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











Publication year range
1.
J Assist Reprod Genet ; 35(10): 1831-1841, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30043336

ABSTRACT

OBJECTIVE: To determine whether recombinant AMH (rAMH) could prevent post-transplant follicular depletion by acting on the stemness markers Oct-4, Sox2, and NANOG. MATERIALS AND METHODS: This was an experimental study where 12 ovariectomized nude mice were xenotransplanted with vitrified/warmed ovarian cortex obtained from a pre-pubertal girl and Alzet pumps delivering rAMH, or placebo (control), were inserted intra-abdominally. Previously vitrified/warmed ovarian cortex fragments were transplanted after 7 days and then harvested after 14 days from pump placement. We performed real-time RT-PCR analyses, ELISA for AMH, FSH, and estradiol, histologic measurement of ovarian follicles, and immunohistochemistry for Ki67 and TUNEL. The main outcome measures were serum levels and tissue expression of the parameters under investigation and follicle count. RESULTS: Serum AMH, FSH, and estradiol reflected post-ovariectomy profiles and were mildly influenced by rAMH administration. Ovarian cortex expression of AMH, AMH-R2, VEGF, GDF9, Oct-4, and Sox2 was lower in rAMH mice than in controls, while NANOG was upregulated. There was a non-significant decrease in primordial follicles after vitrification-warming, and xenotransplantation further decreased this number. There were lower cell replication and depressed apoptosis in the rAMH group. CONCLUSIONS: Administration of recombinant AMH in the peri-transplant period did not protect the initial follicular depletion but decreased apoptosis and cellular activation and regulated stem cell markers' tissue expression. These results aid our understanding of the inhibitory effects of AMH on follicular development and show the benefit of administering exogenous AMH at the time of pre-pubertal ovarian cortex transplant to protect the follicles from pre-activation and premature depletion.


Subject(s)
Anti-Mullerian Hormone/genetics , Heterografts/metabolism , Ovarian Follicle/transplantation , Ovary/transplantation , Animals , Anti-Mullerian Hormone/administration & dosage , Anti-Mullerian Hormone/blood , Apoptosis/genetics , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Developmental , Heterografts/drug effects , Heterografts/growth & development , Humans , Mice , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovariectomy , Ovary/drug effects , Ovary/growth & development , Ovary/metabolism , SOXB1 Transcription Factors/genetics , Transplantation, Heterologous , Vitrification
2.
Nucleic Acid Ther ; 23(6): 379-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24083396

ABSTRACT

Catalytic oligonucleotides, known as DNAzymes, are a new class of nucleic acid-based gene therapy that have recently been used in preclinical animal studies to treat various cancers. In this study the systemic distribution, pharmacokinetics, and safety of intravenously administered anti-MMP (matrix metalloproteinase)-9 DNAzyme (AM9D) were determined in healthy FVB and in MMTV-polyoma virus middle T (PyMT) transgenic mice bearing mammary tumors. MMP-9 is known to be involved in tumor cell development, angiogenesis, invasion, and metastasis. Sulfur-35 ((35)S) labeled ([(35)S]-AM9D) administered intravenously, without the use of carrier molecules, to healthy and mammary tumor bearing MMTV-PyMT transgenic mice distributed to all major organs. The order of percentages of [(35)S]-AM9D accumulation in different organs of healthy and MMTV-PyMT mice were blood>liver>kidney>lung>spleen>heart and mammary tumor>blood≈liver>kidney>spleen>lung>heart, respectively. The amount of AM9D accumulated in mammary tumors 2 hours post injection was 0.6% and 0.2% higher than in either blood or liver, respectively, and its rate of initial clearance from mammary tissue was at least 50% slower than the other organs. Approximately 43% of the delivered dosage of [(35)S]-AM9D was cleared from the system via feces and urine over a period of 72 hours. No evidence of acute or chronic cytotoxicity, local or widespread, associated with AM9D treatment (up to 75 mg AM9D /kg of body weight) was observed in the organs examined. These data suggest that DNAzyme in general and AM9D in particular can be used systemically as a therapeutic agent to treat patients with breast cancer or other metastatic and surgically inaccessible tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA, Catalytic/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Matrix Metalloproteinase 9/metabolism , Administration, Intravenous , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , DNA, Catalytic/pharmacokinetics , DNA, Catalytic/toxicity , Drug Screening Assays, Antitumor , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Polyomavirus , Tissue Distribution
3.
Steroids ; 76(1-2): 193-203, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21070794

ABSTRACT

We have synthesized 3ß,21-dihydroxypregna-5,7-dien-20-one (21(OH) 7DHP) and used UVB radiation to induce its photoconversion to analogues of vitamin D (pD), lumisterol (pL) and tachysterol (pT). The number and character of the products and the dynamics of the process were dependent on the UVB dose. The main products: pD and pT compounds were characterized by UV absorption, MS and NMR spectroscopy after RP-HPLC chromatography. In addition, formation of multiple oxidized derivatives of the primary products was detected and one of these derivatives was characterized as oxidized 21-hydroxyisotachysterol compound (21(OH)oxy-piT). These newly synthesized compounds inhibited growth of human melanoma cells in a dose dependent manner, with greater or equal potency to calcitriol. 3ß,21-Dihydroxy-9ß,10α-pregna-5,7-dien-20-one (21(OH)pL) and 21(OH)oxy-piT had higher potency against pigmented melanoma cells, while the EC(50) for compounds 21(OH)7DHP and (5Z,7E)-3ß,21-dihydroxy-9,10-secopregna-5,7,10(19)-trien-20-one (21(OH)pD) were similar in both pigmented and non-pigmented cells. Moreover, 21(OH)7DHP and its derivatives inhibited proliferation of human epidermal HaCaT keratinocytes, albeit at a lower activity compared to melanoma cells. Importantly, 21(OH)7DHP derivatives strongly inhibited the colony formation of human melanoma cells with 21(OH)pD being the most potent. The potential mechanism of action of newly synthesized compounds was similar to that mediated by 1,25(OH)(2)D(3) and involved ligand-induced translocation of vitamin D receptor into the nucleus. In summary, we have characterized for the first time products of UVB-induced conversion of 21(OH)7DHP and documented that these compounds have selective, inhibitory effects on melanoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Pregnadienediols/pharmacology , Secosteroids/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Melanoma/pathology , Molecular Conformation , Photochemistry , Pregnadienediols/chemical synthesis , Pregnadienediols/chemistry , Secosteroids/chemical synthesis , Secosteroids/chemistry , Stereoisomerism , Ultraviolet Rays
4.
Mol Cell Endocrinol ; 307(1-2): 211-6, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19409957

ABSTRACT

Since melatonin production has been documented in extrapineal and extraneuronal tissues, we investigated the expression of molecular elements of the melatoninergic system in human RPE cells (ARPE-19). The expression of key enzymes for melatonin synthesis: tryptophan hydroxylases (TPH1 and TPH2); arylalkylamine N-acetyltransferase (AANAT) and hydroxyindole-O-methyltransferase (HIOMT) was detected in ARPE-19 cells using RT-PCR. TPH1 and AANAT proteins were detected in ARPE by Western blotting, while sequential metabolism of tryptophan, serotonin and N-acetylserotonin to melatonin was shown by RP-HPLC. We also demonstrated, by means of RT-PCR, that ARPE expressed mRNA encoding the melatonin receptors: MT2 (but not MT1), two isoforms of nuclear receptor (RORalpha1 and RORalpha4/RZR1), and quinone oxidoreductase (NQO2). By analogy with other peripheral tissues, for example the skin, the expression of these metabolic elements in RPE cells suggests that the RPE represents an additional source of melatonin in the eye, to regulate local homeostasis and prevent from oxidative damage in intra-, auto- and/or paracrine fashions.


Subject(s)
Melatonin/biosynthesis , Retinal Pigment Epithelium/metabolism , Acetylserotonin O-Methyltransferase/genetics , Acetylserotonin O-Methyltransferase/metabolism , Arylalkylamine N-Acetyltransferase/genetics , Arylalkylamine N-Acetyltransferase/metabolism , Cell Line , Gene Expression Regulation , Humans , Receptors, Melatonin/genetics , Receptors, Melatonin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/analogs & derivatives , Serotonin/chemistry , Serotonin/metabolism , Tryptophan/chemistry , Tryptophan/metabolism , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
5.
Steroids ; 74(2): 218-28, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19028513

ABSTRACT

Pregna-5,7-dienes and their hydroxylated derivatives can be formed in vivo when there is a deficiency in 7-dehydrocholesterol (7-DHC) Delta-reductase function, e.g., Smith-Lemli-Opitz syndrome (SLOS). Ultraviolet B (UVB) radiation induces photoconversion of 7-DHC to vitamin D3, lumisterol3 and tachysterol3. Two epimers (20R and 20S) of pregna-5,7-diene-3beta,17alpha,20-triol (4R and 4S, respectively) were synthesized and their UVB photo-conversion products identified as corresponding 9,10-secosteroids with vitamin D-like and tachysterol-like structures, and 5,7-dienes with inverted configuration at C-9 and C-10 (lumisterol-like). The number and character of the products and the dynamics of the process were dependent on the UVB dose. At high UVB doses, the formation of multiple oxidized derivatives of the primary products, and the formation of 5,7,9(11)-triene, were observed. The production of vitamin D-like, tachysterol-like and lumisterol-like derivatives was also observed in human skin treated with 4R and 4S, and subjected to UV irradiation, as shown by RP-HPLC. Newly synthesized compounds inhibited melanoma growth in dose dependent manner, and some of them showed equal or higher potency than 1,25(OH)2D3. In summary, we have characterized for the first time the products of UV induced conversion of pregna-5,7-diene-3beta,17alpha,20-triols and documented that the newly synthesized compounds have antiproliferative properties against melanoma cells.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Photolysis/radiation effects , Pregnadienes/chemistry , Pregnadienes/pharmacology , Pregnadienetriols/chemistry , Pregnadienetriols/chemical synthesis , Pregnadienetriols/pharmacology , Acetylation , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Pregnadienes/chemical synthesis , Pregnadienes/metabolism , Pregnadienetriols/metabolism , Secosteroids/analysis , Secosteroids/chemistry , Skin/metabolism , Skin/radiation effects , Stereoisomerism , Ultraviolet Rays
6.
Photochem Photobiol Sci ; 7(12): 1570-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19037511

ABSTRACT

Calcitriol (3beta,5Z,7E)-9,10-secocholesta-5,7,10(19)-trien-1alpha,3beta,25-triol) is a powerful oncostatic form of vitamin D3 that is of limited clinical utility due to hypercalcemic (toxic) effects. Since the removal of the side chain reduces or eliminates the calcemic activity of vitamin D3, secosteroidal compounds lacking or with a shortened side chain are good candidates for anti-cancer drugs. In addition, 5,7-steroidal dienes without a side chain can be generated in vivo under pathological conditions. A series of androsta- and pregna-5,7-dienes was efficiently synthesized from their respective 3-acetylated 5-en precursors by bromination-dehydrobromination and deacetylation reactions. Ultraviolet B (UVB) irradiation was used to generate corresponding 9,10-secosteroids with vitamin D-like structures. Additional products with tachysterol-like (T-like) structures or 5,7-dienes with inverted configuration at C-9 and C-10 (lumisterol, L-like) were also detected. Different doses of UVB resulted in formation of various products. At low doses, previtamin D-, T- or L-like compounds were formed as the main products, while higher doses induced further isomerization, with formation of potentially oxidized derivatives. In summary, we describe dynamic UVB induced conversion of androsta- and pregna-5,7-dienes into vitamin D-like compounds and their rearranged analogues; additionally novel T-like and L-like structures were also produced and characterized. Further biological evaluation of newly synthesized compounds should help to select the best candidate(s) for potential treatment of hyperproliferative diseases including cancer.


Subject(s)
Androstadienes/chemistry , Pregnadienes/chemistry , Androstadienes/chemical synthesis , Androstadienes/radiation effects , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Molecular Conformation , Photolysis , Pregnadienes/chemical synthesis , Pregnadienes/radiation effects , Ultraviolet Rays
7.
J Invest Dermatol ; 128(9): 2271-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18368131

ABSTRACT

It has been shown that mammalian cytochrome P450scc can metabolize vitamin D3 to 20-hydroxyvitamin D3 (20(OH)D3) and 20,22(OH)2D3. To define the biological significance of this pathway, we tested the effects of 20(OH)D3 on the differentiation program of keratinocytes and on the expression of enzymes engaged in vitamin D3 metabolism. Immortalized HaCaT and adult human epidermal keratinocytes were used as a model and the effects of 20(OH)D3 were compared with those of 25(OH)D3 and 1,25(OH)2D3. 20(OH)D3 inhibited proliferation and caused G2/M arrest. 20(OH)D3 stimulated involucrin and inhibited cytokeratin 14 expression. The potency of 20(OH)D3 was comparable to that of 1,25(OH)2D3. 20(OH)D3 decreased the expression of cytochrome P450 enzyme (CYP)27A1 and CYP27B1, however, having only slight effect on CYP24. The effect of 20(OH)D3 was dependent on the vitamin D receptor (VDR). As shown by electrophoretic mobility shift assay, 20(OH)D3 stimulated the binding of nuclear proteins to the VDRE. Transfection of cells with VDR-specific siRNA decreased 20(OH)D3-stimulated transcriptional activity of the VDRE promoter and the expression of involucrin and CYP24 mRNA. Therefore, the above studies identify 20(OH)D3 as a biologically active secosteroid that induces keratinocyte differentiation. These data imply that the previously unreported pathway of vitamin D3 metabolism by P450scc may have wider biological implications depending, for example, on the extent of adrenal gland or cutaneous metabolism.


Subject(s)
Calcifediol/analogs & derivatives , Cell Differentiation/drug effects , Cholecalciferol/metabolism , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Adrenal Glands/metabolism , Calcifediol/metabolism , Calcifediol/pharmacology , Calcitriol/metabolism , Cell Differentiation/physiology , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cholestanetriol 26-Monooxygenase/metabolism , Humans , Hydroxylation , Keratin-14/metabolism , Protein Precursors/metabolism , Receptors, Calcitriol/metabolism , Skin/metabolism , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase
8.
J Pharmacol Exp Ther ; 323(2): 658-64, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17693586

ABSTRACT

N-Benzyladriamycin-14-valerate (AD 198) is one of several novel anthracycline protein kinase C (PKC)-activating agents developed in our laboratories that demonstrates cytotoxic superiority over doxorubicin (Adriamycin; DOX) through its circumvention of multiple mechanisms of drug resistance. This characteristic is attributed at least partly to the principal cellular action of AD 198: PKC activation through binding to the C1b (diacylglycerol binding) regulatory domain. A significant dose-limiting effect of DOX is chronic, dose-dependent, and often irreversible cardiotoxicity ascribed to the generation of reactive oxygen species (ROS) from the semiquinone ring structure of DOX. Despite the incorporation of the same ring structure in AD 198, we hypothesized that AD 198 might also be cardioprotective through its ability to activate PKC-epsilon, a key component of protective ischemic preconditioning in cardiomyocytes. Chronic administration of fractional LD(50) doses of DOX and AD 198 to mice results in histological evidence of dose-dependent ventricular damage by DOX but is largely absent from AD 198-treated mice. The absence of significant cardiotoxicity with AD 198 occurs despite the equal ability of DOX and AD 198 to generate ROS in primary mouse cardiomyocytes. Excised rodent hearts perfused with AD 198 prior to hypoxia induced by vascular occlusion are protected from functional impairment to an extent comparable to preconditioning ischemia. AD 198-mediated cardioprotection correlates with increased PKC-epsilon activation and is inhibited in hearts from PKC-epsilon knockout mice. These results suggest that, despite ROS production, the net cardiac effect of AD 198 is protection through activation of PKC-epsilon.


Subject(s)
Heart/drug effects , Protein Kinase C-epsilon/physiology , Animals , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Enzyme Activation , Female , Ischemic Preconditioning, Myocardial , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Ventricular Function, Left/drug effects
9.
Leuk Res ; 31(8): 1085-95, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17187856

ABSTRACT

Bcr-Abl activity in chronic myelogenous leukemia (CML) results in dysregulated cell proliferation and resistance against multiple cytotoxic agents due to the constitutive activation of proliferative signaling pathways. Currently, the most effective treatment of CML is the inhibition of Bcr-Abl activity by imatinib mesylate (Gleevec). Imatinib efficacy is limited by development of resistance through either expression of Bcr-Abl variants that bind imatinib less avidly, increased expression of Bcr-Abl, or expression of multidrug transport proteins. N-Benzyladriamycin-14-valerate (AD 198) is a novel antitumor PKC activating agent that triggers rapid apoptosis through PKC-delta activation and mitochondrial depolarization in a manner that is unaffected by Bcl-2 expression. We demonstrate that Bcr-Abl expression does not confer resistance to AD 198. Further, AD 198 rapidly induces Erk1/2 and STAT5 phosphorylation prior to cytochrome c release from mitochondria, indicating that proliferative pathways are active even as drug-treated cells undergo apoptosis. At sub-cytotoxic doses, AD 198 and its cellular metabolite, N-benzyladriamycin (AD 288) sensitize CML cells to imatinib through a supra-additive reduction in the level of Bcr-Abl protein expression. These results suggest that AD 198 is an effective treatment for CML both in combination with imatinib and alone against imatinib-resistant CML cells.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Fusion Proteins, bcr-abl/metabolism , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Benzamides , Cytochromes c/metabolism , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Drug Synergism , Fusion Proteins, bcr-abl/genetics , HL-60 Cells/drug effects , HL-60 Cells/metabolism , HL-60 Cells/pathology , Humans , Imatinib Mesylate , Immunoblotting , K562 Cells/drug effects , K562 Cells/metabolism , K562 Cells/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism
10.
Pediatr Dent ; 28(4): 357-62, 2006.
Article in English | MEDLINE | ID: mdl-16903446

ABSTRACT

PURPOSE: The purpose of this study was to examine intralingual (IL) and submucosal (SM) delivery offlumazenil as viable alternatives to immediate intravenous (IV) administration for reversing benzodiazepine sedation in an animal model. METHODS: A dog animal model was chosen based upon comparable body weight to children (12-17 kg) and the ease of oral access in this species. Research design was a nonrandomized matched pair study. This type of "before-and-after study" allowed the dogs to receive 3 different routes of flumazenil administration (IV, IL, and SM) following an initial dose of midazolam (0.5 mg/kg IV). Blood samples were obtained (at 0, 2, 4, 8, 15, and 30 minutes) for high performance liquid chromatography (HPLC) analysis of flumazenil and midazolam, and oxygen saturation values were recorded. RESULTS: Both IL and SM delivery of flumazenil were determined to be viable alternatives to immediate IV administration for reversing benzodiazepine-induced oxygen saturation (SaO2) desaturation. For flumazenil to be able to reverse the SaO2 desaturation, the plasma levels must be greater than 5 ng/ml, which was exceeded by IL and SM drug delivery. CONCLUSION: In a benzodiazepine-induced desaturation, the submucosal and intralingual routes are viable alternatives to intravenous administration of flumazenil in an animal model.


Subject(s)
Benzodiazepines/antagonists & inhibitors , Flumazenil/administration & dosage , GABA Modulators/administration & dosage , GABA Modulators/antagonists & inhibitors , Midazolam/antagonists & inhibitors , Oxygen/blood , Administration, Oral , Animals , Dogs , Injections, Intravenous , Male , Models, Animal
11.
FASEB J ; 20(9): 1564-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16793870

ABSTRACT

Melatonin, which can be produced in the skin, exerts a protective effect against damage induced by UV radiation (UVR). We have investigated the effect of UVB, the most damaging component of UVR, on melatonin metabolism in HaCaT keratinocytes and in a cell-free system. Four metabolites were identified by HPLC and LC-MS: 6-hydroxymelatonin, N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK), 2-hydroxymelatonin (the main intermediate between melatonin and AFMK), and 4-hydroxymelatonin. Concentrations of these photoproducts were directly proportional to UVR-dose and to melatonin substrate content, and their accumulation was time-dependent. The UVR-dependent increase of AFMK and 2-hydroxymelatonin was also detected in keratinocytes, where it was accompanied by simultaneous consumption of intracellular melatonin. Of note, melatonin and its two major metabolites, 2-hydroxymelatonin and AFMK, were also detected in untreated keratinocytes, neither irradiated nor preincubated with melatonin. Thus, intracellular melatonin metabolism is enhanced under exposure to UVR. The additional biological activity of these individual melatonin metabolites increases the spectrum of potential actions of the recently identified cutaneous melatoninergic system.


Subject(s)
Keratinocytes/metabolism , Keratinocytes/radiation effects , Melatonin/metabolism , Ultraviolet Rays , Anti-Bacterial Agents/pharmacology , Biological Transport/radiation effects , Cell Line , Cell-Free System , Dose-Response Relationship, Radiation , Humans , Keratinocytes/drug effects , Melatonin/radiation effects
12.
Anticancer Drugs ; 17(5): 495-502, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16702805

ABSTRACT

Unlike nuclear-targeted anthracyclines, the extranuclear-targeted doxorubicin congener, N-benzyladriamycin-14-valerate (AD 198), does not interfere with normal topoisomerase II activity, but binds to the C1b regulatory domain of conventional and novel isoforms of protein kinase C (PKC). The resulting interaction leads to enzyme activation and rapid apoptosis in a variety of mammalian cell lines through a pathway involving mitochondrial events such as membrane depolarization (Deltapsim) and cytochrome c release. Unlike other triggers of apoptosis, AD 198-mediated apoptosis is unimpeded by the expression of Bcl-2 and Bcl-XL. We have further examined AD 198-induced apoptosis in 32D.3 mouse myeloid cells to determine how the anti-apoptotic effects of Bcl-2 are circumvented. The PKC-delta inhibitor, rottlerin, and transfection with a transdominant-negative PKC-delta expression vector both inhibit AD 198 cytotoxicity through inhibition of Deltapsim and cytochrome c release. While the pan-caspase inhibitor Z-VAD-FMK blocks AD 198-induced PKC-delta cleavage, however, it does not inhibit Deltapsim and cytochrome c release, indicating that AD 198 induces PKC-delta holoenzyme activation to achieve apoptotic mitochondrial effects. AD 198-mediated Deltapsim and cytochrome c release are also unaffected by cellular treatment with either the mitochondrial permeability transition pore complex (PTPC) inhibitor cyclosporin A or the Ca chelators EGTA and BAPTA-AM. These results suggest that AD 198 activates PKC-delta holoenzyme, resulting in Deltapsim and cytochrome c release through a mechanism that is independent of both PTPC activation and Ca flux across the mitochondria. PTPC-independent mitochondrial activation by AD 198 is consistent with the inability of Bcl-2 and Bcl-XL expression to block AD 198-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Biological Transport/drug effects , Calcium/metabolism , Cell Line , Cytochromes c/metabolism , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Granulocyte Precursor Cells/drug effects , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/ultrastructure , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Mitochondrial Permeability Transition Pore , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism
13.
Mol Pharmacol ; 65(4): 1038-47, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15044634

ABSTRACT

Nuclear factor kappaB (NF-kappaB) has been implicated in inducible chemoresistance against anthracyclines. In an effort to improve the cytotoxicity of anthracyclines while reducing their cardiotoxic effects, we have developed a novel class of extranuclear-localizing 14-O-acylanthracyclines that bind to the phorbol ester/diacylglycerol-binding C1b domain of conventional and novel protein kinase C (PKC) isoforms, thereby promoting an apoptotic response. Because PKCs have been shown to be involved in NF-kappaB activation, in this report, we determined the mechanism of NF-kappaB activation by N-benzyladriamycin-14-valerate (AD 198) and N-benzyladriamycin-14-pivalate (AD 445), two novel 14-O-acylanthracylines. We show that the induction of NF-kappaB activity in response to drug treatment relies on the activation of PKC-delta and NF-kappaB-activating kinase (NAK), independent of ataxia telengectasia mutated and p53 activities. In turn, NAK activates the IKK complex through phosphorylation of the IKK-2 subunit. We find that neither NF-kappaB activation nor ectopic expression of Bcl-X(L) confers protection from AD 198-induced cell killing. Overall, our data indicate that activation of novel PKC isoforms by cytoplasmic-targeted 14-O-acylanthracyclines promotes an apoptotic response independent of DNA damage, which is unimpeded by inducible activation of NF-kappaB.


Subject(s)
Anthracyclines/pharmacology , NF-kappa B/metabolism , Protein Kinase C/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Line , Cytoplasm/drug effects , Cytoplasm/enzymology , DNA Damage , DNA-Binding Proteins , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Enzyme Activation , I-kappa B Kinase , Mice , Protein Kinase C-delta , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , bcl-X Protein
14.
Mol Cell Biol ; 24(5): 1823-35, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14966265

ABSTRACT

We have identified a novel pathway of ataxia telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNA-PK) signaling that results in nuclear factor kappaB (NF-kappaB) activation and chemoresistance in response to DNA damage. We show that the anthracycline doxorubicin (DOX) and its congener N-benzyladriamycin (AD 288) selectively activate ATM and DNA-PK, respectively. Both ATM and DNA-PK promote sequential activation of the mitogen-activated protein kinase (MAPK)/p90(rsk) signaling cascade in a p53-independent fashion. In turn, p90(rsk) interacts with the IkappaB kinase 2 (IKK-2) catalytic subunit of IKK, thereby inducing NF-kappaB activity and cell survival. Collectively, our findings suggest that distinct members of the phosphatidylinositol kinase family activate a common prosurvival MAPK/IKK/NF-kappaB pathway that opposes the apoptotic response following DNA damage.


Subject(s)
DNA Damage , DNA-Binding Proteins , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/physiology , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Survival , DNA-Activated Protein Kinase , Doxorubicin/analogs & derivatives , Doxorubicin/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , I-kappa B Kinase , Mice , NIH 3T3 Cells , Nuclear Proteins , Protein Subunits/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
15.
J Pediatr Pharmacol Ther ; 9(2): 126-32, 2004 Apr.
Article in English | MEDLINE | ID: mdl-23118699

ABSTRACT

OBJECTIVES: Intravenous Y-site administration of more than one medication through the same in-line catheter is a common practice used in the management of acute seizures. The objective of this study was to determine the compatibility of valproate sodium (Depacon(®); 2 or 20 mg/mL) with 13 medications that are frequently administered to manage seizures or are given to patients with an acute head injury who are at risk for developing post-traumatic epilepsy. METHODS: The study medications included atracurium, dexamethasone, diazepam, fosphenytoin, lorazepam, magnesium sulfate, mannitol, methyl-prednisolone, midazolam, pentobarbital, phenytoin, ranitidine, and thiopental. Equal volumes of valproate and each of the study drugs were admixed and immediately examined using several physiochemical criteria: Tyndall effect, color and pH change, gas evolution, and particle formation (HIAC/Royco liquid particle counter). Samples were also evaluated using HPLC analysis (C(18) column; methanol/tetrahydrofuran/ phosphate buffer; 44/1/55% v/v, at 1.5 mL/min; 50°C) with UV (190-400 nm) photodiode detection. The valproate peak (220 nm) was quantified by both peak area and height. Samples were analyzed within 5 minutes of admixture and were reassessed at 15 and 30 minutes. RESULTS: With the exception of diazepam, midazolam, and phenytoin, all of the remaining drugs were chemically compatible with valproate, both in 5% Dextrose Injection, USP(D5W) and in 0.9% Sodium Chloride Injection, USP (Normal Saline -NS). None of the compatible medications produced a significant pH change, discernible gas, particle formation, reduced valproate titer by HPLC analysis (coefficient of variability < 1.5%), or the temporal formation of unidentified UV absorbing (190-400 nm) peaks. CONCLUSIONS: Intravenous valproate is compatible with most agents employed in seizure management or used in patients at risk for seizures following head injury and is safe for concurrent Y-site drug administration.

16.
Mol Cancer Ther ; 1(7): 469-81, 2002 May.
Article in English | MEDLINE | ID: mdl-12479265

ABSTRACT

Bcl-2 inhibits apoptosis induced by numerous antitumor drugs, including doxorubicin and daunorubicin and is, thus, a major impediment to successful cancer chemotherapy. Here, we report the ability of a novel family of nonnuclear targeted anthracyclines to induce rapid apoptosis in cells despite Bcl-2 or Bcl-X(L) expression. Typified by N-benzyladriamycin-14-valerate (AD 198) and N-benzyladriamycin-14-pivalate (AD 445), this family of compounds binds to the C1 regulatory domain of protein kinase C (PKC), competitively inhibits phorbol ester binding in cell-free studies, and induces PKC translocation in intact cells. PKC-delta has an established role as a pro-apoptotic protein through the association of the holoenzyme or catalytic fragment with mitochondria. In proliferating 32D.3 myeloid cells, or in 32D.3 cells engineered to overexpress Bcl-2, substantial levels of PKC-delta are associated with mitochondria. However, after a 1-h exposure to 5 microM AD 198, cytochrome c release, caspase-3 activation, poly(ADP-ribose) polymerase (PARP) cleavage, PKC-delta cleavage, and DNA fragmentation are observed. Pretreatment of 32D.3 cells with the selective PKC-delta inhibitor, rottlerin, but not the general PKC inhibitor, GF 109203X, or PKC-alpha and -beta inhibitor, Gö 6976, delayed the 50% cell kill to >24 h for control and Bcl-2 overexpressing 32D.3 cells treated with 5 microM AD 198. Rottlerin delayed PKC-delta and PARP cleavage to >20 h post-drug exposure and also delayed mitochondrial membrane depolarization. In contrast, the pan-caspase inhibitor Z-Val-Ala-Asp-CH2F blocked PKC-delta and PARP cleavage, but not mitochondrial membrane depolarization. These results suggest that AD 198 induces mitochondrial-dependent apoptosis in 32D.3 cells by activating PKC-delta holoenzyme on mitochondria, which, in turn, overrides the antiapoptotic effects of Bcl-2.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Biological Transport , Caspase 3 , Caspases/metabolism , Cell Line , Cell Membrane/metabolism , Cell Survival , Cytochrome c Group/metabolism , DNA Fragmentation , Dose-Response Relationship, Drug , Doxorubicin/analogs & derivatives , Humans , Immunoblotting , Intracellular Membranes/metabolism , Isoenzymes/metabolism , Mice , Microscopy, Fluorescence , Mitochondria/metabolism , Models, Chemical , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase C-alpha , Protein Kinase C-delta , Protein Structure, Tertiary , Signal Transduction , Subcellular Fractions , Time Factors , Transfection , Tumor Cells, Cultured
17.
Mol Cancer Ther ; 1(7): 483-92, 2002 May.
Article in English | MEDLINE | ID: mdl-12479266

ABSTRACT

Anthracycline antibiotics like doxorubicin (DOX) are known to exert their antitumor effects primarily via DNA intercalation and topoisomerase II inhibition. By contrast, the noncross-resistant cytoplasmically localizing DOX analogue, N-benzyladriamycin-14-valerate (AD 198), only weakly binds DNA and does not inhibit topoisomerase II, yet it displays superior antitumor activity, strongly suggesting a distinct cytotoxic mechanism. In recent modeling studies, we reported a structural similarity between AD 198 and commonly accepted ligands for the C1-domain of protein kinase C (PKC), and we hypothesized that the unique biological activity of AD 198 may derive, in part, through this kinase. Consistent with this hypothesis, the present biochemical studies demonstrate that AD 198 competes with [3H]phorbol-12,13-dibutyrate ([3H]PDBu) for binding to phorbol-responsive PKC isoforms, the isolated C1b domain of PKC-delta (delta C1b), and the nonkinase phorbol ester receptor, beta2-chimaerin. In NIH/3T3 cells, AD 198 competitively blocks PKC activation by C1-ligands. Importantly, neither DOX nor N-benzyladriamycin, the principal AD 198 metabolite, inhibits basal or phorbol-stimulated PKC activity or appreciably competes for [3H]PDBu binding. In CEM cells, structure activity studies with 14-acyl congeners indicate that the rapid induction of apoptosis correlates with competition for [3H]PDBu binding, strongly implicating phorbol-binding proteins in drug activity. Collectively, these studies support the conclusion that AD 198 is a C1-ligand and that C1-ligand receptors are selective drug targets. These studies provide the impetus for continuing efforts to understand the molecular basis for the unique biological activity of AD 198 and provide for the design of analogues with improved affinity for C1-domains and potentially greater antitumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Protein Kinase C/chemistry , 3T3 Cells , Amino Acid Sequence , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Doxorubicin/analogs & derivatives , Kinetics , Mice , Models, Chemical , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Isoforms , Protein Kinase C/metabolism , Protein Structure, Tertiary , Rats , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL