Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Theriogenology ; 203: 82-88, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36989544

ABSTRACT

Bacterial contamination in the semen deteriorates spermatozoa function. One mechanism through which this may occur is by inducing a premature form of the acrosome reaction (spontaneous acrosome reaction (sAR)) which has been shown to abrogate fertilization. To understand the mechanism by which bacteria affect sperm functions, we determined the effects of bacteria on sperm sAR and on other parameters involved in sperm capacitation. Sperm cells undergo biochemical changes in the female reproductive tract collectively called capacitation. Only capacitated sperm can undergo the physiological acrosomal exocytosis process near or on the oocyte, which allows the spermatozoon to penetrate and fertilize the egg. Bovine sperm incubated with the bacteria Escherichia coli (E. coli), Staphylococcus aureus (S. aureus) or Pseudomonas aeruginosa (P. aeruginosa), revealed a sperm-bacteria interaction, however only E. coli and P. aeruginosa caused an increase in sperm sAR. This effect was seen only when the bacteria were present with the sperm during the full incubation under capacitation conditions but not when the bacteria were added to capacitated sperm. These results indicate that bacteria affect sperm during capacitation and not at the AR step. In addition, Ca2+ influx, protein kinase A, and protein tyrosine phosphorylation activities, three essential processes that promote capacitation, were inhibited by the bacteria. Moreover, increasing intracellular cAMP, which also occur during sperm capacitation, caused significant reverse of sAR induced by the bacteria.


Subject(s)
Acrosome Reaction , Semen , Male , Cattle , Animals , Female , Escherichia coli , Staphylococcus aureus , Spermatozoa , Sperm Capacitation , Acrosome/physiology
2.
Lasers Surg Med ; 50(5): 535-551, 2018 07.
Article in English | MEDLINE | ID: mdl-29333608

ABSTRACT

BACKGROUND: Staphylococcus aureus is a major pathogen in clinical microbiology. It is known to cause infections at various body sites and can be life-threatening. The development of resistance to many well-established antibiotic treatments and the prevalence of methicillin-resistant S. aureus (MRAS) among hospital patients and the general community pose challenges in treating the pathogen. The antimicrobial effect of photodynamic therapy (PDT) has been a subject of study for a long time and can offer new strategies for dealing with resistant strains. OBJECTIVE: In our study, we searched for a positive synergistic relationship between PDT and the standard antibiotics used to treat S. aureus and MRSA infections. MATERIALS AND METHODS: The phototoxic profile of deuteroporphyrin (DP) in both resistant and susceptible clinical strains of S. aureus was determined by plating of treated and untreated broth cultures. Electron microscopy imaging was done to explore possible sites of damage and free-radical accumulation in the cells during DP-PDT. Minimal inhibitory concentration (MIC) of oxacillin, gentamicin, vancomycin, rifampin, and fusidic acid was determined using the broth dilution method, and the checkerboard method was used to detect and evaluate the synergistic potential of DP-PDT and antibiotic combinations. A synergistic combination was further characterized using broth cultures and plating. RESULTS: DP-PDT using a light dose of 15 J/cm2 showed a bactericidal effect even with a small concentration of 17 µM DP. Transmission electron microscopy indicated profound damage in the cell wall and cell membrane, and the appearance of mesosome-like structures. Free radicals tend to localize in the cell membrane and inside the mesosome. No synergistic effect was detected by combining PDT with gentamicin, vancomycin, rifampin, and fusidic acid treatments. A positive synergistic effect was observed only in DP-PDT-oxacillin combined treatment using the checkerboard method. The effect was observed in clinical antibiotic-resistant isolates after DP-PDT using a light dose of 46 J/cm2 and small concentrations of DP. Oxacillin MIC decreased below 2 µg/ml in resistant strains under such conditions. Cultures which did not undergo new cycles of DP-PDT recovered their original oxacillin resistance after a few generations. CONCLUSIONS: PDT with porphyrins shows possible new therapeutic options in treating drug-resistant S. aureus at body sites suitable for irradiation. The synergistic effect of DP-PDT with oxacillin on clinical strains illustrates the potential of PDT to augment traditional antibiotic treatment based on cell wall inhibitors. Lasers Surg. Med. 50:535-551, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Anti-Bacterial Agents/pharmacology , Deuteroporphyrins/pharmacology , Oxacillin/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Staphylococcus aureus/drug effects , Drug Synergism , Microbial Sensitivity Tests
3.
Eur J Pharm Sci ; 91: 91-7, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27283485

ABSTRACT

Anemia is a major cause of morbidity and mortality worldwide resulting from a wide variety of pathological conditions. In severe cases it is treated by blood transfusions or injection of erythroid stimulating agents, e.g., erythropoietin (Epo), which can be associated with serious adverse effects. Therefore, there is a need to develop new treatment modalities. We recently reported that treatment of erythroleukemic cells with the novel the bi-functional prodrugs of 5-aminolevulinic acid (ALA) and butyric acid (BA), AN233 and AN908, enhanced hemoglobin (Hb) synthesis to a substantially higher level than did ALA and BA individually or their mixture. Herein, we describe that these prodrugs when given orally to mice induced histone deacetylase inhibition in the kidneys, bone marrow and spleen, thus, indicating good penetrability to the tissues. In mice where anemia was chemically induced, treatment with the prodrugs increased the Hb, the number of red blood cells (RBCs) and the percentage of reticulocytes to normal levels. The prodrugs had no adverse effects even after repeated treatment at 100-200mg/kg for 50days. The lack of increased levels of Epo in the blood of mice that were treated with the prodrugs suggests that AN233 and AN908 affected the Hb and RBC levels in an Epo-independent manner. Taken together with our previous studies, we propose that the prodrugs increase globin expression by BA inhibition of histone deacetylase and elevation heme synthesis by ALA. These results support an Epo-independent approach for treating anemia with these prodrugs.


Subject(s)
Anemia/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Levulinic Acids/therapeutic use , Prodrugs/therapeutic use , Acetylation/drug effects , Aminolevulinic Acid/metabolism , Anemia/blood , Anemia/metabolism , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Butyric Acid/metabolism , Erythrocyte Count , Erythropoiesis/drug effects , Erythropoietin/blood , Erythropoietin/pharmacology , Hemoglobins/analysis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases , Histones/metabolism , Kidney/drug effects , Kidney/metabolism , Levulinic Acids/pharmacology , Male , Mice, Inbred BALB C , Prodrugs/pharmacology , Spleen/drug effects , Spleen/metabolism
4.
Photochem Photobiol Sci ; 13(1): 38-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24173598

ABSTRACT

Photodynamic therapy (PDT) of cancer involves inflicting lethal damage to the cells of malignant tumors, primarily by singlet oxygen that is generated following light-absorption in a photosensitizer molecule. Dysfunction of cells is manifested in many ways, including peroxidation of cellular components, membrane rupture, depolarization of electric potentials, termination of mitochondrial activity, onset of apoptosis and necrosis and eventually cell lysis. These events do not necessarily occur in linear fashion and different types of damage to cell components occur, most probably, in parallel. In this report we measured the relative rates of damage to two cellular membranes: the plasma membrane and the mitochondrial membrane. We employed photosensitizers of diverse hydrophobicities and used different incubation procedures, which lead to their different intra-cellular localizations. We monitored the damage that was inflicted on these membranes, by employing optical probes of membrane integrity, in a multi-color FACS experiment. The potentiometric indicator JC-1 monitored the electric cross-membrane potential of the mitochondria and the fluorometric indicator Draq7 monitored the rupture of the plasma membrane. We show that the electric depolarization of the mitochondrial membrane and the damage to the enveloping plasma membrane proceed with different kinetics that reflect the molecular character and intracellular location of the sensitizer: PpIX that is synthesized in the cells from ALA causes rapid mitochondrial damage and very slow damage to the plasma membrane, while externally added PpIX has an opposite effect. The hydrophilic sensitizer HypS4 can be taken up by the cells by different incubation conditions, and these affect its intracellular location, and as a consequence either the plasma membrane or the mitochondria is damaged first. A similar correlation was found for additional extracellularly-provided photosensitizers HP and PpIX.


Subject(s)
Aminolevulinic Acid/pharmacology , Cell Membrane/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membranes/drug effects , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacology , Anthracyclines , Benzimidazoles , Carbocyanines , Cell Line, Tumor , Cell Survival/drug effects , Flow Cytometry , Fluorescent Dyes , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Light , Neurons/drug effects , Neurons/pathology
5.
Invest New Drugs ; 30(3): 1028-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21509470

ABSTRACT

Herein we describe a series of multifunctional 5-aminolevulinic-acid (ALA) prodrugs for photodynamic dependent and independent cancer therapy (PDT). We studied the cell-death mechanisms in glioblastoma U251 cells treated with four ALA-prodrugs: (1) AlaAcBu, that releases ALA, acetaldehyde, and butyric acid; (2) AlaFaBu, that releases ALA, formaldehyde, and butyric acid; (3) AlaFaPi, that releases ALA, formaldehyde and pivalic acid (4) AlaAcPi that releases ALA, acetaldehyde and pivalic acid. We examined the light-activated and dark cell-death mechanisms of the active metabolites released from the prodrugs by unspecific cellular hydrolases. The active moieties accelerated biosynthesis of protoporphyrin IX (PpIX) due to upregulated porphobilinogen deaminase (PBGD) activity. AlaAcBu was found to be the superior prodrug for PDT due to its ability to induce the highest PpIX synthesis. Photo-irradiation of AlaAcBu-treated cells led to dissipation of the mitochondrial membrane potential and reduction in the mitochondria metabolic activities; apoptosis and necrosis. Electron microscopy analyses of these cells revealed mitochondrial and endoplasmic reticulum swelling, membrane blebbing, apoptotic bodies and necrotic cell rupture. The formaldehyde-releasing prodrugs AlaFaBu and AlaFaPi induced low PDT efficacy, moreover sequestering the formaldehyde with semicarbazide resulted in high PpIX synthesis, suggesting that formaldehyde inhibited its synthesis. ALA and AlaAcBu phototherapy resulted in a dramatic accumulation of ubiquitinated proteins due to reduced proteasome activity and expression. In conclusion, the PDT potency of the prodrugs was in the order: AlaAcBu, AlaAcPi > AlaFaBu ≥ ALA > AlaFaPi, and the superiority of AlaAcBu stems from lower molar concentrations of AlaAcBu and lower light intensity needed to activate cell death following PDT.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/pharmacology , Cell Death/drug effects , Glioblastoma/drug therapy , Photochemotherapy , Prodrugs/pharmacology , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/ultrastructure , Humans , Hydroxymethylbilane Synthase/metabolism , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism
6.
Photochem Photobiol Sci ; 10(12): 1926-33, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22020364

ABSTRACT

Multi-drug resistance of breast cancer is a major obstacle in chemotherapy of cancer treatments. Recently it was suggested that photodynamic therapy (PDT) can overcome drug resistance of tumors. ALA-PDT is based on the administration of 5-aminolevulinic acid (ALA), the natural precursor for the PpIX biosynthesis, which is a potent natural photosensitizer. In the present study we used the AlaAcBu, a multifunctional ALA-prodrug for photodynamic inactivation of drug resistant MCF-7/DOX breast cancer cells. Supplementation of low doses (0.2mM) of AlaAcBu to the cells significantly increased accumulation of PpIX in both MCF-7/WT and MCF-7/DOX cells in comparison to ALA, or ALA + butyric acid (BA). In addition, our results show that MCF-7/DOX cells are capable of producing higher levels of porphyrins than MCF-7/WT cells due to low expression of the enzyme ferrochelatase, which inserts iron into the tetra-pyrrol ring to form the end product heme. Light irradiation of the AlaAcBu treated cells activated efficient photodynamic killing of MCF-7/DOX cells similar to the parent MCF-7/WT cells, depicted by low mitochondrial enzymatic activity, LDH leakage and decreased cell survival following PDT. These results indicate that the pro-drug AlaAcBu is an effective ALA derivative for PDT treatments of multidrug resistant tumors.


Subject(s)
Aminolevulinic Acid/pharmacology , Levulinic Acids/pharmacology , Photosensitizing Agents/pharmacology , Prodrugs/pharmacology , Aminolevulinic Acid/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Levulinic Acids/chemistry , Levulinic Acids/therapeutic use , Microscopy, Fluorescence , Photochemotherapy , Photosensitizing Agents/therapeutic use , Prodrugs/therapeutic use , Protoporphyrins/metabolism
7.
Photochem Photobiol Sci ; 10(8): 1310-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21655622

ABSTRACT

Successful 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) is dependent on efficient porphyrin synthesis in the inflicted cancer tissue, which is regulated by several enzymes. Irradiation of the tumor excites the light-sensitive porphyrins and results in ROS production and cell death. In this study we investigated the effect of the expression levels of two main enzymes in heme biosynthesis, ALA dehydratase (ALAD) and porphobilinogen deaminase (PBGD), on the capacity of K562 cells to undergo cell death following ALA-PDT. We manipulated PBGD and ALAD expression levels by shRNAs and PBGD overexpressing plasmid. PBGD down-regulation induced an elevation in ALAD activity, while overexpression of PBGD reduced ALAD activity, indicating a novel regulation feedback of PBGD on ALAD activity. This feedback mechanism enabled partial PpIX synthesis under PBGD silencing, whereas ALAD silencing reduced PpIX production to a minimum. ALA-PDT efficacy was directly correlated to PpIX levels. Thus, only ALAD-silenced cells were not affected by ALA+ irradiation, while following PBGD silencing, the accumulated PpIX, though decreased, was sufficient for successful ALA-PDT. The alterations in ALAD activity level initiated by changes in PBGD expression indicates PBGD's central role in heme synthesis. This enables efficient ALA-PDT, even when PBGD is not fully active. Conversely, ALAD loss resulted in reduced PpIX synthesis and consequently failure in ALA-PDT, due to the absence of compensation mechanism for ALAD.


Subject(s)
Aminolevulinic Acid/pharmacology , Gene Expression Regulation, Neoplastic , Hydroxymethylbilane Synthase/metabolism , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/therapeutic use , Apoptosis , Humans , Hydroxymethylbilane Synthase/antagonists & inhibitors , Hydroxymethylbilane Synthase/genetics , K562 Cells , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/enzymology , Light , Photochemotherapy , Porphobilinogen Synthase/antagonists & inhibitors , Porphobilinogen Synthase/genetics , Porphobilinogen Synthase/metabolism , Protoporphyrins/metabolism , RNA Interference , RNA, Small Interfering/metabolism
8.
J Environ Pathol Toxicol Oncol ; 29(2): 137-58, 2010.
Article in English | MEDLINE | ID: mdl-20932248

ABSTRACT

Environmental pollution of heavy metals is very abundant nowadays from industry, chemicals, old paints, and pipes or resulting from previous contaminants accumulating in the food chain. Most of the iron demands of the body are needed for heme synthesis and assembly, but iron is also required for Fe-S cluster proteins and other redox enzymes. Heme is an essential, iron-binding molecule used as a prosthetic group of hemoproteins or as a regulator in multiple cellular pathways. In this review, we focused on the effect of exposure to heavy metals, such as Pb, Ga, Cu, Kd, Hg and Al, on heme synthesis as the main iron-sequestering process of the human body. These metals compete with iron on transporters, reduce the cellular iron pool and moreover, bind to proteins, and cause physical and mental disturbances. Heavy metals mainly impair various aspects of the heme synthesis pathway: gene expression, enzyme activity, and iron integration into protoporphyrin IX. Main risk factors are described as well as effects on iron dependent processes in order to increase public awareness to the distribution of heavy metals in our close environment and the harsh consequences of exposure, even in low doses.


Subject(s)
Gene Expression Regulation/drug effects , Heme/biosynthesis , Metals, Heavy/toxicity , 5-Aminolevulinate Synthetase/genetics , Animals , Cadmium/toxicity , Copper/toxicity , Ferrochelatase/genetics , Heme Oxygenase (Decyclizing)/genetics , Homeostasis , Humans , Hydroxymethylbilane Synthase/genetics , Iron/metabolism , Lead Poisoning/metabolism , Photochemotherapy , Porphobilinogen Synthase/genetics
9.
J Environ Pathol Toxicol Oncol ; 28(3): 209-21, 2009.
Article in English | MEDLINE | ID: mdl-19888908

ABSTRACT

Wilson's disease (Wd) is a genetic disorder resulting in Cu2+ accumulation, and is caused by mutations in the ATP7B gene, the copper transporter. In vivo studies show a correlation between Cu2+ accumulation and malfunction of the heme biosynthesis pathway. In this study, we describe multiple effects of Cu2+ accumulation on heme synthesis, which, in turn, affect proteasomal activity. Cu2+ toxicity was examined in two hepatocellular carcinoma cell lines, HepG2 and Hep3B, with Hep3B cells containing an integrated hepatitis B virus genome. Exposure of HepG2 and Hep3B cells to Cu2+ inhibited the enzymes PBGD and ALAD of the heme synthesis pathway and, in parallel, upregulated heme oxygenase-1 (HO-1). Proto-porphyrin IX (PpIX) and the heme pool were reduced as a result of these processes. PpIX synthesis was found to be lower in cells expressing the mutant ATP7B (P1134P), compared to those expressing the WT enzyme. Proteasomal activity was inhibited under Cu2+ treatment in HepG2 cells; however, Cu2+ induced marked proteosomal acceleration in Hep3B cells. Under these conditions, Ub-conjugated proteins were gradually accumulated, whereas treatment with bathocuproine disulfonic acid (BCS), a Cu2+ chelator, reversed this effect. In conclusion, our data suggest that copper downregulates the heme synthesis pathway in hepatocellular cells and further reduces it in the presence of mutated ATP7B.


Subject(s)
Copper/toxicity , Enzyme Inhibitors/toxicity , Heme/biosynthesis , Hepatocytes/drug effects , Hepatolenticular Degeneration , Proteasome Endopeptidase Complex/drug effects , Adenosine Triphosphatases/biosynthesis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cation Transport Proteins/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Copper-Transporting ATPases , Disulfiram/pharmacology , Down-Regulation/drug effects , Hepatocytes/metabolism , Humans , Hydroxymethylbilane Synthase/antagonists & inhibitors , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Porphobilinogen Synthase/antagonists & inhibitors , Proteasome Endopeptidase Complex/metabolism , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin/metabolism
10.
Photochem Photobiol Sci ; 8(10): 1461-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19789817

ABSTRACT

Synthesis of protoporphyrin IX (PpIX) by malignant cells is essential for the success of ALA-based photodynamic therapy (PDT). Two key enzymes that were described as affecting PpIX accumulation during ALA treatment are porphobilinogen deaminase (PBGD) and ferrochelatase. Here, we show that down regulation of ALA dehydratase (ALAD) expression and activity by specific shRNA induced a marked decrease in PpIX synthesis in K562 erythroleukemic cells. Photo-inactivation efficacy following ALA-PDT was directly correlated with ALAD-silencing and cellular levels of PpIX. MTT metabolism following ALA-PDT was shown to be 60% higher in ALAD-silenced cells in comparison to control cells, indicating that mitochondria were protected in the silenced cells. Morphological analysis by scanning electron microscopy (SEM) of cells treated by ALA-PDT showed no morphological changes in ALAD-silenced cells, in contrast to controls exhibiting cell deformations and lysis. Membrane integrity following ALA-PDT was kept intact and undamaged in ALAD-silenced cells as examined by Annexin V-FITC/PI staining and LDH-L leakage. We conclude that ALAD, although it is present in the cell at abundant levels, has a major and limiting role in regulating PpIX synthesis and ALA-PDT outcome.


Subject(s)
Aminolevulinic Acid/pharmacology , Gene Silencing , Leukemia, Erythroblastic, Acute/pathology , Photochemotherapy , Porphobilinogen Synthase/deficiency , Porphobilinogen Synthase/genetics , Aminolevulinic Acid/therapeutic use , Cell Death/drug effects , Cell Death/radiation effects , Humans , K562 Cells , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Protoporphyrins/biosynthesis
11.
Article in English | MEDLINE | ID: mdl-19392651

ABSTRACT

The involvement of environmental heavy metals in Parkinson's disease (PD) has been suggested by epidemiologic studies; however, the mechanism of this effect is unknown. PD is characterized by the aggregation of alpha-synuclein in Lewy bodies. We previously showed that Pb2+ accelerates proteasomal activity. Therefore, we examined the effect of Pb2+, Ga3+, and Cu2+ on alpha-synuclein in human SH-SY5Y cells. The heavy metals induced an increase in heme-oxygenase-1 levels without significant cell death or ROS generation. The metals inhibited ALA-dehydratase, which is the inhibitory subunit of the proteasome, thereby accelerating proteasomal activity and decreasing protein levels of CDK-1 and PBGD. However, alpha-synuclein protein levels increased after exposure to metals, similar to the effect obtained with the proteasome inhibitor, hemin, suggesting that alpha-synuclein is inaccessible to proteasomal degradation. Indeed, electron microscopy revealed the formation of aggresomes in Pb2+- or hemin-treated cells. Thus, although heavy metals enhance proteasomal activity, alpha-synuclein is protected from degradation, and its protein levels and aggregation are increased.


Subject(s)
Copper/toxicity , Gallium/toxicity , Lead/toxicity , Proteasome Endopeptidase Complex/drug effects , alpha-Synuclein/metabolism , Apoptosis/drug effects , Cell Line , Cytoplasmic Structures/drug effects , Cytoplasmic Structures/ultrastructure , Heme Oxygenase-1/metabolism , Hemin/pharmacology , Humans , Mutation , Porphobilinogen Synthase/antagonists & inhibitors , Porphobilinogen Synthase/metabolism , Proteasome Endopeptidase Complex/ultrastructure , Reactive Oxygen Species/metabolism
12.
J Med Chem ; 51(23): 7356-69, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-19007111

ABSTRACT

Multifunctional acyloxyalkyl ester prodrugs of 5-aminolevulinic acid in cancer cell lines inhibited the proteasome and induced apoptosis and heme synthesis. The most potent prodrug was butyryloxymethyl 5-amino-4-oxopentanoate (1a). The metabolically released formaldehyde from the prodrugs was the dominant factor affecting cell viability by a ROS-dependent mechanism and was responsible for rapid phosphorylation of H2AX, suppression of the cell survival protein c-myc, and transient elevation in the expression of p21. 1a, which differs from 2a by releasing butyric instead of pivalic acid, was a more potent inducer of heme and acetylated H4 expression and induced apoptosis through activation of caspase 9. 1a and 1b specifically increased the level of the photosensitizer protoporphyrin 9, leading to enhancement of cell death by photodynamic therapy (PDT). The advantage of these multifunctional prodrugs over 5-ALA is their greater potency in the non-PDT mechanism of cancer cell killing and their ability to also augment PDT.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/pharmacology , Antineoplastic Agents/pharmacology , Prodrugs/pharmacology , Aminolevulinic Acid/chemical synthesis , Aminolevulinic Acid/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Butyric Acid/metabolism , Butyric Acid/pharmacology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , HL-60 Cells , Heme/metabolism , Histones/drug effects , Histones/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Phosphorylation , Photochemistry , Porphyrins/metabolism , Prodrugs/chemical synthesis , Prodrugs/chemistry , Proteasome Inhibitors , Proto-Oncogene Proteins c-myc/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Reactive Oxygen Species/metabolism , Stereoisomerism , Tumor Cells, Cultured
13.
Cancer Lett ; 256(1): 39-48, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17611019

ABSTRACT

The antiangiogenic and antineoplastic activities of the butyric acid prodrugs AN-7 and AN-9 were demonstrated in vitro with HUVEC by inhibition of proliferation and vascular tubes formation, enhanced apoptosis, and inhibition of 22Rv-1 cells migration. In the sc implanted human prostate tumors (22Rv-1) in nude mice, AN-7 significantly inhibited Ki-67, HIF-1alpha, HER-2/neu, bFGF and increased PTEN level. AN-7 and AN-9 reduced hemoglobin accumulation in matrigel plugs implanted sc in Balb-c mice. Herein, we show that the anticancer activity of AN-7 and AN-9 can be attributed in part to their antiangiogenic activities suggesting potential therapeutic benefits for prostate cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Butyrates/pharmacology , Neovascularization, Pathologic/drug therapy , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured , Umbilical Veins/cytology
14.
Biochim Biophys Acta ; 1762(9): 819-27, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16935474

ABSTRACT

Acute intermittent porphyria (AIP) is a neuropathic disease caused by a dominant inherited deficiency in porphobilinogen deaminase (PBGD). We investigated the expression and the degradation of the human PBGD-mutations G748A, G748C and 887insA following transfection into human SH-SY5Y neuroblastoma cells. Mutant proteins exhibited reduced protein expression compared to transfected wild-type (wt) PBGD as revealed by Western blotting. The transcription levels assessed by real-time PCR of these mutant species were identical to those of the wild type. Immuno-fluorescence microscopy revealed reduced cellular distribution of the mutated PBGDs in the cytosol and the nucleus in comparison to the wild-type PBGD. Enhanced cellular accumulation of the mutated and wild-type PBGDs was detected following inhibition of the proteasome by the inhibitors CLBL and hemin. Elevated expression of wt and mutated PBGD protein levels was either achieved by hemin or heme-arginate treatment. On the other hand, enhanced PBGD degradation was achieved by lead poisoning of ALAD in the SH-SY5Y cells concomitant with acceleration of proteasomal activity, most probably by ALAD participation in proteasomal regulation [G.G. Guo, M. Gu, J.D. Etlinger, 240-kDa proteasome inhibitor (CF-2) is identical to delta-aminolevulinic acid dehydratase. J Biol Chem 1994; 269:12399-402.] Our results suggest that the difference in expression between the wild-type and mutant proteins appears to be regulated on the level of protein degradation. In conclusion, we demonstrate that the PBGD cellular pool is controlled by the proteasome activity, which in turn is down regulated by hemin or up-regulated by Pb-ALAD.


Subject(s)
Hydroxymethylbilane Synthase/genetics , Hydroxymethylbilane Synthase/metabolism , Neuroblastoma/enzymology , Porphyria, Acute Intermittent/genetics , Proteasome Endopeptidase Complex/genetics , RNA, Messenger/metabolism , Aminolevulinic Acid , Cell Line, Tumor , Cell Nucleus/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/metabolism , Gene Expression Regulation , Hemin/pharmacology , Humans , Lactones/pharmacology , Lead Poisoning , Mutant Proteins , Transfection
15.
Photochem Photobiol ; 82(5): 1319-25, 2006.
Article in English | MEDLINE | ID: mdl-16740058

ABSTRACT

Photodynamic therapy (PDT) is being evaluated in clinical trials for treatment of various oncologic and ophthalmic diseases. The main cause for cell inactivation and retardation of tumor growth after photoactivation of sensitizers is very short-lived singlet oxygen molecules that are produced and have limited diffusion distances. In this paper we show that the extent of biological damage can be modulated by using protoporphyrin, which was modified to increase its lipophilicity, and which also places the tetrapyrrole core deeper within the membrane by the carboxylate groups being anchored at the lipid:water interface. The uptake of the parent molecule (PPIX) and its diheptanoic acid analogue (PPIXC6) by WiDR and CT26 cells was investigated by fluorescence microscopy and by fluorescence intensity from the cells. The uptake of PPIXC6 increased almost linearly with incubation length for over 24 h, whereas for PPIX only 1 h was needed to reach maximal intracellular concentration. Fluorescence microscopy of both cell lines indicated that both drugs were distributed diffusely in the plasma membrane and cytoplasm, but remained outside the nucleus. The efficiency of in vitro inactivation of WiDr and CT26 cells increased with the length of the alkylcarboxylic chain. Tumors in mice that were treated with PPIX-PDT grew more slowly than control tumors. However, tumors that were given PPIXC6 followed by light exposure showed a significant delay in their growth.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Survival/drug effects , Protoporphyrins/chemistry , Protoporphyrins/pharmacokinetics , Biological Transport , Cell Line, Tumor , Colonic Neoplasms , Humans , Kinetics , Photochemotherapy , Photosensitivity Disorders , Photosensitizing Agents/toxicity , Protoporphyrins/toxicity
16.
J Environ Pathol Toxicol Oncol ; 25(1-2): 145-58, 2006.
Article in English | MEDLINE | ID: mdl-16566714

ABSTRACT

Protoporphyrin IX (PpIX) synthesis by malignant cells is successfully exploited for photodynamic therapy (PDT) following administration of 5-aminolevulinic acid (ALA) and light irradiation. The influence of two environmental heavy metal poisons, lead and gallium, on PpIX-synthesis and ALA-PDT was studied in two neu-ronal cell lines, SH-SY5Y neuroblastoma and PC12 pheochromocytoma. The heavy metal intoxication affected two of the heme-synthesis enzymes, ALA-dehydratase (ALAD) and porphobilinogen deaminase (PBGD). The present results show that lead poisoning significantly decreased the PBGD cellular level and inhibited its enzymatic activity, whereas the effects of gallium were less prominent. Although, the protein levels were reduced, the mRNA levels of PBGD remained unchanged during metal intoxication. These findings show additional inhibitory activity of lead on top of its classical effect on ALAD. Proteasome activity was enhanced during lead treatment, as measured by the AMC fluorigenic proteasome assay. The reduction in PBGD levels was not a consequence of PBGD mRNA reduced synthesis, which remained unchanged as shown by RT-PCR analysis. As a result of the lead poisoning, marked alterations in the cell cycle were observed, including a decreased G1 phase and an increased number of S phase cells. The efficacy of ALA-PDT was reduced in correlation with decreased activities of the enzymes during lead intoxication. We may conclude that lead poisoning adversely affects the outcome of ALA photodynamic therapy of cancer.


Subject(s)
Aminolevulinic Acid/pharmacology , Lead/toxicity , Photochemotherapy , Porphobilinogen Synthase/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Enzyme Inhibitors/toxicity , Gallium/toxicity , Humans , Hydroxymethylbilane Synthase/genetics , Hydroxymethylbilane Synthase/metabolism , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Photosensitizing Agents/pharmacology , Porphobilinogen Synthase/metabolism , Proteasome Endopeptidase Complex/metabolism , Protoporphyrins/metabolism , RNA, Messenger/metabolism , Rats
17.
Int J Cancer ; 116(2): 226-35, 2005 Aug 20.
Article in English | MEDLINE | ID: mdl-15800932

ABSTRACT

AN-7, a prodrug of butyric acid, induced histone hyperacetylation and differentiation and inhibited proliferation of human prostate 22Rv1 cancer cells in vitro and in vivo. In nude mice implanted with these cells, 50 mg/kg AN-7 given orally thrice a week led to inhibition of tumor growth and metastasis, tumor regression in >25% of animals and increased survival. Median time to the experimental end point (tumor volume 2 cm3 or death) in the untreated was 52 days, and average tumor volume was 0.8 +/- 0.18 cm3. At the same time, 94.4% of AN-7-treated mice survived and had average tumor volumes of 0.37 +/- 0.1 cm3. PSA expression was a useful marker for 22Rv1 lung metastasis detection. Sizeable metastases positively stained for PSA and limited air gaps were found in lungs of untreated mice. In animals treated with AN-7, lung morphology appeared normal. Primary tumors of treated animals were highly positive for PSA and had an elevated level of p21 and the proapoptotic protein Bax. Sections taken from AN-7-treated animals, examined under an electron microscope, exhibited condensed chromatin and apoptotic bodies. PSA serum levels were higher in untreated compared to treated animals and correlated with tumor volume. Since prolonged oral administration with 50 mg/kg or a single oral dose of 1.2 g/kg AN-7 did not cause adverse effects and the former exhibited significant anticancer activity, AN-7 is likely to display a high therapeutic index and may be beneficial for prostate cancer patients.


Subject(s)
Butyrates/pharmacology , Organophosphorus Compounds/pharmacology , Prodrugs , Prostatic Neoplasms/pathology , Acetylation , Administration, Oral , Animals , Cell Differentiation , Cell Proliferation , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Prostate-Specific Antigen/blood , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Photochem Photobiol Sci ; 3(5): 423-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15122359

ABSTRACT

The mechanistic aspects of Escherichia coli photodynamic inactivation (PDI) have been studied in bacteria expressing the reporter protein GFP, following transfection with wild type pGFP plasmid and treatment with the hydrophilic cationic sensitizer tetra-meso(N-methyl-4-pyridyl)porphine tetratosylate (TMPyP). Cell survival and morphology during PDI were correlated with plasmid-GFP degradation in comparison to DNA and RNA strand-breaks, while photobleaching of the GFP chromophore was used to monitor protein photodamage. Singlet oxygen generated upon TMPyP photoactivation interacted with target nucleic acid polymers in a drug-and light-dose dependent manner. The hierarchy and cascade of the photodamage was in the order: genomic-DNA > total RNA > plasmid-DNA, as revealed by specific extraction and agarose electrophoresis. The notable resistance of the plasmid DNA in comparison to genomic DNA has implications for PDI of antibiotic-resistant bacteria. Re-growth of the treated cells in fresh medium showed structural features of an SOS response. Under these conditions, DNA repair machinery was initiated by typical alignment of DNA-protein co-aggregates accompanied by lateral assembly of ribosomes, apart from damaged DNA-arrays, as depicted by electron microscopy. GFP-TMPyP interactions were demonstrated by double green and red fluorescence on electrophoresis plates analyzed by spectral imaging. Photobleaching measurements revealed specific GFP photodamage directly related to PDI of the E. coli. The kinetics of both the GFP photobleaching and the K(+) efflux, representing photodamage to cytosolic proteins and membrane damage, respectively, were found to be similar. The survival curves were correlated to chromosomal degradation and ultrastructural damage. We conclude that TMPyP-dependent PDI of E. coli is primarily dependent on genomic DNA photodamage rather than on protein or membrane malfunctions.


Subject(s)
Escherichia coli/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Escherichia coli/genetics , Escherichia coli/radiation effects , Escherichia coli/ultrastructure , Green Fluorescent Proteins , Luminescent Proteins/genetics , Microscopy, Electron
19.
Photochem Photobiol Sci ; 3(5): 430-5, 2004 May.
Article in English | MEDLINE | ID: mdl-15122360

ABSTRACT

In the present study we examined the production of high amounts of porphyrins upon induction by delta-aminolevulinic acid (ALA) in 9 bacterial strains. This was performed by solely inducing the porphyrin biosynthesis pathway. Four of the strains were Gram positive bacteria and five were Gram negative strains. All strains, except Streptococcus faecalis, produced porphyrins when incubated in PBS with 0.38 mM ALA for 4 h. Excess porphyrin production was excreted to the medium. Gram positive bacteria exhibited fluorescent emission peaks at 622 nm for the endogenous and 617 nm for the excreted porphyrins. Gram negative bacteria exhibited a 630 nm emission peak for the endogenous and a 615 nm emission peak for the excreted extracellular porphyrins. Upon illumination of the ALA induced Staphylococcal strains with 407-420 nm blue light, a decrease of five orders of magnitude was demonstrated with a light dose of 50 J cm(-2). Total eradication of the Staphylococcal strains could be achieved with a 100 J cm(-2) dose, which resulted in a decrease in viability of seven orders of magnitude. The viability of all the induced Gram negative strains and B. cereus decreased by one or two orders of magnitude upon illumination with 50 and 100 J cm(-2), respectively. This difference in the photoinactivation rate was found to be due to the distribution and amounts of the various porphyrins in the bacterial strains. The predominant porphyrin in the Staphylococcal strains was coproporphyrin (68.3-74.6%). In the Gram negative strains there was no predominant porphyrin and the porphyrins found were mostly 5-carboxyporphyrin, uroporphyrin, 7- carboxyporphyrin, coproporphyrin and protoporphyrin. In the B. cereus(Gram positive) strain the predominant porphyrin was uroporphyrin (75.8%). Although the total production of porphyrins in the Gram negative bacteria was higher than in the Staphylococcal strains, the amount of coproporphyrin produced by the latter was twice to three times higher than in the Gram negative strains. The extracellular excreted porphyrins did not contribute to the photoinactivation in any of the tested strains. Significant decreases in the Na(+) and K(+) content were detected in induced S. aureus after illumination while only small changes were observed in E. coli B. The green fluorescent protein within the cytoplasm of induced E. coli strains was only partially disrupted (by 60% only). These results indicate a partial yield of the effects generated by (1)O(2) radicals resulting from the photoinactivation of Gram negative bacteria and a successful generation of the same effects in the Staphylococcal strains.


Subject(s)
Aminolevulinic Acid/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Spectrometry, Fluorescence
20.
Oncogene ; 22(34): 5221-8, 2003 Aug 14.
Article in English | MEDLINE | ID: mdl-12917623

ABSTRACT

Porphobilinogen deaminase (PBGD) is a rate-limiting enzyme of the heme biosynthesis pathway, whose level is elevated in various human tumors. PBGD was observed in both nuclear and cytoplasmic fractions of C6 glioma cells by immunostaining. During mitosis, chromatids were intensely stained for PBGD in comparison to the interphase chromatin. Using the yeast two-hybrid system, we identified RanBPM, the nuclear Ran-binding protein, as an interacting partner of PBGD. During butyrate-induced differentiation of C6, both nuclear and cytoplasmic PBGD levels declined as did Ran protein and its nucleotide exchange factor RCC1. N,N'-hexamethylene bis-acetamide-dependent differentiation resulted in an increase of the cytoplasmic PBGD, whereas nuclear PBGD, Ran protein and RCC1 remained unchanged. mRNA levels of PBGD remained unchanged during stimulation with both butyrate and N,N'-hexamethylene bis-acetamide. The enzymatic activity of PBGD and protoporphyrin IX synthesis in C6 cells were dependent on the differentiation induction agent. We conclude that PBGD possibly has a nuclear role in addition to its cytosolic enzymatic activity required for heme synthesis, which is related to cell transformation and differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Nucleus/metabolism , Glioma/metabolism , Hydroxymethylbilane Synthase/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Animals , HeLa Cells , Humans , Molecular Sequence Data , Porphyrins/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...