Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32295913

ABSTRACT

Histone deacetylase inhibitors (HDACi) are the most widely studied HIV latency-reversing agents (LRAs). The HDACi suberoylanilide hydroxamic acid (vorinostat [VOR]) has been employed in several clinical HIV latency reversal studies, as well as in vitro models of HIV latency, and has been shown to effectively induce HIV RNA and protein expression. Despite these findings, response to HDACi can vary, particularly with intermittent dosing, and information is lacking on the relationship between the host transcriptional response and HIV latency reversal. Here, we report on global gene expression responses to VOR and examine the longevity of the transcriptional response in various cellular models. We found that many genes are modulated at 6 h post-VOR treatment in HCT116, Jurkat, and primary resting CD4 T cells, yet return to baseline levels after an 18-h VOR-free period. With repeat exposure to VOR in resting CD4 T cells, we found similar and consistent transcriptional changes at 6 h following each serial treatment. In addition, serial exposure in HIV-infected suppressed donor CD4 T cells showed consistent transcriptional changes after each exposure to VOR. We identified five host genes that were strongly and consistently modulated following histone deacetylase (HDAC) inhibition; three (H1F0, IRGM, and WIPI49) were upregulated, and two (PHF15 and PRDM10) were downregulated. These genes demonstrated consistent modulation in peripheral blood mononuclear cell (PBMC) samples from HIV-positive (HIV+) participants who received either single or multiple doses of 400 mg of VOR. Interestingly, the host transcriptional response did not predict induction of cell-associated HIV RNA, suggesting that other cellular factors play key roles in HIV latency reversal in vivo despite robust HDACi pharmacological activity.IMPORTANCE Histone deacetylase inhibitors are widely studied HIV latency-reversing agents (LRAs). VOR, an HDACi, induces histone acetylation and chromatin remodeling and modulates host and HIV gene expression. However, the relationship between these events is poorly defined, and clinical studies suggest diminished HIV reactivation in resting CD4 T cells with daily exposure to VOR. Our study provides evidence that VOR induces a consistent level of host cell gene transcription following intermittent exposure. In addition, in response to VOR exposure a gene signature that was conserved across single and serial exposures both in vitro and in vivo was identified, indicating that VOR can consistently and reproducibly modulate transcriptional host responses. However, as the HIV response to HDACi declines over time, other factors modulate viral reactivation in vivo despite robust HDAC activity. The identified host gene VOR biomarkers can be used for monitoring the pharmacodynamic activity of HDAC inhibitors.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , HIV Infections/drug therapy , Vorinostat/pharmacology , Acetylation , CD4-Positive T-Lymphocytes/drug effects , HIV-1/metabolism , HIV-1/pathogenicity , Histone Deacetylase Inhibitors/pharmacology , Humans , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Primary Cell Culture , Virus Activation/drug effects , Virus Latency/drug effects , Vorinostat/metabolism
2.
Clin Pharmacol Ther ; 107(6): 1325-1333, 2020 06.
Article in English | MEDLINE | ID: mdl-31868916

ABSTRACT

Fine-needle aspiration (FNA) for serial hepatic sampling may be an efficient and less invasive alternative to core needle biopsy (CNB), the current standard for liver tissue sampling. In this randomized, open-label trial in 31 participants with hepatitis C virus genotype 1 infection (NCT01678131/Merck protocol PN048), we evaluated the feasibility of using FNA to obtain human liver tissue samples appropriate for measuring hepatic pharmacokinetics (PK), using vaniprevir as a tool compound. The primary end point was successful retrieval of liver tissue specimens with measurable vaniprevir concentrations at two of three specified FNA time points. Twenty-nine patients met the primary end point and, therefore, were included in the PK analyses. Hepatic vaniprevir concentrations obtained with FNA were consistent with known vaniprevir PK properties. The shape of liver FNA and CNB concentration-time profiles were comparable. In conclusion, FNA may be effective for serial tissue sampling to assess hepatic drug exposure in patients with liver disease.


Subject(s)
Antiviral Agents/pharmacokinetics , Biopsy, Fine-Needle/methods , Cyclopropanes/pharmacokinetics , Hepatitis C, Chronic/drug therapy , Isoindoles/pharmacokinetics , Lactams, Macrocyclic/pharmacokinetics , Leucine/analogs & derivatives , Liver/metabolism , Proline/analogs & derivatives , Sulfonamides/pharmacokinetics , Adult , Antiviral Agents/administration & dosage , Cyclopropanes/administration & dosage , Female , Genotype , Hepacivirus/genetics , Hepacivirus/isolation & purification , Hepatitis C, Chronic/virology , Humans , Isoindoles/administration & dosage , Lactams, Macrocyclic/administration & dosage , Leucine/administration & dosage , Leucine/pharmacokinetics , Liver/virology , Male , Middle Aged , Proline/administration & dosage , Proline/pharmacokinetics , Sulfonamides/administration & dosage , Tissue Distribution , Young Adult
3.
Antiviral Res ; 139: 161-170, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27889530

ABSTRACT

DESIGN: The HIV latent CD4+ T cell reservoir is broadly recognized as a barrier to HIV cure. Induction of HIV expression using protein kinase C (PKC) agonists is one approach under investigation for reactivation of latently infected CD4+ T cells (Beans et al., 2013; Abreu et al., 2014; Jiang et al., 2014; Jiang and Dandekar, 2015). We proposed that an increased understanding of the molecular mechanisms of action of PKC agonists was necessary to inform on biological signaling and pharmacodynamic biomarkers. RNA sequencing (RNA Seq) was applied to identify genes and pathways modulated by PKC agonists. METHODS: Human CD4+ T cells were treated ex vivo with Phorbol 12-myristate 13-acetate, prostatin or ingenol-3-angelate. At 3 h and 24 h post-treatment, cells were harvested and RNA-Seq was performed on RNA isolated from cell lysates. The genes differentially expressed across the PKC agonists were validated by quantitative RT-PCR (qPCR). A subset of genes was evaluated for their role in HIV reactivation using siRNA and CRISPR approaches in the Jurkat latency cell model. RESULTS: Treatment of primary human CD4+ T cells with PKC agonists resulted in alterations in gene expression. qPCR of RNA Seq data confirmed upregulation of 24 genes, including CD69, Egr1, Egr2, Egr3, CSF2, DUSP5, and NR4A1. Gene knockdown of Egr1 and Egr3 resulted in reduced expression and decreased HIV reactivation in response to PKC agonist treatment, indicating a potential role for Egr family members in latency reversal. CONCLUSION: Overall, our results offer new insights into the mechanism of action of PKC agonists, biomarkers of pathway engagement, and the potential role of EGR family in HIV reactivation.


Subject(s)
HIV-1/physiology , Protein Kinase C/metabolism , Virus Activation/drug effects , Virus Latency/drug effects , Biomarkers , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Diterpenes/chemistry , Diterpenes/pharmacology , Drug Agonism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 3/genetics , Gene Expression , HIV Infections/virology , Humans , Jurkat Cells , Male , Phorbols/pharmacology , Sequence Analysis, RNA
4.
J Med Chem ; 59(7): 3489-98, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27011007

ABSTRACT

Herein, we describe the development of a functionally selective liver X receptor ß (LXRß) agonist series optimized for Emax selectivity, solubility, and physical properties to allow efficacy and safety studies in vivo. Compound 9 showed central pharmacodynamic effects in rodent models, evidenced by statistically significant increases in apolipoprotein E (apoE) and ATP-binding cassette transporter levels in the brain, along with a greatly improved peripheral lipid safety profile when compared to those of full dual agonists. These findings were replicated by subchronic dosing studies in non-human primates, where cerebrospinal fluid levels of apoE and amyloid-ß peptides were increased concomitantly with an improved peripheral lipid profile relative to that of nonselective compounds. These results suggest that optimization of LXR agonists for Emax selectivity may have the potential to circumvent the adverse lipid-related effects of hepatic LXR activity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoproteins E/cerebrospinal fluid , Benzamides/chemistry , Benzamides/pharmacology , Orphan Nuclear Receptors/agonists , Piperidines/chemistry , Piperidines/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Dogs , Hep G2 Cells , Humans , Lipids/analysis , Liver/drug effects , Liver/metabolism , Liver X Receptors , Locomotion/drug effects , Macaca mulatta , Madin Darby Canine Kidney Cells , Mice , Mice, Transgenic
5.
J Neurosci ; 34(8): 2884-97, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24553930

ABSTRACT

A hallmark of Alzheimer's disease (AD) brain is the amyloid ß (Aß) plaque, which is comprised of Aß peptides. Multiple lines of evidence suggest that Aß oligomers are more toxic than other peptide forms. We sought to develop a robust assay to quantify oligomers from CSF. Antibody 19.3 was compared in one-site and competitive ELISAs for oligomer binding specificity. A two-site ELISA for oligomers was developed using 19.3 coupled to a sensitive, bead-based fluorescent platform able to detect single photons of emitted light. The two-site ELISA was >2500× selective for Aß oligomers over Aß monomers with a limit of detection ∼ 0.09 pg/ml in human CSF. The lower limit of reliable quantification of the assay was 0.18 pg/ml and the antibody pairs recognized Aß multimers comprised of either synthetic standards, or endogenous oligomers isolated from confirmed human AD and healthy control brain. Using the assay, a significant 3- to 5-fold increase in Aß oligomers in human AD CSF compared with comparably aged controls was demonstrated. The increase was seen in three separate human cohorts, totaling 63 AD and 54 controls. CSF oligomers ranged between 0.1 and 10 pg/ml. Aß oligomer levels did not strongly associate with age or gender, but had an inverse correlation with MMSE score. The C statistic for the Aß oligomer ROC curve was 0.86, with 80% sensitivity and 88% specificity to detect AD, suggesting reasonable discriminatory power for the AD state and the potential for utility as a diagnostic marker.


Subject(s)
Aging/cerebrospinal fluid , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Amyloid beta-Peptides/immunology , Antibodies/immunology , Antibody Specificity , Biomarkers/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay , False Positive Reactions , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Peptide Fragments/cerebrospinal fluid , ROC Curve , Reproducibility of Results , Scattering, Radiation
6.
J Med Chem ; 54(7): 2351-8, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21395312

ABSTRACT

Agonists of somatostatin receptor subtype 2 (sst(2)) have been proposed as therapeutics for the treatment of proliferative diabetic retinopathy and exudative age-related macular degeneration. An HTS screen identified 2-quinolones as weak agonists of sst(2), and these were optimized to provide small molecules with sst(2) binding and functional potency comparable to peptide agonists. Agonist 21 was shown to inhibit rat growth hormone secretion following systemic administration and to inhibit ocular neovascular lesion formation after local administration.


Subject(s)
Drug Design , Quinolines/chemical synthesis , Quinolines/pharmacology , Receptors, Somatostatin/agonists , Animals , CHO Cells , Choroidal Neovascularization/drug therapy , Cricetinae , Cricetulus , Dogs , Female , Humans , Male , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Rats , Substrate Specificity
7.
Clin Cancer Res ; 14(15): 4859-68, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676759

ABSTRACT

PURPOSE: A major mechanism of resistance to methylating agents, including temozolomide, is the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT). Preclinical data indicates that defective DNA mismatch repair (MMR) results in tolerance to temozolomide regardless of AGT activity. The purpose of this study was to determine the role of MMR deficiency in mediating resistance in samples from patients with both newly diagnosed malignant gliomas and those who have failed temozolomide therapy. EXPERIMENTAL DESIGN: The roles of AGT and MMR deficiency in mediating resistance in glioblastoma multiforme were assessed by immunohistochemistry and microsatellite instability (MSI), respectively. The mutation status of the MSH6 gene, a proposed correlate of temozolomide resistance, was determined by direct sequencing and compared with data from immunofluorescent detection of MSH6 protein and reverse transcription-PCR amplification of MSH6 RNA. RESULTS: Seventy percent of newly diagnosed and 78% of failed-therapy glioblastoma multiforme samples expressed nuclear AGT protein in > or = 20% of cells analyzed, suggesting alternate means of resistance in 20% to 30% of cases. Single loci MSI was observed in 3% of patient samples; no sample showed the presence of high MSI. MSI was not shown to correlate with MSH6 mutation or loss of MSH6 protein expression. CONCLUSIONS: Although high AGT levels may mediate resistance in a portion of these samples, MMR deficiency does not seem to be responsible for mediating temozolomide resistance in adult malignant glioma. Accordingly, the presence of a fraction of samples exhibiting both low AGT expression and MMR proficiency suggests that additional mechanisms of temozolomide resistance are operational in the clinic.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Base Pair Mismatch , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , DNA Mismatch Repair , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioma/drug therapy , Glioma/genetics , Adult , Aged , Aged, 80 and over , Dacarbazine/pharmacology , Female , Glioblastoma/genetics , Humans , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/genetics , Temozolomide
9.
Mol Cancer Ther ; 5(10): 2531-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17041097

ABSTRACT

Promoter hypermethylation of the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) has been associated with an enhanced response to chloroethylating and methylating agents in patients with malignant glioma. The purpose of this study was to compare three distinct yet related indices for measuring AGT to determine if these assays could be used interchangeably when AGT status is to be used to guide chemotherapeutic decisions. Real-time methylation-specific PCR (MSP), assessed as the ratio of methylated AGT copies to internal beta-actin control, was used to quantitate AGT hypermethylation in 32 glioma samples. Data were compared with AGT enzyme activity as well as immunohistochemical detection of AGT protein from the same samples. Hypermethylation of the AGT promoter was detected in 19 of 31 (61%) samples evaluable by MSP. Low-level AGT, defined as <20% nuclear AGT staining by immunohistochemistry, was found in 10 of 32 samples (31%), whereas 12 of 32 (38%) had low levels of AGT activity. Correlation of immunohistochemistry to AGT activity was statistically significant (P = 0.014) as was the correlation of immunohistochemistry to MSP (P = 0.043), whereas MSP compared with AGT activity (P = 0.246) was not significant. Cross-tabulation of immunohistochemistry and MSP data based on prognostic groups, where good prognosis was represented by an immunohistochemistry of <20% and an MSP ratio >12, showed no significant relationship (P = 0.214), suggesting that one assay cannot be used interchangeably for another. The observed discordance between respective measures of AGT based on prognosis supports further standardization of AGT assays designed to guide therapeutic practice. The data also suggest that consideration be given to the large population of AGT-expressing cells within samples when therapeutic strategies based on tumor methylation are used.


Subject(s)
Brain Neoplasms/enzymology , Glioma/enzymology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Adult , Aged , Aged, 80 and over , DNA Methylation , Female , Humans , Immunohistochemistry , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic
10.
J Clin Oncol ; 23(28): 7178-87, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16192602

ABSTRACT

PURPOSE: We conducted a two-phase clinical trial in patients with progressive malignant glioma (MG). The first phase of this trial was designed to determine the dose of O6-BG effective in producing complete depletion of tumor AGT activity for 48 hours. The second phase of the trial was designed to define the maximum tolerated dose (MTD) of a single dose of temozolomide when combined with O6-BG. In addition, plasma concentrations of O6-BG and O6-benzyl-8-oxoguanine were evaluated after O6-BG. PATIENTS AND METHODS: For our first phase of the clinical trial, patients were scheduled to undergo craniotomy for AGT determination after receiving a 1-hour O6-BG infusion at 120 mg/m2 followed by a continuous infusion at an initial dose of 30 mg/m2/d for 48 hours. The dose of the continuous infusion of O6-BG escalated until tumor AGT was depleted. Once the O6-BG dose was established a separate group of patients was enrolled in the second phase of clinical trial, in which temozolomide, administered as a single dose at the end of the 1-hour O6-BG infusion, was escalated until the MTD was determined. RESULTS: The O6-BG dose found to be effective in depleting tumor AGT activity at 48 hours was an IV bolus of 120 mg/m2 over 1 hour followed by a continuous infusion of 30 mg/m2/d for 48 hours. On enrolling 38 patients in six dose levels of temozolomide, the MTD was established at 472 mg/m2 with dose-limiting toxicities limited to myelosuppression. CONCLUSION: This study provides the foundation for a phase II trial of O6-BG plus temozolomide in temozolomide-resistant MG.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Brain Neoplasms/pathology , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacokinetics , Disease Progression , Female , Glioma/pathology , Guanine/administration & dosage , Guanine/adverse effects , Guanine/analogs & derivatives , Guanine/pharmacokinetics , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Temozolomide
11.
J Biol Chem ; 279(50): 52200-9, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15469933

ABSTRACT

Secreted protein acidic, rich in cysteine (SPARC), is an extracellular matrix protein expressed in many advanced cancers, including malignant gliomas. We and others have previously shown that human glioma cell lines engineered to overexpress SPARC adopt an invasive phenotype. We now show that SPARC expression increases cell survival under stress initiated by serum withdrawal through a decrease in apoptosis. Phosphatidylinositol 3-OH kinase/AKT is a potent pro-survival pathway that contributes to the malignancy of gliomas. Cells expressing SPARC display increased AKT activation with decreased caspase 3/7 activity. Exogenous SPARC rapidly induces AKT phosphorylation, an effect that is blocked by a neutralizing SPARC antibody. Furthermore, AKT activation is essential for the anti-apoptotic effects of SPARC as the decreased apoptosis and caspase activity associated with SPARC expression can be blocked with dominant-negative AKT or a specific AKT inhibitor. As tumor cells face stressful microenvironments particularly during the process of invasion, these results suggest that SPARC functions, in part, to promote tumor progression by enabling tumor cells to survive under stressful conditions.


Subject(s)
Cell Survival/physiology , Glioma/pathology , Glioma/physiopathology , Osteonectin/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/physiology , Caspase 3 , Caspase 7 , Caspases/metabolism , Cattle , Cell Line, Tumor , Culture Media, Serum-Free , Enzyme Activation , Humans , Osteonectin/genetics , Osteonectin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
12.
J Biol Chem ; 277(34): 30469-76, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12063265

ABSTRACT

NF-kappaB is an important transcription factor that has a role in a variety of responses such as inflammation, oncogenesis, apoptosis, and viral replication. Oxidative stress is well known to induce the activation of NF-kappaB. Cells can be exposed to either endogenously produced oxidants or oxidants produced by surrounding cells. In addition, ischemia reperfusion and certain cancer therapies such as chemotherapy and photodynamic therapy are thought to result in oxygen radical production. Because of the important role that NF-kappaB has in multiple responses, it is critical to determine the mechanisms by which oxidative stress induces NF-kappaB activity. We report that the calmodulin antagonist W-7 and the calcium/calmodulin-dependent (CaM) kinase inhibitors KN-93 and K252a, can block oxidative stress-induced IkappaB phosphorylation in Jurkat T lymphocytes. Furthermore, KN-93 but not KN-92 can block hydrogen peroxide-induced Akt and IKK phosphorylation. In addition, we found that expression of a kinase-dead CaM-KIV construct in two cell lines inhibits IkappaB phosphorylation or degradation and that expression of CaM-KIV augments hydrogen peroxide-induced IkappaB phosphorylation and degradation. Although the CaM kinases appear to be required for this response, increases in intracellular calcium do not appear to be required. These results identify the CaM kinases as potential targets that can be used to minimize NF-kappaB activation in response to oxidative stress.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Hydrogen Peroxide/pharmacology , I-kappa B Proteins/metabolism , T-Lymphocytes/metabolism , Benzylamines/pharmacology , Humans , I-kappa B Kinase , Jurkat Cells , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...