Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Toxicol Sci ; 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39302733

ABSTRACT

Molnupiravir is registered or authorized in several countries as a 5-day oral COVID-19 treatment for adults. Molnupiravir is a prodrug of the antiviral ribonucleoside ß-D-N4-hydroxycytidine (NHC) that distributes into cells, where it is phosphorylated to its pharmacologically active ribonucleoside triphosphate (NHC-TP) form. NHC-TP incorporates into SARS-CoV-2 RNA by the viral RNA-dependent RNA polymerase, resulting in an accumulation of errors in the viral genome, leading to inhibition of viral replication and loss of infectivity. The potential of molnupiravir to induce genomic mutations and DNA damage was comprehensively assessed in several in vitro and in vivo genotoxicity assays and a carcinogenicity study, in accordance with international guideline recommendations and expert opinion. Molnupiravir and NHC induced mutations in vitro in bacteria and mammalian cells but did not induce chromosome damage in in vitro or in vivo assays. The in vivo mutagenic and carcinogenic potential of molnupiravir was tested in a series of in vivo mutagenicity studies in somatic and germ cells (Pig-a Assay and Big Blue® TGR Mutation Assay) and in a carcinogenicity study (transgenic rasH2-Tg mouse), using durations of exposure and doses exceeding those used in clinical therapy. In vitro genotoxicity results are superseded by robustly conducted in vivo studies. Molnupiravir did not increase mutations in somatic or germ cells in the in vivo animal studies and was negative in the carcinogenicity study. The interpretation criteria for each study followed established regulatory guidelines. Taken together, these data indicate that molnupiravir use does not present a genotoxicity or carcinogenicity risk for patients.

2.
Toxicol Res (Camb) ; 13(1): tfad120, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38223529

ABSTRACT

Fialuridine (FIAU) is a nucleoside-based drug that caused liver failure and deaths in a human clinical trial that were not predicted by nonclinical safety studies. A recent report concluded that a TK-NOG humanized liver (hu-liver) mouse model detected human-specific FIAU liver toxicity, and broader use of that model could improve drug safety testing. We further evaluated this model at similar dose levels to assess FIAU sensitivity and potential mechanistic biomarkers. Although we were unable to reproduce the marked acute liver toxicity with two separate studies (including one with a "sensitized" donor), we identified molecular biomarkers reflecting the early stages of FIAU mitochondrial toxicity, which were not seen with its stereoisomer (FIRU). Dose dependent FIAU-induced changes in hu-liver mice included more pronounced reductions in mitochondrial to nuclear DNA (mtDNA/nucDNA) ratios in human hepatocytes compared to mouse hepatocytes and kidneys of the same animals. FIAU treatment also triggered a p53 transcriptional response and opposing changes in transcripts of nuclear- and mitochondrial-encoded mitochondrial proteins. The time dependent accumulation of FIAU into mtDNA is consistent with the ≥9-week latency of liver toxicity observed for FIAU in the clinic. Similar changes were observed in an in vitro micro-patterned hepatocyte coculture system. In addition, FIAU-dependent mtDNA/nucDNA ratio and transcriptional alterations, especially reductions in mitochondrially encoded transcripts, were seen in livers of non-engrafted TK-NOG and CD-1 mice dosed for a shorter period. Conclusion: These mechanistic biomarker findings can be leveraged in an in vitro model and in a more routine preclinical model (CD-1 mice) to identify nucleosides with such a FIAU-like mitochondrial toxicity mechanistic liability potential. Further optimization of the TK-NOG hu-liver mouse model is necessary before broader adoption for drug safety testing.

3.
Toxicol Sci ; 181(2): 148-159, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33837425

ABSTRACT

A new safety testing paradigm that relies on gene expression biomarker panels was developed to easily and quickly identify drug-induced injuries across tissues in rats prior to drug candidate selection. Here, we describe the development, qualification, and implementation of gene expression signatures that diagnose tissue degeneration/necrosis for use in early rat safety studies. Approximately 400 differentially expressed genes were first identified that were consistently regulated across 4 prioritized tissues (liver, kidney, heart, and skeletal muscle), following injuries induced by known toxicants. Hundred of these "universal" genes were chosen for quantitative PCR, and the most consistent and robustly responding transcripts selected, resulting in a final 22-gene set from which unique sets of 12 genes were chosen as optimal for each tissue. The approach was extended across 4 additional tissues (pancreas, gastrointestinal tract, bladder, and testes) where toxicities are less common. Mathematical algorithms were generated to convert each tissue's 12-gene expression values to a single metric, scaled between 0 and 1, and a positive threshold set. For liver, kidney, heart, and skeletal muscle, this was established using a training set of 22 compounds and performance determined by testing a set of approximately 100 additional compounds, resulting in 74%-94% sensitivity and 94%-100% specificity for liver, kidney, and skeletal muscle, and 54%-62% sensitivity and 95%-98% specificity for heart. Similar performance was observed across a set of 15 studies for pancreas, gastrointestinal tract, bladder, and testes. Bundled together, we have incorporated these tissue signatures into a 4-day rat study, providing a rapid assessment of commonly seen compound liabilities to guide selection of lead candidates without the necessity to perform time-consuming histopathologic analyses.


Subject(s)
Gene Expression Profiling , Pharmaceutical Preparations , Animals , Liver , Rats , Risk Assessment , Transcriptome
4.
Toxicol Sci ; 156(2): 422-427, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28115646

ABSTRACT

The skeletal muscle (SKM) injury biomarkers, skeletal troponin I (sTnI), myosin light chain 3 (Myl3), and creatine kinase muscle isoform (Ckm) have been shown recently to be more sensitive and specific for monitoring drug-induced SKM injury than the conventional biomarkers, aspartate transaminase (AST) and creatine kinase (CK) enzymatic assays in rat toxicology studies. To evaluate the utility of these SKM biomarkers across species, they were assessed in 2 dog models: a drug-induced injury study in Beagle dogs and a 160 km endurance exercise run completed by Alaskan sled dogs. In the drug-induced injury model, mean sTnI and Myl3 plasma levels were 6- and 18-fold, respectively, compared with baseline as early as Study Day (SD) 15, while mean plasma AST and CK levels did not increase, and biopsy samples were non-remarkable for histopathology prior to SD 29 when degeneration was first noted. Peak group mean plasma responses over baseline for sTnI, Myl3, and Ckm biomarkers were 96-, 103-, and 11-fold, respectively, compared with 2.5-fold for AST and 3.8-fold for CK-enzymatic (CK-enz) assay. In the sled dog sustained exercise model, the peak response for all biomarkers was observed at the first sampling (2 h) after the completion of the run. The sTnI, Myl3, and Ckm mean fold peak values compared with baseline were 170-, 120-, and 150-fold, respectively, while AST increased 7-fold and CK-enz increased 29-fold. These findings support the conclusion that sTnI, Myl3, and Ckm are sensitive early tissue leakage biomarkers for monitoring SKM injury and effects of exercise in dog, extending their utility across preclinical species beyond the rat, and provide further support to investigate their translational utility to clinical trial settings to monitor for drug-induced SKM injury and ensure patient safety.


Subject(s)
Biomarkers/blood , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscular Diseases/blood , Physical Endurance , Animals , Creatine Kinase, MM Form/blood , Dogs , Male , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/etiology , Muscular Diseases/pathology , Myosin Light Chains/blood , Species Specificity , Troponin I/blood
5.
Toxicol Sci ; 150(1): 247-56, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26721300

ABSTRACT

Novel skeletal muscle (SKM) injury biomarkers that have recently been identified may outperform or add value to the conventional SKM injury biomarkers aspartate transaminase (AST) and creatine kinase (CK). The relative performance of these novel biomarkers of SKM injury including skeletal troponin I (sTnI), myosin light chain 3 (Myl3), CK M Isoform (Ckm), and fatty acid binding protein 3 (Fabp3) was assessed in 34 rat studies including both SKM toxicants and compounds with toxicities in tissues other than SKM. sTnI, Myl3, Ckm, and Fabp3 all outperformed CK or AST and/or added value for the diagnosis of drug-induced SKM injury (ie, myocyte degeneration/necrosis). In addition, when used in conjunction with CK and AST, sTnI, Myl3, CKm, and Fabp3 individually and collectively improved diagnostic sensitivity and specificity, as well as diagnostic certainty, for SKM injury and responded in a sensitive manner to low levels of SKM degeneration/necrosis in rats. These findings support the proposal that sTnI, Myl3, Ckm, and Fabp3 are suitable for voluntary use, in conjunction with CK and AST, in regulatory safety studies in rats to monitor drug-induced SKM injury and the potential translational use of these exploratory biomarkers in early clinical trials to ensure patient safety.


Subject(s)
Biomarkers/blood , Muscle, Skeletal/drug effects , Muscular Diseases/blood , Muscular Diseases/chemically induced , Animals , Creatine Kinase, MM Form/blood , Dose-Response Relationship, Drug , Fatty Acid Binding Protein 3 , Fatty Acid-Binding Proteins/blood , Female , Male , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Muscular Diseases/enzymology , Muscular Diseases/metabolism , Myosin Light Chains/blood , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Wistar , Research Design , Sensitivity and Specificity , Troponin I/blood
6.
Toxicol Sci ; 138(1): 3-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24361871

ABSTRACT

Novel urinary kidney safety biomarkers have been identified recently that may outperform or add value to the conventional renal function biomarkers, blood urea nitrogen (BUN) and serum creatinine (SCr). To assess the relative performance of the growing list of novel biomarkers, a comprehensive evaluation was conducted for 12 urinary biomarkers in 22 rat studies including 12 kidney toxicants and 10 compounds with toxicities observed in organs other than kidney. The kidney toxicity studies included kidney tubular toxicants and glomerular toxicants. The 12 urinary biomarkers evaluated included Kim-1, clusterin, osteopontin, osteoactivin, albumin, lipocalin-2, GST-α, ß2-microglobulin, cystatin C, retinol binding protein 4, total protein, and N-acetyl-ß-D-glucosaminidase. Receiver operator characteristic (ROC) curves were generated for each biomarker and for BUN and SCr to compare the relative performance of the 12 biomarkers in individual animals against the microscopic histomorphologic changes observed in the kidney. Among the kidney toxicity biomarkers analyzed, Kim-1, clusterin, and albumin showed the highest overall performance for detecting drug-induced renal tubular injury in the rat in a sensitive and specific manner, whereas albumin showed the highest performance in detecting drug-induced glomerular injury. Although most of the evaluated kidney biomarkers were more sensitive in detecting kidney toxicity compared with BUN and SCr, all biomarkers demonstrated some lack of specificity, most notably NGAL and osteopontin, illustrating the need for caution when interpreting urinary biomarker increases in rat samples when organ toxicity is unknown.


Subject(s)
Biomarkers/urine , Kidney Diseases/chemically induced , Kidney Diseases/urine , Kidney/drug effects , Toxicity Tests , Animals , Enzyme-Linked Immunosorbent Assay , Female , Kidney/pathology , Kidney Diseases/blood , Kidney Diseases/pathology , Limit of Detection , Male , ROC Curve , Rats , Rats, Sprague-Dawley , Rats, Wistar
7.
Toxicol Sci ; 130(2): 229-44, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872058

ABSTRACT

Alanine aminotransferase (ALT) activity is the most frequently relied upon reference standard for monitoring liver injury in humans and nonclinical species. However, limitations of ALT include a lack of specificity for diagnosing liver injury (e.g., present in muscle and the gastrointestinal tract), its inability to monitor certain types of hepatic injury (e.g., biliary injury), and ambiguity with respect to interpretation of modest or transient elevations (< 3× upper limit of normal). As an initial step to both understand and qualify additional biomarkers of hepatotoxicity that may add value to ALT, three novel candidates have been evaluated in 34 acute toxicity rat studies: (1) alpha-glutathione S-transferase (GSTA), (2) arginase 1 (ARG1), and (3) 4-hydroxyphenylpyruvate dioxygenase (HPD). The performance of each biomarker was assessed for its diagnostic ability to accurately detect hepatocellular injury (i.e., microscopic histopathology), singularly or in combination with ALT. All three biomarkers, either alone or in combination with ALT, improved specificity when compared with ALT alone. Hepatocellular necrosis and/or degeneration were detected by all three biomarkers in the majority of animals. ARG1 and HPD were also sensitive in detecting single-cell necrosis in the absence of more extensive hepatocellular necrosis/degeneration. ARG1 showed the best sensitivity for detecting biliary injury with or without ALT. All the biomarkers were able to detect biliary injury with single-cell necrosis. Taken together, these novel liver toxicity biomarkers, GSTA, ARG1, and HPD, add value (both enhanced specificity and sensitivity) to the measurement of ALT alone for monitoring drug-induced liver injury in rat.


Subject(s)
4-Hydroxyphenylpyruvate Dioxygenase/metabolism , Arginase/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Glutathione Transferase/metabolism , Isoenzymes/metabolism , Liver/enzymology , Alanine Transaminase/metabolism , Animals , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Linear Models , Liver/drug effects , Liver/pathology , Logistic Models , Male , Predictive Value of Tests , ROC Curve , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sensitivity and Specificity , Tissue Distribution
8.
Nat Biotechnol ; 28(5): 470-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20458317

ABSTRACT

The capacities of urinary trefoil factor 3 (TFF3) and urinary albumin to detect acute renal tubular injury have never been evaluated with sufficient statistical rigor to permit their use in regulated drug development instead of the current preclinical biomarkers serum creatinine (SCr) and blood urea nitrogen (BUN). Working with rats, we found that urinary TFF3 protein levels were markedly reduced, and urinary albumin were markedly increased in response to renal tubular injury. Urinary TFF3 levels did not respond to nonrenal toxicants, and urinary albumin faithfully reflected alterations in renal function. In situ hybridization localized TFF3 expression in tubules of the outer stripe of the outer medulla. Albumin outperformed either SCr or BUN for detecting kidney tubule injury and TFF3 augmented the potential of BUN and SCr to detect kidney damage. Use of urinary TFF3 and albumin will enable more sensitive and robust diagnosis of acute renal tubular injury than traditional biomarkers.


Subject(s)
Albuminuria/urine , Biomarkers, Pharmacological/urine , Kidney Diseases , Kidney Tubules/drug effects , Neuropeptides/urine , Animals , Carbapenems/toxicity , Cisplatin/toxicity , Gentamicins/toxicity , Histocytochemistry , Iridoid Glycosides , Iridoids/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/diagnosis , Kidney Tubules/pathology , Logistic Models , ROC Curve , Rats , Trefoil Factor-3
9.
Nat Biotechnol ; 28(5): 478-85, 2010 May.
Article in English | MEDLINE | ID: mdl-20458318

ABSTRACT

Kidney toxicity accounts both for the failure of many drug candidates as well as considerable patient morbidity. Whereas histopathology remains the gold standard for nephrotoxicity in animal systems, serum creatinine (SCr) and blood urea nitrogen (BUN) are the primary options for monitoring kidney dysfunction in humans. The transmembrane tubular protein kidney injury molecule-1 (Kim-1) was previously reported to be markedly induced in response to renal injury. Owing to the poor sensitivity and specificity of SCr and BUN, we used rat toxicology studies to compare the diagnostic performance of urinary Kim-1 to BUN, SCr and urinary N-acetyl-beta-D-glucosaminidase (NAG) as predictors of kidney tubular damage scored by histopathology. Kim-1 outperforms SCr, BUN and urinary NAG in multiple rat models of kidney injury. Urinary Kim-1 measurements may facilitate sensitive, specific and accurate prediction of human nephrotoxicity in preclinical drug screens. This should enable early identification and elimination of compounds that are potentially nephrotoxic.


Subject(s)
Biomarkers, Pharmacological/urine , Cell Adhesion Molecules/urine , Kidney Function Tests/methods , Kidney , Acetylglucosaminidase/urine , Animals , Biomarkers, Pharmacological/metabolism , Blood Urea Nitrogen , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cisplatin/toxicity , Creatinine/blood , Cyclosporine/toxicity , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions , Gentamicins/toxicity , Histocytochemistry , Kidney/drug effects , Kidney/injuries , Kidney Function Tests/standards , Male , Oligonucleotide Array Sequence Analysis , ROC Curve , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reperfusion Injury , Thioacetamide/toxicity
10.
Nat Biotechnol ; 28(5): 486-94, 2010 May.
Article in English | MEDLINE | ID: mdl-20458319

ABSTRACT

The Predictive Safety Testing Consortium's first regulatory submission to qualify kidney safety biomarkers revealed two deficiencies. To address the need for biomarkers that monitor recovery from agent-induced renal damage, we scored changes in the levels of urinary biomarkers in rats during recovery from renal injury induced by exposure to carbapenem A or gentamicin. All biomarkers responded to histologic tubular toxicities to varied degrees and with different kinetics. After a recovery period, all biomarkers returned to levels approaching those observed in uninjured animals. We next addressed the need for a serum biomarker that reflects general kidney function regardless of the exact site of renal injury. Our assay for serum cystatin C is more sensitive and specific than serum creatinine (SCr) or blood urea nitrogen (BUN) in monitoring generalized renal function after exposure of rats to eight nephrotoxicants and two hepatotoxicants. This sensitive serum biomarker will enable testing of renal function in animal studies that do not involve urine collection.


Subject(s)
Biomarkers, Pharmacological , Cystatin C/blood , Kidney Diseases/diagnosis , Kidney Function Tests/methods , Animals , Biomarkers, Pharmacological/blood , Biomarkers, Pharmacological/metabolism , Biomarkers, Pharmacological/urine , Blood Urea Nitrogen , Carbapenems/toxicity , Creatinine/blood , Drug-Related Side Effects and Adverse Reactions , Female , Gentamicins/toxicity , Kidney/drug effects , Kidney/metabolism , Male , ROC Curve , Rats , Rats, Sprague-Dawley , Rats, Wistar
11.
Clin Chem ; 55(11): 1977-83, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19745058

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are endogenous, small noncoding RNAs. Because of their size, abundance, tissue specificity, and relative stability in plasma, miRNAs hold promise as unique accessible biomarkers to monitor tissue injury. METHODS: We investigated the use of liver-, muscle- and brain-specific miRNAs as circulating biomarkers of tissue injury. We used a highly sensitive quantitative PCR assay to measure specific miRNAs (miR-122, miR-133a, and miR-124) in plasma samples from rats treated with liver or muscle toxicants and from a rat surgical model of stroke. RESULTS: We observed increases in plasma concentrations of miR-122, miR-133a, and miR-124 corresponding to injuries in liver, muscle, and brain, respectively. miR-122 and miR-133a illustrated specificity for liver and muscle toxicity, respectively, because they were not detectable in the plasma of animals with toxicity to the other organ. This result contrasted with the results for alanine aminotransferase (ALT) and aspartate aminotransferase, which were both increased with either organ toxicity. Furthermore, miR-122 exhibited a diagnostic sensitivity superior to that of ALT when the results were correlated to the liver histopathologic results. The miR-124 concentration increased in the plasma of rats 8 h after surgery to produce brain injury and peaked at 24 h, while the miR-122 and miR-133a concentrations remained at baseline values. CONCLUSIONS: These results demonstrate that tissue-specific miRNAs may serve as diagnostically sensitive plasma biomarkers of tissue injury.


Subject(s)
MicroRNAs/blood , Wounds and Injuries/diagnosis , Wounds and Injuries/genetics , Animals , Biomarkers/blood , Female , Heart Injuries/diagnosis , Heart Injuries/genetics , Kidney/injuries , Kidney/pathology , Liver/injuries , Liver/pathology , Male , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Stroke/diagnosis , Stroke/genetics
12.
Chem Res Toxicol ; 19(8): 986-98, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16918237

ABSTRACT

The identification of biomarkers for disease state, drug efficacy, and toxicity is becoming increasingly important for drug discovery and development. We have used two-dimensional differential in-gel electrophoresis and mass spectrometry to identify proteomic markers associated with hepatocellular steatosis in rats after dosing with a compound (CDA) in preclinical development. Rats were dosed daily for up to 5 days with CDA for measurement of blood biochemical parameters, histological, and proteomic analysis. Alterations in plasma glucose and liver transaminases were detected from dosing day 3 onward, and livers showed trace levels of hepatocellular vacuolation from 6 h which increased in extent and severity over the 5 day time course. The number of significantly altered protein spots increased over the 5 day time course, and Ingenuity Pathway Analysis showed that the predominant functions altered by CDA treatment were cell death and cellular assembly and organization. This included alterations in secreted proteins, endoplasmic reticulum and mitochondrial chaperones, antioxidant proteins, and enzymes involved in fatty acid biosynthesis. Comparative in vitro dosing studies showed similar alterations to the proteome, neutral lipid accumulation, and mitochondrial dehydrogenase activity in response to CDA treatment of cultured rat hepatocytes. The finding that several proteins showed significant changes in abundance before the onset of overt toxicity in vivo suggested that these could serve as predictive biomarkers of compounds with a propensity to induce liver steatosis. These markers underwent further direct analysis in the in vitro hepatocyte toxicity model to determine their utility in the development of high throughput assays for drug-induced steatosis.


Subject(s)
Fatty Liver/metabolism , Hepatocytes/metabolism , Proteome/biosynthesis , Proteomics/methods , Animals , Biomarkers/analysis , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Fatty Liver/chemically induced , Fatty Liver/pathology , Female , Fluorescent Dyes , Gene Expression , Hepatocytes/drug effects , Hepatocytes/pathology , Mass Spectrometry , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Proteome/analysis , Proteome/genetics , Rats , Time Factors
13.
Chem Res Toxicol ; 17(5): 605-12, 2004 May.
Article in English | MEDLINE | ID: mdl-15144217

ABSTRACT

A significant problem faced by pharmaceutical companies today is the failure of lead compounds in the later stages of development due to unexpected toxicities. We have used two-dimensional differential in-gel electrophoresis and mass spectrometry to identify a proteomic signature associated with hepatocellular steatosis in rats after dosing with a compound in preclinical development. Liver toxicity was monitored over a 5 day dosing regime using blood biochemical parameter measurements and histopathological analysis. As early as 6 h postdosing, livers showed hepatocellular vacuolation, which increased in extent and severity over the course of the study. Alterations in plasma glucose, alanine aminotransferase, and aspartate aminotransferase were not detected until the third day of dosing and changed in magnitude up to the final day. The proteomic changes were observed at the earliest time point, and many of these could be associated with known toxicological mechanisms involved in liver steatosis. This included up-regulation of pyruvate dehydrogenase, phenylalanine hydroxylase, and 2-oxoisovalerate dehydrogenase, which are involved in acetyl-CoA production, and down-regulation of sulfite oxidase, which could play a role in triglyceride accumulation. In addition, down-regulation of the chaperone-like protein, glucose-regulated protein 78, was consistent with the decreased expression of the secretory proteins serum paraoxonase, serum albumin, and peroxiredoxin IV. The correlation of these protein changes with the clinical and histological data and their occurrence before the onset of the biochemical changes suggest that they could serve as predictive biomarkers of compounds with a propensity to induce liver steatosis.


Subject(s)
Fatty Liver/metabolism , Proteomics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Acetyl Coenzyme A/metabolism , Animals , Aryldialkylphosphatase/metabolism , Biomarkers , Electrophoresis, Gel, Two-Dimensional/methods , Fatty Liver/chemically induced , Fatty Liver/enzymology , Fatty Liver/pathology , Female , Glucose/metabolism , Mass Spectrometry/methods , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Peroxidases/metabolism , Peroxiredoxins , Phenylalanine Hydroxylase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Rats , Serum Albumin/metabolism , Transaminases/metabolism , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL