Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Digit Biomark ; 4(Suppl 1): 28-49, 2020.
Article in English | MEDLINE | ID: mdl-33442579

ABSTRACT

Innovative tools are urgently needed to accelerate the evaluation and subsequent approval of novel treatments that may slow, halt, or reverse the relentless progression of Parkinson disease (PD). Therapies that intervene early in the disease continuum are a priority for the many candidates in the drug development pipeline. There is a paucity of sensitive and objective, yet clinically interpretable, measures that can capture meaningful aspects of the disease. This poses a major challenge for the development of new therapies and is compounded by the considerable heterogeneity in clinical manifestations across patients and the fluctuating nature of many signs and symptoms of PD. Digital health technologies (DHT), such as smartphone applications, wearable sensors, and digital diaries, have the potential to address many of these gaps by enabling the objective, remote, and frequent measurement of PD signs and symptoms in natural living environments. The current climate of the COVID-19 pandemic creates a heightened sense of urgency for effective implementation of such strategies. In order for these technologies to be adopted in drug development studies, a regulatory-aligned consensus on best practices in implementing appropriate technologies, including the collection, processing, and interpretation of digital sensor data, is required. A growing number of collaborative initiatives are being launched to identify effective ways to advance the use of DHT in PD clinical trials. The Critical Path for Parkinson's Consortium of the Critical Path Institute is highlighted as a case example where stakeholders collectively engaged regulatory agencies on the effective use of DHT in PD clinical trials. Global regulatory agencies, including the US Food and Drug Administration and the European Medicines Agency, are encouraging the efficiencies of data-driven engagements through multistakeholder consortia. To this end, we review how the advancement of DHT can be most effectively achieved by aligning knowledge, expertise, and data sharing in ways that maximize efficiencies.

2.
Toxicol Sci ; 161(1): 58-75, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28973697

ABSTRACT

Pharmaceuticals and chemicals produce hemangiosarcomas (HS) in mice, often by nongenotoxic, proliferative mechanisms. A mode-of-action (MOA) for hemangiosarcoma was proposed based on information presented at an international workshop (Cohen et al., Hemangiosarcoma in rodents: Mode-of-action evaluation and human relevance. Toxicol. Sci. 111, 4-18.). Five key elements of the MOA were articulated and included hypoxia, macrophage activation, increased angiogenic growth factors, dysregulated angiogenesis/erythropoiesis, and endothial cell proliferation. The goal of the current study was to add to the weight-of-evidence for the proposed MOA by assessing these key elements with 3 different compounds of varying potency for HS induction: fenretinide (high), troglitazone (intermediate), and elmiron (low). Multiple endpoints, including hypoxia (hyproxyprobe, transcriptomics), endothelial cell (EC) proliferation, and clinical and anatomic pathology, were assessed after 2, 4, and 13-weeks of treatment in B6C3F1 mice. All 3 compounds demonstrated strong evidence for dysregulated erythropoiesis (decrease in RBC and a failure to increase reticulocytes) and macrophage activation (4- to 11-fold increases); this pattern of hematological changes in mice might serve as an early biomarker to evaluate EC proliferation in suspected target organs for potential HS formation. Fenretinide demonstrated all 5 key elements, while troglitazone demonstrated 4 and elmiron demonstrated 3. Transcriptomics provided support for the 5 elements of the MOA, but was not any more sensitive than hypoxyprobe immunohistochemistry for detecting hypoxia. The overall transcriptional evidence for the key elements of the proposed MOA was also consistent with the potency of HS induction. These data, coupled with the previous work with 2-butoxyethanol and pregablin, increase the weight-of-evidence for the proposed MOA for HS formation.


Subject(s)
Fenretinide/toxicity , Hemangiosarcoma/chemically induced , Neovascularization, Pathologic/chemically induced , Pentosan Sulfuric Polyester/toxicity , Troglitazone/toxicity , Animals , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Hemangiosarcoma/metabolism , Hemangiosarcoma/pathology , Macrophage Activation/drug effects , Male , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Organ Specificity
3.
Toxicol Sci ; 159(1): 42-49, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28903491

ABSTRACT

Drug-induced vascular injury (DIVI) in preclinical studies can delay, if not terminate, a drug development program. Clinical detection of DIVI can be very difficult as there are no definitive biomarkers known to reliably detect this disorder in all instances. The preclinical identification of DIVI requires detailed microscopic examination of a wide range of tissues although one of the most commonly affected areas in rats is the mesenteric vasculature. The reason for this predisposition of mesenteric arteries in rats as well as the exact mechanism and cell types involved in the initial development of these lesions have not been fully elucidated. We hypothesized that by using a mixed culture of cells from rat mesenteric tissue, we would be able to identify an RNA expression signature that could predict the invivo development of DIVI. Five compounds designed to inhibit Phosphodiesterase 4 activity (PDE4i) were chosen as positive controls. PDE4i's are well known to induce DIVI in the mesenteric vasculature of rats and there is microscopic evidence that this is associated, at least in part, with a proinflammatory mechanism. We surveyed, by qRT-PCR, the expression of 96 genes known to be involved in inflammation and using a Random-Forest model, identified 12 genes predictive of invivo DIVI outcomes in rats. Using these genes, we were able to cross-validate the ability of the Random-Forest modeling to predict the concentration at which PDE4i caused DIVI invivo.


Subject(s)
Mesenteric Arteries/cytology , Phosphodiesterase 4 Inhibitors/toxicity , Vascular System Injuries/chemically induced , Animals , Male , Rats , Rats, Sprague-Dawley
4.
Toxicol Pathol ; 42(4): 709-24, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24705881

ABSTRACT

Previously we found that regulation of eNOS is an important part of the pathogenic process of Drug-induced vascular injury (DIVI) for PDE4i. The aims of the current study were to examine the phosphorylation of eNOS in mesentery versus aorta at known regulatory sites across DIVI-inducing drug classes and to compare changes across species. We found that phosphorylation at S615 in rats was elevated 35-fold 2 hr after the last dose of CI-1044 in mesentery versus 3-fold in aorta. Immunoprecipitation studies revealed that many of the upstream regulators of eNOS activation were associated with eNOS in 1 or more signalosome complexes. Next rats were treated with drugs from 4 other classes known to cause DIVI. Each drug was given alone and in combination with SIN-1 (NO donor) or L-NAME (eNOS inhibitor), and the level of eNOS phosphorylation in mesentery and aorta tissue was correlated with the extent of vascular injury and measured serum nitrite. Drugs or combinations produced altered serum nitrite levels as well as vascular injury score in the mesentery. The results suggested that phosphorylation of S615 may be associated with DIVI activity. Studies with the species-specific A2A adenosine agonist CI-947 in rats versus primates showed a similar pattern.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Nitric Oxide Synthase Type III/metabolism , Vascular System Injuries/chemically induced , Vascular System Injuries/pathology , Adenosine/administration & dosage , Adenosine/adverse effects , Adenosine/analogs & derivatives , Animals , Aorta/metabolism , Azepines/administration & dosage , Azepines/adverse effects , Dose-Response Relationship, Drug , Male , Niacinamide/administration & dosage , Niacinamide/adverse effects , Niacinamide/analogs & derivatives , Nitric Oxide/blood , Nitric Oxide Synthase Type III/genetics , Nitrites/blood , Phosphorylation , Rats , Rats, Sprague-Dawley
5.
Toxicol Pathol ; 42(4): 635-57, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24777748

ABSTRACT

Better biomarkers are needed to identify, characterize, and/or monitor drug-induced vascular injury (DIVI) in nonclinical species and patients. The Predictive Safety Testing Consortium (PSTC), a precompetitive collaboration of pharmaceutical companies and the U.S. Food and Drug Administration (FDA), formed the Vascular Injury Working Group (VIWG) to develop and qualify translatable biomarkers of DIVI. The VIWG focused its research on acute DIVI because early detection for clinical and nonclinical safety monitoring is desirable. The VIWG developed a strategy based on the premise that biomarkers of DIVI in rat would be translatable to humans due to the morphologic similarity of vascular injury between species regardless of mechanism. The histomorphologic lexicon for DIVI in rat defines degenerative and adaptive findings of the vascular endothelium and smooth muscles, and characterizes inflammatory components. We describe the mechanisms of these changes and their associations with candidate biomarkers for which advanced analytical method validation was completed. Further development is recommended for circulating microRNAs, endothelial microparticles, and imaging techniques. Recommendations for sample collection and processing, analytical methods, and confirmation of target localization using immunohistochemistry and in situ hybridization are described. The methods described are anticipated to aid in the identification and qualification of translational biomarkers for DIVI.


Subject(s)
Biomarkers/blood , Drug-Related Side Effects and Adverse Reactions , Vascular System Injuries/chemically induced , Vascular System Injuries/pathology , Animals , Drug Evaluation, Preclinical/trends , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , United States , United States Food and Drug Administration
6.
Toxicol Pathol ; 42(4): 784-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24670818

ABSTRACT

Several classes of drugs have been shown to cause drug-induced vascular injury (DIVI) in preclinical toxicity studies. Measurement of blood flow and vessel diameter in numerous vessels and across various tissues by ultrasound imaging has the potential to be a noninvasive translatable biomarker of DIVI. Our objective was to demonstrate the utility of high-frequency ultrasound imaging for measuring changes in vascular function by evaluating blood flow and vessel diameter in the superior mesenteric arteries (SMA) of rats treated with compounds that are known to cause DIVI and are known vasodilators in rat: fenoldopam, CI-1044, and SK&F 95654. Blood flow, vessel diameter, and other parameters were measured in the SMA at 4, 8, and 24 hr after dosing. Mild to moderate perivascular accumulations of mononuclear cells, neutrophils in tunica adventitia, and superficial tunica media as well as multifocal hemorrhage and necrosis in the tunica media were found in animals 24 hr after treatment with fenoldopam and SK&F 95654. Each compound caused marked increases in blood flow and shear stress as early as 4 hr after dosing. These results suggest that ultrasound imaging may constitute a functional correlate for the microscopic finding of DIVI in the rat.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Ultrasonography/methods , Vascular System Injuries/chemically induced , Vascular System Injuries/pathology , Animals , Azepines/adverse effects , Fenoldopam/adverse effects , Hemodynamics , Male , Mesenteric Arteries/diagnostic imaging , Mesenteric Arteries/drug effects , Niacinamide/adverse effects , Niacinamide/analogs & derivatives , Pyridazines/adverse effects , Pyridines/adverse effects , Rats , Rats, Sprague-Dawley , Vascular System Injuries/diagnostic imaging
7.
Toxicol Appl Pharmacol ; 274(1): 156-67, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24126418

ABSTRACT

Pretreatment of mice with a low hepatotoxic dose of acetaminophen (APAP) results in resistance to a subsequent, higher dose of APAP. This mouse model, termed APAP autoprotection was used here to identify differentially expressed genes and cellular pathways that could contribute to this development of resistance to hepatotoxicity. Male C57BL/6J mice were pretreated with APAP (400mg/kg) and then challenged 48h later with 600mg APAP/kg. Livers were obtained 4 or 24h later and total hepatic RNA was isolated and hybridized to Affymetrix Mouse Genome MU430_2 GeneChip. Statistically significant genes were determined and gene expression changes were also interrogated using the Causal Reasoning Engine (CRE). Extensive literature review narrowed our focus to methionine adenosyl transferase-1 alpha (MAT1A), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), flavin-containing monooxygenase 3 (Fmo3) and galectin-3 (Lgals3). Down-regulation of MAT1A could lead to decreases in S-adenosylmethionine (SAMe), which is known to protect against APAP toxicity. Nrf2 activation is expected to play a role in protective adaptation. Up-regulation of Lgals3, one of the genes supporting the Nrf2 hypothesis, can lead to suppression of apoptosis and reduced mitochondrial dysfunction. Fmo3 induction suggests the involvement of an enzyme not known to metabolize APAP in the development of tolerance to APAP toxicity. Subsequent quantitative RT-PCR and immunochemical analysis confirmed the differential expression of some of these genes in the APAP autoprotection model. In conclusion, our genomics strategy identified cellular pathways that might further explain the molecular basis for APAP autoprotection.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Gene Expression Regulation/physiology , Models, Animal , Animals , Chemical and Drug Induced Liver Injury/genetics , Gene Expression Regulation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL
8.
Clin Cancer Res ; 17(21): 6641-5, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22046025

ABSTRACT

There has been an explosion of technology-enabled scientific insight into the basic biology of the causes of adverse events. This has been driven, in part, by the development of the various "omics" tools (e.g., genomics, proteomics, and metabolomics) and associated bioinformatics platforms. Meanwhile, for decades, changes in preclinical testing protocols and guidelines have been limited. Preclinical safety testing currently relies heavily on the use of outdated animal models. Application of systems biology methods to evaluation of toxicities in oncology treatments can accelerate the introduction of safe, effective drugs. Systems biology adds insights regarding the causes and mechanisms of adverse effects, provides important and actionable information to help understand the risks and benefits to humans, focuses testing on methods that add value to the safety testing process, and leads to modifications of chemical entities to reduce liabilities during development. Leveraging emerging technologies, such as genomics and proteomics, may make preclinical safety testing more efficient and accurate and lead to better safety decisions. The development of a U.S. Food and Drug Administration guidance document on the use of systems biology in clinical testing would greatly benefit the development of drugs for oncology by communicating the potential application of specific methodologies, providing a framework for qualification and application of systems biology outcomes, and providing insight into the challenges and limitations of systems biology in the regulatory decision-making process.


Subject(s)
Antineoplastic Agents/adverse effects , Drug-Related Side Effects and Adverse Reactions/diagnosis , Systems Biology/methods , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Translational Research, Biomedical
9.
Am J Pathol ; 179(1): 240-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703406

ABSTRACT

Macrophage colony-stimulating factor (M-CSF) is a hematopoietic growth factor that is responsible for the survival and proliferation of monocytes and the differentiation of monocytes into macrophages, including Kupffer cells (KCs) in the liver. KCs play an important role in the clearance of several serum enzymes, including aspartate aminotransferase and creatine kinase, that are typically elevated as a result of liver or skeletal muscle injury. We used three distinct animal models to investigate the hypothesis that increases in the levels of serum enzymes can be the result of decreases in KCs in the apparent absence of hepatic or skeletal muscle injury. Specifically, neutralizing M-CSF activity via a novel human monoclonal antibody reduced the CD14(+)CD16(+) monocyte population, depleted KCs, and increased aspartate aminotransferase and creatine kinase serum enzyme levels in cynomolgus macaques. In addition, the treatment of rats with clodronate liposomes depleted KCs and led to increased serum enzyme levels, again without evidence of tissue injury. Finally, in the osteopetrotic (Csf1(op)/Csf1(op)) mice lacking functional M-CSF and having reduced levels of KCs, the levels of serum enzymes are higher than in wild-type littermates. Together, these findings support a mechanism for increases in serum enzyme levels through M-CSF regulation of tissue macrophage homeostasis without concomitant histopathological changes in either the hepatic or skeletal system.


Subject(s)
Aspartate Aminotransferases/blood , Creatine Kinase/blood , Kupffer Cells/pathology , Liver/enzymology , Muscle, Skeletal/enzymology , Osteopetrosis/pathology , Animals , Antibodies, Monoclonal/pharmacology , Bone Density Conservation Agents/pharmacology , Clodronic Acid/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Liver/injuries , Liver/pathology , Macaca fascicularis , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/physiology , Male , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Osteopetrosis/metabolism , Rats , Rats, Sprague-Dawley , Receptors, IgG/metabolism
10.
Toxicol Sci ; 122(1): 7-15, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21498876

ABSTRACT

Drug-induced vascular injury (DIVI) is observed in rat mesenteric arterioles in response to treatment with phosphodiesterase-4 inhibitors (PDE4i). However, the mechanisms responsible for causing the characteristic vascular lesions are unclear. Nitrotyrosine (NT) adducts, markers of local nitric oxide (NO) production, have been observed in close proximity to the arterial lesions and in the inflammatory cells associated with DIVI. To determine if NO has a direct role in DIVI, rats were treated with the PDE4i CI-1044 at 10, 20, or 40 mg/kg alone or in combination with the nitric oxide synthase inhibitor L-NAME (60 mg/kg) or the nitric oxide donor SIN-1 (30 mg/kg). Mesenteries were collected and processed for microscopic evaluation. NT formation was evaluated in situ via immunohistochemical staining. Serum nitrite (SN), a marker of in vivo NO production, was measured. Compared with vehicle controls, treatment with CI-1044 alone resulted in dose-related increases in the frequency and severity of vascular injury, SN levels, and NT residues. SIN-1 coadministration caused vascular injury to occur at lower doses of CI-1044, compared with CI-1044 alone, with the overall incidence and severity of injury being greater across all CI-1044-dose groups. Following administration of 20 or 40 mg/kg CI-1044, there were also increases in NT immunoreactivity when SIN-1 was coadministered and significant increases in SN. Conversely, coadministration of L-NAME resulted in marked reduction of injury, NT, and SN when compared with CI-1044 alone. The present study suggests that NO production is closely linked to PDE4i-induced vascular injury.


Subject(s)
Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Phosphodiesterase 4 Inhibitors/metabolism , Vascular System Injuries/chemically induced , Animals , Azepines/adverse effects , Biomarkers/analysis , Dose-Response Relationship, Drug , Enzyme Inhibitors/adverse effects , Immunohistochemistry , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , Molsidomine/analogs & derivatives , Molsidomine/metabolism , NG-Nitroarginine Methyl Ester/adverse effects , Niacinamide/adverse effects , Niacinamide/analogs & derivatives , Nitric Oxide/analysis , Nitric Oxide Synthase/metabolism , Nitrites/blood , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/analysis
11.
Toxicol Sci ; 113(1): 254-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812364

ABSTRACT

To understand the molecular mechanisms underlying compound-induced hemangiosarcomas in mice, and therefore, their human relevance, a systems biology approach was undertaken using transcriptomics and Causal Network Modeling from mice treated with 2-butoxyethanol (2-BE). 2-BE is a hemolytic agent that induces hemangiosarcomas in mice. We hypothesized that the hemolysis induced by 2-BE would result in local tissue hypoxia, a well-documented trigger for endothelial cell proliferation leading to hemangiosarcoma. Gene expression data from bone marrow (BM), liver, and spleen of mice exposed to a single dose (4 h) or seven daily doses of 2-BE were used to develop a mechanistic model of hemangiosarcoma. The resulting mechanistic model confirms previous work proposing that 2-BE induces macrophage activation and inflammation in the liver. In addition, the model supports local tissue hypoxia in the liver and spleen, coupled with increased erythropoeitin signaling and erythropoiesis in the spleen and BM, and suppression of mechanisms that contribute to genomic stability, events that could be contributing factors to hemangiosarcoma formation. Finally, an immunohistochemistry method (Hypoxyprobe) demonstrated that tissue hypoxia was present in the spleen and BM. Together, the results of this study identify molecular mechanisms that initiate hemangiosarcoma, a key step in understanding safety concerns that can impact drug decision processes, and identified hypoxia as a possible contributing factor for 2-BE-induced hemangiosarcoma in mice.


Subject(s)
Bone Marrow/metabolism , Cell Transformation, Neoplastic/metabolism , Hemangiosarcoma/metabolism , Liver/metabolism , Models, Biological , Signal Transduction , Spleen/metabolism , Systems Biology , Animals , Bone Marrow/pathology , Cell Cycle , Cell Differentiation , Cell Hypoxia , Cell Proliferation , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Erythropoiesis , Erythropoietin/metabolism , Ethylene Glycols , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genomic Instability , Hemangiosarcoma/chemically induced , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Hematopoietic Stem Cells/metabolism , Hemolysis , Hepatitis/metabolism , Hepatitis/pathology , Immunohistochemistry , Liver/pathology , Macrophage Activation , Male , Mice , Spleen/pathology , Time Factors
12.
Toxicol Appl Pharmacol ; 222(2): 169-79, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17585979

ABSTRACT

Pretreatment of mice with the peroxisome proliferator clofibrate (CFB) protects against acetaminophen (APAP)-induced hepatotoxicity. Previous studies have shown that activation of the nuclear peroxisome proliferator activated receptor-alpha (PPARalpha) is required for this effect. The present study utilizes gene expression profile analysis to identify potential pathways contributing to PPARalpha-mediated hepatoprotection. Gene expression profiles were compared between wild type and PPARalpha-null mice pretreated with vehicle or CFB (500 mg/kg, i.p., daily for 10 days) and then challenged with APAP (400 mg/kg, p.o.). Total hepatic RNA was isolated 4 h after APAP treatment and hybridized to Affymetrix Mouse Genome MGU74 v2.0 GeneChips. Gene expression analysis was performed utilizing GeneSpring software. Our analysis identified 53 genes of interest including vanin-1, cell cycle regulators, lipid-metabolizing enzymes, and aldehyde dehydrogenase 2, an acetaminophen binding protein. Vanin-1 could be important for CFB-mediated hepatoprotection because this protein is involved in the synthesis of cysteamine and cystamine. These are potent antioxidants capable of ameliorating APAP toxicity in rodents and humans. HPLC-ESI/MS/MS analysis of liver extracts indicates that enhanced vanin-1 gene expression results in elevated cystamine levels, which could be mechanistically associated with CFB-mediated hepatoprotection.


Subject(s)
Clofibrate/pharmacology , Gene Expression Profiling/methods , Liver/drug effects , PPAR alpha/genetics , Acetaminophen/administration & dosage , Acetaminophen/toxicity , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Amidohydrolases , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chemical and Drug Induced Liver Injury , Clofibrate/therapeutic use , Cluster Analysis , Cystamine/chemistry , Cystamine/metabolism , Cysteamine/chemistry , Cysteamine/metabolism , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , GPI-Linked Proteins , Liver/metabolism , Liver/pathology , Liver Diseases/genetics , Liver Diseases/prevention & control , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Oligonucleotide Array Sequence Analysis/methods , PPAR alpha/metabolism , Pantetheine/chemistry , Pantetheine/metabolism , Pantothenic Acid/chemistry , Pantothenic Acid/metabolism , Peroxisome Proliferators/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Drug Metab Dispos ; 35(3): 493-500, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17194716

ABSTRACT

CYP51 fulfills an essential requirement for all cells, by catalyzing three sequential mono-oxidations within the cholesterol biosynthesis cascade. Inhibition of fungal CYP51 is used as a therapy for treating fungal infections, whereas inhibition of human CYP51 has been considered as a pharmacological approach to treat dyslipidemia and some forms of cancer. To predict the interaction of inhibitors with the active site of human CYP51, a three-dimensional quantitative structure-activity relationship model was constructed. This pharmacophore model of the common structural features of CYP51 inhibitors was built using the program Catalyst from multiple inhibitors (n = 26) of recombinant human CYP51-mediated lanosterol 14alpha-demethylation. The pharmacophore, which consisted of one hydrophobe, one hydrogen bond acceptor, and two ring aromatic features, demonstrated a high correlation between observed and predicted IC(50) values (r = 0.92). Validation of this pharmacophore was performed by predicting the IC(50) of a test set of commercially available (n = 19) and CP-320626-related (n = 48) CYP51 inhibitors. Using predictions below 10 microM as a cutoff indicative of active inhibitors, 16 of 19 commercially available inhibitors (84%) and 38 of 48 CP-320626-related inhibitors (79.2%) were predicted correctly. To better understand how inhibitors fit into the enzyme, potent CYP51 inhibitors were used to build a Cerius(2) receptor surface model representing the volume of the active site. This study has demonstrated the potential for ligand-based computational pharmacophore modeling of human CYP51 and enables a high-throughput screening system for drug discovery and data base mining.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/chemistry , Models, Molecular , Oxidoreductases/antagonists & inhibitors , Amides/chemistry , Humans , Indoles/chemistry , Quantitative Structure-Activity Relationship , Sterol 14-Demethylase
14.
Bioinformatics ; 22(16): 2028-36, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16809386

ABSTRACT

MOTIVATION: Although numerous methods have been developed to better capture biological information from microarray data, commonly used single gene-based methods neglect interactions among genes and leave room for other novel approaches. For example, most classification and regression methods for microarray data are based on the whole set of genes and have not made use of pathway information. Pathway-based analysis in microarray studies may lead to more informative and relevant knowledge for biological researchers. RESULTS: In this paper, we describe a pathway-based classification and regression method using Random Forests to analyze gene expression data. The proposed methods allow researchers to rank important pathways from externally available databases, discover important genes, find pathway-based outlying cases and make full use of a continuous outcome variable in the regression setting. We also compared Random Forests with other machine learning methods using several datasets and found that Random Forests classification error rates were either the lowest or the second-lowest. By combining pathway information and novel statistical methods, this procedure represents a promising computational strategy in dissecting pathways and can provide biological insight into the study of microarray data. AVAILABILITY: Source code written in R is available from http://bioinformatics.med.yale.edu/pathway-analysis/rf.htm.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Pattern Recognition, Automated , Algorithms , Animals , Computer Simulation , Data Interpretation, Statistical , Humans , Mice , Models, Biological , Models, Statistical , Rats , Regression Analysis , Software
15.
Toxicol Pathol ; 34(1): 27-32, 2006.
Article in English | MEDLINE | ID: mdl-16507541

ABSTRACT

Acute vascular injury that leads to vascular inflammation is a common finding in the preclinical toxicity testing of drugs in rats and dogs. However, the relevance of this finding for risk to humans is unclear. Concern about the safety of these drugs is heightened by the current lack of noninvasive clinical methods to predict the onset of vascular damage in animals or humans. Determining the relevance of this poorly understood preclinical outcome for humans requires a better understanding of the molecular mechanisms of injury in addition to the development of sensitive and specific leading biomarkers for the clinical diagnosis of acute vascular damage. Most molecular research on this toxicity has been performed in rats, but recent development of canine gene expression microarrays makes transcriptomic studies now possible in the dog. In this study, we investigated the molecular mechanisms of drug-induced vascular injury in dogs using gene arrays. After treating Beagles with toxic doses of CI-947, an adenosine receptor agonist, we profiled gene expression in the coronary arteries and correlated those changes with histopathology at 16 and 24 hours after dosing. The results demonstrated that pathobiological processes such as stimulation of the innate immune response, increased extracellular matrix turnover and oxidative stress were active at times of very early injury.


Subject(s)
Adenosine/analogs & derivatives , Gene Expression Profiling/methods , Purinergic P1 Receptor Agonists , Toxicity Tests/methods , Vasculitis/chemically induced , Acute Disease , Adenosine/toxicity , Administration, Oral , Animals , Biomarkers , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Dogs , Dose-Response Relationship, Drug , Gene Expression/drug effects , Genomics , Male , Oligonucleotide Array Sequence Analysis , Vasculitis/genetics , Vasculitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...