Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Behav Brain Res ; 409: 113263, 2021 07 09.
Article in English | MEDLINE | ID: mdl-33775776

ABSTRACT

Total sleep deprivation (TSD) causes a decline in almost all cognitive domains, especially working memory. However, we do not have a clear understanding of the degree working memory is impaired under prolonged TSD, nor do we know the underlying neurophysiological mechanism. In this study, we recorded EEG data from 64 subjects while they performed a working memory task during resting wakefulness, after 24 h TSD, and after 30 h TSD. ANOVA was used to verify performance differences between 24 h and 30 h TSD in working memory tasks: (1) reaction time and accuracy hit rates, (2) P200, N200, and P300 amplitude and latency in measurements of event-related potential, as well as (3) effective connectivity strength between brain areas associated with working memory. Compared to 24 h TSD, 30 h TSD significantly decreased accuracy hit rates and induced a larger N200 difference waveform. The effective connectivity analysis showed that 30 h TSD also decreased beta frequency in effective connection strength from the right insular lobe to the left anterior cingulate cortex (ACC). Effective connection from the left ventrolateral prefrontal cortex to the left dorsolateral prefrontal cortex increased in the match condition of the 2-back task. In conclusion, 30 h TSD had a greater negative impact on working memory than 24 h TSD. This impairment of working memory is associated with decreased strength in the effective connection from the right insula to the left ACC.


Subject(s)
Connectome , Evoked Potentials/physiology , Gyrus Cinguli/physiopathology , Insular Cortex/physiopathology , Memory, Short-Term/physiology , Sleep Deprivation/physiopathology , Adult , Electroencephalography , Humans , Male , Young Adult
2.
Breast Cancer Res Treat ; 141(1): 43-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23959396

ABSTRACT

Trastuzumab is effective in the treatment of HER2/neu over-expressing breast cancer, but not all patients benefit from it. In vitro data suggest a role for HER3 in the initiation of signaling activity involving the AKT­mTOR pathway leading to trastuzumab insensitivity. We sought to investigate the potential of HER3 alone and in the context of p95HER2 (p95), a trastuzumab resistance marker, as biomarkers of trastuzumab escape. Using the VeraTag® assay platform, we developed a dual antibody proximity-based assay for the precise quantitation of HER3 total protein (H3T) from formalin-fixed paraffin-embedded (FFPE) breast tumors. We then measured H3T in 89 patients with metastatic breast cancer treated with trastuzumab-based therapy, and correlated the results with progression-free survival and overall survival using Kaplan­Meier and decision tree analyses that also included HER2 total (H2T) and p95 expression levels. Within the sub-population of patients that over-expressed HER2, high levels of HER3 and/or p95 protein expression were significantly associated with poor clinical outcomes on trastuzumab-based therapy. Based on quantitative H3T, p95, and H2T measurements, multiple subtypes of HER2-positive breast cancer were identified that differ in their outcome following trastuzumab therapy. These data suggest that HER3 and p95 are informative biomarkers of clinical outcomes on trastuzumab therapy, and that multiple subtypes of HER2-positive breast cancer may be defined by quantitative measurements of H3T, p95, and H2T.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/secondary , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Neoplasm Proteins/biosynthesis , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/classification , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cohort Studies , Decision Trees , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Neoplasm Proteins/genetics , Peptide Fragments/analysis , Peptide Fragments/immunology , Prognosis , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-3/genetics , Receptor, ErbB-3/immunology , Retrospective Studies , Single-Blind Method , Trastuzumab , Treatment Outcome
3.
Clin Cancer Res ; 16(16): 4226-35, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20664024

ABSTRACT

PURPOSE: p95HER2 is an NH(2)-terminally truncated form of HER2 that lacks the trastuzumab binding site and is therefore thought to confer resistance to trastuzumab treatment. In this report, we introduce a new antibody that has enabled the first direct quantitative measurement of p95HER2 in formalin-fixed paraffin-embedded (FFPE) breast cancer tissues. We sought to show that quantitative p95HER2 levels would correlate with outcome in trastuzumab-treated HER2-positive metastatic breast cancer. EXPERIMENTAL DESIGN: The novel p95HER2 antibody used here was characterized for sensitivity, specificity, and selectivity over full-length HER2. Quantitative p95HER2 levels were measured in 93 metastatic breast tumors using a VeraTag FFPE assay to determine the correlation of p95HER2 levels with outcomes. RESULTS: Within a cohort of trastuzumab-treated metastatic breast cancer patients, high levels of p95HER2 were found to correlate with shorter progression-free survival [hazard ratio (HR), 1.9; P = 0.017] and overall survival (HR, 2.2; P = 0.012) in patients with tumors selected to be HER2 positive by the VeraTag HER2 assay. For those with tumors found to be fluorescence in situ hybridization positive, elevated p95HER2 correlated similarly with shorter progression-free survival (HR, 1.8; P = 0.022) and overall survival (HR, 2.2; P = 0.009). CONCLUSIONS: We have successfully generated an antibody that can specifically detect p95HER2, and developed an assay to quantify expression in FFPE tumor specimens. Using this novel assay, we have identified a group of HER2-positive patients expressing p95HER2 that have a worse outcome while on trastuzumab. As p95HER2 retains sensitivity to kinase inhibitors, measurement of p95HER2 in breast tumor sections may be useful in guiding treatment for patients with HER2-positive breast cancer.


Subject(s)
Antibodies, Monoclonal , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptor, ErbB-2/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Blotting, Western , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Separation , Cohort Studies , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Formaldehyde , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Paraffin Embedding , Peptide Fragments/analysis , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , Sensitivity and Specificity , Tissue Fixation , Trastuzumab , Treatment Outcome
4.
BMC Cancer ; 10: 56, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20178580

ABSTRACT

BACKGROUND: We have recently described the correlation between quantitative measures of HER2 expression or HER2 homodimers by the HERmark assay and objective response (RR), time-to progression (TTP), and overall survival (OS) in an expanded access cohort of trastuzumab-treated HER2-positive patients with metastatic breast cancer (MBC) who were stringently selected by fluorescence in situ hybridization (FISH). Multivariate analyses suggested a continuum of HER2 expression that correlated with outcome following trastuzumab. Here we investigate the relationship between HER2 expression or HER2 homodimers and OS in a clinic-based population of patients with MBC selected primarily by IHC. METHODS: HERmark, a proximity-based assay designed to detect and quantitate protein expression and dimerization in formalin-fixed paraffin-embedded (FFPE) tissues, was used to measure HER2 expression and HER2 homodimers in FFPE samples from patients with MBC. Assay results were correlated with OS using univariate Kaplan-Meier, hazard function plots, and multivariate Cox regression analyses. RESULTS: Initial analyses revealed a parabolic relationship between continuous measures of HER2 expression and risk of death, suggesting that the assumption of linearity for the HER2 expression measurements may be inappropriate in subsequent multivariate analyses. Cox regression analyses using the categorized variable of HER2 expression level demonstrated that higher HER2 levels predicted better survival outcomes following trastuzumab treatment in the high HER2-expressing group. CONCLUSIONS: These data suggest that the quantitative amount of HER2 expression measured by Hermark may be a new useful marker to identify a more relevant target population for trastuzumab treatment in patients with MBC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/chemistry , Antibodies, Monoclonal, Humanized , Breast Neoplasms/pathology , Cohort Studies , Dimerization , Disease Progression , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Multivariate Analysis , Neoplasm Metastasis , Proportional Hazards Models , Trastuzumab , Treatment Outcome
5.
Diagn Mol Pathol ; 18(1): 22-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19214112

ABSTRACT

The selection of patients with HER2-positive breast cancer for treatment with trastuzumab is based on the measurement of HER2 protein expression by immunohistochemistry, or the presence of HER2 gene amplification by fluorescence in situ hybridization (FISH). By using multivariate analyses, we investigate the relationship between quantitative measurements of HER2 expression or HER2:HER2 dimers and objective response (Response Evaluation Criteria in Solid Tumors), time to progression, and breast cancer survival after trastuzumab treatment in a cohort of patients with metastatic breast cancer who were primarily selected for treatment by FISH. The VeraTag assay, a proximity-based assay designed to quantitate protein expression and dimerization in formalin-fixed, paraffin-embedded tissue specimens, was used to measure HER2 protein expression and HER2:HER2 dimer levels. In a Cox proportional hazards analysis, higher HER2 expression or HER2:HER2 dimer levels were both correlated with longer survival (P=0.0058 and P=0.016, respectively) after treatment with trastuzumab in a population of patients that were either FISH-positive (90%) or immunohistochemistry 3+ (10%). Patients with higher levels of HER2 expression or HER2:HER2 dimers seemed to derive little benefit from the addition of concomitant chemotherapy to trastuzumab, whereas those with lower levels benefited significantly [interaction test P=0.43 (HER2 expression), P=0.27 (HER2:HER2 dimers)]. These data suggest that more quantitative or functional measurements of HER2 status may facilitate the development of more personalized treatment strategies for patients with metastatic breast cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/secondary , Breast/chemistry , Receptor, ErbB-2/analysis , Antibodies, Monoclonal, Humanized , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Female , Humans , In Situ Hybridization, Fluorescence/methods , Middle Aged , Protein Multimerization , Survival Analysis , Trastuzumab , Treatment Outcome
6.
Diagn Mol Pathol ; 18(1): 11-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19214113

ABSTRACT

The availability of drugs targeting the EGFR/HER/erbB signaling pathway has created a need for diagnostics that accurately predict treatment responses. We have developed and characterized a novel assay to provide sensitive and quantitative measures of HER proteins and homodimers in formalin-fixed, paraffin-embedded (FFPE) cell lines and breast tumor tissues, to test these variables. In the VeraTag assay, HER proteins and homodimers are detected through the release of fluorescent tags conjugated to specific HER antibodies, requiring proximity to a second HER antibody. HER2 protein quantification was normalized to tumor area, and compared to receptor numbers in 12 human tumor cell lines determined by fluorescence-activated cell sorting (FACS), and with HER immunohistochemistry (IHC) test categories and histoscores in cell lines and 170 breast tumors. HER1 and HER2 expression levels determined by the VeraTag assay are proportional to receptor number over more than a 2 log10 range, and HER homodimer levels are consistent with crosslinking and immunoprecipitation results. VeraTag HER2 measurements of breast tumor tissue and cell lines correlate with standard IHC test categories (P<0.001). VeraTag HER2 levels also agree with IHC histoscores at lower HER2 protein levels, but are continuous and overlapping between IHC test categories, extending the dynamic range 5-fold to 10-fold at higher HER2 levels. The VeraTag assay specifically and reproducibly measures HER1 and HER2 protein and homodimers in FFPE tissues. The continuous measure of HER2 protein levels over a broad dynamic range, and the novel HER2 homodimer measure, are presently being assessed as predictive markers for responses to targeted HER2 therapy.


Subject(s)
Breast Neoplasms/pathology , Breast/chemistry , ErbB Receptors/analysis , Pathology, Clinical/methods , Receptor, ErbB-2/analysis , ErbB Receptors/metabolism , Female , Humans , Protein Multimerization , Receptor, ErbB-2/metabolism , Staining and Labeling/methods
7.
Biochemistry ; 44(8): 2982-92, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15723541

ABSTRACT

The E. coli isozyme of gamma-aminobutyrate aminotransferase (GABA-AT) is a tetrameric pyridoxal phosphate-dependent enzyme that catalyzes transamination between primary amines and alpha-keto acids. The roles of the active site residues V241, E211, and I50 in the GABA-AT mechanism have been probed by site-directed mutagenesis. The beta-branched side chain of V241 facilitates formation of external aldimine intermediates with primary amine substrates, while E211 provides charge compensation of R398 selectively in the primary amine half-reaction and I50 forms a hydrophobic lid at the top of the substrate binding site. The structures of the I50Q, V241A, and E211S mutants were solved by X-ray crystallography to resolutions of 2.1, 2.5, and 2.52 A, respectively. The structure of GABA-AT is similar in overall fold and active site structure to that of dialkylglycine decarboxylase, which catalyzes both transamination and decarboxylation half-reactions in its normal catalytic cycle. Therefore, an attempt was made to convert GABA-AT into a decarboxylation-dependent aminotransferase similar to dialkylglycine decarboxylase by systematic mutation of E. coli GABA-AT active site residues. Two of the twelve mutants presented, E211S/I50G/C77K and E211S/I50H/V80D, have approximately 10-fold higher decarboxylation activities than the wild-type enzyme, and the E211S/I50H/V80D has formally changed the reaction specificity to that of a decarboxylase.


Subject(s)
4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Escherichia coli/enzymology , Amino Acid Substitution , Binding, Competitive , Crystallography, X-Ray , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...