoptotic proteins Bcl-2 and Bcl-XL was detected. We also found that the photosensitizer, mainly retained in lysosomes and endoplasmic reticulum (ER), promotes the permeabilization of lysosomal membranes and induces ER stress. Lysosomal membrane permeabilization was demonstrated by the reduction of acridine orange lysosome fluorescence, the release of Cathepsin D into the cytosol and ∼50% decrease of Hsp70, a chaperone recognized as a lysosomal stabilizer. Cathepsin D also contributed to Bid cleavage and caspase 8 activation. The oxidative damage to the ER induced an unfolded protein response characterized, 3 h after irradiation, by a 3-fold increase in cytosolic Ca2+ levels and 3-4 times higher expression of ER chaperones GRP78/BIP, calnexin, Hsp90 and Hsp110. The cell death signaling promoted by cytosolic Ca2+, calpains and lysosomal proteases was partially abolished by the Ca2+ chelator BAPTA-AM, the calpain inhibitor PD 150606 and proteases inhibitors. Furthermore, Bax down-regulation observed in Pc9-treated cells was undetectable in the presence of PD 150606, indicating that calpains contribute to Bax proteolytic damage. In summary, our results indicate that photoactivation of Pc9-T1107 led to lysosomal membrane permeabilization, induction of ER stress and activation of a caspase-dependent apoptotic cell death.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/metabolism , Endoplasmic Reticulum Stress/drug effects , Indoles/pharmacology , Lysosomes/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Endoplasmic Reticulum Chaperone BiP , Isoindoles , Lysosomes/pathology , Mice , Neoplasm Proteins/metabolism , Permeability/drug effects , Phototherapy
8.
J Photochem Photobiol B ; 170: 140-151, 2017 May.
Article in English | MEDLINE | ID: mdl-28432944

ABSTRACT

Photodynamic therapy is emerging as a hopeful method for the treatment of oncological diseases. In the search of novel therapeutic strategies for colorectal cancer, in this work we reported the photocytotoxic activity of a lipophilic zinc(II) phthalocyanine on a murine colon adenocarcinoma cell line (CT26 cells). The 2,9(10),16(17),23(24) tetrakis[(2-dimethylamino)ethylsulfanyl]phthalocyaninatozinc(II), named Pc9, was encapsulated into Tetronic® 1107 polymeric poloxamine micelles (T1107) and assayed in 2D and 3D cell cultures. We showed that the formulation Pc9-T1107 was efficient to reduce cell viability after photodynamic treatment both in 2D cultures (IC50 10±2nM) as well as in CT26 spheroids (IC50 370±11nM). Cellular uptake of Pc9-T1107 was a time- and concentration-dependent process, being the phthalocyanine formulation mainly incorporated into lysosomal vesicles and endoplasmic reticulum cisterns, but not in mitochondria. Pc9-T1107 also induced the formation of reactive oxygen species immediately after cell irradiation. We also found that the phototoxic action of Pc9-T1107 was partially reversed in the presence of antioxidants, such as TROLOX and N-acetyl-cysteine. In addition, we showed that Pc9-T1107 treatment triggered an apoptotic cell death, as suggested by the detection of pyknotic nuclei, the reduction in the expression levels of procaspase-3 and the increase in caspase-3 enzymatic activity.


Subject(s)
Apoptosis/drug effects , Drug Carriers/chemistry , Indoles/chemistry , Indoles/toxicity , Micelles , Organometallic Compounds/toxicity , Photosensitizing Agents/toxicity , Antioxidants/chemistry , Antioxidants/metabolism , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Endoplasmic Reticulum/metabolism , Humans , Isoindoles , Light , Lysosomes/metabolism , Organometallic Compounds/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Zinc Compounds
9.
Int J Biochem Cell Biol ; 45(11): 2553-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23994488

ABSTRACT

In order to find a novel photosensitizer to be used in photodynamic therapy for cancer treatment, we have previously showed that the cationic zinc(II) phthalocyanine named Pc13, the sulfur-linked dye 2,9(10),16(17),23(24)-tetrakis[(2-trimethylammonium) ethylsulfanyl]phthalocyaninatozinc(II) tetraiodide, exerts a selective phototoxic effect on human nasopharynx KB carcinoma cells and induces an apoptotic response characterized by an increase in the activity of caspase-3. Since the activation of an apoptotic pathway by chemotherapeutic agents contributes to the elimination of malignant cells, in this study we investigated the molecular mechanisms underlying the antitumor action of Pc13. We found that after light exposure, Pc13 induced the production of reactive oxygen species (ROS), which are mediating the resultant cytotoxic action on KB cells. ROS led to an early permeabilization of lysosomal membranes as demonstrated by the reduction of lysosome fluorescence with acridine orange and the release of lysosomal proteases to cytosol. Treatment with antioxidants inhibited ROS generation, preserved the integrity of lysosomal membrane and increased cell proliferation in a concentration-dependent manner. Lysosome disruption was followed by mitochondrial depolarization, cytosolic release of cytochrome C and caspases activation. Although no change in the total amount of Bax was observed, the translocation of Bax from cytosol to mitochondria, the cleavage of the pro-apoptotic protein Bid, together with the decrease of the anti-apoptotic proteins Bcl-XL and Bcl-2 indicated the involvement of Bcl-2 family proteins in the induction of the mitochondrial pathway. It was also demonstrated that cathepsin D, but not caspase-8, contributed to Bid cleavage. In conclusion, Pc13-induced cell photodamage is triggered by ROS generation and activation of the mitochondrial apoptotic pathway through the release of lysosomal proteases. In addition, our results also indicated that Pc13 induced a caspase-dependent apoptotic response, being activation of caspase-8, -9 and -3 the result of a post-mitochondrial event.


Subject(s)
Dermatitis, Phototoxic/metabolism , Dermatitis, Phototoxic/pathology , Indoles/toxicity , Lysosomes/metabolism , Mitochondria/metabolism , Organometallic Compounds/toxicity , Caspases/metabolism , Cathepsins/antagonists & inhibitors , Cathepsins/metabolism , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Humans , Indoles/chemistry , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/radiation effects , Isoindoles , Lysosomes/drug effects , Lysosomes/radiation effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/drug effects , Mitochondria/radiation effects , Models, Biological , Organometallic Compounds/chemistry , Permeability/drug effects , Permeability/radiation effects , Photochemotherapy , Protein Transport/drug effects , Protein Transport/radiation effects , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Zinc Compounds , bcl-2-Associated X Protein/metabolism
10.
Photochem Photobiol ; 89(2): 492-500, 2013.
Article in English | MEDLINE | ID: mdl-22924690

ABSTRACT

A major difficulty in photodynamic therapy is the poor solubility of the photosensitizer (PS) under physiological conditions which correlates with low bioavailability. PS aggregation leads to a decrease in the photodynamic efficiency and a more limited activity in vitro and in vivo. To improve the aqueous solubility and reduce the aggregation of 2,9(10),16(17),23(24)-tetrakis[(2-dimethylamino)ethylsulfanyl]phthal-ocyaninatozinc(II) (Pc9), the encapsulation into four poloxamine polymeric micelles (T304, T904, T1107 and T1307) displaying a broad spectrum of molecular weight and hydrophilic-lipophilic balance was investigated. The aqueous solubility of Pc9 was increased up to 30 times. Morphological evaluation showed the formation of Pc9-loaded spherical micelles in the nanosize range. UV/Vis and fluorescence studies indicated that Pc9 is less aggregated upon encapsulation in comparison with Pc9 in water-DMSO 2% and remained photostable. Pc9-loaded micelles generated singlet molecular oxygen in high yields. Photocytotoxicity assays using human nasopharynx KB carcinoma cells confirmed that the encapsulation of Pc9 in T1107 and T1307 increases its photocytotoxicity by 10 times in comparison with the free form in water-DMSO. In addition, Pc9 incorporated into cells was mainly localized in lysosomes.


Subject(s)
Cell Survival/drug effects , Cytotoxins/pharmacology , Ethylenediamines/chemistry , Indoles/pharmacology , Organometallic Compounds/pharmacology , Photosensitizing Agents/pharmacology , Polymers/chemistry , Biological Transport , Carcinoma , Cell Survival/radiation effects , Cytotoxins/chemistry , Drug Compounding , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/chemistry , Isoindoles , KB Cells , Light , Micelles , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Nasopharynx/drug effects , Nasopharynx/pathology , Organometallic Compounds/chemistry , Photochemotherapy , Photosensitizing Agents/chemistry , Singlet Oxygen , Solubility , Water , Zinc Compounds
11.
Eur J Med Chem ; 46(11): 5532-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21955680

ABSTRACT

The synthesis and photophysical parameters of two novel isosteric cationic zinc(II) phthalocyanines: 2,9(10),16(17),23(24)-tetrakis[(N-butyl-N-methylammoniumethylsulfanyl]phthalocyaninatozinc(II) tetraiodide (6) and 2,9(10),16(17),23(24)-tetrakis[(N-dibutyl-N-methylammonium)ethoxy]phthalocyaninatozinc(II) tetraiodide (7) were investigated. Maximum absorption values were 686.5 nm and 678 nm for 6 and 7, respectively, whereas singlet molecular oxygen generation was 0.42 and 0.67, respectively. The photodynamic effect and the cellular uptake of both phthalocyanines were evaluated on human nasopharynx KB carcinoma cells. After light exposure, phthalocyanine 6 showed a higher cytotoxic activity than 7. In addition, a higher intracellular uptake of 6 and a preferential localization within lysosomes were demonstrated. The production of a greater amount of reactive oxygen species after phthalocyanine 6 irradiation would be responsible for its potent phototoxic action on KB cells.


Subject(s)
Alkanes/chemistry , Chemistry Techniques, Synthetic , Indoles/chemical synthesis , Indoles/pharmacology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacology , Biological Transport , Humans , Indoles/chemistry , Indoles/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Isoindoles , KB Cells , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Reactive Oxygen Species/metabolism , Zinc Compounds
12.
Eur J Med Chem ; 45(9): 4129-39, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20599298

ABSTRACT

The photodynamic activity of water-soluble cationic zinc(II) phthalocyanines using human nasopharynx carcinoma (KB cells) was investigated. A sulfur-linked cationic dye, named: 2,9(10),16(17),23(24)-tetrakis[(2-trimethylammonium)ethylsulfanyl]phthalocyaninatozinc(II) tetraioidide (13) is the most active of four sensitizer assays and shows a singlet oxygen quantum yield of 0.58 and a higher bathochromic shift of 10 nm for the Q-band as compared with the oxygen-linked cationic aliphatic phthalocyanine: 2,9(10),16(17),23(24)-tetrakis[(2-trimethylammonium)ethoxy]phthalocyaninatozinc(II) tetraioidide (11) and the best photo-stability in water in comparison with their tetra-alpha-substituted counterparts 1,8(11),15(18),22(25)-tetrakis[(2-trimethylammonium)ethoxy]phthalocyaninatozinc(II) tetraioidide (12) and 1,8(11),15(18),22(25)-tetrakis[(2-trimethylammonium)ethylsulfanyl]phthalocyaninatozinc(II) tetraioidide (14). Phthalocyanine 13, partially localized in lysosomes, led to cell photoinactivation in a concentration- and light dose-dependent manner. After photodynamic treatment, compound 13 induced an apoptotic response--as indicated by morphological cell changes--an increase in the activity of caspase-3 and the cleavage of poly-ADP-ribose-polymerase substrate (PARP).


Subject(s)
Indoles/chemistry , Indoles/pharmacology , Nasopharyngeal Neoplasms/pathology , Water/chemistry , Apoptosis/drug effects , Apoptosis/radiation effects , Biological Transport , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Humans , Indoles/metabolism , Intracellular Space/metabolism , Isoindoles , KB Cells , Photochemical Processes , Photochemotherapy , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...