105 results on '"Scott CW"'
Search Results
2. A study of a patient with chronic lymphocytic leukemia (CLL) which demonstrates that proliferation of the lymphocytic clone in CLL does not include T lymphocytes
- Author
-
C.W. Scott, Josef T. Prchal, G. Lucivero, A.J. Carroll, Alexander R. Lawton, Prchal, Jt, Lucivero, Giacomo, Carroll, Aj, Lawton, Ar, and Scott, Cw
- Subjects
Electrophoresis ,CD20 ,T-Lymphocytes ,Chronic lymphocytic leukemia ,Immunology ,Clone (cell biology) ,Glucosephosphate Dehydrogenase ,Middle Aged ,Biology ,Lymphocyte Activation ,medicine.disease ,Virology ,Antibodies, Anti-Idiotypic ,Leukemia, Lymphoid ,Pathology and Forensic Medicine ,Phenotype ,biology.protein ,medicine ,Humans ,Immunology and Allergy ,Female - Published
- 1979
3. A microfluidic system that replicates pharmacokinetic (PK) profiles in vitro improves prediction of in vivo efficacy in preclinical models.
- Author
-
Singh D, Deosarkar SP, Cadogan E, Flemington V, Bray A, Zhang J, Reiserer RS, Schaffer DK, Gerken GB, Britt CM, Werner EM, Gibbons FD, Kostrzewski T, Chambers CE, Davies EJ, Montoya AR, Fok JHL, Hughes D, Fabre K, Wagoner MP, Wikswo JP, and Scott CW
- Subjects
- Animals, DNA, Humans, Mice, Models, Biological, Cell Culture Techniques, Microfluidics
- Abstract
Test compounds used on in vitro model systems are conventionally delivered to cell culture wells as fixed concentration bolus doses; however, this poorly replicates the pharmacokinetic (PK) concentration changes seen in vivo and reduces the predictive value of the data. Herein, proof-of-concept experiments were performed using a novel microfluidic device, the Microformulator, which allows in vivo like PK profiles to be applied to cells cultured in microtiter plates and facilitates the investigation of the impact of PK on biological responses. We demonstrate the utility of the device in its ability to reproduce in vivo PK profiles of different oncology compounds over multiweek experiments, both as monotherapy and drug combinations, comparing the effects on tumour cell efficacy in vitro with efficacy seen in in vivo xenograft models. In the first example, an ERK1/2 inhibitor was tested using fixed bolus dosing and Microformulator-replicated PK profiles, in 2 cell lines with different in vivo sensitivities. The Microformulator-replicated PK profiles were able to discriminate between cell line sensitivities, unlike the conventional fixed bolus dosing. In a second study, murine in vivo PK profiles of multiple Poly(ADP-Ribose) Polymerase 1/2 (PARP) and DNA-dependent protein kinase (DNA-PK) inhibitor combinations were replicated in a FaDu cell line resulting in a reduction in cell growth in vitro with similar rank ordering to the in vivo xenograft model. Additional PK/efficacy insight into theoretical changes to drug exposure profiles was gained by using the Microformulator to expose FaDu cells to the DNA-PK inhibitor for different target coverage levels and periods of time. We demonstrate that the Microformulator enables incorporating PK exposures into cellular assays to improve in vitro-in vivo translation understanding for early therapeutic insight., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: DS, AB, TK, DH are employees of CN Bio and hold stock or options in the company. CN Bio licenses intellectual property from Vanderbilt University related to the microfluidic device described in this study. SPD, EC, VF, JZ, FG, ARM, CEC, EJD, JHLF, KF, MPW, CWS are or were employees of AstraZeneca at the time of conducting these studies. EC, VF, JZ, ARM, CEC, CS hold stock in AstraZeneca.
- Published
- 2022
- Full Text
- View/download PDF
4. Cross-ministry data on service use and limitations faced by children in special education.
- Author
-
Russell MJ, Michael Scott CW, Murias K, Ben Gibbard W, Cui X, Tough S, and Zwicker JD
- Subjects
- Child, Education, Special, Humans, Autistic Disorder, Disabled Children, Intellectual Disability
- Abstract
Background: Children with disabilities often face limitations that cross support sectors., Objective: Our aim was to measure cross-ministry service use, outcomes, and functional limitations faced by children who qualified for special education., Methods: We used longitudinal British Columbia ministry data linked to children (0-18y) registered in K-12 education. Children were grouped by special education funding (most to least; Level 1, Level 2, Level 3, Unfunded, and no special education), and related to 1) service use patterns, 2) the age they first used disability services, and 3) functional limitations reported in health visits. We also reported how length of special education use related to disability service use., Results: Of 111,274 children, 154(0.1%) were Level 1, 4427(4.0%) Level 2, 2897(2.6%) Level 3, 13472(12.1%) Unfunded, and 90324(81.2%) not in special education. Children with higher funding levels, compared to lower levels of funding, generally were more likely to experience poorer outcomes, have functional limitations, have service needs, and receive early support. One exception was children with serious behavioral/mental health special education coding, which had poorer outcomes for their level of funding. Children received child disability supports early (about half of users started by 4y), but use was mostly limited to those with many years (9+years) of funded special education (70.7% of the all users) and biased to certain special education codes (i.e., Level 1, severe intellectual disability, and autism)., Conclusions: This study provides evidence of the long-term, diverse needs of children in special education and may be used to inform decisions surrounding their support., (Crown Copyright © 2021. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
5. Human ileal organoid model recapitulates clinical incidence of diarrhea associated with small molecule drugs.
- Author
-
Belair DG, Visconti RJ, Hong M, Marella M, Peters MF, Scott CW, and Kolaja KL
- Subjects
- Drug-Related Side Effects and Adverse Reactions, Humans, Pharmaceutical Preparations, Diarrhea chemically induced, Ileum, Models, Biological, Organoids
- Abstract
Drug-induced gastrointestinal toxicity (GIT) is a common treatment-emergent adverse event that can negatively impact dosing, thereby limiting efficacy and treatment options for patients. An in vitro assay of GIT is needed to address patient variability, mimic the microphysiology of the gut, and accurately predict drug-induced GIT. Primary human ileal organoids (termed 'enteroids') have proven useful for stimulating intestinal stem cell proliferation and differentiation to multiple cell types present in the gut epithelium. Enteroids have enabled characterization of gut biology and the signaling involved in the pathogenesis of disease. Here, enteroids were differentiated from four healthy human donors and assessed for culture duration-dependent differentiation status by immunostaining for gut epithelial markers lysozyme, chromogranin A, mucin, and sucrase isomaltase. Differentiated enteroids were evaluated with a reference set of 31 drugs exhibiting varying degrees of clinical incidence of diarrhea, a common manifestation of GIT that can be caused by drug-induced thinning of the gut epithelium. An assay examining enteroid viability in response to drug treatment demonstrated 90% accuracy for recapitulating the incidence of drug-induced diarrhea. The human enteroid viability assay developed here presents a promising in vitro model for evaluating drug-induced diarrhea., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
6. Developing in vitro assays to transform gastrointestinal safety assessment: potential for microphysiological systems.
- Author
-
Peters MF, Choy AL, Pin C, Leishman DJ, Moisan A, Ewart L, Guzzie-Peck PJ, Sura R, Keller DA, Scott CW, and Kolaja KL
- Subjects
- Caco-2 Cells, Humans, Intestinal Mucosa, Intestines, Microfluidics, Organoids
- Abstract
Drug-induced gastrointestinal toxicities (DI-GITs) are among the most common adverse events in clinical trials. High prevalence of DI-GIT has persisted among new drugs due in part to the lack of robust experimental tools to allow early detection or to guide optimization of safer molecules. Developing in vitro assays for the leading GI toxicities (nausea, vomiting, diarrhoea, constipation, and abdominal pain) will likely involve recapitulating complex physiological properties that require contributions from diverse cell/tissue types including epithelial, immune, microbiome, nerve, and muscle. While this stipulation may be beyond traditional 2D monocultures of intestinal cell lines, emerging 3D GI microtissues capture interactions between diverse cell and tissue types. These interactions give rise to microphysiologies fundamental to gut biology. For GI microtissues, organoid technology was the breakthrough that introduced intestinal stem cells with the capability of differentiating into each of the epithelial cell types and that self-organize into a multi-cellular tissue proxy with villus- and crypt-like domains. Recently, GI microtissues generated using miniaturized devices with microfluidic flow and cyclic peristaltic strain were shown to induce Caco2 cells to spontaneously differentiate into each of the principle intestinal epithelial cell types. Second generation models comprised of epithelial organoids or microtissues co-cultured with non-epithelial cell types can successfully reproduce cross-'tissue' functional interactions broadening the potential of these models to accurately study drug-induced toxicities. A new paradigm in which in vitro assays become an early part of GI safety assessment could be realized if microphysiological systems (MPS) are developed in alignment with drug-discovery needs. Herein, approaches for assessing GI toxicity of pharmaceuticals are reviewed and gaps are compared with capabilities of emerging GI microtissues (e.g., organoids, organ-on-a-chip, transwell systems) in order to provide perspective on the assay features needed for MPS models to be adopted for DI-GIT assessment.
- Published
- 2020
- Full Text
- View/download PDF
7. Human 3D Gastrointestinal Microtissue Barrier Function As a Predictor of Drug-Induced Diarrhea.
- Author
-
Peters MF, Landry T, Pin C, Maratea K, Dick C, Wagoner MP, Choy AL, Barthlow H, Snow D, Stevens Z, Armento A, Scott CW, and Ayehunie S
- Subjects
- Caco-2 Cells, Cell Differentiation, Electric Impedance, Humans, Pharmaceutical Preparations, Primary Cell Culture, Diarrhea chemically induced, Drug Evaluation methods, Drug-Related Side Effects and Adverse Reactions, Epithelial Cells physiology, Epithelial Cells ultrastructure
- Abstract
Drug-induced gastrointestinal toxicities (GITs) rank among the most common clinical side effects. Preclinical efforts to reduce incidence are limited by inadequate predictivity of in vitro assays. Recent breakthroughs in in vitro culture methods support intestinal stem cell maintenance and continual differentiation into the epithelial cell types resident in the intestine. These diverse cells self-assemble into microtissues with in vivo-like architecture. Here, we evaluate human GI microtissues grown in transwell plates that allow apical and/or basolateral drug treatment and 96-well throughput. Evaluation of assay utility focused on predictivity for diarrhea because this adverse effect correlates with intestinal barrier dysfunction which can be measured in GI microtissues using transepithelial electrical resistance (TEER). A validation set of widely prescribed drugs was assembled and tested for effects on TEER. When the resulting TEER inhibition potencies were adjusted for clinical exposure, a threshold was identified that distinguished drugs that induced clinical diarrhea from those that lack this liability. Microtissue TEER assay predictivity was further challenged with a smaller set of drugs whose clinical development was limited by diarrhea that was unexpected based on 1-month animal studies. Microtissue TEER accurately predicted diarrhea for each of these drugs. The label-free nature of TEER enabled repeated quantitation with sufficient precision to develop a mathematical model describing the temporal dynamics of barrier damage and recovery. This human 3D GI microtissue is the first in vitro assay with validated predictivity for diarrhea-inducing drugs. It should provide a platform for lead optimization and offers potential for dose schedule exploration., (© The Author(s) 2018. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2019
- Full Text
- View/download PDF
8. Toward β-Secretase-1 Inhibitors with Improved Isoform Selectivity.
- Author
-
Johansson P, Kaspersson K, Gurrell IK, Bäck E, Eketjäll S, Scott CW, Cebers G, Thorne P, McKenzie MJ, Beaton H, Davey P, Kolmodin K, Holenz J, Duggan ME, Budd Haeberlein S, and Bürli RW
- Subjects
- Amyloid Precursor Protein Secretases chemistry, Amyloid beta-Peptides metabolism, Animals, Aspartic Acid Endopeptidases chemistry, Brain metabolism, Catalytic Domain, Dogs, Female, Humans, Madin Darby Canine Kidney Cells, Male, Mice, Inbred C57BL, Molecular Structure, Oxazoles chemical synthesis, Oxazoles chemistry, Oxazoles pharmacokinetics, Oxazoles pharmacology, Peptide Fragments metabolism, Protease Inhibitors chemical synthesis, Protease Inhibitors chemistry, Protease Inhibitors pharmacokinetics, Protein Isoforms antagonists & inhibitors, Protein Isoforms chemistry, Rats, Spiro Compounds chemical synthesis, Spiro Compounds chemistry, Spiro Compounds pharmacokinetics, Spiro Compounds pharmacology, Structure-Activity Relationship, gp100 Melanoma Antigen metabolism, Amyloid Precursor Protein Secretases antagonists & inhibitors, Aspartic Acid Endopeptidases antagonists & inhibitors, Protease Inhibitors pharmacology
- Abstract
BACE1 is responsible for the first step in APP proteolysis, leading to toxic Aβ production, and has been indicated to play a key role in the pathogenesis of Alzheimer's disease. The related isoform BACE2 is thought to be involved in processing of the pigment cell-specific melanocyte protein. To avoid potential effects on pigmentation, we investigated the feasibility for developing isoform-selective BACE1 inhibitors. Cocrystal structures of 47 compounds were analyzed and clustered according to their selectivity profiles. Selective BACE1 inhibitors were found to exhibit two distinct conformational features proximal to the flap and the S3 subpocket. Several new molecules were designed and tested to make use of this observation. The combination of a pyrimidinyl C-ring and a methylcyclohexyl element resulted in lead molecule 28, which exhibited ∼50-fold selectivity. Compared to a nonselective BACE1/2 inhibitor, 28 showed significantly less inhibition of PMEL processing in human melanocytes, indicating good functional selectivity of this inhibitor class.
- Published
- 2018
- Full Text
- View/download PDF
9. Susto, Coraje, y Fatalismo : Cultural-Bound Beliefs and the Treatment of Diabetes Among Socioeconomically Disadvantaged Hispanics.
- Author
-
Moreira T, Hernandez DC, Scott CW, Murillo R, Vaughan EM, and Johnston CA
- Abstract
Hispanics are disparately affected by diabetes. Treating socioeconomically disadvantaged Hispanics is challenging due to economic and cultural barriers. Health care providers must understand that cultural beliefs about medicine and disease may have an impact on how diabetes treatment is viewed. Concepts such as susto (fright), coraje (anger), and fatalismo (fatalism) are common cultural beliefs. If these beliefs are not well understood by the health care provider, recommendations for treatment are likely to be discarded. To dismantle cultural barriers between the patient and the health care provider, there are several strategies that a health care provider can implement. For instance, a health care provider must develop trust with the patient. The health care provider could also engage a family member or promotora or promotor (community health worker) in the conversation. Furthermore, if the cultural barriers are significant, the patient may be best served by receiving treatment from someone with a better understanding of his or her background. Thus, a referral may be appropriate., Competing Interests: Declaration of Conflicting Interests The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2018
- Full Text
- View/download PDF
10. Deconvoluting Kinase Inhibitor Induced Cardiotoxicity.
- Author
-
Lamore SD, Ahlberg E, Boyer S, Lamb ML, Hortigon-Vinagre MP, Rodriguez V, Smith GL, Sagemark J, Carlsson L, Bates SM, Choy AL, Stålring J, Scott CW, and Peters MF
- Subjects
- Calcium metabolism, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells enzymology, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Myocytes, Cardiac enzymology, Myocytes, Cardiac metabolism, Protein Kinases metabolism, Reverse Transcriptase Polymerase Chain Reaction, Heart drug effects, Protein Kinase Inhibitors toxicity
- Abstract
Many drugs designed to inhibit kinases have their clinical utility limited by cardiotoxicity-related label warnings or prescribing restrictions. While this liability is widely recognized, designing safer kinase inhibitors (KI) requires knowledge of the causative kinase(s). Efforts to unravel the kinases have encountered pharmacology with nearly prohibitive complexity. At therapeutically relevant concentrations, KIs show promiscuity distributed across the kinome. Here, to overcome this complexity, 65 KIs with known kinome-scale polypharmacology profiles were assessed for effects on cardiomyocyte (CM) beating. Changes in human iPSC-CM beat rate and amplitude were measured using label-free cellular impedance. Correlations between beat effects and kinase inhibition profiles were mined by computation analysis (Matthews Correlation Coefficient) to identify associated kinases. Thirty kinases met criteria of having (1) pharmacological inhibition correlated with CM beat changes, (2) expression in both human-induced pluripotent stem cell-derived cardiomyocytes and adult heart tissue, and (3) effects on CM beating following single gene knockdown. A subset of these 30 kinases were selected for mechanistic follow up. Examples of kinases regulating processes spanning the excitation-contraction cascade were identified, including calcium flux (RPS6KA3, IKBKE) and action potential duration (MAP4K2). Finally, a simple model was created to predict functional cardiotoxicity whereby inactivity at three sentinel kinases (RPS6KB1, FAK, STK35) showed exceptional accuracy in vitro and translated to clinical KI safety data. For drug discovery, identifying causative kinases and introducing a predictive model should transform the ability to design safer KI medicines. For cardiovascular biology, discovering kinases previously unrecognized as influencing cardiovascular biology should stimulate investigation of underappreciated signaling pathways., (© The Author 2017. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2017
- Full Text
- View/download PDF
11. Food Insecurity and Pre-diabetes in Adults: Race/Ethnic and Sex Differences.
- Author
-
Murillo R, Reesor LM, Scott CW, and Hernandez DC
- Subjects
- Adolescent, Adult, Cross-Sectional Studies, Female, Humans, Male, Middle Aged, Risk, Sex Factors, Young Adult, Black or African American ethnology, Food Supply statistics & numerical data, Hispanic or Latino statistics & numerical data, Poverty ethnology, Prediabetic State ethnology, White People ethnology
- Abstract
Objective: We examined sex and race/ethnicity differences in the association between food insecurity status and prediabetes among adults., Method: We used cross-sectional 2011 and 2012 National Health Interview Survey data on non-Hispanic white, non-Hispanic black, and Hispanic adults aged 18-59 years whose household income was ≤ 299% Federal Poverty Line (N = 19,048). Food insecurity status was determined by 3 or more affirmative responses on the 10-item USDA Food Security Scale. Pre-diabetes was self-reported. Logistic regression analyses were used to estimate associations of food insecurity with pre-diabetes and adjusted for several demographic characteristics. All models were stratified by sex and race/ethnicity., Results: In adjusted models, food insecure non-Hispanic white women and non-Hispanic black women had 53% and over 200% higher odds of being pre-diabetic, respectively. Food insecurity was not related to pre-diabetes for Hispanic women or men., Conclusion: Limited food resources appear to place non-Hispanic white and non-Hispanic black women at risk for pre-diabetes. Linking food assistance programs with community-based health education programs may be a comprehensive approach to support those who are food insecure with diabetes prevention.
- Published
- 2017
- Full Text
- View/download PDF
12. Fragment-assisted hit investigation involving integrated HTS and fragment screening: Application to the identification of phosphodiesterase 10A (PDE10A) inhibitors.
- Author
-
Varnes JG, Geschwindner S, Holmquist CR, Forst J, Wang X, Dekker N, Scott CW, Tian G, Wood MW, and Albert JS
- Subjects
- Crystallography, X-Ray, Dose-Response Relationship, Drug, Humans, Models, Molecular, Molecular Structure, Phosphodiesterase Inhibitors chemical synthesis, Phosphodiesterase Inhibitors chemistry, Structure-Activity Relationship, Drug Evaluation, Preclinical, High-Throughput Screening Assays, Phosphodiesterase Inhibitors analysis, Phosphodiesterase Inhibitors pharmacology, Phosphoric Diester Hydrolases metabolism
- Abstract
Fragment-based drug design (FBDD) relies on direct elaboration of fragment hits and typically requires high resolution structural information to guide optimization. In fragment-assisted drug discovery (FADD), fragments provide information to guide selection and design but do not serve as starting points for elaboration. We describe FADD and high-throughput screening (HTS) campaign strategies conducted in parallel against PDE10A where fragment hit co-crystallography was not available. The fragment screen led to prioritized fragment hits (IC50's ∼500μM), which were used to generate a hypothetical core scaffold. Application of this scaffold as a filter to HTS output afforded a 4μM hit, which, after preparation of a small number of analogs, was elaborated into a 16nM lead. This approach highlights the strength of FADD, as fragment methods were applied despite the absence of co-crystallographical information to efficiently identify a lead compound for further optimization., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
13. Reversible and Species-Specific Depigmentation Effects of AZD3293, a BACE Inhibitor for the Treatment of Alzheimer's Disease, Are Related to BACE2 Inhibition and Confined to Epidermis and Hair.
- Author
-
Cebers G, Lejeune T, Attalla B, Soderberg M, Alexander RC, Budd Haeberlein S, Kugler AR, Ingersoll EW, Platz S, and Scott CW
- Abstract
Background: AZD3293 (also known as LY3314814) is a novel, potent, non-selective BACE1/BACE2 inhibitor currently in Phase 3 clinical development for the treatment of Alzheimer's disease., Objectives: The purpose of these studies was to characterize the effects, putative mechanism, and reversibility of hypopigmentation following treatment with AZD3293 in pigmented Long-Evans rats, Beagle dogs, human cell cultures, and humans., Design: Nonclinical studies were conducted in Long-Evans pigmented rats, and both young and older Beagle dogs using a variety of oral dose levels of AZD3293 or AZD3839 (BACE inhibition reference compound; used in older dogs only) for dosing durations of 13 to 26 weeks. In vitro studies of normal human epidermal melanocytes and reconstituted human epidermis were also conducted. Skin biopsy data from a multiple-dose Phase 1 clinical study of AZD3293 (NCT01795339) are also reported., Setting: Nonclinical in vivo and in vitro studies were conducted in laboratory settings in the US, Canada, and France; the multiple dose clinical study was conducted in a specialized inpatient setting in the US., Participants: Beagle dogs: 13-week study N=36 young (8-10 mo) animals; 39-week study N=64 young animals; and a second 13-week study N=32 older (30-32 mo) animals. Long-Evans rats: N=68 animals. Multiple-dose clinical study: only data for subjects enrolled in Part 2 of this study are included in this report (N=16)., Interventions: AZD3293 was the primary intervention used in these studies. AZD3839, a relatively BACE1-selective reference inhibitor compound was used in one group in the 13 week study in older Beagle dogs and one in vitro assessment. Finally, AZ1340, another relatively BACE1-selective reference inhibitor compound was used only in one in vitro assessment., Measurements: Measurements for the nonclinical studies in dogs and rats included macroscopic observation and assessment of skin biopsies via histopathology, immunochemistry, and electron microscopy. Measurements for the in vitro studies included melanocyte premelanosome protein (PMEL) processing, cytotoxicity, melanin synthesis, Pmel17 labeling, and melanocyte dendricity. Measurements in the clinical study included scoring of melanin content in skin biopsies taken before and after dosing with AZD3293 over 14 days at dose levels up to 150 mg., Results: Depigmentation in rats and dogs was limited to skin, hair, and mucosa with no effects on other pigmented tissues. At a cellular level depigmentation was observed within a week of treatment, whereas the appearance of depigmentation in skin and hair did not become apparent until, at earliest, 4 weeks of treatment. The depigmentation effects were reversible, not associated with degenerative or inflammatory changes, and were dose- and species-dependent in severity. Full recovery of melanization was observed at the microscopic (cellular) level and at least partial recovery was seen in the macroscopic appearance of animals by the end of the 12-week recovery period in both rats and dogs. Interestingly, no changes in melanin production or melanocyte morphology were seen in human primary melanocytes or reconstituted human epidermis in vitro. Finally, there were no changes in melanization level in skin biopsies following 12 days of daily AZD3293 treatment at doses of AZD3293 up to 150 mg/day in human subjects., Conclusions: AZD3293, a novel, potent, non-selective BACE1/BACE2 inhibitor is in development as a potentially disease-modifying treatment for Alzheimer's disease. Chronic nonclinical studies in Beagle dogs and pigmented rats showed macroscopic and microscopic hypopigmentation effects of AZD3293 that were limited to skin, hair, and mucosa. These effects were shown to be reversible in both species. Analysis of data from nonclinical and in vitro studies suggests that hypopigmentation is caused by BACE2 inhibition resulting in accumulation of a premelanosome protein fragment, which interrupts the normal production of melanin. No macroscopic or microscopic reports of hypopigmentation were observed in a Phase 1 clinical study following 13 doses of AZD3293 over 14 days at dose levels up to 150 mg/day. These data suggest that hypopigmentation is species-specific and humans appear to be least sensitive to the depigmentation effect caused by BACE2 inhibition., Competing Interests: Gvido Cebers, MD, PhD: GC was an AstraZeneca employee at the time the primary manuscript work was completed. GC is no longer an employee of AstraZeneca. GC has no financial disclosures. Typhaine Lejeune, DVM, DECVP: TL is an employee of Charles River Laboratories, which conducted in vivo non-clinical studies reported in this paper as a paid contractor for AstraZeneca. TL has no financial disclosures. Bassem Attalla, BS: Dr. BA is an employee of Charles River Laboratories, which conducted in vivo non-clinical studies reported in this paper as a paid contractor for AstraZeneca. BA has no financial disclosures. Magnus Soderberg, MD, PhD: MS is an employee of AstraZeneca and owns AstraZeneca stock. MS has no other financial disclosures. Robert C. Alexander, MD: RCA was an AstraZeneca employee at the time the primary manuscript work was completed. RCA is no longer an employee of AstraZeneca but remains a participant in AstraZeneca’s long term incentive program and owns AstraZeneca stock. RCA is currently an employee of Pfizer. RCA has no other financial disclosures. Samantha Budd Haeberlein, PhD: SBH was an AstraZeneca employee at the time the primary manuscript work was completed. SBA is no longer an employee of AstraZeneca but remains a participant in AstraZeneca’s long term incentive program and owns AstraZeneca stock. SBA is currently an employee of Biogen. SBA has no other financial disclosures. Alan R. Kugler, PhD: ARK was an AstraZeneca employee at the time the primary manuscript work was completed. ARK is no longer an employee of AstraZeneca but remains a participant in AstraZeneca’s long term incentive program and owns AstraZeneca stock. ARK has no other financial disclosures. Evan W. Ingersoll, PhD: EWI was a paid independent contractor for AstraZeneca during the time these studies were conducted and throughout this manuscript development. He provided program management, scientific support, and medical writing services including serving as coordinating editor for this manuscript. EWI has no other financial disclosures. Stefan Platz, DVM, PhD: SP is an AstraZeneca employee, is a participant in AstraZeneca’s long term incentive program, and owns AstraZeneca stock. SP has no other financial disclosures. Clay W. Scott, PhD: CWS is an AstraZeneca employee, is a participant in AstraZeneca’s long term incentive program, and owns AstraZeneca stock. CWS has no other financial disclosures.
- Published
- 2016
- Full Text
- View/download PDF
14. Structural Insights Lead to a Negamycin Analogue with Improved Antimicrobial Activity against Gram-Negative Pathogens.
- Author
-
McKinney DC, Basarab GS, Cocozaki AI, Foulk MA, Miller MD, Ruvinsky AM, Scott CW, Thakur K, Zhao L, Buurman ET, and Narayan S
- Abstract
Negamycin is a natural product with antibacterial activity against a broad range of Gram-negative pathogens. Recent revelation of its ribosomal binding site and mode of inhibition has reinvigorated efforts to identify improved analogues with clinical potential. Translation-inhibitory potency and antimicrobial activity upon modification of different moieties of negamycin were in line with its observed ribosomal binding conformation, reaffirming stringent structural requirements for activity. However, substitutions on the N6 amine were tolerated and led to N6-(3-aminopropyl)-negamycin (31f), an analogue showing 4-fold improvement in antibacterial activity against key bacterial pathogens. This represents the most potent negamycin derivative to date and may be a stepping stone toward clinical development of this novel antibacterial class.
- Published
- 2015
- Full Text
- View/download PDF
15. Human stem cell-derived cardiomyocytes in cellular impedance assays: bringing cardiotoxicity screening to the front line.
- Author
-
Peters MF, Lamore SD, Guo L, Scott CW, and Kolaja KL
- Subjects
- Animals, Cardiotoxicity diagnosis, Cardiotoxicity pathology, Cells, Cultured, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical methods, Electric Impedance, Humans, Myocardial Contraction drug effects, Myocardial Contraction physiology, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, Stem Cells pathology, Stem Cells physiology, Cardiotoxins toxicity, Myocytes, Cardiac drug effects, Stem Cells drug effects
- Abstract
Cardiovascular (CV) toxicity is a leading cause of drug attrition and withdrawal. Introducing in vitro assays with higher throughput should permit earlier CV hazard identification and enable medicinal chemists to design-out liabilities. Heretofore, development of in vitro CV assays has been limited by the challenge of replicating integrated cardiovascular physiology while achieving the throughput and consistency required for screening. These challenges appear to be met with a combination of human stem cell-derived cardiomyocytes (CM) which beat spontaneously and monitoring the response with technology that can assess drug-induced changes in voltage dependent contraction such as cellular impedance which has been validated with excellent predictivity for drug-induced arrhythmia and contractility. Here, we review advances in cardiomyocyte impedance with emphasis on stem cell-derived cardiomyocyte models for toxicity screening. Key perspectives include: the electrical principles of impedance technology, impedance detection of cardiomyocyte beating, beat parameter selection/analysis, validation in toxicity and drug discovery, and future directions. As a conclusion, an in vitro screening cascade is proffered using the downstream, inclusive detection of CM impedance assays as a primary screen followed by complementary CM assays chosen to enable mechanism-appropriate follow-up. The combined approach will enhance testing for CV liabilities prior to traditional in vivo models.
- Published
- 2015
- Full Text
- View/download PDF
16. An impedance-based cellular assay using human iPSC-derived cardiomyocytes to quantify modulators of cardiac contractility.
- Author
-
Scott CW, Zhang X, Abi-Gerges N, Lamore SD, Abassi YA, and Peters MF
- Subjects
- Animals, Animals, Newborn, Cardiotoxicity, Dogs, Dose-Response Relationship, Drug, Drug Discovery, Electric Impedance, High-Throughput Screening Assays, Humans, Induced Pluripotent Stem Cells cytology, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, Predictive Value of Tests, Rats, Sarcomeres drug effects, Sarcomeres pathology, Drug-Related Side Effects and Adverse Reactions pathology, Drug-Related Side Effects and Adverse Reactions physiopathology, Induced Pluripotent Stem Cells drug effects, Myocardial Contraction drug effects, Myocytes, Cardiac drug effects, Pharmaceutical Preparations analysis
- Abstract
Cardiovascular toxicity, a prominent reason for late-stage failures in drug development, has resulted in a demand for in vitro assays that can predict this liability in early drug discovery. Current in vitro cardiovascular safety testing primarily focuses on ion channel modulation and low throughput cardiomyocyte (CM) contractility measurements. We evaluated both human induced pluripotent stem cell-derived CMs (hiPSC-CMs) and rat neonatal CMs (rat CMs) on the xCELLigence Cardio system which uses impedance technology to quantify CM beating properties in a 96-well format. Forty-nine compounds were tested in concentration-response mode to determine potency for modulation of CM beating, a surrogate biomarker for contractility. These compounds had previously been tested in vivo and in a low throughput in vitro optical-based contractility assay that measures sarcomere shortening in electrically paced dog CMs. In comparison with in vivo contractility effects, hiPSC-CM impedance had assay sensitivity, specificity, and accuracy values of 90%, 74%, and 82%, respectively. These values compared favorably to values reported for the dog CM optical assay (83%, 84%, and 82%) and were slightly better than impedance using rat CMs (77%, 74%, and 74%). The potency values from the hiPSC-CM and rat CM assays spanned four orders of magnitude and correlated with values from the dog CM optical assay (r(2 )= 0.76 and 0.70, respectively). The Cardio system assay has >5× higher throughput than the optical assay. Thus, hiPSC-CM impedance testing can help detect the human cardiotoxic potential of novel therapeutics early in drug discovery, and if a hazard is identified, has sufficient throughput to support the design-make-test-analyze cycle to mitigate this liability., (© The Author 2014. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
17. Cellular impedance assays for predictive preclinical drug screening of kinase inhibitor cardiovascular toxicity.
- Author
-
Lamore SD, Kamendi HW, Scott CW, Dragan YP, and Peters MF
- Subjects
- Animals, Dogs, Male, Mitogen-Activated Protein Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinases metabolism, Rats, Rats, Wistar, Cardiovascular System drug effects, Drug Evaluation, Preclinical, Protein Kinase Inhibitors pharmacology
- Abstract
Cardiovascular (CV) toxicity is a leading contributor to drug attrition. Implementing earlier testing has successfully reduced human Ether-à-go-go-Related Gene-related arrhythmias. How- ever, analogous assays targeting functional CV effects remain elusive. Demand to address this gap is particularly acute for kinase inhibitors (KIs) that suffer frequent CV toxicity. The drug class also presents some particularly challenging requirements for assessing functional CV toxicity. Specifically, an assay must sense a downstream response that integrates diverse kinase signaling pathways. In addition, sufficient throughput is essential for handling inherent KI nonselectivity. A new opportunity has emerged with cellular impedance technology, which detects spontaneous beating cardiomyocytes. Impedance assays sense morphology changes downstream of cardiomyocyte contraction. To evaluate cardiomyocyte impedance assays for KI screening, we investigated two distinct KI classes where CV toxicity was discovered late and target risks remain unresolved. Microtubule-associated protein/microtubule affinity regulating kinase (MARK) inhibitors decrease blood pressure in dogs, whereas checkpoint kinase (Chk) inhibitors (AZD7762, SCH900776) exhibit dose-limiting CV toxicities in clinical trials. These in vivo effects manifested in vitro as cardiomyocyte beat cessation. MARK effects were deemed mechanism associated because beat inhibition potencies correlated with kinase inhibition, and gene knockdown and microtubule-targeting agents suppressed beating. MARK inhibitor impedance and kinase potencies aligned with rat blood pressure effects. Chk inhibitor effects were judged off-target because Chk and beat inhibition potencies did not correlate and knockdowns did not alter beating. Taken together, the data demonstrate that cardiomyocyte impedance assays can address three unmet needs-detecting KI functional cardiotoxicity in vitro, determining mechanism of action, and supporting safety structure-activity relationships.
- Published
- 2013
- Full Text
- View/download PDF
18. Human induced pluripotent stem cells and their use in drug discovery for toxicity testing.
- Author
-
Scott CW, Peters MF, and Dragan YP
- Subjects
- Cell Culture Techniques, Central Nervous System cytology, Central Nervous System drug effects, Drug Evaluation, Preclinical, Humans, Liver cytology, Liver drug effects, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Regenerative Medicine trends, Drug Discovery methods, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells drug effects, Toxicity Tests methods
- Abstract
Predicting human safety risks of novel xenobiotics remains a major challenge, partly due to the limited availability of human cells to evaluate tissue-specific toxicity. Recent progress in the production of human induced pluripotent stem cells (hiPSCs) may fill this gap. hiPSCs can be continuously expanded in culture in an undifferentiated state and then differentiated to form most cell types. Thus, it is becoming technically feasible to generate large quantities of human cell types and, in combination with relatively new detection methods, to develop higher-throughput in vitro assays that quantify tissue-specific biological properties. Indeed, the first wave of large scale hiSC-differentiated cell types including patient-derived hiPSCS are now commercially available. However, significant improvements in hiPSC production and differentiation processes are required before cell-based toxicity assays that accurately reflect mature tissue phenotypes can be delivered and implemented in a cost-effective manner. In this review, we discuss the promising alignment of hiPSCs and recently emerging technologies to quantify tissue-specific functions. We emphasize liver, cardiovascular, and CNS safety risks and highlight limitations that must be overcome before routine screening for toxicity pathways in hiSC-derived cells can be established., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
19. Development of a plate-based optical biosensor fragment screening methodology to identify phosphodiesterase 10A inhibitors.
- Author
-
Geschwindner S, Dekker N, Horsefield R, Tigerström A, Johansson P, Scott CW, and Albert JS
- Subjects
- Biosensing Techniques, Drug Design, Drug Evaluation, Preclinical methods, Surface Plasmon Resonance, Phosphodiesterase Inhibitors isolation & purification, Phosphoric Diester Hydrolases drug effects
- Abstract
We describe the development of a novel fragment screening methodology employing a plate-based optical biosensor system that can operate in a 384-well format. The method is based on the "inhibition in solution assay" (ISA) approach using an immobilized target definition compound (TDC) that has been specifically designed for this purpose by making use of available structural information. We demonstrate that this method is robust and is sufficiently sensitive to detect fragment hits as weak as KD 500 μM when confirmed in a conventional surface plasmon resonance approach. The application of the plate-based screen, the identification of fragment inhibitors of PDE10A, and their structural characterization are all discussed in a forthcoming paper.
- Published
- 2013
- Full Text
- View/download PDF
20. Evaluation of cellular impedance measures of cardiomyocyte cultures for drug screening applications.
- Author
-
Peters MF, Scott CW, Ochalski R, and Dragan YP
- Subjects
- Animals, Animals, Newborn, Biological Assay methods, Cardiovascular Agents pharmacology, Cells, Cultured, Culture Media, Data Interpretation, Statistical, Dimethyl Sulfoxide pharmacology, Electric Impedance, Ether-A-Go-Go Potassium Channels genetics, Heart Rate drug effects, Myocardial Contraction drug effects, Patch-Clamp Techniques, Rats, Rats, Wistar, Structure-Activity Relationship, Drug Evaluation, Preclinical methods, Myocytes, Cardiac drug effects, Myocytes, Cardiac physiology
- Abstract
Cardiovascular toxicity is a leading contributor to drug withdrawal and late-stage attrition. Earlier and broader screening is a validated approach to build-in cardiovascular safety as demonstrated with human Ether-à-go-go-related gene (hERG) screening to reduce drug-induced arrhythmia. There is an urgent need for novel in vitro assays to address other mechanistic aspects of cardiovascular function, including contractility, heart rate, toxicity, hypertrophy, and non-hERG arrhythmia. Recent advances in label-free cellular impedance technology now enable tracking of spontaneous, synchronized beating of cultured cardiomyocytes. Analysis of beating allows integrated detection that is downstream of electrical and mechanical aspects of contraction. Here, we evaluate impedance-based cardiomyocyte responses against criteria required for drug screening. The throughput and sensitivity allowed for rapid assay development. Critical variables for rat neonatal cardiomyocyte assays included cell density and serum levels. Once optimized, consistent, stable beating for at least 3 days was straight-forward to achieve. In tests of compounds spanning a breadth of target classes, the potency values showed excellent precision, wide dynamic range, and consistency across multiple experiments. Cardiomyocyte impedance assays can extract multiple beat-related parameters. In these experiments, rate, amplitude, and rise slope were examined and each yielded acceptable precision. Potency values calculated by beat rate and amplitude were highly correlated for most compounds although selected compounds displayed unique profiles indicative of different mechanisms. Tests with known cardiovascular active drugs revealed concordance with clinical findings. Thus, impedance assays combine novel features including sensitivity to contractile activity, versatile data analysis, and robust/translatable data in a format with sufficient throughput to become a valuable addition to the cardiovascular in vitro screening arsenal.
- Published
- 2012
- Full Text
- View/download PDF
21. Azepines and piperidines with dual norepinephrine dopamine uptake inhibition and antidepressant activity.
- Author
-
Brown DG, Bernstein PR, Wu Y, Urbanek RA, Becker CW, Throner SR, Dembofsky BT, Steelman GB, Lazor LA, Scott CW, Wood MW, Wesolowski SS, Nugiel DA, Koch S, Yu J, Pivonka DE, Li S, Thompson C, Zacco A, Elmore CS, Schroeder P, Liu J, Hurley CA, Ward S, Hunt HJ, Williams K, McLaughlin J, Hoesch V, Sydserff S, Maier D, and Aharony D
- Abstract
Herein, we describe the discovery of inhibitors of norepinephrine (NET) and dopamine (DAT) transporters with reduced activity relative to serotonin transporters (SERT). Two compounds, 8b and 21a, along with nomifensine were tested in a rodent receptor occupancy study and demonstrated dose-dependent displacement of radiolabeled NET and DAT ligands. These compounds were efficacious in a rat forced swim assay (model of depression) and also had activity in rat spontaneous locomotion assay.
- Published
- 2012
- Full Text
- View/download PDF
22. Biochemical characterization and in vitro activity of AZ513, a noncovalent, reversible, and noncompetitive inhibitor of fatty acid amide hydrolase.
- Author
-
Scott CW, Tian G, Yu XH, Paschetto KA, Wilkins DE, Meury L, Cao CQ, Varnes J, and Edwards PD
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, HEK293 Cells, Humans, In Vitro Techniques, Male, Patch-Clamp Techniques, Rats, Spinal Cord drug effects, Spinal Cord physiology, Synapses drug effects, Amidohydrolases antagonists & inhibitors, Benzamides chemistry, Benzamides pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Pyrimidines chemistry, Pyrimidines pharmacology, Pyrroles chemistry, Pyrroles pharmacology
- Abstract
Fatty acid amide hydrolase (FAAH) hydrolyzes several bioactive lipids including the endocannabinoid anandamide. Synthetic FAAH inhibitors are being generated to help define the biological role(s) of this enzyme, the lipids it degrades in vivo, and the disease states that might benefit from its pharmacological modulation. AZ513 inhibits human FAAH (IC(50)=551 nM), is 20-fold more potent against rat FAAH (IC(50)=27 nM), and is inactive at 10 μM against the serine hydrolases acetylcholinesterase, thrombin, and trypsin. In contrast to most other potent FAAH inhibitors, AZ513 showed no evidence of covalently modifying the enzyme and displayed reversible inhibition. In an enzyme cross-competition assay, AZ513 did not compete with OL-135, an inhibitor that binds to the catalytic site in FAAH, which indicates that AZ513 does not bind to the catalytic site and is therefore noncompetitive with respect to substrate. AZ513 has good cell penetration as demonstrated by inhibition of anandamide hydrolysis in human FAAH-transfected HEK293 cells (IC(50)=360 nM). AZ513 was tested in a rat spinal cord slice preparation where CB(1) activation reduces excitatory post-synaptic currents (EPSCs). In this native tissue assay of synaptic activity, AZ513 reduced EPSCs, which is consistent with inhibiting endogenous FAAH and augmenting endocannabinoid tone. AZ513 has a unique biochemical profile compared with other published FAAH inhibitors and will be a useful tool compound to further explore the role of FAAH in various biological processes., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
23. Mechanism of inhibition of fatty acid amide hydrolase by sulfonamide-containing benzothiazoles: long residence time derived from increased kinetic barrier and not exclusively from thermodynamic potency.
- Author
-
Tian G, Paschetto KA, Gharahdaghi F, Gordon E, Wilkins DE, Luo X, and Scott CW
- Subjects
- Animals, Benzothiazoles chemistry, CHO Cells, Cricetinae, Cricetulus, Enzyme Inhibitors chemistry, Humans, Kinetics, Recombinant Proteins chemistry, Recombinant Proteins pharmacology, Spectrometry, Mass, Electrospray Ionization, Thermodynamics, Amidohydrolases antagonists & inhibitors, Benzothiazoles pharmacology, Enzyme Inhibitors pharmacology, Sulfonamides chemistry
- Abstract
Fatty acid amide hydrolase (FAAH) has emerged as a potential target for developing analgesic, anxiolytic, antidepressant, sleep-enhancing, and anti-inflammatory drugs, and tremendous efforts have been made to discover potent and selective inhibitors of FAAH. Most known potent FAAH inhibitors described to date employ covalent mechanisms, inhibiting the enzyme either reversibly or irreversibly. Recently, a benzothiazole-based analogue (1) has been described possessing a high potency against FAAH yet lacking a structural feature previously known to interact with FAAH covalently. However, covalent inhibition of FAAH by 1 has not been fully ruled out, and the issue of reversibility has not been addressed. Confirming previous reports, 1 inhibited recombinant human FAAH (rhFAAH) with high potency with IC(50) ~2 nM. It displayed an apparently noncompetitive and irreversible inhibition, titrating rhFAAH stoichiometrically within normal assay times. The inhibition appeared to be time dependent, but the time dependence only improved potency by a small degree (from ~8 to ~2 nM). However, mass spectrometric analyses of the reaction mixture failed to reveal any cleavage product or covalent adduct and showed full recovery of the parent compound, ruling out covalent, irreversible inhibition. Dialysis revealed recovery of enzyme activity from enzyme-inhibitor complex over a prolonged time (>10 h), demonstrating that 1 is indeed a reversible, albeit slowly dissociating inhibitor of FAAH. Molecular docking indicated that the sulfonamide group of 1 could form hydrogen bonds with several residues involved in catalysis, thereby mimicking the transition state. The long residence time displayed by 1 does not appear to derive exclusively from great thermodynamic potency and is consistent with an increased kinetic energy barrier that prevents dissociation from happening quickly.
- Published
- 2011
- Full Text
- View/download PDF
24. 2,6-Disubstituted pyrazines and related analogs as NR2B site antagonists of the NMDA receptor with anti-depressant activity.
- Author
-
Brown DG, Maier DL, Sylvester MA, Hoerter TN, Menhaji-Klotz E, Lasota CC, Hirata LT, Wilkins DE, Scott CW, Trivedi S, Chen T, McCarthy DJ, Maciag CM, Sutton EJ, Cumberledge J, Mathisen D, Roberts J, Gupta A, Liu F, Elmore CS, Alhambra C, Krumrine JR, Wang X, Ciaccio PJ, Wood MW, Campbell JB, Johansson MJ, Xia J, Wen X, Jiang J, Wang X, Peng Z, Hu T, and Wang J
- Subjects
- Animals, Antidepressive Agents chemistry, Antidepressive Agents pharmacology, Binding Sites, Binding, Competitive, Dose-Response Relationship, Drug, Inhibitory Concentration 50, Mice, Molecular Structure, Motor Activity drug effects, Protein Binding drug effects, Pyrazines chemistry, Pyrazines pharmacology, Antidepressive Agents chemical synthesis, Pyrazines chemical synthesis, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors
- Abstract
Herein we describe the discovery of compounds that are competitive antagonists of the CP101-606 binding site within the NR2B subtype of the NMDA receptor. The compounds identified do not possess phenolic functional groups such as those in ifenprodil and related analogs. Initial identification of hits in this series focused on a basic, secondary amine side chain which led to good potency, but also presented a hERG liability. Further modifications led to examples of non-basic replacements which demonstrated much less liability in this regard. Finally, one compound in the series, 6a, was tested in the mouse forced swim depression assay and found to show activity (s.c. 60 mg/kg)., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
25. Label-free whole-cell assays: expanding the scope of GPCR screening.
- Author
-
Scott CW and Peters MF
- Subjects
- Animals, Drug Discovery methods, Humans, Biological Assay instrumentation, Biosensing Techniques instrumentation, Drug Discovery instrumentation, Receptors, G-Protein-Coupled drug effects
- Abstract
A new class of instruments offers an unprecedented combination of label-free detection with exquisite sensitivity to live-cell responses. These instruments can quantify G-protein-coupled receptor (GPCR) signaling through G(s), G(i) and G(q) pathways and in some cases distinguish G-protein coupling, with sensitivity high enough to detect endogenous receptors. Here, we review emerging data evaluating impedance- and optical-based label-free instruments for GPCR drug discovery. In comparison with traditional GPCR assays, we highlight strengths, weaknesses and future opportunities for label-free biosensors. The ability to qualitatively distinguish G-protein coupling has groundbreaking potential for assessing functional selectivity, a concept that is changing the way GPCR pharmacology is defined and screening strategies are designed., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
26. Comparing label-free biosensors for pharmacological screening with cell-based functional assays.
- Author
-
Peters MF, Vaillancourt F, Heroux M, Valiquette M, and Scott CW
- Subjects
- Animals, CHO Cells, Cells, Corticotropin-Releasing Hormone pharmacology, Cricetinae, Cricetulus, Culture Media, Dose-Response Relationship, Drug, Drug Discovery, Humans, Muscarinic Agonists chemistry, Muscarinic Agonists pharmacology, Receptor, Muscarinic M1 agonists, Receptor, Muscarinic M2 agonists, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled metabolism, Reproducibility of Results, Signal Transduction drug effects, TRPV Cation Channels agonists, Biological Assay methods, Biosensing Techniques, Drug Evaluation, Preclinical methods
- Abstract
The diversity and impact of label-free technologies continues to expand in drug discovery. Two classes of label-free instruments, using either an electrical impedance-based or an optical-based biosensor, are now available for investigating the effects of ligands on cellular targets. Studies of GPCR function have been especially prominent with these instruments due to the importance of this target class in drug discovery. Although both classes of biosensors share similar high sensitivity to changes in cell shape and structure, it is unknown whether these biosensors yield similar results when comparing the same GPCR response. Furthermore, since cell morphology changes induced by GPCRs differ depending on which G-protein is activated, there is potential for these instruments to have differential sensitivities to G-protein signaling. Here 1 impedance (CellKey)- and 2 optical-based instruments (BIND and Epic) are compared using Gi-coupled (ACh M2), Gq-coupled (ACh M1), and Gs-coupled (CRF1) receptors. All 3 instruments were robust in agonist and antagonist modes yielding comparable potencies and assay variance. Both the impedance and optical biosensors showed similar high sensitivity for detecting an endogenous D1/D5 receptor response and a melanocortin-4 receptor inverse agonist (agouti-related protein). The impedance-based biosensor was uniquely able to qualitatively distinguish G-protein coupling and reveal dual signaling by CRF1. Finally, responses with a ligand-gated ion channel, TRPV1, were similarly detectable in each instrument. Thus, despite some differences, both impedance- and optical-based platforms offer robust live-cell, label-free assays well suited to drug discovery and typically yield similar pharmacological profiles for GPCR ligands.
- Published
- 2010
- Full Text
- View/download PDF
27. After-action reviews: a venue for the promotion of safety climate.
- Author
-
Allen JA, Baran BE, and Scott CW
- Subjects
- Humans, Organizational Culture, Safety Management organization & administration, Task Performance and Analysis, Workload, Accidents, Occupational prevention & control, Fires, Safety Management methods
- Abstract
This study investigated the role of after-action reviews on perceptions of safety climate at the group and organizational levels. Moderated and mediated regression analyses of data from 67 firefighting crews suggest that after-action review frequency positively influenced both levels of safety climate. Safety-oriented group norms fully mediated the relationship between after-action review frequency and group-level safety climate. Fire-station busyness moderated the relationship between after-action review frequency and organizational-level safety climate, such that the relationship was non-existent for highly busy stations. These findings suggest that after-action reviews constitute a specific venue through which managers can promote safety climate in high-risk environments., (Copyright 2009 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
28. Evaluating cellular impedance assays for detection of GPCR pleiotropic signaling and functional selectivity.
- Author
-
Peters MF and Scott CW
- Subjects
- Animals, Brain Neoplasms metabolism, Brain Neoplasms pathology, CHO Cells, Cell Culture Techniques, Cell Line, Tumor, Cells, Cultured, Cricetinae, Cricetulus, Cytochalasin D pharmacology, Dopamine Agonists pharmacology, Dose-Response Relationship, Drug, Electric Impedance, GTP-Binding Protein alpha Subunits, Gi-Go drug effects, GTP-Binding Protein alpha Subunits, Gq-G11 drug effects, GTP-Binding Protein alpha Subunits, Gs drug effects, Humans, Inhibitory Concentration 50, Kidney cytology, Neuroectodermal Tumors, Primitive, Peripheral metabolism, Neuroectodermal Tumors, Primitive, Peripheral pathology, Osteosarcoma metabolism, Osteosarcoma pathology, Pertussis Toxin pharmacology, Receptor, Muscarinic M1 metabolism, Receptors, Dopamine D2 metabolism, Receptors, Dopamine D5 metabolism, Sensitivity and Specificity, Signal Transduction drug effects, Signal Transduction physiology, alpha-MSH agonists, alpha-MSH analogs & derivatives, Biological Assay methods, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, GTP-Binding Protein alpha Subunits, Gs metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
G-protein-coupled receptors can couple to different signal transduction pathways in different cell types (termed cell-specific signaling) and can activate different signaling pathways depending on the receptor conformation(s) stabilized by the activating ligand (functional selectivity). These concepts offer potential for developing pathway-specific drugs that increase efficacy and reduce side effects. Despite significant interest, functional selectivity has been difficult to exploit in drug discovery, in part due to the burden of multiple assays. Cellular impedance assays use an emerging technology that can qualitatively distinguish Gs, Gi/o, and Gq signaling in a single assay and is thereby suited for studying these pharmacological concepts. Cellular impedance confirmed cell-specific Gs and Gq coupling for the melanocortin-4 receptor and dual Gi and Gs signaling with the cannabinoid-1 (CB1) receptor. The balance of Gi versus Gs signaling depended on the cell line. In CB1-HEKs, Giand Gs-like responses combined to yield a novel impedance profile demonstrating the dynamic nature of these traces. Cellspecific signaling was observed with endogenous D1 receptor in U-2 cells and SK-N-MC cells, yet the pharmacological profile of partial and full agonists was similar in both cell lines. We conclude that the dynamic impedance profile encodes valuable relative signaling information and is sufficiently robust to help evaluate cell-specific signaling and functional selectivity.
- Published
- 2009
- Full Text
- View/download PDF
29. Evaluation of cellular dielectric spectroscopy, a whole-cell, label-free technology for drug discovery on Gi-coupled GPCRs.
- Author
-
Peters MF, Knappenberger KS, Wilkins D, Sygowski LA, Lazor LA, Liu J, and Scott CW
- Subjects
- Allosteric Regulation, Animals, CHO Cells, Cricetinae, Cricetulus, Cyclic AMP metabolism, Guanosine 5'-O-(3-Thiotriphosphate) metabolism, Humans, Receptor, Muscarinic M4 metabolism, Receptors, Dopamine D2 agonists, Receptors, Dopamine D2 metabolism, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled antagonists & inhibitors, Reproducibility of Results, Structure-Activity Relationship, Drug Evaluation, Preclinical methods, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Receptors, G-Protein-Coupled metabolism, Spectrum Analysis methods
- Abstract
Cellular dielectric spectroscopy (CDS) is an emerging technology capable of detecting a range of whole-cell responses in a label-free manner. A new CDS-based instrument, CellKey, has been developed that is optimized for G-protein coupled receptor (GPCR) detection and has automated liquid handling in microplate format, thereby making CDS accessible to lead generation/optimization drug discovery. In addition to having sufficient throughput, new assay technologies must pass rigorous standards for assay development, signal window, dynamic range, and reproducibility to effectively support drug discovery SAR studies. Here, the authors evaluated CellKey with 3 different G(i)-coupled GPCRs for suitability in supporting SAR studies. Optimized assay conditions compatible with the precision, reproducibility, and throughput required for routine screening were quickly achieved for each target. Across a 1000-fold range in compound potencies, CellKey results correlated with agonist and antagonist data obtained using classical methods ([(35)S]GTPgammaS binding and cAMP production). For partial agonists, relative efficacy measurements also correlated with GTPgammaS data. CellKey detection of positive allosteric modulators appeared superior to GTPgammaS methodology. Agonist and antagonist activity could be accurately quantified under conditions of low receptor expression. CellKey is a new technology platform that uses label-free detection in a homogeneous assay that is unaffected by color quenching and is easily integrated into existing microtiter-based compound testing and data analysis procedures for drug discovery.
- Published
- 2007
- Full Text
- View/download PDF
30. Neurokinin-3 receptor-specific antagonists talnetant and osanetant show distinct mode of action in cellular Ca2+ mobilization but display similar binding kinetics and identical mechanism of binding in ligand cross-competition.
- Author
-
Tian G, Wilkins D, and Scott CW
- Subjects
- Animals, Binding, Competitive, CHO Cells, Cricetinae, Cricetulus, Kinetics, Neurokinin B analogs & derivatives, Neurokinin B metabolism, Piperidines metabolism, Quinolines metabolism, Calcium metabolism, Piperidines pharmacology, Quinolines pharmacology, Receptors, Neurokinin-3 antagonists & inhibitors
- Abstract
Talnetant and osanetant, two structurally diverse antagonists of neurokinin-3 receptor (NK3), displayed distinct modes of action in Ca2+ mobilization. Although talnetant showed a normal Schild plot with a slope close to unity and a Kb similar to its Ki value in binding, osanetant presented an aberrant Schild with a steep slope (3.3 +/- 0.5) and a Kb value (12 nM) significantly elevated compared with its Ki value (0.8 nM) in binding. Kinetic binding experiments indicated a simple one-step binding mechanism with relatively fast on- and off-rates for both antagonists, arguing against slow onset of antagonism as the reason for abnormal Schild. This conclusion was supported by prolonged preincubation of antagonist that failed to improve the observed aberrant Schild. In ligand cross-competition binding, both talnetant and osanetant displayed linear reciprocal plots of identical slope when [MePhe7]neurokinin B (NKB) was used as the other competition partner with 125I-[MePhe7]NKB as the radioligand, indicating competitive binding of either antagonist with regard to [MePhe7]NKB. Similar patterns were obtained when talnetant was tested against osanetant, indicating competitive binding between the two antagonists as well. These results were reproduced when [3H]4-quinolinecarboxamide (SB222200), a close derivative of talnetant, was used as the radioligand. Taken together, these data strongly suggest binding of both talnetant and osanetant at the orthosteric binding site with similar kinetic properties and do not support the hypothesis that the aberrant Schild observed in functional assays for osanetant is derived from differences in the mechanism of binding for these NK3 antagonists.
- Published
- 2007
- Full Text
- View/download PDF
31. Mechanism of gamma-secretase cleavage activation: is gamma-secretase regulated through autoinhibition involving the presenilin-1 exon 9 loop?
- Author
-
Knappenberger KS, Tian G, Ye X, Sobotka-Briner C, Ghanekar SV, Greenberg BD, and Scott CW
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Amino Acid Sequence, Amyloid Precursor Protein Secretases, Amyloid beta-Peptides metabolism, Aspartic Acid Endopeptidases, Binding Sites, Cell Line, Endopeptidases chemistry, Enzyme Activation, Enzyme Inhibitors chemistry, Humans, Ligands, Membrane Proteins genetics, Membrane Proteins metabolism, Models, Molecular, Molecular Sequence Data, Molecular Structure, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Presenilin-1, Endopeptidases metabolism, Enzyme Inhibitors metabolism, Exons, Membrane Proteins chemistry, Protein Structure, Secondary
- Abstract
Maturation of gamma-secretase requires an endoproteolytic cleavage in presenilin-1 (PS1) within a peptide loop encoded by exon 9 of the corresponding gene. Deletion of the loop has been demonstrated to cause familial Alzheimer's disease. A synthetic peptide corresponding to the loop sequence was found to inhibit gamma-secretase in a cell-free enzymatic assay with an IC(50) of 2.1 microM, a value similar to the K(m) (3.5 microM) for the substrate C100. Truncation at either end, single amino acid substitutions at certain residues, sequence reversal, or randomization reduced its potency. Similar results were also observed in a cell-based assay using HEK293 cells expressing APP. In contrast to small-molecule gamma-secretase inhibitors, kinetic inhibition studies demonstrated competitive inhibition of gamma-secretase by the exon 9 peptide. Consistent with this finding, inhibitor cross-competition kinetics indicated noncompetitive binding between the exon 9 peptide and L685458, a transition-state analogue presumably binding at the catalytic site, and ligand competition binding experiments revealed no competition between L685458 and the exon 9 peptide. These data are consistent with the proposed gamma-secretase mechanism involving separate substrate-binding and catalytic sites and binding of the exon 9 peptide at the substrate-binding site, but not the catalytic site of gamma-secretase. NMR analyses demonstrated the presence of a loop structure with a beta-turn in the middle of the exon 9 peptide and a loose alpha-helical conformation for the rest of the peptide. Such a structure supports the hypothesis that this exon 9 peptide can adopt a distinct conformation, one that is compact enough to occupy the putative substrate-binding site without necessarily interfering with binding of small molecule inhibitors at other sites on gamma-secretase. We hypothesize that gamma-secretase cleavage activation may be a result of a cleavage-induced conformational change that relieves the inhibitory effect of the intact exon 9 loop occupying the substrate-binding site on the immature enzyme. It is possible that the DeltaE9 mutation causes Alzheimer's disease because cleavage activation of gamma-secretase is no longer necessary, alleviating constraints on Abeta formation.
- Published
- 2004
- Full Text
- View/download PDF
32. Kaposi's sarcoma-associated herpesvirus K7 protein targets a ubiquitin-like/ubiquitin-associated domain-containing protein to promote protein degradation.
- Author
-
Feng P, Scott CW, Cho NH, Nakamura H, Chung YH, Monteiro MJ, and Jung JU
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Autophagy-Related Proteins, Carrier Proteins genetics, Cell Line, Cysteine Endopeptidases metabolism, Dimerization, Herpesvirus 8, Human genetics, Humans, I-kappa B Proteins metabolism, Mitochondrial Proteins genetics, Multienzyme Complexes metabolism, NF-KappaB Inhibitor alpha, Proteasome Endopeptidase Complex, Protein Structure, Tertiary, Recombinant Fusion Proteins metabolism, Two-Hybrid System Techniques, Ubiquitins genetics, Viral Proteins genetics, Carrier Proteins metabolism, Cell Cycle Proteins, Herpesvirus 8, Human metabolism, Mitochondrial Proteins metabolism, Ubiquitins metabolism, Viral Proteins metabolism
- Abstract
Pathogens exploit host machinery to establish an environment that favors their propagation. Because of their pivotal roles in cellular physiology, protein degradation pathways are common targets for viral proteins. Protein-linking integrin-associated protein and cytoskeleton 1 (PLIC1), also called ubiquilin, contains an amino-terminal ubiquitin-like (UBL) domain and a carboxy-terminal ubiquitin-associated (UBA) domain. PLIC1 is proposed to function as a regulator of the ubiquitination complex and proteasome machinery. Kaposi's sarcoma-associated herpesvirus (KSHV) contains a small membrane protein, K7, that protects cells from apoptosis induced by various stimuli. We report here that cellular PLIC1 is a K7-interacting protein and that the central hydrophobic region of K7 and the carboxy-terminal UBA domain of PLIC1 are responsible for their interaction. Cellular PLIC1 formed a dimer and bound efficiently to polyubiquitinated proteins through its carboxy-terminal UBA domain, and this activity correlated with its ability to stabilize cellular I kappa B protein. In contrast, K7 interaction prevented PLIC1 from forming a dimer and binding to polyubiquitinated proteins, leading to the rapid degradation of I kappa B. Furthermore, K7 expression promoted efficient degradation of the p53 tumor suppressor, resulting in inhibition of p53-mediated apoptosis. These results indicate that KSHV K7 targets a regulator of the ubiquitin- and proteasome-mediated degradation machinery to deregulate cellular protein turnover, which potentially provides a favorable environment for viral reproduction.
- Published
- 2004
- Full Text
- View/download PDF
33. Parkin suppresses wild-type alpha-synuclein-induced toxicity in SHSY-5Y cells.
- Author
-
Oluwatosin-Chigbu Y, Robbins A, Scott CW, Arriza JL, Reid JD, and Zysk JR
- Subjects
- Adenosine Triphosphate metabolism, Adenoviridae genetics, Cell Line, Cell Survival, Cytoprotection, Gene Expression, Genetic Vectors, Ligases genetics, Mutation, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Neurons cytology, Synucleins, Transfection, alpha-Synuclein, Ligases metabolism, Nerve Tissue Proteins antagonists & inhibitors, Neurons metabolism, Ubiquitin-Protein Ligases
- Abstract
Current hypotheses concerning the mechanism of neuronal cell death in Parkinson's disease (PD) and related synucleopathies propose a functional interaction between parkin and alpha-synuclein (alphaS). Recently parkin was shown to suppress mutant alphaS-induced toxicity in primary neurons, providing a basis for an association between these proteins and neuronal loss [Neuron 36 (2000) 1007-1019]. We have asked if a similar association could be made between wild-type (wt) alphaS and parkin. We examined inducible over-expression of alphaS in SHSY-5Y cells through adenoviral infection under conditions which produce cellular toxicity through a reduction in ATP levels. We demonstrate that parkin suppresses toxicity induced by mutant (A53T) and wt alphaS. Parkin over-expression was also associated with the appearance of higher molecular weight alphaS-immunoreactive bands by Western blot analysis. These data, consistent with a protective role for parkin, extend previous findings to include a functional association between parkin and the wt form of alphaS.
- Published
- 2003
- Full Text
- View/download PDF
34. A medium-throughput functional assay of KCNQ2 potassium channels using rubidium efflux and atomic absorption spectrometry.
- Author
-
Scott CW, Wilkins DE, Trivedi S, and Crankshaw DJ
- Subjects
- Anthracenes pharmacology, Carbamates pharmacology, Cell Line, Clone Cells, Dose-Response Relationship, Drug, Humans, Indoles pharmacology, KCNQ2 Potassium Channel, Kinetics, Phenylenediamines pharmacology, Potassium Channel Blockers pharmacology, Potassium Channels drug effects, Potassium Channels genetics, Potassium Channels, Voltage-Gated, Potassium Chloride pharmacology, Pyridines pharmacology, Quaternary Ammonium Compounds pharmacology, Rubidium analysis, Spectrophotometry, Atomic, Tetraethylammonium pharmacology, Transfection, Potassium Channels metabolism, Rubidium metabolism
- Abstract
Heterologous expression of KCNQ2 (Kv7.2) results in the formation of a slowly activating, noninactivating, voltage-gated potassium channel. Using a cell line that stably expresses KCNQ2, we developed a rubidium flux assay to measure the functional activity and pharmacological modulation of this ion channel. Rubidium flux was performed in a 96-well microtiter plate format; rubidium was quantified using an automated atomic absorption spectrometer to enable screening of 1000 data points/day. Cells accumulated rubidium at 37 degrees C in a monoexponential manner with t(1/2)=40min. Treating cells with elevated extracellular potassium caused membrane depolarization and stimulation of rubidium efflux through KCNQ2. The rate of rubidium efflux increased with increasing extracellular potassium: the t(1/2) at 50mM potassium was 5.1 min. Potassium-stimulated efflux was potentiated by the anticonvulsant drug retigabine (EC(50)=0.5 microM). Both potassium-induced and retigabine-facilitated efflux were blocked by TEA (IC(50)s=0.4 and 0.3mM, respectively) and the neurotransmitter release enhancers and putative cognition enhancers linopirdine (IC(50)s=2.3 and 7.1 microM, respectively) and XE991 (IC(50)s=0.3 and 0.9 microM, respectively). Screening a collection of ion channel modulators revealed additional inhibitors including clofilium (IC(50) = 27 microM). These studies extend the pharmacological profile of KCNQ2 and demonstrate the feasibility of using this assay system to rapidly screen for compounds that modulate the function of KCNQ2.
- Published
- 2003
- Full Text
- View/download PDF
35. A homogeneous in vitro functional assay for estrogen receptors: coactivator recruitment.
- Author
-
Liu J, Knappenberger KS, Kack H, Andersson G, Nilsson E, Dartsch C, and Scott CW
- Subjects
- Amino Acid Sequence, Cyclic AMP Response Element-Binding Protein metabolism, Estradiol metabolism, Estrogen Antagonists pharmacology, Estrogen Receptor alpha, Estrogen Receptor beta, Europium metabolism, Fluorescence Resonance Energy Transfer methods, Histone Acetyltransferases, Humans, Ligands, Molecular Sequence Data, Nuclear Receptor Coactivator 1, Oligopeptides, Peptides metabolism, Protein Binding, Raloxifene Hydrochloride pharmacology, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Estrogen agonists, Tamoxifen pharmacology, Transcription Factors metabolism, Transcriptional Activation physiology, Receptors, Estrogen metabolism
- Abstract
Estrogen receptor (ER)-mediated gene transcription occurs via the formation of a multimeric complex including ligand-activated receptors and nuclear coactivators. We have developed a homogeneous in vitro functional assay to help study the ligand-dependent interaction of ERs with various nuclear coactivators. The assay consists of FLAG-tagged ERalpha or ERbeta ligand binding domain (LBD), a biotinylated coactivator peptide, europium-labeled anti-FLAG antibody, and streptavidin-conjugated allophycocyanin. Upon agonist binding, the biotinylated coactivator peptide is recruited to FLAG-tagged ER LBD to form a complex and thus allow fluorescence resonance energy transfer (FRET) to occur between europium and allophycocyanin. Compounds with estrogen antagonism block the agonist-mediated recruitment of a coactivator and prevent FRET. The assay was used to evaluate the preference of ERs for various coactivators and ligands. Both ERalpha and ERbeta exhibited strong preferences for coactivator peptides corresponding to steroid receptor coactivator-1 and PPARgamma coactivor-1 vs. peroxisome proliferator-activated receptor-interacting protein and cAMP response element binding protein-binding protein. 17beta-Estradiol acted as a nonselective agonist for ERalpha and ERbeta. Genistein showed full agonism for ERalpha and only partial agonism for ERbeta, but with higher potency for ERbeta than ERalpha. Both raloxifene and tamoxifen behaved as full antagonists in this functional assay. The results obtained using compounds with a wide range of potency correlated well with those from a cell-based reporter gene assay. Therefore, this simple in vitro functional assay is predictive of ligand-dependent transactivation function of the receptor and, as such, is useful in nuclear receptor applications including mechanistic studies.
- Published
- 2003
- Full Text
- View/download PDF
36. Novel small molecule inhibitors of caspase-3 block cellular and biochemical features of apoptosis.
- Author
-
Scott CW, Sobotka-Briner C, Wilkins DE, Jacobs RT, Folmer JJ, Frazee WJ, Bhat RV, Ghanekar SV, and Aharony D
- Subjects
- Aniline Compounds chemical synthesis, Animals, Apoptosis physiology, Caspase 3, Cell Line, Coloring Agents, DNA Fragmentation drug effects, Enzyme Inhibitors chemical synthesis, Fluorescent Dyes, Humans, In Situ Nick-End Labeling, Kinetics, Nerve Growth Factor pharmacology, PC12 Cells, Phenotype, Quinazolines chemical synthesis, Rats, Recombinant Proteins metabolism, Staurosporine pharmacology, Structure-Activity Relationship, Swine, Aniline Compounds pharmacology, Apoptosis drug effects, Caspase Inhibitors, Enzyme Inhibitors pharmacology, Quinazolines pharmacology
- Abstract
Caspase-3 is an intracellular cysteine protease, activated as part of the apoptotic response to cell injury. Its interest as a therapeutic target has led many to pursue the development of inhibitors. To date, only one series of nonpeptidic inhibitors have been described, and these have limited selectivity within the caspase family. Here we report the properties of a series of anilinoquinazolines (AQZs) as potent small molecule inhibitors of caspase-3. The AQZs inhibit human caspase-3 with Ki values in the 90 to 800 nM range. A subset of AQZs are equipotent against caspase-6, although most lack activity against this isoform and caspase-1, -2, -7, and -8. The AQZs inhibit endogenous caspase-3 activity toward a cell permeable, exogenously added substrate in staurosporine-treated SH-SY5Y cells. The AQZs reduce biochemical and cellular features of apoptosis that are thought to be a consequence of caspase-3 activation including DNA fragmentation, TUNEL staining, and the various morphological features that define the terminal stages of apoptotic cell death. Moreover, the AQZs also inhibit apoptosis induced by nerve growth factor withdrawal from differentiated PC12 cells. Thus, the AQZs represent a new and structurally novel class of inhibitors, some of which selectively inhibit caspase-3 and will thereby allow evaluation of the role of caspase-3 activity in various cellular models of apoptosis.
- Published
- 2003
- Full Text
- View/download PDF
37. Microplate gel-filtration method for radioligand-binding assays.
- Author
-
Liu J, Zacco A, Piser TM, and Scott CW
- Subjects
- Binding, Competitive, Centrifugation, Humans, Iodine Radioisotopes metabolism, Kinetics, Ligands, Protein Binding, Recombinant Proteins metabolism, Reticulocytes metabolism, Substrate Specificity, Chromatography, Gel methods, Radioligand Assay methods, Receptors, Estrogen metabolism
- Abstract
A thin-layer gel-filtration chromatographic method has been developed in a 96-well format to separate free and protein-bound ligand in radioligand-binding assays. The mobile phase in the gel-filtration plate is removed via quick centrifugation before samples are applied. Protein-bound ligand is recovered via centrifugation into another 96-well plate for radioactivity measurements. The method exhibits excellent recovery of protein-ligand complexes and less opportunity for dissociation of the complexes since it eliminates major dilution effects from the mobile phase of a column and from elution steps in conventional gel-filtration chromatography. It offers other advantages: simple, rapid, inexpensive, quantitative, and able to handle a large number of samples as required in drug discovery and clinical settings. This microplate gel-filtration method was optimized in studies of receptor-ligand interactions using estrogen receptors as examples and can be used in other radioligand-binding assays.
- Published
- 2002
- Full Text
- View/download PDF
38. Interactions between GSK3beta and caspase signalling pathways during NGF deprivation induced cell death.
- Author
-
Bhat RV, Leonov S, Luthman J, Scott CW, and Lee CM
- Subjects
- Animals, Apoptosis physiology, Glycogen Synthase Kinase 3 beta, PC12 Cells, Rats, Caspases physiology, Cell Death physiology, Glycogen Synthase Kinase 3 physiology, Nerve Growth Factor physiology, Signal Transduction physiology, Tumor Cells, Cultured physiology
- Abstract
Withdrawal of NGF (NGF-W) in PC12 cells leads to caspase and GSK3beta activation which results in cell death. Our recent findings suggest that inhibition of GSK3beta promotes PC12 cell survival after NGF-W. To determine whether these pathways interact from a signalling perspective, we compared the effects of BAF (a general caspase inhibitor), Li+ (a GSK3beta inhibitor) and insulin on NGF-W induced PC12 cell death. Maximal increase in DNA fragmentation was observed 3 h after NGF-W and was inhibited by BAF (7.5 microM), Li+ (IC(50) = 2 mM) and insulin (IC(50) = 100 nM). BAF inhibited caspase-3 activity and delayed cell death up to 6 h after NGF-W indicating that caspase inhibition is sufficient to prevent apoptosis. BAF had no major effect on GSK3betaactive site phosphorylation or activity suggesting the caspase pathway does not regulate GSK3beta activity. Conversely, Li+ inhibited caspase activity by only 20% but promoted cell survival for 24 h after NGF-W. Overexpression of dominant negative mutants of GSK3beta also inhibited apoptosis, but had only a minor effect on caspase activity after NGF-W. Taken together, these results suggest that GSK3beta is upstream of caspase signalling, and exerts a small effect on the caspase pathway.
- Published
- 2002
- Full Text
- View/download PDF
39. Synthesis and enzymatic evaluation of a P1 arginine aminocoumarin substrate library for trypsin-like serine proteases.
- Author
-
Edwards PD, Mauger RC, Cottrell KM, Morris FX, Pine KK, Sylvester MA, Scott CW, and Furlong ST
- Subjects
- Humans, Oligopeptides chemistry, Oligopeptides metabolism, Protein Conformation, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Structure-Activity Relationship, Substrate Specificity, Arginine, Coumarins, Oligopeptides chemical synthesis, Peptide Library, Serine Endopeptidases metabolism, Thrombin metabolism, Trypsin metabolism
- Abstract
A method for the solid-phase synthesis of P1 arginine containing peptides via attachment of the arginine side-chain guanidine group is described. This procedure is applied to the preparation of a tetrapeptide, P1 arginine aminocoumarin PS-SCL. This library was validated by using it to determine the P4-P2 specificity for thrombin and comparing the results to the known thrombin subsite specificity. This is the first reported example of a PS-SCL library containing a P1 arginine.
- Published
- 2000
- Full Text
- View/download PDF
40. Regulation and localization of tyrosine216 phosphorylation of glycogen synthase kinase-3beta in cellular and animal models of neuronal degeneration.
- Author
-
Bhat RV, Shanley J, Correll MP, Fieles WE, Keith RA, Scott CW, and Lee CM
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinases genetics, Cell Death, Glycogen Synthase Kinase 3, Glycogen Synthase Kinases, Humans, PC12 Cells, Phosphorylation, Rats, Signal Transduction, Tyrosine, Brain Ischemia metabolism, Brain Ischemia pathology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Neurons metabolism, Neurons pathology
- Abstract
Inactivation of glycogen synthase kinase-3beta (GSK3beta) by S(9) phosphorylation is implicated in mechanisms of neuronal survival. Phosphorylation of a distinct site, Y(216), on GSK3beta is necessary for its activity; however, whether this site can be regulated in cells is unknown. Therefore we examined the regulation of Y(216) phosphorylation on GSK3beta in models of neurodegeneration. Nerve growth factor withdrawal from differentiated PC12 cells and staurosporine treatment of SH-SY5Y cells led to increased phosphorylation at Y(216), GSK3beta activity, and cell death. Lithium and insulin, agents that lead to inhibition of GSK3beta and adenoviral-mediated transduction of dominant negative GSK3beta constructs, prevented cell death by the proapoptotic stimuli. Inhibitors induced S(9) phosphorylation and inactivation of GSK3beta but did not affect Y(216) phosphorylation, suggesting that S(9) phosphorylation is sufficient to override GSK3beta activation by Y(216) phosphorylation. Under the conditions examined, increased Y(216) phosphorylation on GSK3beta was not an autophosphorylation response. In resting cells, Y(216) phosphorylation was restricted to GSK3beta present at focal adhesion sites. However, after staurosporine, a dramatic alteration in the immunolocalization pattern was observed, and Y(216)-phosphorylated GSK3beta selectively increased within the nucleus. In rats, Y(216) phosphorylation was increased in degenerating cortical neurons induced by ischemia. Taken together, these results suggest that Y(216) phosphorylation of GSK3beta represents an important mechanism by which cellular insults can lead to neuronal death.
- Published
- 2000
- Full Text
- View/download PDF
41. C3 activation is inhibited by analogs of compstatin but not by serine protease inhibitors or peptidyl alpha-ketoheterocycles.
- Author
-
Furlong ST, Dutta AS, Coath MM, Gormley JJ, Hubbs SJ, Lloyd D, Mauger RC, Strimpler AM, Sylvester MA, Scott CW, and Edwards PD
- Subjects
- Complement C3 metabolism, Complement C3-C5 Convertases antagonists & inhibitors, Complement Factor B analysis, Complement Factor B antagonists & inhibitors, Hemolysis drug effects, Humans, Immunoenzyme Techniques, Peptides, Cyclic chemical synthesis, Complement Activation drug effects, Complement C3 antagonists & inhibitors, Complement Inactivator Proteins pharmacology, Peptides, Cyclic pharmacology, Serine Proteinase Inhibitors pharmacology
- Abstract
C3 convertase is a key enzyme in the complement cascade and is an attractive therapeutic target for drug design. Recent studies have demonstrated that this enzyme is inhibited by compstatin (Morikis, D. , Assa-Munt, N., Sahu, A., Lambris, J.D., 1998. Solution structure of Compstatin, a potent complement inhibitor. Protein Sci. (7) 619-627; Sahu, A., Kay, B.K., Lambris, J.D., 1996. Inhibition of human complement by a C3-binding peptide isolated from a phage-displayed random peptide library. J. Immunol. (157) 884-891), a 13 amino acid cyclic peptide that binds to C3. Since the enzyme exhibits some homology to serine proteases, substrate-based design could be another avenue for drug design. In this study, we confirm the activity of compstatin using different sources of enzyme and different assay systems. We also tested the activity of substituted compstatin analogs and compared the selectivity and toxicity of these compounds to peptidyl alpha-ketoheterocyclic compounds. Our work confirms the activity of compstatin in both alternative and classical complement pathways, describes 11 new active analogs of this cyclic peptide, and provides evidence for key segments of the peptide for activity. Compstatin and related active analogs showed little or no inhibition of clotting or key enzymes in the clotting cascade nor did they appear to have significant cytotoxicity. The characteristics of compstatin suggest that this peptide and its analogs could be attractive candidates for further clinical development. By contrast, known serine protease inhibitors, including peptidyl alpha-ketoheterocycles, did not inhibit C3 convertase illustrating the atypical nature of this enzyme.
- Published
- 2000
- Full Text
- View/download PDF
42. Molecular cloning of the guinea-pig IL-5 receptor alpha and beta subunits and reconstitution of a high affinity receptor.
- Author
-
Scott CW, Logsdon NJ, Wilkins DE, Norris TE, Sobotka-Briner C, Hubbs S, and Graham A
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Cloning, Molecular, Gene Expression, Guinea Pigs, Humans, Molecular Sequence Data, Protein Binding, Protein Isoforms genetics, Protein Isoforms metabolism, Receptors, Interleukin metabolism, Receptors, Interleukin-5, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Sequence Analysis, DNA, Spodoptera cytology, Interleukin-5 metabolism, Receptors, Interleukin genetics
- Abstract
The functional IL-5 receptor is a heteromeric complex consisting of an alpha and beta subunit. The cloning, sequencing and expression of guinea-pig IL-5Ralpha and beta subunits is described. The guinea-pig IL-5Ralpha subunit cDNA encodes a protein of M(r)47 kDa, which is 72 and 66% homologous to the human and murine orthologs, respectively. Three guinea-pig IL-5Rbeta subunit cDNA clones were isolated, which differ in the N-terminus and are 56-64% homologous to the human and murine IL-5Rbeta subunits. Expressing human IL-5Ralphabeta and guinea-pig IL-5Ralphabeta(1)in the baculovirus-insect cell system resulted in recombinant receptors which bound hIL-5 with high affinity (K(d)=0.19 and 0.11 nM, respectively). Expressing just gpIL-5Ralpha was not sufficient to demonstrate binding. This contrasts with the human receptor, where hIL-5Ralpha alone can bind hIL-5 with high affinity. gpIL-5Ralphabeta(1)bound both hIL-5 and mIL-5 with comparable affinity (K(i)=0.10 and 0.06 nM), similar to that seen with hIL-5Ralphabeta. Thus, both the heteromeric hIL-5R and gpIL-5Ralphabeta(1)can bind multiple IL-5 orthologs with high affinity whereas the murine IL-5R is selective for the murine ligand., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
43. Expression and in vitro properties of guinea pig IL-5: comparison to human and murine orthologs.
- Author
-
Scott CW, Budzilowicz C, Hubbs SJ, Stein M, Sobotka-Briner C, and Wilkins DE
- Subjects
- Animals, Base Sequence, Cell Line, Cloning, Molecular, DNA Primers genetics, Gene Expression, Guinea Pigs, Humans, In Vitro Techniques, Inflammation Mediators metabolism, Inflammation Mediators pharmacology, Interleukin-5 genetics, Kinetics, Mice, Receptors, Interleukin metabolism, Receptors, Interleukin-5, Recombinant Proteins genetics, Recombinant Proteins metabolism, Recombinant Proteins pharmacology, Species Specificity, Interleukin-5 metabolism, Interleukin-5 pharmacology
- Abstract
Interleukin-5 (IL-5) is a key mediator of eosinophilic inflammation. The biological role of this cytokine in an allergic airway inflammatory response has been widely demonstrated in guinea pigs, yet the interaction of guinea pig IL-5 (gpIL-5) with its receptor has not been studied. Experiments were performed to quantitate the interaction of gpIL-5 with gpIL-5r and to compare this affinity with that of hIL-5 and mIL-5 and their cognate receptors. The cross-species affinity and agonist efficacy were evaluated to see if gpIL-5r had a restricted species reactivity (as is the case with mIL-5r) or did not distinguish between IL-5 orthologs (similar to hIL-5r). gpIL-5 was cloned using mRNA isolated from cells obtained by bronchoalveolar lavage. Recombinant gpIL-5 was expressed in T. ni insect cells and purified from spent media. Binding assays were performed using insect cells expressing hIL-5ralphabeta or gpIL-5ralphabeta1 as previously described (Cytokine, 12:858-866, 2000) or using B13 cells which express mIL-5r. The agonist potency and efficacy properties of each IL-5 ortholog were evaluated by quantitating the proliferative response of human TF-1 cells and murine B13 cells. gpIL-5 bound with high affinity to recombinant gpIL-5r as demonstrated by displacing [125I]hIL-5 (Ki = 160 pM). gpIL-5 also bound to hIL-5r with high affinity (Ki = 750 pM). hIL-5 and mIL-5 showed similar, high-affinity binding profiles to both gpIL-5r and hIL-5r. In contrast, gpIL-5 and hIL-5 did not bind to the mIL-5r as demonstrated by an inability to displace [125I]mIL-5, even at 1000-fold molar excess. These differences in affinity for IL-5r orthologs correlated with bioassay results: human TF-1 cells showed roughly comparable proliferative responses to guinea pig, human and murine IL-5 whereas murine B13 cells showed a strong preference for murine over guinea pig and human IL-5 (EC50 = 1.9, 2200 and 720 pM, respectively). Recombinant gpIL-5 binds to the gpIL-5r with high affinity, similar to that seen with the human ligand-receptor pair. gpIL-5r and hIL-5r do not distinguish between the three IL-5 orthologs whereas mIL-5r has restricted specificity for its cognate ligand.
- Published
- 2000
- Full Text
- View/download PDF
44. Acupuncture: does it have a place in military general practice?
- Author
-
Scott CW
- Subjects
- Female, Humans, Male, Treatment Outcome, United Kingdom, Acupuncture Therapy statistics & numerical data, Family Practice, Military Medicine
- Abstract
Acupuncture can be learnt by doctors in a short space of time. Its mode of action is becoming increasingly understood and attempts are being made for statistical evaluation to allow for Western medical acceptance. After attending a basic course in acupuncture, the author describes his first one hundred cases. The preponderance of military patients, chronicity of the presenting complaints and the promising results obtained, illustrates the potential use of such a simple technique in military general practice.
- Published
- 1998
- Full Text
- View/download PDF
45. Stages of change: interactions with treatment compliance and involvement.
- Author
-
DiClemente CC and Scott CW
- Subjects
- Humans, Models, Psychological, Motivation, Psychotherapy, Substance-Related Disorders psychology, Time Factors, Patient Compliance, Patient Participation, Substance-Related Disorders therapy
- Abstract
Current perspectives on compliance and involvement in treatment often overlook the fact that treatment occurs in the context of a process of change and not vice versa. Each individual moves at a unique pace through a series of stages of change and in a cyclical fashion over a substantial period of time. Treatment personnel and programs should recognize the diversity of stage status in their clients and address each one in a manner compatible with the client's current stage of change, the tasks needed to move forward in the process of change, and an understanding of the course of change. Such considerations should assist the therapist in developing strategies to increase the engagement of a wide variety of clients, to improve retention of these clients in a realistic course of treatment, and to foster participation in stage-appropriate tasks that promote successful movement through the stages to sustained, long-term change.
- Published
- 1997
46. Specific cleavage of recombinant protein tau3 between valine-220 and tyrosine-221 (val-309 and tyr-310 of tau4) by a double-stranded DNA-stimulated protease.
- Author
-
Wu JM, An S, Ng C, Scott CW, and Caputo CB
- Subjects
- Amino Acid Sequence, Enzyme Activation, HL-60 Cells, Humans, Hydrolysis, Molecular Sequence Data, DNA metabolism, Endopeptidases metabolism, Tyrosine metabolism, Valine metabolism, tau Proteins metabolism
- Abstract
The protein tau was degraded to distinct products by a DNA-stimulated protease isolated from human leukemia HL-60 cell extracts. The enzyme partially purified by sequential chromatography on GTP-agarose, DEAE-cellulose, and TSK 3000 (0.6 X 60 mm) columns eluted as a 300-450 kDa protein which appeared as 60-90 kDa polypeptides on SDS-PAGE. Protease activity was stimulated by synthetic and natural DNAs and was most active at pH 8.5. Human recombinant tau3 was degraded by the DEAE-cellulose-eluted protease to a 26-kDa and several 14- to 16-kDa peptides. Degradation of tau was partially blocked by preincubation with tubulin, suggesting that the DNA-stimulated cleavage of tau occurred at the carboxyl-terminus, at or near the "tubulin-interactive" domains. The 26-kDa fragment was shown by amino acid sequencing to correspond to the N-terminus of tau whereas sequencing of the 16-kDa fragment yielded YKPVDLSKVT. These results show the existence of a DNA-stimulated protease capable of cleaving tau3 between valine-220 and tyrosine-221 (equivalent to valine 309 and tyrosine-310 of tau4).
- Published
- 1996
- Full Text
- View/download PDF
47. A filtration-based assay to quantitate granulocyte-macrophage colony-stimulating factor binding.
- Author
-
Scott CW, Gomes BC, Hubbs SJ, and Koenigbauer HC
- Subjects
- Cell Line, Humans, Radioligand Assay methods, Sensitivity and Specificity, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
Filtration-based binding assays have numerous advantages over centrifugation-based assays, yet they have not been established for many protein ligands due to the high nonspecific binding of the protein to the membrane filter. This paper describes a vacuum filtration method that permits quantitative evaluation of [125I]GM-CSF binding to its receptor on intact cells. The method includes the use of glass fiber filters presoaked in a solution of polyvinylpyrrolidone and Tween 20 to greatly reduce nonspecific binding of the protein ligand. The ratio of specific:nonspecific binding observed with this filtration assay was comparable to values reported for centrifugation assays. [125I] GM-CSF binding to HL-60 cells was shown to be time-dependent, saturable, and specific. The estimated Kd (70 pM) and Bmax (160 r/cell) were similar to values reported using centrifugation assays. This filtration method is much less labor-intensive, has greater sample throughput, and allows for a more rapid determination of GM-CSF binding compared to the centrifugation-based assay. Although developed to quantitate the binding of GM-CSF to its receptor on intact cells, this assay is also applicable to other cytokines and can be used with both intact cells and isolated plasma membrane preparations.
- Published
- 1995
- Full Text
- View/download PDF
48. Glycated tau protein in Alzheimer disease: a mechanism for induction of oxidant stress.
- Author
-
Yan SD, Chen X, Schmidt AM, Brett J, Godman G, Zou YS, Scott CW, Caputo C, Frappier T, and Smith MA
- Subjects
- Alzheimer Disease pathology, Brain pathology, Glycation End Products, Advanced metabolism, Humans, Immunohistochemistry, Neuroblastoma metabolism, Neurofibrillary Tangles ultrastructure, Recombinant Proteins metabolism, Tumor Cells, Cultured, tau Proteins genetics, tau Proteins metabolism, Alzheimer Disease metabolism, Glycation End Products, Advanced isolation & purification, Reactive Oxygen Species metabolism, tau Proteins isolation & purification
- Abstract
The stability of proteins that constitute the neurofibrillary tangles and senile plaques of Alzheimer disease suggests that they would be ideal substrates for nonenzymatic glycation, a process that occurs over long times, even at normal levels of glucose, ultimately resulting in the formation of advanced glycation end products (AGEs). AGE-modified proteins aggregate, and they generate reactive oxygen intermediates. Using monospecific antibody to AGEs, we have colocalized these AGEs with paired helical filament tau in neurofibrillary tangles in sporadic Alzheimer disease. Such neurons also exhibited evidence of oxidant stress: induction of malondialdehyde epitopes and heme oxygenase 1 antigen. AGE-recombinant tau generated reactive oxygen intermediates and, when introduced into the cytoplasm of SH-SY5Y neuroblastoma cells, induced oxidant stress. We propose that in Alzheimer disease, AGEs in paired helical filament tau can induce oxidant stress, thereby promoting neuronal dysfunction.
- Published
- 1994
- Full Text
- View/download PDF
49. Casein kinase II preferentially phosphorylates human tau isoforms containing an amino-terminal insert. Identification of threonine 39 as the primary phosphate acceptor.
- Author
-
Greenwood JA, Scott CW, Spreen RC, Caputo CB, and Johnson GV
- Subjects
- Amino Acid Sequence, Casein Kinase II, Humans, In Vitro Techniques, Mass Spectrometry, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Mapping, Phosphothreonine metabolism, Polyethyleneimine metabolism, Protein Binding, tau Proteins classification, Protein Serine-Threonine Kinases metabolism, tau Proteins metabolism
- Abstract
The in vitro phosphorylation of the microtubule-associated protein tau by casein kinase II was studied. Purified human brain tau was phosphorylated by casein kinase II to a stoichiometry of 0.7 mol of 32P/mol of tau. Individual recombinant human tau isoforms were phosphorylated to stoichiometries ranging from 0.2 to 0.8 mol of 32P/mol of tau. Casein kinase II catalyzed a 4-fold greater incorporation of phosphate into the tau isoform containing a 58-amino acid insert near its amino terminus (T4L) than the isoforms without the 58-amino acid insert (T3 and T4). Phosphopeptide mapping of casein kinase II phosphorylated human tau and recombinant tau isoforms suggested that the isoforms containing an amino-terminal insert constitute the major substrates for casein kinase II within the tau family. The sites of phosphorylation on T4L were identified by digesting phosphorylated T4L with the protease Asp-N, separating the peptides by reversed phase high performance liquid chromatography, and analyzing the isolated peptides by liquid-secondary ion mass spectrometry and solid-phase amino-terminal sequencing. Thr39 was identified as the predominant phosphorylation site, which is located 5 residues from the amino-terminal insert in T4L. Phosphopeptide mapping of tau isolated from LA-N-5 neuroblastoma cells indicates that Thr39 is phosphorylated in situ. To our knowledge, this is the first demonstration of a differential phosphorylation of the human tau isoforms, with the isoforms containing the acidic amino-terminal insert being the preferred substrates of casein kinase II.
- Published
- 1994
50. Aggregation of tau protein by aluminum.
- Author
-
Scott CW, Fieles A, Sygowski LA, and Caputo CB
- Subjects
- 3T3 Cells, Animals, Cattle, Deferoxamine pharmacology, Electrophoresis, Polyacrylamide Gel, Humans, Mice, Microscopy, Electron, Phosphorylation, Recombinant Proteins drug effects, Transfection, Aluminum pharmacology, Neurofilament Proteins drug effects, tau Proteins drug effects
- Abstract
Aluminum has been detected in Alzheimer neurofibrillary tangles, but the significance of its presence is unknown. The principal component of tangles is the paired helical filament (PHF), comprised of tau protein. We investigated whether aluminum could induce tau protein to form filaments or aggregate. When 10 microM bovine tau or non-phosphorylated recombinant human tau was combined with 400 microM or more aluminum, tau protein appeared to aggregate, observed as a dose-dependent decrease in electrophoretic mobility on SDS-PAGE. Tau appeared as a smear above the region of the expected tau bands and, at higher aluminum doses, failed to enter the gel. A tau fragment encompassing the microtubule binding domains did not show decreased mobility in the presence of aluminum, but did form aggregates that failed to electrophorese. However no fibrillar structures were observed in the aluminum-treated tau samples when observed by electron microscopy. The effect of aluminum on tau mobility was reversed by incubating with 1 mM deferoxamine. In contrast, the morphology of PHF fibrils was unaffected by deferoxamine treatment and the characteristic abnormal mobility of PHF-tau was not reduced by deferoxamine. This suggests that aluminum is not, by itself, a significant factor in maintaining the assembly of PHF-tau as fibrils or in its abnormal mobility on SDS gels. Aluminum treatment of 3T3 fibroblasts transfected with human tau resulted in toxicity, but did not change tau expression levels or induce tau aggregation. In conclusion, aluminum appears to induce isolated tau protein to aggregate in a phosphate-independent way, without the formation of fibrils.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.