Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Radiat Res ; 174(5): 574-81, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20954859

ABSTRACT

For repair of damaged DNA, cells increase de novo synthesis of deoxyribonucleotide triphosphates through the rate-limiting, p53-regulated ribonucleotide reductase (RNR) enzyme. In this study we investigated whether pharmacological inhibition of RNR by 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, NSC #663249) enhanced chemoradiation sensitivity through a mechanism involving sustained DNA damage. RNR inactivation by 3-AP and resulting chemoradiosensitization were evaluated in human cervical (CaSki, C33-a) cancer cells through study of DNA damage (γ-H2AX signal) by flow cytometry, RNR subunit p53R2 and p21 protein steady-state levels by Western blot analysis and laser scanning imaging cytometry, and cell survival by colony formation assays. 3-AP treatment led to sustained radiation- and cisplatin-induced DNA damage (i.e. increased γ-H2AX signal) in both cell lines through a mechanism of inhibited RNR activity. Radiation, cisplatin and 3-AP exposure resulted in significantly elevated numbers and persistence of γ-H2AX foci that were associated with reduced clonogenic survival. DNA damage was associated with a rise in p53R2 but not p21 protein levels 6 h after treatment with radiation and/or cisplatin plus 3-AP. We conclude that blockage of RNR activity by 3-AP impairs DNA damage responses that rely on deoxyribonucleotide production and thereby may substantially increase chemoradiosensitivity of human cervical cancers.


Subject(s)
Enzyme Inhibitors/pharmacology , Radiation Tolerance/drug effects , Ribonucleotide Reductases/antagonists & inhibitors , Uterine Cervical Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Female , Humans , Pyridines/pharmacology , Radiation Tolerance/radiation effects , Ribonucleotide Reductases/metabolism , Thiosemicarbazones/pharmacology , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
2.
Photochem Photobiol ; 86(5): 1161-73, 2010.
Article in English | MEDLINE | ID: mdl-20553412

ABSTRACT

Photodynamic therapy (PDT) with lysosome-targeted photosensitizers induces the intrinsic pathway of apoptosis via the cleavage and activation of the BH3-only protein Bid by proteolytic enzymes released from photodisrupted lysosomes. To investigate the role of Bid in apoptosis induction and the role of damaged lysosomes on cell killing by lysosome-targeted PDT, we compared the responses of wild type and Bid-knock-out murine embryonic fibroblasts toward a mitochondrion/endoplasmic reticulum-binding photosensitizer, Pc 4, and a lysosome-targeted sensitizer, Pc 181. Whereas apoptosis and overall cell killing were induced equally well by Pc 4-PDT in both cell lines, Bid(-/-) cells were relatively resistant to induction of apoptosis and to overall killing following PDT with Pc 181, particularly at low PDT doses. Thus, Bid is critical for the induction of apoptosis caused by PDT with the lysosome-specific sensitizers, but dispensable for PDT targeted to other membranes.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/physiology , Drug Delivery Systems , Muramidase/radiation effects , Photochemotherapy , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/genetics , Blotting, Western , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Knockout Techniques , Mitochondria/drug effects , Molecular Structure , Muramidase/drug effects , Photosensitizing Agents/radiation effects
3.
Autophagy ; 6(2): 248-55, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20083906

ABSTRACT

Photodynamic therapy (PDT) uses a photosensitizer, light and oxygen to produce extensive oxidative damage to organelles housing the photosensitizer. Although PDT is an efficient trigger of apoptosis, it also induces autophagy in many kinds of cells. Autophagy can serve as both a cell survival and a cell death mechanism. Our previous study indicates that autophagy contributes to cell death after PDT, especially in apoptosis-deficient cells. Here, we provide further evidence to support the role of autophagy in cell killing after PDT. Autophagy was blocked by knockdown of one essential factor, LC3 or Atg7, in MCF-7 cells. The cells were exposed to a range of doses of PDT sensitized by the phthalocyanine Pc 4; steps in autophagy were monitored by western blotting for LC3-II and by fluorescence microscopy for the uptake of monodansylcadaverine or for the distribution of transfected GFP-LC3; and overall cell death was monitored by MTT assay and by clonogenic assay. We find that blocking autophagy increased the survival of MCF-7 cells after PDT and increased the shoulder on the dose-response curve. In response to Pc 4-PDT, Atg7-deficient MCF-7 cells remained capable of robust accumulation of LC3-II, but were defective in comparison to Atg7(+) cells in the formation of autophagosomes. We conclude that apoptosis-deficient cells rely on autophagy for cell death after Pc 4-PDT and that the strong activation of LC3 maturation in response to PDT could occur even in cells with limited or no Atg7 expression.


Subject(s)
Breast Neoplasms , Cell Line, Tumor/radiation effects , Photochemotherapy , Ubiquitin-Activating Enzymes/deficiency , Apoptosis/physiology , Autophagy/physiology , Autophagy-Related Protein 7 , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Female , Humans , Indoles/therapeutic use , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Photosensitizing Agents/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
4.
J Nucl Med ; 51(1): 130-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008981

ABSTRACT

UNLABELLED: Photodynamic therapy (PDT) is a relatively new therapy that has shown promise for treating various cancers in both preclinical and clinical studies. The present study evaluated the potential use of PET with radiolabeled choline to monitor early tumor response to PDT in animal models. METHODS: Two human prostate cancer models (PC-3 and CWR22) were studied in athymic nude mice. A second-generation photosensitizer, phthalocyanine 4 (Pc 4), was delivered to each animal by a tail vein injection 48 h before laser illumination. Small-animal PET images with (11)C-choline were acquired before PDT and at 1, 24, and 48 h after PDT. Time-activity curves of (11)C-choline uptake were analyzed before and after PDT. The percentage of the injected dose per gram of tissue was quantified for both treated and control tumors at each time point. In addition, Pc 4-PDT was performed in cell cultures. Cell viability and (11)C-choline uptake in PDT-treated and control cells were measured. RESULTS: For treated tumors, normalized (11)C-choline uptake decreased significantly 24 and 48 h after PDT, compared with the same tumors before PDT (P < 0.001). For the control tumors, normalized (11)C-choline uptake increased significantly. For mice with CWR22 tumors, the prostate-specific antigen level decreased 24 and 48 h after PDT. Pc 4-PDT in cell culture showed that the treated tumor cells, compared with the control cells, had less than 50% (11)C-choline activity at 5, 30, and 45 min after PDT, whereas the cell viability test showed that the treated cells were viable longer than 7 h after PDT. CONCLUSION: PET with (11)C-choline is sensitive for detecting early changes associated with Pc 4-PDT in mouse models of human prostate cancer. Choline PET has the potential to determine whether a PDT-treated tumor responds to treatment within 48 h after therapy.


Subject(s)
Choline , Indoles/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/therapeutic use , Radiopharmaceuticals , Algorithms , Animals , Cell Line, Tumor , Cell Survival/drug effects , Choline/chemical synthesis , Choline/pharmacokinetics , Humans , Image Processing, Computer-Assisted , Isoindoles , Isotope Labeling , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/pathology , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics
5.
Radiat Res ; 172(6): 666-76, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19929413

ABSTRACT

Therapeutic ionizing radiation damages DNA, increasing p53-regulated ribonucleotide reductase (RNR) activity required for de novo synthesis of the deoxyribonucleotide triphosphates used during DNA repair. This study investigated the pharmacological inhibition of RNR in cells of virally or mutationally silenced p53 cancer cell lines using 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, Triapine(R), NSC #663249), a chemotherapeutic radiosensitizer that equally inhibits RNR M2 and p53R2 small subunits. The effects of 3-AP on RNR inhibition and resulting radiosensitization were evaluated in cervical (CaSki, HeLa and C33-a) and colon (RKO, RKO-E6) cancer cells. 3-AP treatment significantly enhanced radiation-related cytotoxicity in cervical and colon cancer cells. 3-AP treatment significantly decreased RNR activity, caused prolonged radiation-induced DNA damage, and resulted in an extended G(1)/S-phase cell cycle arrest in all cell lines. Similar effects were observed in both RKO and RKO-E6 cells, suggesting a p53-independent mechanism of radiosensitization. We conclude that inhibition of ribonucleotide reductase by 3-AP enhances radiation-mediated cytotoxicity independent of p53 regulation by impairing repair processes that rely on deoxyribonucleotide production, thereby substantially increasing the radiation sensitivity of human cancers.


Subject(s)
Gene Silencing , Neoplasms/enzymology , Pyridines/pharmacology , Radiation Tolerance , Ribonucleotide Reductases/antagonists & inhibitors , Thiosemicarbazones/pharmacology , Tumor Suppressor Protein p53/genetics , Base Sequence , Blotting, Western , Cell Line , DNA Damage , DNA Primers , DNA Repair , Flow Cytometry , Humans , Neoplasms/genetics , Neoplasms/pathology
6.
Photochem Photobiol ; 85(5): 1189-200, 2009.
Article in English | MEDLINE | ID: mdl-19508642

ABSTRACT

The phthalocyanine photosensitizer Pc 4 has been shown to bind preferentially to mitochondrial and endoplasmic reticulum membranes. Upon photoirradiation of Pc 4-loaded cells, membrane components, especially Bcl-2, are photodamaged and apoptosis, as indicated by activation of caspase-3 and cleavage of poly(ADP-ribose) polymerase, is triggered. A series of analogs of Pc 4 were synthesized, and the results demonstrate that Pcs with the aminopropylsiloxy ligand of Pc 4 or a similar one on one side of the Pc ring and a second large axial ligand on the other side of the ring have unexpected properties, including enhanced cell uptake, greater monomerization resulting in greater intracellular fluorescence and three-fold higher affinity constants for liposomes. The hydroxyl-bearing axial ligands tend to reduce aggregation of the Pc and direct it to lysosomes, resulting in four to six times more killing of cells, as defined by loss of clonogenicity, than with Pc 4. Whereas Pc 4-PDT photodamages Bcl-2 and Bcl-xL, Pc 181-PDT causes much less photodamage to Bcl-2 over the same dose-response range relative to cell killing, with earlier cleavage of Bid and slower caspase-3-dependent apoptosis. Therefore, within this series of photosensitizers, these hydroxyl-bearing axial ligands are less aggregated than is Pc 4, tend to localize to lysosomes and are more effective in overall cell killing than is Pc 4, but induce apoptosis more slowly and by a modified pathway.


Subject(s)
Indoles/pharmacology , Lysosomes/drug effects , Mitochondria/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Silicon/chemistry , Isoindoles , Magnetic Resonance Spectroscopy
7.
Mitochondrion ; 8(3): 237-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18514589

ABSTRACT

10-N-Nonyl acridine orange (NAO) is used as a mitochondrial probe because of its high affinity for cardiolipin (CL). Targeting of NAO may also depend on mitochondrial membrane potential. As the nonyl group has been considered essential for targeting, a systematic study of alkyl chain length was undertaken; three analogues (10-methyl-, 10-hexyl-, and 10-hexadecyl-acridine orange) were synthesized and their properties studied in phospholipid monolayers and breast cancer cells. The shortest and longest alkyl chains reduced targeting, whereas the hexyl group was superior to the nonyl group, allowing very clear and specific targeting to mitochondria at concentrations of 20-100 nM, where no evidence of toxicity was apparent. Additional studies in wild-type and cardiolipin-deficient yeast cells suggested that cellular binding was not absolutely dependent upon cardiolipin.


Subject(s)
Acridine Orange/analogs & derivatives , Acridine Orange/metabolism , Cells/metabolism , Fluorescent Dyes/metabolism , Mitochondria/metabolism , Acridine Orange/chemical synthesis , Acridine Orange/chemistry , Acridine Orange/pharmacology , Breast Neoplasms/pathology , Cardiolipins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Humans , Molecular Structure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Temperature , Time Factors
8.
Autophagy ; 4(1): 125-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18025862

ABSTRACT

Photodynamic therapy (PDT) induces apoptosis in many cell types. Recent reports identified autophagy as an alternative cell-death process following PDT. Here we investigated the occurrence of autophagy after PDT with the photosensitizer Pc 4 in human cancer cells that are deficient in the pro-apoptotic factor Bax (human prostate cancer DU145) or the apoptosis mediator caspase-3 (human breast cancer MCF-7v) and in apoptosis-competent cells (MCF-7c3 stably overexpressing human pro-caspase-3 and Chinese hamster ovary CHO 5A100). Further, each cell line was also studied with and without stably overexpressed Bcl-2. By electron microscopy and immunoblot analysis, autophagy was observed in all cells studied, whether or not they were capable of typical apoptosis or overexpressed Bcl-2. Bcl-2 overexpression protected against PDT-induced apoptosis and loss of clonogenicity in apoptosis-competent cells (MCF-7c3 and CHO); however, it did not protect against the development of autophagy or against loss of clonogenicity in apoptosis-deficient cells (MCF-7v and DU145). The results show that autophagy may be the dominant cell death pathway following PDT in cells that are incapable of undergoing normal apoptosis. In such cells, Bcl-2 does not protect against autophagic death.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Photochemotherapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Phagosomes/metabolism , Photosensitizing Agents/metabolism
9.
Photochem Photobiol ; 83(5): 1016-23, 2007.
Article in English | MEDLINE | ID: mdl-17880494

ABSTRACT

Photodynamic therapy (PDT) is an efficient inducer of apoptosis in many types of cells, except in cells deficient in one or more of the factors that mediate apoptosis. Recent reports have identified autophagy as a potential alternative cell death process following PDT. Here we investigated the occurrence of autophagy after PDT with the photosensitizer Pc 4 in human cancer cells that are deficient in the pro-apoptotic factor Bax (human prostate cancer DU145 cells) or the apoptosis mediator caspase-3 (human breast cancer MCF-7v cells) and in apoptosis-competent cells (MCF-7c3 cells that stably overexpress human pro-caspase-3 and Chinese hamster ovary CHO 5A100 cells). Further, each of the cell lines was also studied with and without stably overexpressed Bcl-2. Autophagy was identified by electron microscopic observation of the presence of double-membrane-delineated autophagosomal vesicles in the cytosol and by immunoblot observation of the Pc 4-PDT dose- and time-dependent increase in the level of LC3-II, a component of the autophagosomal membrane. Autophagy was observed in all of the cell lines studied, whether or not they were capable of typical apoptosis and whether or not they overexpressed Bcl-2. The presence of stably overexpressed Bcl-2 in the cells protected against PDT-induced apoptosis and loss of clonogenicity in apoptosis-competent cells (MCF-7c3 and CHO 5A100 cells). In contrast, Bcl-2 overexpression did not protect against the development of autophagy in any of the cell lines or against loss of clonogenicity in apoptosis-deficient cells (MCF-7v and DU145 cells). Furthermore, 3-methyladenine and wortmannin, inhibitors of autophagy, provided greater protection against loss of viability to apoptosis-deficient than to apoptosis-competent cells. The results show that autophagy occurs during cell death following PDT in human cancer cells competent or not for normal apoptosis. Only the apoptosis-competent cells are protected by Bcl-2 against cell death.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Photochemotherapy , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Microscopy, Electron, Transmission , Photosensitizing Agents/pharmacology
10.
Oncogene ; 24(46): 6987-92, 2005 Oct 20.
Article in English | MEDLINE | ID: mdl-16007152

ABSTRACT

The antiapoptotic Bcl-2-family proteins, Bcl-2 and Bcl-xL, are recognized phototargets of photodynamic therapy (PDT) with the mitochondrion-targeting phthalocyanine photosensitizer Pc 4. In the present study, we found that myeloid cell leukemia 1 (Mcl-1), another antiapoptotic member of the Bcl-2 family, was not photodamaged in Pc 4-PDT-treated human carcinoma cells MCF-7c3, MDA-MB468, DU145, and A431, although Mcl-1 turnover was observed after exposure of HeLa or MCF-7c3 cells to a supralethal dose of UVC. In contrast, when human lymphoma U937 and Jurkat cells were treated with Pc 4-PDT, staurosporine (STS) or UVC, Mcl-1 was cleaved to generate a 28-kDa fragment over a 2-4 h period. The cleavage of Mcl-1 was accompanied by the activation of caspases-3, -9, and -8. The broad-specificity caspase inhibitor z-VAD-fmk completely blocked Mcl-1 cleavage induced by PDT, STS or UVC, providing evidence for Mcl-1 as a substrate for caspases. Western blot analysis localized Mcl-1 to mitochondria, ER, and cytosol of both MCF-7c3 and U937 cells, suggesting that Mcl-1 protein, unlike Bcl-2 and Bcl-xL, is not a target for Pc 4-PDT, probably due to its localization to sites removed from those of Pc 4 binding. The 28-kDa cleaved fragment of Mcl-1, which has proapoptotic activity, was produced in PDT-treated lymphoid-derived cells, but not in cells of epithelial origin, suggesting that PDT-induced rapid and extensive apoptosis in lymphoma cells may result in part from the sensitivity of their Mcl-1 to caspase cleavage, removing an important negative control on apoptosis.


Subject(s)
Epithelial Cells/drug effects , Indoles/pharmacology , Lymphoid Tissue/drug effects , Neoplasm Proteins/metabolism , Photosensitizing Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Caspases/metabolism , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/radiation effects , Humans , Lymphoid Tissue/cytology , Lymphoid Tissue/radiation effects , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Staurosporine/pharmacology , Substrate Specificity , Ultraviolet Rays
11.
Oncogene ; 22(58): 9197-204, 2003 Dec 18.
Article in English | MEDLINE | ID: mdl-14681679

ABSTRACT

The antiapoptotic oncoprotein Bcl-2 is now a recognized phototarget of photodynamic therapy (PDT) with the phthalocyanine Pc 4 and with other mitochondrion-targeting photosensitizers. Photodamage, observed on Western blots as the loss of the native 26-kDa Bcl-2 protein, is PDT dose dependent and occurs in multiple cell lines, in the cold, and immediately upon photoirradiation. In our initial study, no photochemical damage was observed to Bcl-xL, in spite of its similarity in size, sequence, location and function to Bcl-2. The original study used a commercial anti-Bcl-xS/L antibody. We have revisited this issue by examining Western blots developed using one of three epitope-specific anti-Bcl-xL antibodies from commercial sources, a polyclonal antibody generated to the entire protein, as well as the antibody used previously. All five Bcl-xL antibodies recognized bacterially expressed Bcl-xL, but not Bcl-2, whereas an anti-Bcl-2 antibody recognized Bcl-2 and not Bcl-xL. All five Bcl-xL antibodies recognized at least one protein migrating at approximately 30 kDa; two of the antibodies recognized an additional band, migrating at approximately 33 or approximately 24 kDa. We now observe Pc 4-PDT-induced photodamage to all Bcl-xL-related proteins, except the 33-kDa species, in several human cancer cell lines. The results indicate that, in addition to the expected quantitative differences that may reflect exposure of individual epitopes, the antibodies also detect proteins of different apparent molecular weights that may be distinct isoforms or post-translationally modified forms of Bcl-xL. No evidence for PDT-induced phosphorylation or degradation was observed. Bcl-xL localized to mitochondria was considerably more sensitive to photodamage than was Bcl-xL in the cytosol, indicating that as previously found for Bcl-2, Bcl-xL must be membrane localized to be photosensitive.


Subject(s)
Indoles/pharmacology , Light , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/chemistry , Radiation-Sensitizing Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cytosol/metabolism , Dose-Response Relationship, Drug , Epitopes , Humans , Mitochondria/metabolism , Precipitin Tests , Protein Isoforms , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/chemistry , bcl-2-Associated X Protein , bcl-X Protein
12.
Photochem Photobiol ; 78(1): 1-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12929741

ABSTRACT

We have reported that photodynamic therapy (PDT) using the photosensitizer phthalocyanine (Pc) 4 and red light damages the antiapoptotic protein Bcl-2. Recently, using transient transfection of Bcl-2 deletion mutants, we identified the membrane anchorage domains of Bcl-2 as necessary to form the photosensitive target. However, it is not clear how Bcl-2 photodamage sensitizes cells to Pc 4-PDT-induced apoptosis, whether overall cell killing is also sensitized or how up-regulation of Bcl-2 in tumors might make them more or less responsive to Pc 4-PDT. In this study we report on MCF-7c3 cells (human breast cancer cells expressing stably transfected procaspase-3) overexpressing wild-type Bcl-2 or certain deletion mutants in either a transient or a stable mode. By flow cytometric analysis of transiently transfected cells, we found that wild-type Bcl-2, Bcl-2delta33-54 and Bcl-2delta37-63 (each of which can be photodamaged) protected cells from apoptosis caused by Pc 4-PDT. In contrast, Bcl-2delta210-239, which lacks the C-terminal transmembrane domain and cannot be photodamaged, afforded no protection. We then evaluated the PDT sensitivity of transfected cell lines stably overexpressing high levels of wild-type Bcl-2 or one of the Bcl-2 mutants. Overexpression of wild-type Bcl-2, Bcl-2delta33-54 or Bcl-2delta37-63 resulted in relative resistance of cells to Pc 4-PDT, as assessed by morphological apoptosis or loss of clonogenicity. Furthermore, overexpression of Bcl-2 also inhibited the activation-associated conformational change of the proapoptotic protein Bax, and higher doses of Pc 4 and light were required to activate Bax in cells expressing high levels of Bcl-2. Many advanced cancer cells have elevated amounts of Bcl-2. Our results show that increasing the dose of Pc 4-PDT can overcome the resistance afforded by either Bcl-2 or the two mutants. PDT regimens that photodamage Bcl-2 lead to activation of Bax, induction of apoptosis and elimination of the otherwise resistant tumor cells.


Subject(s)
Apoptosis , Indoles/pharmacology , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/chemistry , Female , Green Fluorescent Proteins , Humans , Indoles/therapeutic use , Luminescent Proteins/metabolism , Membrane Proteins/analysis , Mitochondria/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Envelope/chemistry , Photosensitizing Agents/therapeutic use , Protein Conformation , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-bcl-2/genetics , Transfection , Tumor Stem Cell Assay , Up-Regulation , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
13.
Exp Cell Res ; 283(2): 135-45, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12581734

ABSTRACT

To test the role of caspase 3 in apoptosis and in overall cell lethality caused by the protein kinase inhibitor staurosporine, we compared the responses of MCF-7c3 cells that express a stably transfected CASP-3 gene to parental MCF-7:WS8 cells transfected with vector alone and lacking procaspase-3 (MCF-7v). Cells were exposed to increasing doses (0.15-1 microM) of staurosporine for periods up to 19 h. Apoptosis was efficiently induced in MCF-7c3 cells, as demonstrated by cytochrome c release, processing of procaspase-3, procaspase-8, and Bid, increase in caspase-3-like DEVDase activity, cleavage of the enzyme poly(ADP-ribose) polymerase, DNA fragmentation, changes in nuclear morphology, and TUNEL assay and flow cytometry. For all of these measures except cytochrome c release, little or no activity was detected in MCF-7v cells, confirming that caspase-3 is essential for efficient induction of apoptosis by staurosporine, but not for mitochondrial steps that occur earlier in the pathway. MCF-7c3 cells were more sensitive to staurosporine than MCF-7v cells when assayed for loss of viability by reduction of a tetrazolium dye. However, the two cell lines were equally sensitive to killing by staurosporine when evaluated by a clonogenic assay. A similar distinction between apoptosis and loss of clonogenicity was observed for the cancer chemotherapeutic agent VP-16. These results support our previous conclusions with photodynamic therapy: (a) assessing overall reproductive death of cancer cells requires a proliferation-based assay, such as clonogenicity; and (b) the critical staurosporine-induced lethal event is independent of those mediated by caspase-3.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Caspases/physiology , Enzyme Inhibitors/pharmacology , Staurosporine/pharmacology , Caspase 3 , Caspases/genetics , Cell Death/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Etoposide/pharmacology , Female , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Tumor Cells, Cultured
14.
J Biol Chem ; 278(3): 2021-9, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12379660

ABSTRACT

Photodynamic therapy using the photosensitizer Pc 4 and red light photochemically destroys the antiapoptotic protein Bcl-2 and induces apoptosis. To characterize the requirements for photodamage, we transiently transfected epitope-tagged Bcl-2 deletion mutants into DU-145 cells. Using confocal microscopy and Western blots, wild-type Bcl-2 and mutants with deletions near the N terminus were found in mitochondria, endoplasmic reticulum, and nuclear membranes and were photodamaged. A mutant missing the C terminus, including the transmembrane domain, spread diffusely in cells and was not photodamaged. Bcl-2 missing alpha-helices 5/6 was also not photodamaged. Bcl-2 missing only one of those alpha-helices, with or without substitutions of the singlet oxygen-targeted amino acids, behaved like wild-type Bcl-2 with respect to localization and photodamage. Using green fluorescent protein (GFP)-tagged Bcl-2 or mutants in live cells, no change in either the localization or the intensity of GFP fluorescence was observed in response to Pc 4 photodynamic therapy. Western blot analysis of either GFP- or Xpress-tagged Bcl-2 revealed that the photodynamic therapy-induced disappearance of the Bcl-2 band was accompanied by the appearance of bands indicative of heavily cross-linked Bcl-2 protein. Therefore, the alpha(5)/alpha(6) region of Bcl-2 is required for photodamage and cross-linking, and domain-dependent photodamage to Bcl-2 offers a unique mechanism for activation of apoptosis.


Subject(s)
Photosensitizing Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/radiation effects , Base Sequence , Cell Membrane/metabolism , DNA Primers , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Luminescent Proteins/radiation effects , Male , Microscopy, Confocal , Mitochondria/metabolism , Photochemotherapy , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Singlet Oxygen/metabolism , Tumor Cells, Cultured
15.
Photochem Photobiol ; 76(2): 217-23, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12194220

ABSTRACT

Photodynamic therapy (PDT) using the second-generation photosensitizer phthalocyanine (Pc) 4 causes mitochondrial damage and induces apoptosis through the release of cytochrome c to the cytosol. Another protein of the mitochondrial intermembrane space, Smac/DIABLO (second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI), is also released to the cytosol in response to apoptotic stimuli and promotes caspase activation by binding IAP. To investigate the possible role of Smac/DIABLO in apoptosis induced by Pc 4-PDT, we transfected Smac/DIABLO (tagged at its C-terminus with green fluorescent protein [GFP]) into MCF-7c3 cells (human breast cancer MCF-7 cells stably transfected with procaspase-3) and DU-145 cells (human prostate cancer cells that express no Bax because of a frameshift insertion mutation). Confocal microscopy showed that recombinant Smac/DIABLO, like cytochrome c, localized to mitochondria and colocalized with MitoTracker Red. Three hours after exposure of MCF-7c3 cells to PDT (200 nM Pc 4 and 150 mJ/cm2 red light), Smac/DIABLO-GFP, as well as cytochrome c, was found largely in the cytosol. In contrast, for DU-145 cells, both Smac/DIABLO-GFP and cytochrome c remained in the mitochondria after PDT. By staining with Hoechst 33,342, typical apoptotic nuclei were observed in MCF-7c3 cells, but not in DU-145 cells, after Pc 4-PDT. These results suggest that the release of Smac/DIABLO from mitochondria may be regulated by a Bax-mediated mechanism and that Smac/DIABLO may cooperate with the cytochrome c-dependent apoptosis pathway. In addition, in MCF-7c3 cells transfected by Smac/DIABLO-GFP, apoptosis induced by Pc 4-PDT was greater than in cells transfected with the GFP vector alone or in untransfected cells, as determined by flow cytometry. Thus, Smac/DIABLO promotes apoptosis after Pc 4-PDT in a Bax-dependent manner and may facilitate the passage of PDT-treated cells through the late steps of apoptosis.


Subject(s)
Apoptosis/drug effects , Photochemotherapy , Proto-Oncogene Proteins c-bcl-2 , Apoptosis/physiology , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Cytochrome c Group/metabolism , Female , Green Fluorescent Proteins , Humans , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/genetics , Male , Mitochondrial Proteins/metabolism , Photobiology , Proto-Oncogene Proteins/metabolism , Transfection , Tumor Cells, Cultured , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...