Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Dev Neurosci ; 34(6): 525-32, 2012.
Article in English | MEDLINE | ID: mdl-23428405

ABSTRACT

In a rat model of perinatal inflammation and hypoxia, we investigated the impact of hyperthermia on the deleterious events which are commonly associated with chorioamnionitis. Late-pregnancy gestational day 20 rats received a single injection of either lipopolysaccharide (LPS) Escherichia coli endotoxin or saline. The offspring were born 24-36 h later at full term. The pups underwent hypoxia on the first postnatal day (PND1) immediately after which they were maintained at a planned target temperature for 2 h, before being returned to the dams. The pups were sacrificed on PND5 and the brain tissue was examined. Results showed that LPS alone or in combination with hypoxia was well tolerated. The additional stress of moderate hyperthermia (39°C for 2 h) on PND1 resulted in (a) a significant increase in brain reactive nitrogen species (RNS), (b) a significant increase in caspase-3 activity, (c) a significant increase in c-jun, bax and bcl-2 gene expression and (d) a significant increase in apoptotic cells in the CA1 region of the hippocampus. Hyperthermia was also associated with reduced growth over the ensuing 4 days in a small number of pups. In this model of perinatal inflammation, we demonstrated that brief hyperthermia when superimposed on a perinatal inflammation stimulus and hypoxia led to brain injury while either inflammation alone, or combined inflammatory stimulus and hypoxia did not cause significant damage.


Subject(s)
Brain Injuries/etiology , Fever/complications , Hypoxia-Ischemia, Brain/complications , Inflammation/complications , Animals , Brain Injuries/physiopathology , Disease Models, Animal , Female , Hypoxia-Ischemia, Brain/physiopathology , Immunohistochemistry , In Situ Nick-End Labeling , Lipopolysaccharides/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
2.
Amyloid ; 8(3): 182-93, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11676295

ABSTRACT

Fibril formation is the basis of amyloid production in a number of disease states, such as Alzheimer's disease, diabetes and immunocytic dyscrasias. Compounds that inhibit fibril formation could be directly relevant to the treatment of amyloid diseases, and may also provide a foundation for the development of interventions in other molecular condensation diseases ranging from sickle cell anemia to atherosclerosis. We developed an economical and convenient high-throughput method for screening compounds against fibril formation in microwell plates. Chalcones, flavonoids and biflavonoids were screened against fibril formation by a recombinant antibody variable domain (V1). Chalcones 6 and 14 were found to demonstrate inhibition at 0.1 microM in 79 microM of protein solution in both test tube and microwell plate assays. The concentration of protein in the microwell plate assay could be as low as 5 microM using ThT as a monitoring agent. Molecular modeling studies indicated that both compounds could be individually docked into a binding site at the monomer-monomer interface of the V(L) protein dimer. These studies suggested that these compounds could potentially stabilize the VL dimer and therefore reduce its tendency to form fibrils. These findings may provide the basis for a new therapeutic approach to prevent or treat amyloid diseases.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Chalcone/pharmacology , Drug Evaluation, Preclinical/methods , Flavonoids/pharmacology , Immunoglobulin Variable Region/metabolism , Binding Sites , Chalcone/metabolism , Dimerization , Flavonoids/metabolism , Humans , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
3.
Bioorg Med Chem Lett ; 11(16): 2101-4, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11514148

ABSTRACT

A series of naturally occurring and synthetic biflavonoids was evaluated for inhibitory activity against Mycobacterium tuberculosis H37Rv (Mtb). Compounds 6, 24, and 25 demonstrated 96, 95, and 87% inhibition, respectively, at a screening concentration of 12.5 microg/mL. The type of linkage and the presence of methoxy- and nitro-substituents in biflavonoids may contribute to the observed inhibitory activity. The results of this study represent the discovery of biflavonoids as a potential new class of antituberculosis agent.


Subject(s)
Antitubercular Agents/pharmacology , Flavonoids/pharmacology , Mycobacterium tuberculosis/drug effects , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Flavonoids/chemical synthesis , Flavonoids/chemistry , Microbial Sensitivity Tests
4.
Oncogene ; 20(24): 3028-38, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11420718

ABSTRACT

We are studying the mechanisms of transcriptional activation by nuclear receptors and we focus our studies on the glucocorticoid regulation of the model tyrosine aminotransferase gene. Rather than using in vitro biochemical approaches, we determine the actual events occurring in the cells. Our experimental approaches include genomic footprinting, chromatin immunoprecipitation, in situ hybridization and transgenic mice. Our results show that the glucocorticoid receptor uses a dynamic multistep mechanism to recruit successively accessory DNA binding proteins that assist in the activation process. Chromatin is first remodelled, DNA is then demethylated, and the synthesis of an accessory factor is induced. Efficient transcription induction is finally achieved upon the formation of a 'stable' multiprotein complex interacting with the regulatory element. We discuss: the relative contribution of histone acetyltransferases and ATP-dependent remodelling machines to the chromatin remodelling event; the nature of the remodelled state; the contribution of regulated DNA demethylation to gene memory during development; the mechanisms of regulated DNA demethylation; the dynamics of protein recruitment at regulatory elements; the control of the frequency of transcription pulses and the control levels of the cell-type specificity of the glucocorticoid response.


Subject(s)
Receptors, Glucocorticoid/physiology , Tyrosine Transaminase/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , Humans , Models, Biological , Transcriptional Activation , Tyrosine Transaminase/metabolism
5.
EMBO J ; 20(8): 1974-83, 2001 Apr 17.
Article in English | MEDLINE | ID: mdl-11296230

ABSTRACT

Glucocorticoid hormones were found to regulate DNA demethylation within a key enhancer of the rat liver-specific tyrosine aminotransferase (Tat) gene. Genomic footprinting analysis shows that the glucocorticoid receptor uses local DNA demethylation as one of several steps to recruit transcription factors in hepatoma cells. Demethylation occurs within 2-3 days following rapid (< 1 h) chromatin remodeling and recruitment of a first transcription factor, HNF-3. Upon demethylation, two additional transcription factors are recruited when chromatin is remodeled. In contrast to chromatin remodeling, the demethylation is stable following hormone withdrawal. As a stronger subsequent glucocorticoid response is observed, demethylation appears to provide memory of the first stimulation. During development, this demethylation occurs before birth, at a stage where the Tat gene is not yet inducible, and it could thus prepare the enhancer for subsequent stimulation by hypoglycemia at birth. In vitro cultures of fetal hepatocytes recapitulate the regulation analyzed in hepatoma cells. There fore, demethylation appears to contribute to the fine-tuning of the enhancer and to the memorization of a regulatory event during development.


Subject(s)
Chromatin/metabolism , DNA Methylation , Dexamethasone/pharmacology , Gene Expression Regulation , Glucocorticoids/pharmacology , Liver/drug effects , Receptors, Glucocorticoid/genetics , Tyrosine Transaminase/genetics , 5-Methylcytosine , Animals , Cytosine/analogs & derivatives , Cytosine/metabolism , Enhancer Elements, Genetic , Gene Silencing , Liver/cytology , Liver/embryology , Liver/pathology , Rats , Transcription Factors/metabolism , Tumor Cells, Cultured
6.
Antimicrob Agents Chemother ; 45(5): 1379-86, 2001 May.
Article in English | MEDLINE | ID: mdl-11302799

ABSTRACT

(+)-Calanolide A is a novel, naturally occurring, nonnucleoside inhibitor of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase first isolated from a tropical tree (Calophyllum lanigerum) in the Malaysian rain forest. Previous studies have demonstrated that (+)-calanolide A has specific activity against the reverse transcriptase of HIV-1 and a favorable safety profile in animals. In addition, (+)-calanolide A exhibits a unique HIV-1 resistance profile in vitro. The safety and pharmacokinetics of (+)-calanolide A was examined in four successive single-dose cohorts (200, 400, 600, and 800 mg) in healthy, HIV-negative volunteers. In this initial phase I study, the toxicity of (+)-calanolide A was minimal in the 47 subjects treated. Dizziness, taste perversion, headache, eructation, and nausea were the most frequently reported adverse events. These events were not all judged to be related to study medication nor were they dose related. While 51% of subjects reported mild and transient dizziness, in many cases this appeared to be temporally related to phlebotomy. Calculation of the terminal-phase half-life (t(1/2)) was precluded by intrasubject variability in the 200-, 400-, and 600-mg dose cohorts but was approximately 20 h for the 800-mg dose group. (+)-Calanolide A was rapidly absorbed following administration, with time to maximum concentration of drug in plasma (T(max)) values occurring between 2.4 and 5.2 h postdosing depending on the dose. Plasma levels of (+)-calanolide A at all dosing levels were quite variable; however, both the mean concentration in plasma (C(max)), and the area under the plasma concentration-time curve increased proportionately in relation to the dose. Although raw plasma drug levels were higher in women than in men, when doses were normalized for body mass, the pharmacokinetic profiles were virtually identical with those observed for males. In general, levels of (+)-calanolide A in human plasma were higher than would have been predicted from animal studies, yet the safety profile remained benign. In conclusion, this study demonstrated the safety and favorable pharmacokinetic profile of single doses of (+)-calanolide A in healthy, HIV-negative individuals.


Subject(s)
Coumarins/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Adolescent , Adult , Age Factors , Aged , Area Under Curve , Cohort Studies , Coumarins/adverse effects , Female , Humans , Male , Middle Aged , Pyranocoumarins , Reverse Transcriptase Inhibitors/adverse effects
7.
J Neurosci Res ; 63(5): 388-94, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11223913

ABSTRACT

We have previously shown that tissue plasminogen activator (tPA) participates in the neurotoxicity of microglial conditioned medium (MgCM). Killing of hippocampal neurons by MgCM was prevented by both plasminogen activator inhibitor-1 (PAI-1) and anti-tPA antibody. An N-methyl-D-aspartate (NMDA) receptor blocker protected neurons from MgCM, suggesting that this subtype of glutamate receptor is involved. Whereas glutamate receptor-mediated events are important in cerebral ischemia and tPA has previously been shown to enhance excitotoxicity in hippocampus, we hypothesized that tPA would exaggerate oxygen glucose deprivation (OGD) injury in cultures of hippocampal neurons. Dissociated rat hippocampal cells were grown under conditions designed to optimize neuronal growth while minimizing glial replication. At 7--10 days, cultures were subjected to OGD for 2.5 hr. Recombinant human tPA (1,000 IU) was added immediately after OGD. Viability was assessed 24 hr later. Viable, apoptotic, and necrotic cells were classified and quantified based on staining patterns of acridine orange and ethidium bromide under fluorescence microscopy. tPA alone did not alter neuronal integrity. OGD produced significant neuronal death (viability reduced by 45%, P < 0.001). tPA completely protected OGD-exposed cultures. Potential mechanisms of tPA protection were explored. Whereas tPA antibody abolished the protective effect of tPA, its proteolytic inhibitor PAI-1 did not alter the effect. The effect of tPA was tested in separate free radical and excitatory amino acid insults. It did not protect neurons from hydrogen peroxide (1 microM), S-nitro-acetylpenicillamine (10 microM), glutamate (50 microM), or NMDA (10 microM) damage but significantly attenuated injury caused by 250 microM kainate. We conclude that tPA is capable of protecting hippocampal neurons from OGD by a nonproteolytic action. The mechanism of protection was not defined, although attenuation of AMPA/kainate glutamate receptors may play a role.


Subject(s)
Glucose/pharmacology , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Oxygen/pharmacology , Tissue Plasminogen Activator/pharmacology , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Cell Hypoxia , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media, Conditioned/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Fibrinolysin/metabolism , Glutamic Acid/pharmacology , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hydrogen Peroxide/pharmacology , Kainic Acid/pharmacology , Microglia/metabolism , Microscopy, Fluorescence , N-Methylaspartate/pharmacology , Nitric Oxide Donors/pharmacology , Oxidants/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Tissue Plasminogen Activator/immunology
8.
J Chromatogr B Biomed Sci Appl ; 742(2): 267-75, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10901131

ABSTRACT

A HPLC method was validated for quantification of (+)-calanolide A (1), a novel anti-HIV agent, in rat, dog and human plasma. The synthetic intermediate (+/-)-12-oxocalanolide A (2) was found to be a suitable internal standard. Compounds were extracted from plasma using a solid-phase C(18) cartridge and quantified over the assay range of 12.5 to 800 ng/ml. The method was utilized to determine (+)-calanolide A pharmacokinetics in rats, dogs and humans. This is the first report of a validated HPLC assay for determination of (+)-calanolide A concentrations in rat and dog plasma as well as human plasma obtained from clinical trials. There was no evidence of in vivo epimerization of (+)-calanolide A to its inactive epimer (+)-calanolide B (3).


Subject(s)
Anti-HIV Agents/blood , Chromatography, High Pressure Liquid/methods , Coumarins/blood , Administration, Oral , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Cohort Studies , Coumarins/administration & dosage , Coumarins/pharmacokinetics , Dogs , Female , HIV Seronegativity , Humans , Male , Pyranocoumarins , Rats , Reference Standards , Reproducibility of Results , Spectrometry, Fluorescence
9.
Neurosci Lett ; 285(3): 177-80, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10806315

ABSTRACT

Selective vulnerability of hippocampal neurons to ischemia is felt to relate to intense glutamatergic input and glutamate receptor expression. Since astrocytes are thought to have a neuroprotective role we speculated that hippocampal astrocyte sensitivity to insult could also contribute to this regional vulnerability. The purpose of the study was to determine if there is a differential sensitivity of cultured hippocampal and cortical astrocytes to oxygen-glucose deprivation (OGD). Hippocampal and cortical astrocytes were grown to confluence at matching cell density. Cultures were exposed to OGD for 2, 4, 6, 8 and 10 h. Progressive reduction in viability occurred in hippocampal astrocytes beginning at 2 h OGD. Reduction in cortical astrocyte viability was not observed until 4 h OGD. Death of hippocampal astrocytes was significantly greater than that of cortical astrocytes at each period of OGD. Based on acid phosphatase activity data the LD(50) for OGD duration in hippocampal astrocytes was 2 h compared to 8 h in cortical astrocytes. Regional differences in sensitivity of astrocytes to OGD implies that this may contribute to regional differences in neuronal vulnerability to ischemia.


Subject(s)
Astrocytes/physiology , Cerebral Cortex/cytology , Glucose/deficiency , Hippocampus/cytology , Acid Phosphatase/metabolism , Animals , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/metabolism , Hippocampus/metabolism , L-Lactate Dehydrogenase/metabolism , Rats , Rats, Sprague-Dawley
10.
Glia ; 29(4): 347-54, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10652444

ABSTRACT

Several CNS disorders feature microglial activation. Microglia are known to have both restorative and cytotoxic capabilities. Neuronal apoptosis has been noted after an acute insult such as ischemia. Microglia may participate in this event. We previously showed that conditioned medium (CM) harvested from peritoneal macrophages or from activated microglia triggered apoptosis in rat hippocampal neurons in culture. We wished to characterize the factor responsible for triggering neuronal death. Quiescent microglia produced CM that did not disrupt hippocampal neurons. Lipopolysaccharide-activated microglia produced CM which resulted in neuronal death. This effect was blocked by plasminogen activator inhibitor-1, by tPA STOP, and by co-incubation with tPA antibody. Recombinant human tPA exaggerated the neurotoxic effects of microglial CM, while tPA alone was toxic only at very high concentrations. This in vitro system, which probably excludes any significant impact of microglial free radicals, suggests that microglial tPA may contribute significantly to hippocampal neuronal death.


Subject(s)
Apoptosis/drug effects , Microglia/chemistry , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Tissue Plasminogen Activator/pharmacology , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , Electrophoresis, Polyacrylamide Gel , Hippocampus/cytology , Humans , In Situ Nick-End Labeling , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microtubule-Associated Proteins/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Precipitin Tests , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/pharmacology , Tissue Plasminogen Activator/immunology
11.
Antivir Chem Chemother ; 11(1): 23-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10693651

ABSTRACT

Plant-derived and semi-synthetic calanolide compounds with anti-human immunodeficiency virus type 1 (HIV-1) activity were tested for anti-human cytomegalovirus (HCMV) activity in both cytopathic effect inhibition and plaque reduction assays. The results indicated that the anti-HCMV activity of calanolide compounds does not correlate with their activity against HIV-1. The semi-synthetic 12-keto derivatives tended to be more active against HCMV than the corresponding 12-OH congeners, which were more active against HIV-1. It appeared that the 7,8-unsaturated double bond in the chromene ring played a certain role in maintaining activities against both HCMV and HIV-1. Saturation of the double bond increased the EC50 values against both viruses, with concomitant increase in toxicity. The calanolide compounds reported here are the first non-nucleoside analogues capable of inhibiting both HIV-1 and HCMV and, therefore, may be useful chemoprophylactic agents for HCMV in HIV-infected people or vice versa.


Subject(s)
Anti-HIV Agents/pharmacology , Coumarins/pharmacology , Cytomegalovirus/drug effects , HIV-1/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/isolation & purification , Cells, Cultured , Coumarins/chemical synthesis , Coumarins/isolation & purification , Cytopathogenic Effect, Viral/drug effects , HIV-1/growth & development , HIV-1/pathogenicity , Humans , Microbial Sensitivity Tests , Pyranocoumarins , Viral Plaque Assay
12.
Antivir Chem Chemother ; 11(5): 321-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11142630

ABSTRACT

The natural product (+)-calanolide A, a unique non-nucleoside reverse transcriptase inhibitor (NNRTI) of HIV-1 replication, is currently being evaluated in clinical trials in the USA. (+)-Calanolide A, the congeners costatolide and dihydrocostatolide, and (+)-12-oxo(+)-calanolide A, were evaluated in combination with a variety of mechanically diverse inhibitors of HIV replication to define the efficacy and cellular toxicity of potential clinical drug combinations. These assays should be useful in prioritizing the use of different combination drug strategies in a clinical setting. The calanolides exhibited synergistic antiviral interactions with other nucleoside and non-nucleoside reverse transcriptase inhibitors and protease inhibitors. Additive interactions were also observed when the calanolides were used with representative compounds from each of these classes of inhibitors. No evidence of either combination toxicity or antagonistic antiviral activity was detected with any of the tested compounds. The combination antiviral efficacy of three-drug combinations involving the calanolides, and the efficacy of two- and three-drug combinations using a (+)-calanolide A-resistant challenge virus (bearing the T139I amino acid change in the reverse transcriptase), was also evaluated in vitro. These assays suggest that the best combination of agents based on in vitro anti-HIV assay results would include the calanolides in combination with lamivudine and nelfinavir, since this was the only three-drug combination exhibiting a significant level of synergy. Combination assays with the (+)-calanolide A-resistant strain yielded identical results as seen with the wild-type virus, although the concentration of the calanolides had to be increased.


Subject(s)
Antiviral Agents/pharmacology , Coumarins/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Cell Line , Drug Therapy, Combination , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Pyranocoumarins , Virus Replication/drug effects
13.
Curr Opin Drug Discov Devel ; 3(2): 155-66, 2000 Mar.
Article in English | MEDLINE | ID: mdl-19649847

ABSTRACT

Both naturally occurring and semi-synthetic calanolide compounds are potent anti-human immunodeficiency virus (HIV) agents. In fresh human cells, they are highly effective inhibitors against low passage clinical virus strains, including those representative of the various HIV-1 clade strains (A through F), syncytium-inducing (SI) and non-syncytium-inducing (NSI) isolates, and T-tropic and monocyte-tropic isolates. These compounds also exhibit an enhanced antiviral activity against one of the most prevalent non-nucleoside reverse transcriptase inhibitor (NNRTI)-resistant viruses that is engendered by the Y181C amino acid change in reverse transcriptase (RT). Further enhancement of activity is observed with RTs that possess the Y181C change together with AZT-resistant mutations. Moreover, when challenged with viruses containing Y181C and K103N dual mutations, calanolide compounds remain active. These dual mutations are highly resistant to all approved NNRTIs (eg, delavirdine, nevirapine and efavirenz). In cell culture assays, calanolide compounds, especially (+)-calanolide A, select primarily resistant viruses possessing the T139I amino acid change. This mutation appears to be unique to calanolides since it remains susceptible to other NNRTIs. Synergistic effects are observed in both cultured cells and animal models when calanolides are used in combination with other anti-HIV agents. Enzymatic analyses indicate that calanolides inhibit HIV-1 RT through a mechanism that affects both the Km for normal substrate dTTP and the Vmax, resulting in a mixed-type inhibition, which is different from that of other known NNRTIs. Two possible binding modes/sites at the HIV-1 RT enzyme have been suggested for (+)-calanolide A. Taken together, the calanolide compounds represent a novel and distinct subgroup of the NNRTI family and inclusion of a calanolide in a combination therapy may be clinically beneficial. Of particular interest is the use of calanolide in the treatment of patients who have failed other NNRTI therapy and developed the Y181C mutation or the Y181C/K103N dual mutations. Currently, (+)-calanolide A, the most potent in the series of calanolide compounds, is undergoing clinical investigation for safety and efficacy in HIV-infected individuals.

14.
Biopolymers ; 50(7): 679-89, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10547523

ABSTRACT

We have previously observed that double-stranded DNA fragments containing a tract of the tandemly repeated sequence poly(CA). poly(TG) can associate in vitro to form stable complexes of low electrophoretic mobility, which are recognized with high specificity by proteins HMG1 and HMG2. The formation of such complexes has since been observed to depend on interactions of DNA with polypropylene surfaces, with the suggestion that the formation of low mobility complexes might be the result of strand dissociation followed by misaligned reassociation of the repetitive sequences. The data presented here show that at high ionic strength the interactions of DNA with polypropylene are sufficiently strong for DNA to remain bound to the polypropylene surface, which suggests that DNA might also be involved in interactions with hydrophobic molecules in vivo. Under such conditions, low-mobility complexes are found only in the material adsorbed to the polypropylene surface, and all DNA fragments are able to form low-mobility structures, whether or not they contain repetitive sequences. Preventing the separation of strands by ligating hairpin loop oligonucleotides at both ends of the fragments does not prevent the formation of low-mobility complexes. Our results suggest two different pathways for the formation of complexes. In the first, dissociation is followed by misaligned reassociation of repetitive sequences, yielding duplexes with single-stranded end regions that associate to form multimeric complexes. In the second, repetitive as well as nonrepetitive DNA molecules bound to polypropylene adopt a conformation with locally unwound regions, which allows interactions between neighboring duplexes adsorbed on the surface, resulting in the formation of low-mobility complexes.


Subject(s)
DNA/chemistry , Polypropylenes/chemistry , DNA/ultrastructure , Escherichia coli/chemistry , Nucleic Acid Conformation , Plasmids , Poly A/chemistry , Poly C/chemistry , Poly G/chemistry , Poly T/chemistry
15.
J Neurosci Res ; 56(1): 54-9, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10213475

ABSTRACT

We recently demonstrated that conditioned medium (CM) from peritoneal macrophages or activated microglia triggers a predominantly apoptotic death in hippocampal neurons in culture. We tested the effects of propentofylline (ppf), an agent that is neuroprotective in focal ischemia and is also associated with reduced microglial antigen expression after insult. Ppf had no impact on the secretion of neurotoxin from microglia. However, ppf significantly attenuated the effects of macrophage and microglial conditioned medium on neurons. Ppf did not attenuate neuronal hypoxic injury but did reverse the exaggeration of hypoxic injury exerted by subsequent addition of macrophage CM. A1 and A2 adenosine receptor inhibitors and an inhibitor of adenosine uptake each mimicked the effect of ppf. Neither ATP nor a deaminase inhibitor blocked the effect of microglial CM. These findings may be relevant to the neuroprotective effects of ppf in ischemia and dementia.


Subject(s)
Hippocampus/cytology , Macrophages, Peritoneal/physiology , Microglia/physiology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Xanthines/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine/physiology , Adenosine Triphosphate/pharmacology , Animals , Cell Death/drug effects , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned , Fetus , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Microglia/cytology , Microglia/metabolism , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley
16.
Planta Med ; 65(2): 120-5, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10193201

ABSTRACT

Biflavonoids such as amentoflavone (1), agathisflavone (2), robustaflavone (3), hinokiflavone (4), volkensiflavone (5), rhusflavanone (7), succedaneflavanone (9), all isolated from Rhus succedanea and Garcinia multiflora, as well as their methyl ethers and acetates, volkensiflavone hexamethyl ether (6), rhusflavanone hexaacetate (8), and succedaneflavanone hexaacetate (10) were evaluated for their antiviral activities. The inhibitory activities against a number of viruses including respiratory viruses (influenza A, influenza B, respiratory syncytial, parainfluenza type 3, adenovirus type 5, and measles) and herpes viruses (HSV-1, HSV-2, HCMV, and VZV) were investigated. The results indicated that robustaflavone exhibited strong inhibitory effects against influenza A and influenza B viruses with EC50 values of 2.0 micrograms/ml and 0.2 microgram/ml, respectively, and selectivity index values (SI) of 16 and 454, respectively. Amentoflavone and agathisflavone also demonstrated significant activity against influenza A and B viruses. Amentoflavone and robustaflavone exhibited moderate anti-HSV-1 anti-HSV-2 activities with EC50 values of 17.9 micrograms/ml (HSV-1) and 48.0 micrograms/ml (HSV-2) and SI values of > 5.6 (HSV-1) and > 2.1 (HSV-2) for amentoflavone; EC50 values of 8.6 micrograms/ml (HSV-1) and 8.5 micrograms/ml (HSV-2), and SI values of > 11.6 (HSV-1) and > 11.8 (HSV-2) for robustaflavone. Rhusflavanone demonstrated inhibitory activities against influenza B, measles, and HSV-2 viruses with SI values of 9.3, 8 and > 6.4, respectively. Succedaneaflavanone exhibited inhibitory activities against influenza B virus and VZV with SI values of 15 and < 3.0, respectively.


Subject(s)
Antiviral Agents/pharmacology , Flavonoids/pharmacology , Antiviral Agents/isolation & purification , Cell Line , Flavonoids/isolation & purification , Humans , Microbial Sensitivity Tests , Plants/chemistry , Viruses/drug effects
17.
Bioorg Med Chem Lett ; 9(2): 133-8, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-10021914

ABSTRACT

In vivo anti-HIV efficacy of (+)-calanolide A has been evaluated in a hollow fiber mouse model. It was demonstrated that the compound was capable of suppressing virus replication in two distinct and separate physiologic compartments (i.p. and s.c.) following oral or parenteral administration on a once- or twice-daily treatment schedule. A synergistic effect was observed for the combination of (+)-calanolide A and AZT.


Subject(s)
Anti-HIV Agents/pharmacology , Coumarins/pharmacology , Animals , Cell Line , Cell Survival , Cells, Cultured , Coumarins/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , HIV Core Protein p24/blood , HIV Core Protein p24/drug effects , HIV Reverse Transcriptase/drug effects , HIV Reverse Transcriptase/metabolism , Humans , Macrophages/drug effects , Mice , Pyranocoumarins , Time Factors , Zidovudine/pharmacology
18.
Antivir Ther ; 4(4): 203-9, 1999.
Article in English | MEDLINE | ID: mdl-10723499

ABSTRACT

We have tested both wild-type and drug-resistant mutated, recombinant human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) molecules for sensitivity to each of two non-nucleoside RT inhibitors (NNRTI), (+)-calanolide A and nevirapine, in primer extension assays. We found that RT containing either the V106A or Y181C substitutions, associated with NNRTI resistance, displayed approximately 90-fold resistance to nevirapine but remained fully sensitive to (+)-calanolide A and that the Y181C mutation marginally enhanced susceptibility to the latter drug. In contrast, the Y188H substitution in RT resulted in about 30-fold resistance to (+)-calanolide A in these assays but did not result in diminished sensitivity to nevirapine. Tissue culture results indicated that the combination of (+)-calanolide A and nevirapine possessed an additive to weakly synergistic effect in blocking replication of HIV-1 in tissue culture. These results suggest that (+)-calanolide A and nevirapine might have rationale as a combination therapy for HIV disease.


Subject(s)
Coumarins/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , Reverse Transcriptase Inhibitors/pharmacology , Drug Resistance , HIV-1/drug effects , Mutation , Nevirapine/pharmacology , Pyranocoumarins , Virus Replication/drug effects
19.
Antiviral Res ; 39(2): 81-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9806485

ABSTRACT

Robustaflavone, a naturally occurring biflavanoid isolated from Rhus succedanea, was found to be a potent inhibitor of hepatitis B virus (HBV) replication in 2.2.15 cells, with an effective concentration (EC50) of 0.25 microM, and a selectivity index (SI, IC50/EC90) of 153. Robustaflavone hexaacetate inhibited HBV replication with an EC50 of 0.73 microM, but exhibited no cytotoxicity at concentrations up to 1000 microM. Combinations of robustaflavone with penciclovir and lamivudine displayed synergistic anti-HBV activity, having the most pronounced effects when the combination ratios were similar to the ratio of EC50 potencies. Thus, a 1:1 combination of robustaflavone and penciclovir exhibited an EC50 of 0.11 microM and an SI of 684, while a 10:1 combination of robustaflavone and lamivudine exhibited an EC50 of 0.054 microM and an SI of 894. Statistical analyses of the combination data using the Combostat program confirmed that robustaflavone exhibited synergism with both penciclovir and lamivudine.


Subject(s)
Antiviral Agents/pharmacology , Biflavonoids , Flavonoids/pharmacology , Hepatitis B virus/drug effects , Acyclovir/analogs & derivatives , Acyclovir/pharmacology , Cell Line , Drug Synergism , Guanine , Hepatitis B virus/physiology , Lamivudine/pharmacology , Microbial Sensitivity Tests , Virus Replication/drug effects
20.
Bioorg Med Chem Lett ; 8(16): 2179-84, 1998 Aug 18.
Article in English | MEDLINE | ID: mdl-9873509

ABSTRACT

The three chromanone derivatives, (+)-, (-)-, and (+/-)-12-oxocalanolide A (2), were evaluated for in vitro antiviral activities against HIV and simian immunodeficiency virus (SIV). The compounds were determined to be inhibitors of HIV-1 reverse transcriptase (RT) and exhibited activity against a variety of viruses selected for resistance to other HIV-1 nonnucleoside RT inhibitors. They are the first reported calanolide analogues capable of inhibiting SIV.


Subject(s)
Anti-HIV Agents/pharmacology , Chromones/chemical synthesis , Coumarins/chemistry , Coumarins/chemical synthesis , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Acquired Immunodeficiency Syndrome/virology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Cell Line , Chromones/chemistry , Chromones/pharmacology , Coumarins/pharmacology , Drug Design , HIV-1/enzymology , HIV-1/isolation & purification , Humans , In Vitro Techniques , Leukocytes/virology , Macrophages/virology , Monocytes/virology , Pyranocoumarins , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Simian Immunodeficiency Virus/drug effects , Structure-Activity Relationship , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...