Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neuromolecular Med ; 16(2): 292-307, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24272426

ABSTRACT

Multiple sclerosis (MS) is an inflammatory autoimmune disease of the central nervous system (CNS) involving demyelinating and neurodegenerative processes. Several of the major pathological CNS alterations and behavioral deficits of MS are recapitulated in the experimental autoimmune encephalitis (EAE) mouse model in which the disease process is induced by administration of myelin peptides. Development of EAE requires infiltration of inflammatory cytokine-generating monocytes and macrophages, and auto-reactive T cells, into the CNS. Very late antigen-4 (VLA-4, α4ß1) is an integrin molecule that plays a role in inflammatory responses by facilitating the migration of leukocytes across the blood-brain barrier during inflammatory disease, and antibodies against VLA-4 exhibit therapeutic efficacy in mouse and monkey MS models. Here, we report that the tellurium compound AS101 (ammonium trichloro (dioxoethylene-o,o') tellurate) ameliorates EAE by inhibiting monocyte and T cell infiltration into the CNS. CD49d is an alpha subunit of the VLA-4 (α4ß1) integrin. During the peak stage of EAE, AS101 treatment effectively ameliorated the disease process by reducing the number of CD49d(+) inflammatory monocyte/macrophage cells in the spinal cord. AS101 treatment markedly reduced the pro-inflammatory cytokine levels, while increasing anti-inflammatory cytokine levels. In contrast, AS101 treatment did not affect the peripheral populations of CD11b(+) monocytes and macrophages. AS101 treatment reduced the infiltration of CD4(+) and CD49(+)/VLA4 T cells. In addition, treatment of T cells from MS patients with AS101 resulted in apoptosis, while such treatment did not affect T cells from healthy donors. These results suggest that AS101 reduces accumulation of leukocytes in the CNS by inhibiting the activity of the VLA-4 integrin and provide a rationale for the potential use of Tellurium IV compounds for the treatment of MS.


Subject(s)
Cell Movement/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Ethylenes/therapeutic use , Immunologic Factors/therapeutic use , Integrin alpha4beta1/antagonists & inhibitors , Monocytes/drug effects , Spinal Cord/immunology , T-Lymphocyte Subsets/drug effects , Animals , Apoptosis/drug effects , Blood-Brain Barrier/immunology , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Cytokines/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Ethylenes/pharmacology , Female , Humans , Immunologic Factors/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Monocytes/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Spleen/metabolism , T-Lymphocyte Subsets/immunology
2.
PLoS One ; 8(9): e73625, 2013.
Article in English | MEDLINE | ID: mdl-24066060

ABSTRACT

Failure of conventional therapies to alleviate glioblastoma (GBM) fosters search for novel therapeutic strategies. These include epigenetic modulators as histone deacetylase inhibitors (HDACi), which relax abnormally compact tumor cell chromatin organization, enabling cells to overcome blockage in differentiation. However, in clinical settings, HDACi efficacy is confined to subsets of hematologic malignancies. We reasoned that molecules targeting multiple epigenetic mechanisms may exhibit superior anti-cancer activities. We focused on the redox perylene-quinone Hypericin (HYP) and showed that HYP targets Hsp90 for polyubiquitination, degradation and inactivation. Hsp90 is implicated in mediating inheritable epigenetic modifications transferable to progeny. We therefore examined if HYP can induce epigenetic alterations in GBM cells and show here that HYP indeed, targets multiple mechanisms in human glioblastoma tumor cell lines via unique manners. These elicit major epigenetic signature changes in key developmentally regulated genes. HYP induces neuroglial tumor cell differentiation modulating the cytoarchitecture, neuroglial differentiation antigen expression and causes exit from cell proliferation cycles. Such activities characterize HDACi however HYP is not an HDAC inhibitor. Instead, HYP effectively down-regulates expression of Class-I HDACs, creating marked deficiencies in HDACs cellular contents, leading to histones H3 and H4 hyperacetylation. Expression of EZH2, the Polycomb repressor complex-2 catalytic subunit, which trimethylates histone H3K27 is also suppressed. The resulting histone hyperacetylation and diminished H3K27-trimethylation relax chromatin structure, activating gene transcription including differentiation-promoting genes. DNMT profiles are also modulated increasing global DNA methylation. HYP induces unique epigenetic down-regulations of HDACs, EZH2 and DNMTs, remodeling chromatin structure and culminating in tumor cell differentiation. These modulations generate clinically significant anti-GBM effects obtained in a clinical trial performed in patients with recurrent, progressive disease. Despite this advanced disease stage, patients responded to HYP, displaying stable disease and partial responses; patients on compassionate therapy survived for up to 34 months. Hypericin may constitute a novel anti-glioblastoma therapeutic paradigm.


Subject(s)
Glioblastoma/metabolism , Perylene/analogs & derivatives , Anthracenes , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Humans , Perylene/pharmacology
3.
PLoS One ; 6(9): e22849, 2011.
Article in English | MEDLINE | ID: mdl-21949677

ABSTRACT

The perihydroxylated perylene quinone hypericin has been reported to possess potent anti-metastatic and antiangiogenic activities, generated by targeting diverse crossroads of cancer-promoting processes via unique mechanisms. Hypericin is the only known exogenous reagent that can induce forced poly-ubiquitination and accelerated degradation of heat shock protein 90 (Hsp90) in cancer cells. Hsp90 client proteins are thereby destabilized and rapidly degraded. Hsp70 client proteins may potentially be also affected via preventing formation of hsp90-hsp70 intermediate complexes. We show here that hypericin also induces enhanced degradation of hypoxia-inducible factor 1α (HIF-1α) in two human tumor cell lines, U87-MG glioblastoma and RCC-C2VHL-/- renal cell carcinoma and in the non-malignant ARPE19 retinal pigment epithelial cell line. The hypericin-accelerated turnover of HIF-1α, the regulatory precursor of the HIF-1 transcription factor which promotes hypoxic stress and angiogenic responses, overcomes the physiologic HIF-1α protein stabilization which occurs in hypoxic cells. The hypericin effect also eliminates the high HIF-1α levels expressed constitutively in the von-Hippel Lindau protein (pVHL)-deficient RCC-C2VHL-/- renal cell carcinoma cell line. Unlike the normal ubiquitin-proteasome pathway-dependent turnover of HIF-α proteins which occurs in normoxia, the hypericin-induced HIF-1α catabolism can occur independently of cellular oxygen levels or pVHL-promoted ubiquitin ligation of HIF-1α. It is mediated by lysosomal cathepsin-B enzymes with cathepsin-B activity being optimized in the cells through hypericin-mediated reduction in intracellular pH. Our findings suggest that hypericin may potentially be useful in preventing growth of tumors in which HIF-1α plays pivotal roles, and in pVHL ablated tumor cells such as renal cell carcinoma through elimination of elevated HIF-1α contents in these cells, scaling down the excessive angiogenesis which characterizes these tumors.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Perylene/analogs & derivatives , Anthracenes , Antineoplastic Agents/pharmacology , Blotting, Western , Cathepsin B/metabolism , Cell Hypoxia , Cell Line , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen-Ion Concentration/drug effects , Metabolism/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Perylene/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitination/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Immunology ; 128(2): 287-300, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19740385

ABSTRACT

We developed a method for selectively propagating disease-related autoreactive T-cell lines (auTCLs) based on their increased resistance to apoptosis. The generated auTCLs homogeneously co-express CD45RO and CD49a, adhere strongly to extracellular matrix proteins and express high interleukin-17 (IL-17) messenger RNA levels, resembling a T-cell subset proposed to transmigrate into tissues and induce systemic and local inflammation in rheumatoid arthritis. The combinations of T-cell oligoclones that comprise probable multiple sclerosis (pMS) disease-related lines use a unique portfolio of T-cell receptor beta-chain variable allele (BV genes) combinations forming 'disease-specific cluster patterns'. The auTCL derived from different patients and from different myelin epitopes display striking similarities in BV gene allele clusters and are derived primarily from a disease-prone hotspot residing in the BV gene locus between Vbeta6 and Vbeta9. Conversely, healthy subject TCLs use different BV gene allele sets, forming 'healthy responder usage formats'. These formats were absent from the pMS patient V-beta gene allele combinations evaluated in this study. Hierarchical clustering of the BV gene combinations, distinguish three pMS auTCL groups, implying existence of up to three disease-related immune response patterns. These subgroup patterns may reflect different disease subclasses or alternatively they may suggest immune reactivity to different aetiological agents. Analyses of clonal-clustering patterns may potentially aid in subclassification of MS or in characterizing aetiological agents of this disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Multiple Sclerosis/immunology , Adult , Alleles , Apoptosis/immunology , Autoimmunity , Cell Line , Clone Cells/immunology , Cluster Analysis , Histocompatibility Testing , Humans , Lymphocyte Activation/immunology , Multiple Sclerosis/genetics , Myelin Sheath/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Young Adult
5.
Sensors (Basel) ; 9(4): 2538-54, 2009.
Article in English | MEDLINE | ID: mdl-22574031

ABSTRACT

Microbial biofilms are a major impediment to the use of indwelling medical devices, generating device-related infections with high morbidity and mortality. Major efforts directed towards preventing and eradicating the biofilm problem face difficulties because biofilms protect themselves very effectively by producing a polysaccharide coating, reducing biofilm sensitivity to antimicrobial agents. Techniques applied to combating biofilms have been primarily chemical. These have met with partial and limited success rates, leading to current trends of eradicating biofilms through physico-mechanical strategies. Here we review the different approaches that have been developed to control biofilm formation and removal, focusing on the utilization of acoustic energy to achieve these objectives.

6.
Acta Oncol ; 47(6): 1062-70, 2008.
Article in English | MEDLINE | ID: mdl-17934890

ABSTRACT

BACKGROUND: The active components of Cannabis sativa L., Cannabinoids, traditionally used in the field of cancer for alleviation of pain, nausea, wasting and improvement of well-being have received renewed interest in recent years due to their diverse pharmacologic activities such as cell growth inhibition, anti-inflammatory activity and induction of tumor regression. Here we used several experimental approaches, which identified delta-9-tetrahydrocannabinol (Delta(9)-THC) as an essential mediator of cannabinoid antitumoral action. METHODS AND RESULTS: Administration of Delta(9)-THC to glioblastoma multiforme (GBM) cell lines results in a significant decrease in cell viability. Cell cycle analysis showed G(0/1) arrest and did not reveal occurrence of apoptosis in the absence of any sub-G(1) populations. Western blot analyses revealed a THC altered cellular content of proteins that regulate cell progression through the cell cycle. The cell content of E2F1 and Cyclin A, two proteins that promote cell cycle progression, were suppressed in both U251-MG and U87-MG human glioblastoma cell lines, whereas the level of p16(INK4A), a cell cycle inhibitor was upregulated. Transcription of thymidylate synthase (TS) mRNA, which is promoted by E2F1, also declined as evident by QRT-PCR. The decrease in E2F1 levels resulted from proteasome mediated degradation and was prevented by proteasome inhibitors. CONCLUSIONS: Delta(9)-THC is shown to significantly affect viability of GBM cells via a mechanism that appears to elicit G(1) arrest due to downregulation of E2F1 and Cyclin A. Hence, it is suggested that Delta(9)-THC and other cannabinoids be implemented in future clinical evaluation as a therapeutic modality for brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Cycle/drug effects , Dronabinol/pharmacology , E2F1 Transcription Factor/drug effects , E2F1 Transcription Factor/metabolism , Glioblastoma/drug therapy , Blotting, Western , Brain Neoplasms/metabolism , Cell Division/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/drug effects , E2F1 Transcription Factor/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Humans , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymidylate Synthase/drug effects , Thymidylate Synthase/metabolism , Time Factors , Up-Regulation
7.
Cancer J ; 13(5): 335-44, 2007.
Article in English | MEDLINE | ID: mdl-17921733

ABSTRACT

PURPOSE: Glioblastoma multiforme (GBM) is the most frequent and incurable brain tumor in adults. Although temozolomide (TMZ) does not cure GBM, it has demonstrated anti-GBM activity and has improved survival (8-14 months) and quality of life. We studied the mechanisms by which TMZ affects 2 human GBM cell lines; U251-MG and U87-MG, aiming to unravel the drug-activated cascades to enable the development of combination therapies that will improve the efficacy of TMZ. MATERIALS AND METHODS: The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay was used to assess cell viability. Modulation of gene expression by TMZ therapy was assayed by gene profiling and verified by quantitative real-time polymerase chain reaction. Protein levels influenced by the treatment were studied by Western blots and immunocytochemistry. RESULTS: Increasing concentrations of TMZ decreased cell viability in a concentration-dependent manner. The expression of 1,886 genes was altered >2-fold after TMZ treatment. We focused on the 81 genes similarly altered by TMZ treatment in both cell lines to neutralize tissue-specific characteristics. Fourteen target genes of hypoxia-inducible factor (HIF-1), were found to be up-regulated after TMZ treatment including vascular endothelial growth factor (VEGF). HIF-1alpha expression was constant at the mRNA level; however, its post-treatment protein levels increased compared with those of untreated control cells. DISCUSSION: The genetic analyses suggest that treatment with TMZ activates stress mechanisms in GBM cells that include the angiogenesis-inducing proteins HIF-1alpha and VEGF. We propose that treatment with TMZ be supplemented with either an antibody to VEGF or down-regulators of HIF-1alpha to improve clinical results of TMZ in the treatment of GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Glioblastoma/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/pharmacology , Gene Expression Profiling , Glioblastoma/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction , Temozolomide
8.
Photochem Photobiol ; 83(5): 1270-7, 2007.
Article in English | MEDLINE | ID: mdl-17880523

ABSTRACT

Incorporation of photodynamic therapy into clinical practice for induction of vascular photo-occlusion highlights the need to prevent adverse phototoxicity to sensitive juxtaposed tissues, particularly in the retina. We developed a system termed "competitive quenching" to prevent adverse phototoxic damage. It involves differential compartmentalization of a photoactivator to the intravascular compartment for photoexcitation and delivery of phototoxicity to targeted vessels. A different photodynamic agent is partitioned to the extravascular retinal space to quench reactive oxygen species generated by photosensitization, thereby protecting the adjacent retinal tissues from adverse phototoxicity. The absorption spectra of quenchers must span wavelengths that are shorter and excluded from the spectral range of photoexcitation light to prevent photoactivation of the quencher. Perihydroxylated perylenequinones were found to be suitable to function as "competitive quenchers" with the prototype hypericin identified as a potent quencher. Here we examined the mechanisms operative in competitive quenching and suggest that hypericin forms a complex with verteporfin, thereby quenching singlet oxygen formation. Furthermore, we show that hypericin, with six phenolic hydroxyls, protects retinal and endothelial hybridoma cells from phototoxicity more effectively than the dimethyl tetrahydroxy helianthrone structural analog with only four such phenolic hydroxyls. The findings suggest that hydroxyl numbers contribute to the efficacy of competitive quenching.


Subject(s)
Photosensitizing Agents/toxicity , Porphyrins/toxicity , Quinones/pharmacology , Cell Line , Hydroxylation , Retina/drug effects , Verteporfin
9.
Antimicrob Agents Chemother ; 50(12): 4144-52, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16940055

ABSTRACT

Low-energy surface acoustic waves generated from electrically activated piezo elements are shown to effectively prevent microbial biofilm formation on indwelling medical devices. The development of biofilms by four different bacteria and Candida species is prevented when such elastic waves with amplitudes in the nanometer range are applied. Acoustic-wave-activated Foley catheters have all their surfaces vibrating with longitudinal and transversal dispersion vectors homogeneously surrounding the catheter surfaces. The acoustic waves at the surface are repulsive to bacteria and interfere with the docking and attachment of planktonic microorganisms to solid surfaces that constitute the initial phases of microbial biofilm development. FimH-mediated adhesion of uropathogenic Escherichia coli to guinea pig erythrocytes was prevented at power densities below thresholds that activate bacterial force sensor mechanisms. Elevated power densities dramatically enhanced red blood cell aggregation. We inserted Foley urinary catheters attached with elastic-wave-generating actuators into the urinary tracts of male rabbits. The treatment with the elastic acoustic waves maintained urine sterility for up to 9 days compared to 2 days in control catheterized animals. Scanning electron microscopy and bioburden analyses revealed diminished biofilm development on these catheters. The ability to prevent biofilm formation on indwelling devices and catheters can benefit the implanted medical device industry.


Subject(s)
Biofilms , Candida/physiology , Catheters, Indwelling/microbiology , Animals , Bacterial Adhesion/physiology , Cell Adhesion , Colony Count, Microbial , Enterococcus faecalis/physiology , Erythrocytes/metabolism , Escherichia coli/physiology , Guinea Pigs , Male , Microscopy, Electron, Scanning , Proteus mirabilis/physiology , Rabbits , Sound/adverse effects , Urinary Catheterization , Vibration/adverse effects
10.
J Neuroimmunol ; 172(1-2): 145-55, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16387370

ABSTRACT

Autologous stem cell transplantation is being considered as treatment of severe refractory autoimmune disorders, including MS. Stem cell mobilization is achieved with granulocyte-colony stimulating factor (G-CSF), however, G-CSF administration resulted in cases of worsened clinical MS status. We studied autoreactive T-cell properties, which can promote CNS inflammation in MS. We show that G-CSF enhances MS autoreactive T cell line adhesion to the ECM proteins collagen IV and fibronectin as effectively as the proinflammatory IFNgamma and TNFalpha, known to exacerbate MS symptoms. We propose a link between clinical worsening of MS symptoms induced by G-CSF and the hyperstimulation of T cell adhesion to ECM elicited by G-CSF.


Subject(s)
Cell Adhesion/drug effects , Extracellular Matrix/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Multiple Sclerosis/pathology , T-Lymphocytes/drug effects , Blotting, Western/methods , Cell Count/methods , Cytokines/metabolism , Cytokines/pharmacology , Epistasis, Genetic , Focal Adhesion Kinase 2/metabolism , Gene Expression/drug effects , Hematopoietic Stem Cell Mobilization/methods , Humans , Integrin alpha1beta1/metabolism , Myelin Basic Protein/pharmacology , Time Factors
11.
Clin Dev Immunol ; 12(3): 203-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16295526

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease where T-cells activated against myelin antigens are involved in myelin destruction. Yet, healthy subjects also harbor T-cells responsive to myelin antigens, suggesting that MS patient-derived autoimmune T-cells might bear functional differences from T-cells derived from healthy individuals. We addressed this issue by analyzing gene expression patterns of myelin oligodendrocytic glycoprotein (MOG) responsive T-cell lines generated from MS patients and healthy subjects. We identified 150 transcripts that were differentially expressed between MS patients and healthy controls. The most informative 43 genes exhibited > 1.5-fold change in expression level. Eighteen genes were upregulated including BCL2, lifeguard, IGFBP3 and VEGF. Twenty five genes were down-regulated, including apoptotic activators like TNF and heat shock protein genes. This gene expression pattern was unique to MOG specific T-cell lines and was not expressed in T-cell lines reactive to tetanus toxin (TTX). Our results indicate that activation in MS that promotes T-cell survival and expansion, has its own state and that the unique gene expression pattern that characterize autoreactive T-cells in MS represent a constellation of factors in which the chronicity, timing and accumulation of damage make the difference between health and disease.


Subject(s)
Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Myelin-Associated Glycoprotein/immunology , T-Lymphocytes/immunology , Base Sequence , Cell Line , DNA/genetics , Gene Expression , Gene Expression Profiling , Humans , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Oligonucleotide Array Sequence Analysis , Peptide Fragments/immunology , T-Lymphocytes/metabolism
12.
Angiogenesis ; 8(1): 35-42, 2005.
Article in English | MEDLINE | ID: mdl-16132616

ABSTRACT

Hypericin, a perihydroxylated dianthraquinone is shown here to be a highly potent inhibitor of angiogenesis in several ocular models examined in rat eyes. Extensive angiogenesis induced in the cornea and iris by intra-ocular administration of FGF-2 was effectively inhibited by a minimum of four dose regimens of hypericin (2 mg/kg) administered via the intraperitoneal route at 48 h intervals. Maximal inhibition was achieved when animal treatment with hypericin was initiated 48 h prior to inoculation of FGF-2. The molecular basis for the hypericin-mediated inhibition of angiogenesis in the anterior eye compartment appears to involve several sites in the cascade leading to angiogenesis. We show that the activating phosphorylation of extracellular signal-regulated MAP kinases (ERK1/2) is inhibited by hypericin in human retinal pigment epithelial cells and in EA.hy926 cells, an endothelial hybridoma expressing endothelial cell properties. ERK1/2 activity is required for the transactivation of hypoxia-inducible factor 1 alpha (HIF-1alpha) and in VEGF-induced blood vessel sprouting. MT1-MMP activity in human microvascular endothelial cells was also inhibited. The findings identify hypericin as a potentially useful agent in the treatment of ophthalmic neovascularization pathogeneses.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Corneal Neovascularization/prevention & control , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Perylene/analogs & derivatives , Angiogenesis Inducing Agents , Angiogenesis Inhibitors/therapeutic use , Animals , Anthracenes , Cornea/blood supply , Corneal Neovascularization/chemically induced , Disease Models, Animal , Endothelium, Vascular/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2 , Genes, Tumor Suppressor , Humans , Iris/blood supply , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/antagonists & inhibitors , Nuclear Proteins , Perylene/pharmacology , Perylene/therapeutic use , Phosphorylation/drug effects , Rats , Rats, Wistar , Retinal Neovascularization/chemically induced , Retinal Neovascularization/prevention & control , Vascular Endothelial Growth Factor A/physiology
13.
Int J Oncol ; 27(4): 1097-103, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142328

ABSTRACT

Sigma receptors are present in cancer cell lines. The aim of the present study is to evaluate the anti-tumor activity of a series of sigma 1, sigma 2 and sigma 1/2 ligands in B16 melanoma cell lines. Proliferation, apoptosis, intracellular ATP content, cell cycle and molecular regulators were analyzed. Cell growth was determined using the sulforhodamine B (SRB) colorimetric cytotoxicity assay. Apoptosis was assessed by flow cytometry and DNA fragmentation, using ELISA cell death assay. ATP content was measured spectrofluorometrically and cell cycle analysis was performed by flow cytometry. The cytoplasmic and nuclear expression of cell cycle regulatory molecules, cyclin D and CDK2 (cyclin dependent kinase 2) were determined by Western blot analysis and quantified by densitometry. The sigma ligands in single digit micromolar concentrations inhibited B16 and multidrug-resistant B16 COL/R cell growth, leading to cell death at higher concentrations. The potency order was: haloperidol, reduced-haloperidol, ifenprodil tartrate, opipramol and carbetapentane citrate. B16 COL/R cells were to some extent, less sensitive to sigma ligands. Further studies have shown that the growth inhibitory effect of sigma ligands could be attributed to G1 arrest of the cell cycle, mediated by a marked decrease in cytoplasmic and nuclear cyclin D and CDK2 protein expression, though haloperidol induced loss of cell viability due to apoptosis. Sigma ligands induced an early decrease in ATP content. These data stimulated us to examine the combined anti-proliferative activity of haloperidol and the tyrosine kinase inhibitor imatinib mesylate (STI 571), on SK-MEL-28 human melanoma cells. Preliminary experiments demonstrated a marked synergistic interaction between the two agents.


Subject(s)
Haloperidol/pharmacology , Melanoma/drug therapy , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Benzamides , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cyclin D , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , Cytoplasm/metabolism , DNA Fragmentation , Densitometry , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Imatinib Mesylate , Ligands , Lipids , Melanoma/pathology , Melanoma, Experimental , Mice , Piperazines/pharmacology , Protein Binding , Pyrimidines/pharmacology , Rhodamines/pharmacology , Spectrometry, Fluorescence , Time Factors
14.
Curr Eye Res ; 30(4): 269-77, 2005 Apr.
Article in English | MEDLINE | ID: mdl-16020257

ABSTRACT

PURPOSE: The purpose of this study is to demonstrate feasibility of using our novel concept, termed competitive quenching, for protecting the choroidal extravascular compartment and retinal pigment epithelium (RPE) from verteporfin (VP)-induced phototoxicity using hypericin. Furthermore, we aim to achieve partitioning of the quencher, hypericin, in the extravascular space and VP within the microvascular compartment of the chorio-retinal complex in vivo. METHODS: We protect RPE cells from damage inflicted by photoactivated VP by introducing hypericin into these cells prior to photosensitization to quench the photosensitizing activity of VP. Cell protection levels were measured by MTT and Hemacolor viability assays. Wavelength range used for VP photoexcitation (700 +/- 40 nm) excludes the absorption range of hypericin, preventing the latter from photoactivation. Pharmacokinetic conditions, in which hypericin spreads throughout the choroidal and retinal extravascular space while VP is confined to the vasculature, are delineated using double-fluorescence imaging. RESULTS: Cell viability increased 3- to 5-fold when 10-20 microM hypericin were present in RPE cells during photosensitization with 0.1-0.5 microM VP. VP fluorescence intensity was unchanged by the presence of hypericin in the cells. Hypericin administered intravenously to rats was confined to the choroidal vasculature after 15 min to 2 hr. Subsequently, hypericin partitioned to the choroidal and retinal extravascular space. VP administered at this time was confined to the microvasculature. CONCLUSIONS: RPE and choroid may potentially be protected by compartmentalizing hypericin to the extravascular compartment while VP administered shortly before photosensitization is confined to the microvasculature. Adverse photodynamic therapy (PDT) damage to choroidal tissues adjacent to neovasculature targeted for photoablation have the potential of being prevented by competitive quenching with hypericin.


Subject(s)
Perylene/analogs & derivatives , Perylene/pharmacology , Photochemotherapy , Photosensitizing Agents/toxicity , Pigment Epithelium of Eye/drug effects , Porphyrins/toxicity , Animals , Anthracenes , Cell Line , Cell Survival , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Cytoprotection , Male , Oxidation-Reduction , Perylene/pharmacokinetics , Photosensitizing Agents/pharmacokinetics , Pigment Epithelium of Eye/pathology , Porphyrins/pharmacokinetics , Rabbits , Rats , Rats, Wistar , Retina/metabolism , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Verteporfin
15.
Int J Cancer ; 111(4): 596-603, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15239139

ABSTRACT

A unique property of the photodynamic signal transduction inhibitor hypericin (HY) is high functionality in the dark, which has been shown to result in portfolio of anticancer activities both in vitro and in vivo. Here we show that treatment with HY significantly reduces growth rate of metastases in 2 murine models: breast adenocarcinoma (DA3) and squamous cell carcinoma (SQ2). Focus on metastases was achieved by resection of primary tumors at stages in which micrometastases exist in lungs. Long-term animal survival in DA3 tumor-excised groups increased from 15.6% in controls to 34.5% following supplementary treatment with HY. In mice bearing SQ2 tumor metastases, therapy with HY increased animal survival from 17.7% in controls to 46.1%. Using Laser-induced fluorescence and multipixel spectral image analyses, we demonstrate that HY has a high tendency to accumulate in primary and metastatic tumors; HY content in lungs bearing metastases was approximately 2-fold higher than in the lungs of healthy animals. The tendency of HY to preferentially concentrate in lung metastases, combined with its potent antiproliferative activities, may render HY as a useful supplementary modality in the treatment of metastatic cancer irrespective of photoactivation.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Perylene/analogs & derivatives , Perylene/pharmacology , Perylene/pharmacokinetics , Animals , Anthracenes , Breast Neoplasms/veterinary , Carcinoma, Squamous Cell/veterinary , Disease Models, Animal , Female , Humans , Light , Lung Neoplasms/veterinary , Male , Mice , Mice, Inbred BALB C , Signal Transduction , Survival Analysis , Tissue Distribution
16.
Cancer Res ; 63(23): 8241-7, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678981

ABSTRACT

A unique property of the photodynamic signal transduction inhibitor hypericin is functionality in the dark. We show in tumor cells that hypericin targets the heat shock protein (Hsp) 90 chaperone but not Hsp70 (Hsc70) to enhanced ubiquitinylation. As a consequence Hsp90 chaperone functionality is abrogated and the client proteins, mutant p53, Cdk4, Raf-1, and Plk, are displaced from complexes with Hsp90, destabilized, and degraded via a proteasome-independent pathway. Decline in Raf-1 prevents downstream activation of extracellular signal-regulated kinase 1/2 kinases, the Ras/Raf pathway is inhibited, and tumor cell proliferation is arrested. The cells exhibit multiple aberrations including retardation at G(2)-M, increased cell volume, and multinucleation, all of which are hallmarks of mitotic cell death. The studies demonstrate that ubiquitinylation of Hsp90 inactivates the chaperone, destabilizes the plethora of client proteins, and creates deficiencies in multiple unrelated cellular functions. This combination constitutes a mechanism by which hypericin generates mitotic cell death in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Perylene/analogs & derivatives , Perylene/pharmacology , Ubiquitin/metabolism , Animals , Anthracenes , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Death/drug effects , Cell Death/physiology , Cell Division/drug effects , Cell Line, Tumor , Darkness , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mitosis/drug effects , Mitosis/physiology
17.
Photochem Photobiol ; 76(3): 335-40, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12403456

ABSTRACT

Wavelength effects in photodynamic therapy (PDT) with hypericin (HY) were examined in a C26 colon carcinoma model both in vitro and in vivo. Irradiation of HY-sensitized cells in vitro with either 550 or 590 nm caused the loss of cell viability in a drug- and light-dose-dependent manner. The calculated ratio of HY-based PDT (HY-PDT) efficiencies at these two wavelengths was found to correlate with the numerical ratio of absorbed photons at each wavelength. In vivo irradiation of C26-derived tumors, 6 h after intraperitoneal administration of HY (5 mg/kg), caused extensive vascular damage and tumor necrosis. The depth of tumor necrosis (d) was more pronounced at 590 than at 550 nm and increased when the light dose was raised from 60 to 120 J/cm2. The maximal depths of tumor necrosis (at 120 J/cm2) were 7.5+/-1.5 mm at 550 nm and 9.9+/-0.8 mm at 590 nm. Both values are rather high in view of the limited penetration of green-yellow light into the tissue. Moreover, the depth ratio, d590/d550 = 1.3 (P < 0.001), is smaller than expected considering the 2.2-fold lower HY absorbance and the 1.7-fold lower tissue penetration of radiation at 550 than at 590 nm. This finding indicates that in vivo the depth at which HY-PDT elicits tumor necrosis is not only determined by photophysical considerations (light penetration, number of absorbed photons) but is also influenced significantly by other mechanisms such as vascular effects. Therefore, despite the relatively short-wavelength peaks of absorption, our observations suggest that HY is an effective photodynamic agent that can be useful in the treatment of tumors with depths in the range of 1 cm.


Subject(s)
Models, Animal , Perylene/analogs & derivatives , Perylene/pharmacology , Photochemotherapy , Animals , Anthracenes , In Vitro Techniques , Mice , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...