Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Neurol ; 15: 1322647, 2024.
Article in English | MEDLINE | ID: mdl-38523617

ABSTRACT

Vestibular hair cells (HCs) are mechanoreceptors that sense head motions by modulating the firing rate of vestibular ganglion neurons (VGNs), whose central processes project to vestibular nucleus neurons (VNNs) and cerebellar neurons. We explored vestibular function after HC destruction in adult Pou4f3+/DTR (DTR) mice, in which injections of high-dose (50 ng/g) diphtheria toxin (DT) destroyed most vestibular HCs within 2 weeks. At that time, DTR mice had lost the horizontal vestibulo-ocular reflex (aVORH), and their VNNs failed to upregulate nuclear cFos expression in response to a vestibular stimulus (centrifugation). Five months later, 21 and 14% of HCs were regenerated in utricles and horizontal ampullae, respectively. The vast majority of HCs present were type II. This degree of HC regeneration did not restore the aVORH or centrifugation-evoked cFos expression in VNNs. The failure to regain vestibular pathway function was not due to degeneration of VGNs or VNNs because normal neuron numbers were maintained after HC destruction. Furthermore, sinusoidal galvanic stimulation at the mastoid process evoked cFos protein expression in VNNs, indicating that VGNs were able to regulate VNN activity after HC loss. aVORH and cFos responses in VNNs were robust after low-dose (25 ng/g) DT, which compared to high-dose DT resulted in a similar degree of type II HC death and regeneration but spared more type I HCs in both organs. These findings demonstrate that having more type I HCs is correlated with stronger responses to vestibular stimulation and suggest that regenerating type I HCs may improve vestibular function after HC loss.

2.
Clin Pharmacol Ther ; 114(3): 591-603, 2023 09.
Article in English | MEDLINE | ID: mdl-37429825

ABSTRACT

Genome editing holds the potential for curative treatments of human disease, however, clinical realization has proven to be a challenging journey with incremental progress made up until recently. Over the last decade, advances in clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) systems have provided the necessary breakthrough for genome editing in the clinic. The progress of investigational CRISPR therapies from bench to bedside reflects the culmination of multiple advances occurring in parallel, several of which intersect with clinical pharmacology and translation. Directing the CRISPR therapy to the intended site of action has necessitated novel delivery platforms, and this has resulted in special considerations for the complete characterization of distribution, metabolism, and excretion, as well as immunogenicity. Once at the site of action, CRISPR therapies aim to make permanent alterations to the genome and achieve therapeutically relevant effects with a single dose. This fundamental aspect of the mechanism of action for CRISPR therapies results in new considerations for clinical translation and dose selection. Early advances in model-informed development of CRISPR therapies have incorporated key facets of the mechanism of action and have captured hallmark features of clinical pharmacokinetics and pharmacodynamics from phase I investigations. Given the recent emergence of CRISPR therapies in clinical development, the landscape continues to evolve rapidly with ample opportunity for continued innovation. Here, we provide a snapshot of selected topics in clinical pharmacology and translation that has supported the advance of systemically administered in vivo and ex vivo CRISPR-based investigational therapies in the clinic.


Subject(s)
CRISPR-Cas Systems , Pharmacology, Clinical , Humans , CRISPR-Cas Systems/genetics , Gene Editing/methods
3.
J Appl Toxicol ; 42(10): 1570-1584, 2022 10.
Article in English | MEDLINE | ID: mdl-35393688

ABSTRACT

Inhibition of sodium-glucose cotransporter-2 (SGLT2) has been shown to be a safe and efficacious approach to support managing Type 2 diabetes. In the 2-year carcinogenicity study with the SGLT2 inhibitor empagliflozin in CD-1 mice, an increased incidence of renal tubular adenomas and carcinomas was identified in the male high-dose group but was not observed in female mice. An integrated review of available nonclinical data was conducted to establish a mode-of-action hypothesis for male mouse-specific tumorigenesis. Five key events were identified through systematic analysis to form the proposed mode-of-action: (1) Background kidney pathology in CD-1 mice sensitizes the strain to (2) pharmacology-related diuretic effects associated with SGLT2 inhib ition. (3) In male mice, metabolic demand increases with the formation of a sex- and species-specific empagliflozin metabolite. These features converge to (4) deplete oxidative stress handling reserve, driving (5) constitutive cellular proliferation in male CD-1 mice. The proposed mode of action requires all five key events for empagliflozin to present a carcinogenicity risk in the CD-1 mouse. Considering that empagliflozin is not genotoxic in the standard battery of genotoxicity tests, and not all five key events are present in the context of female mice, rats, or humans, nor for other osmotic diuretics or other SGLT2 inhibitors, the observed male mouse renal tumors are not considered relevant to humans.


Subject(s)
Carcinoma, Renal Cell , Diabetes Mellitus, Type 2 , Kidney Neoplasms , Sodium-Glucose Transporter 2 Inhibitors , Animals , Antigens, CD1/metabolism , Benzhydryl Compounds/toxicity , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Female , Glucosides , Humans , Hypoglycemic Agents/toxicity , Kidney , Kidney Neoplasms/chemically induced , Kidney Neoplasms/complications , Kidney Neoplasms/drug therapy , Male , Mice , Rats , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/toxicity
4.
Lab Chip ; 20(3): 468-476, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31989145

ABSTRACT

The human kidney contains approximately one million nephrons. As the functional unit of the kidney, the nephron affords an opportunity to approximate the kidney at a microphysiological scale. Recent emergence of physiologically accurate human tissue models has radically advanced the possibilities of mimicking organ biology and multi-organ combinations in vitro. Anatomically, the nephron is one of the most complex, sequentially integrated microfluidic units in the body making the miniaturized microfluidic systems excellent candidates for capturing the kidney biology in vitro. While these models are promising, there are a number of considerations for practical implementation into a drug development paradigm. Opportunities for pharmaceutical industry applications of new MPS models often start with drug safety testing. As such, the intent of this article is to focus on safety and ADME applications. This article reviews biological functions of the kidney and options for characterizing known roles in nephrotoxicity. The concept of "context-of-use" is introduced as a framework for describing and verifying the specific features of an MPS platform for use in drug development. Overall, we present a perspective on key attributes of microphysiological kidney models, which the pharmaceutical industry could leverage to improve confident safety and ADME evaluations of experimental therapies.


Subject(s)
Kidney/drug effects , Pharmaceutical Preparations/metabolism , Drug Development , Drug Evaluation, Preclinical/adverse effects , Drug Industry , Humans , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Models, Biological , Pharmaceutical Preparations/chemistry
5.
Lab Chip ; 20(3): 446-467, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31932816

ABSTRACT

Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.


Subject(s)
Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Pharmaceutical Preparations/metabolism , Animals , Drug Development , Drug Evaluation, Preclinical , Drug Industry , Humans , Microfluidic Analytical Techniques/instrumentation , Pharmaceutical Preparations/chemistry
6.
Toxicol Pathol ; 46(6): 671-682, 2018 08.
Article in English | MEDLINE | ID: mdl-29945496

ABSTRACT

An increased incidence of renal tubular adenomas and carcinomas was identified in the 2-year CD-1 mouse carcinogenicity study with empagliflozin (sodium-glucose transporter 2 inhibitor) in high dose (1,000 mg/kg/day) male mice. A 13-week mouse renal investigative pathogenesis study was conducted with empagliflozin to evaluate dose dependency and temporal onset of nonneoplastic degenerative/regenerative renal tubular and molecular (genes, pathways) changes which precede neoplasia. Male and female CD-1 mice were given daily oral doses of 0, 100, 300, or 1,000 mg/kg/day (corresponding carcinogenicity study dose levels) for 1, 2, 4, 8, or 13 weeks. The maximum expected pharmacology with secondary osmotic diuresis was observed by week 1 at ≥100 mg/kg/day in both genders. Histopathologic kidney changes were first detected after 4 weeks of dosing in the male 1,000 mg/kg/day dose group, with progressive increases in the incidence and/or number of findings in this dose group so that they were more readily detected during weeks 8 and 13. Changes detected starting on week 4 consisted of minimal single-cell necrosis and minimal increases in mitotic figures. These changes persisted at an increased incidence at weeks 8 and 13 and were accompanied by minimal to mild tubular epithelial karyomegaly, minimal proximal convoluted tubular epithelial cell hyperplasia, and a corresponding increase in Ki-67-positive nuclei in epithelial cells of the proximal convoluted tubules. There were no corresponding changes in serum chemistry or urinalysis parameters indicative of any physiologically meaningful effect on renal function and thus these findings were not considered to be adverse. Similar changes were not identified in lower-dose groups in males nor were they present in females of any dose group. RNA-sequencing analysis revealed male mouse-specific changes in kidney over 13 weeks of dosing at 1,000 mg/kg/day. Treatment-related changes included genes and pathways related to p53-regulated cell cycle and proliferation, transforming growth factor ß, oxidative stress, and renal injury and the number of genes with significant expression change dramatically increased at week 13. These treatment-related changes in genes and pathways were predominant in high-dose males and complemented the observed temporal renal tubular changes. Overall, these mouse investigative study results support the role of early empagliflozin-related degenerative/regenerative changes only observed in high-dose male CD-1 mice as a key contributing feature to a nongenotoxic mode of renal tumor pathogenesis.


Subject(s)
Benzhydryl Compounds/toxicity , Glucosides/toxicity , Kidney Diseases/chemically induced , Kidney Tubules/drug effects , Precancerous Conditions/chemically induced , Sodium-Glucose Transporter 2 Inhibitors/toxicity , Administration, Oral , Animals , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/blood , Dose-Response Relationship, Drug , Female , Glucosides/administration & dosage , Glucosides/blood , Kidney Diseases/pathology , Kidney Function Tests , Kidney Tubules/pathology , Male , Mice, Inbred Strains , Necrosis , Precancerous Conditions/pathology , Sex Factors , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/blood , Toxicity Tests, Subchronic , Toxicokinetics , Transcriptome/drug effects
7.
Int J Toxicol ; 36(6): 440-448, 2017.
Article in English | MEDLINE | ID: mdl-29130831

ABSTRACT

In a previously reported CD-1 mouse 2-year carcinogenicity study with the sodium glucose cotransporter-2 inhibitor empagliflozin, an increased incidence of renal tubular adenomas and carcinomas was identified only in the male high-dose group. Follow-up investigative studies have shown that the renal tumors in male high-dose mice were preceded by a number of renal degenerative/regenerative findings. Prior cross-species in vitro metabolism studies using microsomes identified an oxidative metabolite (M466/2) predominantly formed in the male mouse kidney and which spontaneously degrades to a metabolite (M380/1) and reactive 4-OH crotonaldehyde (CTA). In order to further evaluate potential modes of action for empagliflozin-associated male mouse renal tumors, we report here a series of in vitro investigative toxicology studies conducted to evaluate the cytotoxic and genotoxic potential of empagliflozin and M466/2. To assess the cytotoxic potential of empagliflozin and M466/2, a primary mouse renal tubular epithelial (mRTE) cell model was used. In mRTE cells, M466/2-derived in vitro 4-OH CTA exposure was cytotoxic, while empagliflozin was not cytotoxic or mitogenic. Empagliflozin and M466/2 were not genotoxic, supporting an indirect mode of action for empagliflozin-associated male mouse renal tumorigenesis. In conclusion, these in vitro data show that M466/2-derived 4-OH CTA exposure is associated with cytotoxicity in renal tubule cells and may be involved in promoting compound-related in vivo renal metabolic stress and chronic low-level renal injury, in turn supporting a nongenotoxic mode of tumor pathogenesis specific to the male mouse.


Subject(s)
Benzhydryl Compounds/metabolism , Benzhydryl Compounds/toxicity , Glucosides/metabolism , Glucosides/toxicity , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/toxicity , Kidney Tubules/drug effects , Oxidative Stress/drug effects , Animals , Benzhydryl Compounds/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Glucosides/chemistry , Hypoglycemic Agents/chemistry , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice, Inbred Strains , Micronuclei, Chromosome-Defective/drug effects , Structure-Activity Relationship
8.
Chem Res Toxicol ; 30(10): 1823-1834, 2017 10 16.
Article in English | MEDLINE | ID: mdl-28885000

ABSTRACT

Drug-induced kidney injury (DIKI) is a common toxicity observed in pharmaceutical development. We demonstrated the use of label-free liquid chromatography-mass spectrometry (LC-MS) and multiplex liquid chromatography-single reaction monitoring (LC-SRM) as practical extensions of standard immunoassay based safety biomarker assessments for identification of new toxicity marker candidates and for improved mechanistic understanding. Two different anticancer drugs, doxorubicin (DOX) and cisplatin (cis-diamminedichloridoplatinum, CDDP), were chosen as the toxicants due to their different modes of nephrotoxicity. Analyses of urine samples from toxicant treated and untreated rats were compared to identify biochemical analytes that changed in response to toxicant exposure. A discovery (label-free LC-MS) and targeted proteomics (multiplex LC-SRM) approach was used in combination with well established immunoassay experiments for the identification of a panel of urinary protein markers related to drug induced nephrotoxicity in rats. The initial generation of an expanded set of markers was accomplished using the label-free LC-MS discovery screen and ELISA based analysis of six nephrotoxicity biomarker proteins. Diagnostic performance of the expanded analyte set was statistically compared to conventional nephrotoxicity biomarkers. False discovery rate (FDR) analysis revealed 18 and 28 proteins from the CDDP and DOX groups, respectively, exhibiting significant differences between the vehicle and treated groups. Multiplex SRM assays were constructed to more precisely quantify candidate markers selected from the discovery screen and immunoassay experiments. To evaluate the sensitivity and specificity for each of the candidate biomarkers, histopathology severity scores were used as a benchmark for renal injury followed by receiver-operating characteristic (ROC) curve analysis on selected biomarkers. Further examination of the best performing analytes revealed relevant biological significance after consideration of anatomical localization and functional roles. In summary, the inclusion of mass spectrometry together with conventional ELISA based assays resulted in the identification of an expanded set of biomarkers with a realistic potential for providing additional beneficial information in mechanistic investigations of drug induced kidney injury and with similar responsiveness to conventionally applied indicators of renal injury.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Doxorubicin/toxicity , Drug Discovery , Kidney Diseases/chemically induced , Animals , Antineoplastic Agents/chemistry , Biomarkers/analysis , Chromatography, Liquid , Cisplatin/chemistry , Doxorubicin/chemistry , Enzyme-Linked Immunosorbent Assay , Kidney Diseases/pathology , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley
9.
Exp Biol Med (Maywood) ; 242(16): 1579-1585, 2017 10.
Article in English | MEDLINE | ID: mdl-28622731

ABSTRACT

Tissue chips are poised to deliver a paradigm shift in drug discovery. By emulating human physiology, these chips have the potential to increase the predictive power of preclinical modeling, which in turn will move the pharmaceutical industry closer to its aspiration of clinically relevant and ultimately animal-free drug discovery. Despite the tremendous science and innovation invested in these tissue chips, significant challenges remain to be addressed to enable their routine adoption into the industrial laboratory. This article describes the main steps that need to be taken and highlights key considerations in order to transform tissue chip technology from the hands of the innovators into those of the industrial scientists. Written by scientists from 13 pharmaceutical companies and partners at the National Institutes of Health, this article uniquely captures a consensus view on the progression strategy to facilitate and accelerate the adoption of this valuable technology. It concludes that success will be delivered by a partnership approach as well as a deep understanding of the context within which these chips will actually be used. Impact statement The rapid pace of scientific innovation in the tissue chip (TC) field requires a cohesive partnership between innovators and end users. Near term uptake of these human-relevant platforms will fill gaps in current capabilities for assessing important properties of disposition, efficacy and safety liabilities. Similarly, these platforms could support mechanistic studies which aim to resolve challenges later in development (e.g. assessing the human relevance of a liability identified in animal studies). Building confidence that novel capabilities of TCs can address real world challenges while they themselves are being developed will accelerate their application in the discovery and development of innovative medicines. This article outlines a strategic roadmap to unite innovators and end users thus making implementation smooth and rapid. With the collective contributions from multiple international pharmaceutical companies and partners at National Institutes of Health, this article should serve as an invaluable resource to the multi-disciplinary field of TC development.


Subject(s)
Drug Evaluation, Preclinical/methods , Microchip Analytical Procedures/methods , Microfluidics/methods , Drug Industry , Humans , Lab-On-A-Chip Devices
10.
Toxicol Appl Pharmacol ; 320: 1-7, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28167222

ABSTRACT

The human testis is sensitive to toxicant-induced injury but current methods for detecting adverse effects are limited, insensitive and unreliable. Animal studies use sensitive histopathological endpoints to assess toxicity, but require testicular tissue that is not available during human clinical trials. More sensitive and reliable molecular biomarkers of testicular injury are needed to better monitor testicular toxicity in both clinical and preclinical. Adult male Wistar Han rats were exposed for 4weeks to compounds previously associated with testicular injury, including cisplatin (0, 0.2, 0.3, or 0.4mg/kg/day), BI665915 (0, 20, 70, 100mg/kg/d), BI665636 (0, 20, 100mg/kg/d) or BI163538 (0, 70, 150, 300mg/kg/d) to evaluate reproductive toxicity and assess changes in sperm mRNA levels. None of the compounds resulted in any significant changes in body, testis or epididymis weights, nor were there decreases in testicular homogenization resistant spermatid head counts. Histopathological evaluation found that only BI665915 treatment caused any testicular effects, including minor germ cell loss and disorganization of the seminiferous tubule epithelium, and an increase in the number of retained spermatid heads. A custom PCR-array panel was used to assess induced changes in sperm mRNA. BI665915 treatment resulted in a significant increase in clusterin (Clu) levels and decreases in GTPase, IMAP family member 4 (Gimap4), prostaglandin D2 synthase (Ptgds) and transmembrane protein with EGF like and two follistatin like domains 1 (Tmeff1) levels. Correlation analysis between transcript levels and quantitative histopathological endpoints found a modest association between Clu with retained spermatid heads. These results demonstrate that sperm mRNA levels are sensitive molecular indicators of testicular injury that can potentially be translated into a clinical setting.


Subject(s)
Acetamides/toxicity , Cisplatin/toxicity , Oxadiazoles/toxicity , RNA, Messenger/biosynthesis , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/drug effects , Testis/metabolism , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Male , Organ Size/drug effects , Organ Size/physiology , Rats , Rats, Wistar , Spermatogenesis/drug effects , Spermatogenesis/physiology , Spermatozoa/pathology , Testis/pathology
11.
Toxicol Sci ; 156(1): 11-13, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27815493

ABSTRACT

One of the goals of the Critical Path Institute's Predictive Safety Testing Consortium (PSTC) is to promote best practices for evaluating novel markers of drug induced injury. This includes the use of sound statistical methods. For rat studies, these practices have centered around comparing the area under the receiver-operator characteristic curve for each novel injury biomarker to those for the standard markers. In addition, the PSTC has previously used the net reclassification index (NRI) and integrated discrimination index (IDI) to assess the increased certainty provided by each novel injury biomarker when added to the information already provided by the standard markers. Due to their relatively simple interpretations, NRI and IDI have generally been popular measures of predictive performance. However recent literature suggests that significance tests for NRI and IDI can have inflated false positive rates and thus, tests based on these metrics should not be relied upon. Instead, when parametric models are employed to assess the added predictive value of a new marker, following (Pepe, M. S., Kerr, K. F., Longton, G., and Wang, Z. (2013). Testing for improvement in prediction model performance. Stat. Med. 32, 1467-1482), the PSTC recommends that likelihood based methods be used for significance testing.


Subject(s)
Biomarkers/metabolism , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drugs, Investigational/adverse effects , Models, Statistical , Toxicity Tests , Xenobiotics/toxicity , Animals , Biomarkers/blood , Biomarkers/urine , Drug Evaluation, Preclinical/trends , Drug-Related Side Effects and Adverse Reactions/blood , Drug-Related Side Effects and Adverse Reactions/metabolism , Drug-Related Side Effects and Adverse Reactions/urine , Drugs, Investigational/classification , False Positive Reactions , Humans , Muscular Diseases/chemically induced , Muscular Diseases/diagnosis , Muscular Diseases/metabolism , Organizations, Nonprofit , Predictive Value of Tests , ROC Curve , Renal Insufficiency/chemically induced , Renal Insufficiency/diagnosis , Renal Insufficiency/metabolism , Toxicity Tests/trends , United States , Xenobiotics/classification
12.
Toxicol Sci ; 151(2): 214-23, 2016 06.
Article in English | MEDLINE | ID: mdl-27026710

ABSTRACT

Traditional kidney biomarkers are insensitive indicators of acute kidney injury, with meaningful changes occurring late in the course of injury. The aim of this work was to demonstrate the diagnostic potential of urinary osteopontin (OPN) and neutrophil gelatinase-associated lipocalin (NGAL) for drug-induced kidney injury (DIKI) in rats using data from a recent regulatory qualification submission of translational DIKI biomarkers and to compare performance of NGAL and OPN to five previously qualified DIKI urinary biomarkers. Data were compiled from 15 studies of 11 different pharmaceuticals contributed by Critical Path Institute's Predictive Safety Testing Consortium (PSTC) Nephrotoxicity Working Group (NWG). Rats were given doses known to cause DIKI or other target organ toxicity, and urinary levels of the candidate biomarkers were assessed relative to kidney histopathology and serum creatinine (sCr) and blood urea nitrogen (BUN).OPN and NGAL outperformed sCr and BUN in identifying DIKI manifested as renal tubular epithelial degeneration or necrosis. In addition, urinary OPN and NGAL, when used with sCr and BUN, increased the ability to detect renal tubular epithelial degeneration or necrosis. NGAL and OPN had comparable or improved performance relative to Kim-1, clusterin, albumin, total protein, and beta-2 microglobulin. Given these data, both urinary OPN and NGAL are appropriate for use with current methods for assessing nephrotoxicity to identify and monitor DIKI in regulatory toxicology studies in rats. These data also support exploratory use of urinary OPN and NGAL in safety monitoring strategies of early clinical trials to aid in the assurance of patient safety.


Subject(s)
Acute Kidney Injury/diagnosis , Acute-Phase Proteins/urine , Lipocalins/urine , Osteopontin/urine , Proto-Oncogene Proteins/urine , Acute Kidney Injury/blood , Acute Kidney Injury/chemically induced , Acute Kidney Injury/urine , Animals , Area Under Curve , Biomarkers/blood , Biomarkers/urine , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Lipocalin-2 , Predictive Value of Tests , ROC Curve , Rats , Reproducibility of Results , Urinalysis
13.
Article in English | MEDLINE | ID: mdl-23364888

ABSTRACT

Although histopathology is considered the gold standard for assessing testicular toxicity in the nonclinical setting, identification of noninvasive biomarkers for testicular injury are desirable to improve safety monitoring capabilities for clinical trials. Inhibin B has been investigated as a noninvasive biomarker for testicular toxicity. This study investigates the correlation of Inhibin B in Wistar Han rats with the onset and reversibility of testicular histopathology from classical testicular toxicants carbendazim, cetrorelix acetate (CTX), and 1,2-dibromo-3-chloropropane (DBCP). The dose regimen included Interim (day 8), Drug (day 29), and nondosing Recovery (day 58) Phases. Inhibin B was not effective at predicting the onset of carbendazim- or CTX-mediated testicular pathology in rats. Inhibin B was reduced by DBCP administration at the end of the Drug Phase only, acting as a leading indicator of the onset of testicular toxicity before the onset of germ cell depletion. However, since Inhibin B was only decreased at the end of the Dosing Phase and not at the Recovery Phase, when the onset of testicular pathology occurred, it is unclear if monitoring Inhibin B would provide sufficient advanced warning for the onset of testicular pathology. Furthermore, follicle stimulating hormone was decreased following CTX and DBCP administration in the Interim Phase and CTX in the Drug Phase. Inhibin B has limited predictive capacity as a leading testicular biomarker in rats.


Subject(s)
Benzimidazoles/toxicity , Carbamates/toxicity , Gonadotropin-Releasing Hormone/analogs & derivatives , Inhibins/blood , Propane/analogs & derivatives , Testis/pathology , Animals , Benzimidazoles/administration & dosage , Biomarkers/blood , Body Weight/drug effects , Carbamates/administration & dosage , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/toxicity , Luteinizing Hormone/blood , Male , Organ Size/drug effects , Propane/administration & dosage , Propane/toxicity , Rats , Rats, Wistar , Survival Analysis , Testis/drug effects , Testis/metabolism
14.
Nat Biotechnol ; 28(5): 446-54, 2010 May.
Article in English | MEDLINE | ID: mdl-20458314

ABSTRACT

Application of any new biomarker to support safety-related decisions during regulated phases of drug development requires provision of a substantial data set that critically assesses analytical and biological performance of that biomarker. Such an approach enables stakeholders from industry and regulatory bodies to objectively evaluate whether superior standards of performance have been met and whether specific claims of fit-for-purpose use are supported. It is therefore important during the biomarker evaluation process that stakeholders seek agreement on which critical experiments are needed to test that a biomarker meets specific performance claims, how new biomarker and traditional comparators will be measured and how the resulting data will be merged, analyzed and interpreted.


Subject(s)
Biomarkers , Drug Discovery , Pharmaceutical Preparations , Animals , Drug Discovery/legislation & jurisprudence , Drug Discovery/methods , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/standards
15.
Nat Biotechnol ; 28(5): 455-62, 2010 May.
Article in English | MEDLINE | ID: mdl-20458315

ABSTRACT

The first formal qualification of safety biomarkers for regulatory decision making marks a milestone in the application of biomarkers to drug development. Following submission of drug toxicity studies and analyses of biomarker performance to the Food and Drug Administration (FDA) and European Medicines Agency (EMEA) by the Predictive Safety Testing Consortium's (PSTC) Nephrotoxicity Working Group, seven renal safety biomarkers have been qualified for limited use in nonclinical and clinical drug development to help guide safety assessments. This was a pilot process, and the experience gained will both facilitate better understanding of how the qualification process will probably evolve and clarify the minimal requirements necessary to evaluate the performance of biomarkers of organ injury within specific contexts.


Subject(s)
Biomarkers, Pharmacological , Drug Approval/legislation & jurisprudence , Kidney , Animals , Drug-Related Side Effects and Adverse Reactions , Europe , Humans , Kidney/drug effects , Kidney/injuries , Pharmaceutical Preparations/standards , United States , United States Food and Drug Administration
16.
Matrix Biol ; 27(7): 609-18, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18619537

ABSTRACT

The mammalian skeleton adjusts bone structure and strength in response to changes in mechanical loading, however the molecular and cellular mechanisms governing this process in vivo are unknown. Terminally differentiated osteoblasts, the osteocytes, are presumptive mechanosensory cells for bone, and cell culture studies demonstrate that beta1 integrins participate in mechanical signaling. To determine the role of beta1 integrins in osteoblasts in vivo, we used the Cre-lox system to delete beta1 integrin from cells committed to the osteoblast lineage. While pCol2.3 Cre-mediated recombination was widespread in bones from Colalpha1(I)-Cre+/beta1fl/fl conditional knockout mice (cKO), beta1 integrin protein was depleted from cortical osteocytes, but not from cancellous osteocytes or cells lining bone surfaces in adults. Bones from cKO mice that were normally loaded were similar in structure to WT littermates. However, hindlimb unloading of adult cKO mice for one week intended to cause bone loss (disuse osteopenia), resulted in unexpected, rapid changes in the geometry of cortical bone; hindlimb unloading increased the cross-sectional area, marrow area, and moments of inertia in cKO, but not WT mice. Furthermore, these hindlimb unloading-induced geometric changes in cortical bone of cKO mice resulted in increased whole bone bending stiffness and strength of the femur. Together, these results confirmed the concept that osteocytes are mechanosensory cells and showed beta1 integrins in cortical osteocytes limited changes in cortical geometry in response to disuse, thus providing the first in vivo evidence that beta1 integrins on osteocytes mediate specific aspects of mechanotransduction.


Subject(s)
Integrin beta1/physiology , Osteocytes/metabolism , Acute Disease , Animals , Bone Diseases, Metabolic , Female , Gene Deletion , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Mice, Knockout , Models, Biological , Models, Genetic , Osteoblasts/metabolism , Signal Transduction , Tensile Strength , Tissue Distribution
17.
Methods Mol Biol ; 341: 103-9, 2006.
Article in English | MEDLINE | ID: mdl-16799192

ABSTRACT

Mucosal tissues undergo contraction and relaxation on a continuous basis. In its normal state, the pliable intestinal tract is characterized by a rhythmic pattern of contractions controlled by its intrinsic neuronal innvervation. In chronic inflammatory diseases such as Crohn's disease, the intestine can become stiff and fibrotic, losing much of its normal motility. Although muscle fiber contraction accounts for much of this activity, contraction of nonmuscle tissue is constantly occurring in events associated with chronic inflammation, such as wound healing, scar formation, and tissue remodeling. However, the physiological and pathological mechanisms defining these events are not well defined. Tissue contraction is a dynamic event characterized by both intracellular and extracellular events. A number of cells, such as fibroblasts, epithelial cells, lymphocytes, and eosinophils, normally reside within the gastrointestinal tract. Additionally, the extracellular matrix is composed of a complex infrastructure that includes collagen and other molecules. The manner in which these two components interact is not certain, but the use of recent model systems has provided insights into these processes. The collagen lattice contraction assay provides a model for tissue contraction that takes advantage of the finding that cell-populated collagen hydrogels contract over time in a predictable, consistent manner. This model allows for investigation of the influence of specific agonists on the rate and extent of matrix contraction.


Subject(s)
Biological Assay , Collagen/chemistry , Extracellular Matrix/chemistry , Models, Biological , Biological Assay/methods , Caco-2 Cells , Collagen/metabolism , Crohn Disease/metabolism , Crohn Disease/pathology , Extracellular Matrix/metabolism , Gels/chemistry , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/chemistry , Intestines/pathology
18.
Exp Cell Res ; 310(1): 79-87, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16098964

ABSTRACT

Cell-matrix interactions transmit a wealth of information about the extracellular environment. In return, a variety of responses from the cell are initiated by changes in the matrix. One such response involves the positive regulation of matrix metalloproteinases (MMPs) by alpha2beta1 integrin attaching to a specific extracellular matrix component, collagen. This study explores the relationship between mechanical and biochemical functions of alpha2beta1 integrins as it pertains to regulating matrix remodeling. To understand this relationship, the individual influences of MMP activity and alpha2beta1 integrin function on collagen gel contraction were studied. We have observed little evidence of mutual participation in matrix remodeling by the alpha2beta1 integrin and MMP activity in cell models where alpha2 is minimally expressed. In cells expressing high levels of alpha2, we see an increase in gel contraction that is enhanced by MMP activity. Measuring tension as it builds within the gel reveals that alpha2beta1 integrin presence correlates with force output but is insensitive to MMP activity. These data strongly suggest that alpha2beta1 regulates collagen gel remodeling through multiple simultaneous mechanisms including force generation and modulation of MMP activity.


Subject(s)
Bone Remodeling , Collagen/metabolism , Extracellular Matrix/enzymology , Integrin alpha2beta1/metabolism , Matrix Metalloproteinases/metabolism , Cell Line, Tumor , DNA, Complementary/biosynthesis , Extracellular Matrix/metabolism , Humans , Integrin alpha2beta1/biosynthesis , Integrin alpha2beta1/genetics
19.
Biochem Biophys Res Commun ; 312(3): 725-32, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14680825

ABSTRACT

The extracellular matrix not only provides a structural scaffold for cells to inhabit but also forms a conduit by which mechanical information may be transmitted. Fibroblasts undergo a variety of changes when activated, including upregulating matrix metalloproteinase (MMP) activity and establishing a smooth muscle-like contractile apparatus. The relationship between MMP activity and matrix contraction has yet to be established. Here we report that inhibition of MMP activity correlates with a significant reduction in collagen gel contraction, however, force development does not change respective to MMP activity. These results suggest cellular controls of contractile forces are independent of MMP activity. Our results also raise the possibility that the material properties of the matrix dynamically change during remodeling.


Subject(s)
Cell Culture Techniques/methods , Collagen Type I/physiology , Extracellular Matrix/physiology , Fibroblasts/physiology , Matrix Metalloproteinases/physiology , Mechanotransduction, Cellular/physiology , Molecular Motor Proteins/physiology , Adaptation, Physiological/physiology , Animals , Cell Communication/drug effects , Cell Communication/physiology , Cell Count , Cell Culture Techniques/instrumentation , Cells, Cultured , Cytochalasin B/pharmacology , Dipeptides/pharmacology , Enzyme Activation/drug effects , Extracellular Matrix/drug effects , Fibroblasts/drug effects , HeLa Cells , Humans , Matrix Metalloproteinase Inhibitors , Mechanotransduction, Cellular/drug effects , Molecular Motor Proteins/drug effects , Muscle Contraction/drug effects , Muscle Contraction/physiology , Rats , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...