34 results on '"Lancia Darville"'
Search Results
2. Different mechanisms of serum complement activation in the plasma of common (Chelydra serpentina) and alligator (Macrochelys temminckii) snapping turtles.
- Author
-
Sarah Baker, Ethan Kessler, Lancia Darville-Bowleg, and Mark Merchant
- Subjects
Medicine ,Science - Abstract
Reptiles are declining worldwide yet our understanding of their immune function lags far behind other taxa. The innate immune system is the primary mode of defense in reptiles, and the serum complement cascade is its major component. We assessed serum complement activity of plasma in two closely related aquatic turtle species, the common snapping turtle (CST; Chelydra serpentina) and alligator snapping turtle (AST; Macrochelys temminckii). We used a sheep red blood cell (SRBC) hemolysis assay to assess serum complement activity. Although the antibacterial activities of the plasma of these turtle species are similar, the hemolytic activity was much stronger in CST than AST. Treatment with inhibitors of the serum complement cascade indicated differences in the mechanisms of complement activation between the turtle species. We subjected plasma from both turtle species to mannan affinity chromatography and analyzed the eluate with SDS-PAGE, which revealed that plasma from the CSTs contained only small amounts of one C-type lectin protein while the AST plasma contained high concentrations of two C-type lectins (31.0 and 35.9 kDa). Edman degradation analyses confirmed that the two AST proteins contained identical N-terminal sequences. Thus, the CST appears to rely more heavily on the alternative mechanism of serum complement activation, while the AST appears to rely more on the lectin-mediated pathway, which is a pattern recognition response to prokaryotes not activated by the SRBCs. These results are unique in that the use of serum complement pathways are generally assumed to be conserved within clades.
- Published
- 2019
- Full Text
- View/download PDF
3. Correction: Different mechanisms of serum complement activation in the plasma of common (Chelydra serpentina) and alligator (Macrochelys temminckii) snapping turtles.
- Author
-
Sarah Baker, Ethan Kessler, Lancia Darville, and Mark Merchant
- Subjects
Medicine ,Science - Abstract
[This corrects the article DOI: 10.1371/journal.pone.0217626.].
- Published
- 2019
- Full Text
- View/download PDF
4. Fucosylated Proteome Profiling Identifies a Fucosylated, Non-Ribosomal, Stress-Responsive Species of Ribosomal Protein S3
- Author
-
Gregory Watson, Daniel Lester, Hui Ren, Connor M. Forsyth, Elliot Medina, David Gonzalez Perez, Lancia Darville, Jiqiang Yao, Vince Luca, John Koomen, Ling Cen, and Eric Lau
- Subjects
ribosomal protein S3 ,fucosylation ,melanoma ,Cytology ,QH573-671 - Abstract
Alterations in genes encoding for proteins that control fucosylation are known to play causative roles in several developmental disorders, such as Dowling-Degos disease 2 and congenital disorder of glycosylation type IIc (CDGIIc). Recent studies have provided evidence that changes in fucosylation can contribute to the development and progression of several different types of cancers. It is therefore important to gain a detailed understanding of how fucosylation is altered in disease states so that interventions may be developed for therapeutic purposes. In this report, we find that fucosylation occurs on many intracellular proteins. This is an interesting finding, as the fucosylation machinery is restricted to the secretory pathway and is thought to predominately affect cell-membrane-bound and secreted proteins. We find that Ribosomal protein S3 (RPS3) is fucosylated in normal tissues and in cancer cells, and that the extent of its fucosylation appears to respond to stress, including MAPK inhibitors, suggesting a new role in posttranslational protein function. Our data identify a new ribosome-independent species of fucosylated RPS3 that interacts with proteins involved in posttranscriptional regulation of RNA, such as Heterogeneous nuclear ribonucleoprotein U (HNRNPU), as well as with a predominance of non-coding RNAs. These data highlight a novel role for RPS3, which, given previously reported oncogenic roles for RPS3, might represent functions that are perturbed in pathologies such as cancer. Together, our findings suggest a previously unrecognized role for fucosylation in directly influencing intracellular protein functions.
- Published
- 2021
- Full Text
- View/download PDF
5. Fucosylation of HLA-DRB1 regulates CD4+ T cell-mediated anti-melanoma immunity and enhances immunotherapy efficacy
- Author
-
Daniel K. Lester, Chase Burton, Alycia Gardner, Patrick Innamarato, Krithika Kodumudi, Qian Liu, Emma Adhikari, Qianqian Ming, Daniel B. Williamson, Dennie T. Frederick, Tatyana Sharova, Michael G. White, Joseph Markowitz, Biwei Cao, Jonathan Nguyen, Joseph Johnson, Matthew Beatty, Andrea Mockabee-Macias, Matthew Mercurio, Gregory Watson, Pei-Ling Chen, Susan McCarthy, Carlos MoranSegura, Jane Messina, Kerry L. Thomas, Lancia Darville, Victoria Izumi, John M. Koomen, Shari A. Pilon-Thomas, Brian Ruffell, Vincent C. Luca, Robert S. Haltiwanger, Xuefeng Wang, Jennifer A. Wargo, Genevieve M. Boland, and Eric K. Lau
- Subjects
Cancer Research ,Oncology - Abstract
Despite reports of striking outcomes, immunotherapy efficacy in melanoma is limited to subsets of patients 1, 2. Combining immunotherapies with other modalities has yielded limited improvements but also adverse events requiring cessation of treatment 1. In addition to ineffective patient stratification, efficacy can be impaired by paucity of tumor-infiltrating lymphocytes (TILs). Thus, effective strategies to safely increase TILs are urgently needed to improve immunotherapies 3. Here, we report that dietary administration of the sugar L-fucose triggers CD4+T cell-mediated increases in TILs, anti-tumor immunity, and enhanced immune checkpoint blockade responses. This is induced by the fucosylation and cell surface enrichment of the MHC-II protein HLA-DRB1 in melanoma. Single-cell immunofluorescent staining analysis of patient melanoma specimens demonstrates that fucosylation and fucosylated HLA-DRB1 is associated with intratumoral T cell abundance and anti-PD1 responder status. Our findings demonstrate that fucosylation is a key mediator of anti-tumor immunity, via regulation of melanoma cell surface HLA-DRB1 and induction of anti-tumor immunity, suggesting use of melanoma fucosylation as a novel strategy to stratify patients for immunotherapies. Importantly, our study suggests that L-fucose represents a powerful, non-toxic agent for safely increasing anti-tumor immunity and immunotherapy efficacy in melanoma.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table S4 from Cell Type–specific Adaptive Signaling Responses to KRASG12C Inhibition
- Author
-
Eric B. Haura, Uwe Rix, John M. Koomen, Denis Imbody, Fumi Kinose, Sandip Chavan, Ryan Franzese, Brandon Stone, Lancia Darville, Victoria Izumi, Bin Fang, Eric A. Welsh, and Hitendra S. Solanki
- Abstract
Supplementary Table S4. The complete list of identified phosphopeptides after (pY) enrichment and TMT-based quantitative phosphoproteomics analysis in KRASG12C mutant lung cancer cell lines (n=8) with mean-centered log abundance values.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figures from Cell Type–specific Adaptive Signaling Responses to KRASG12C Inhibition
- Author
-
Eric B. Haura, Uwe Rix, John M. Koomen, Denis Imbody, Fumi Kinose, Sandip Chavan, Ryan Franzese, Brandon Stone, Lancia Darville, Victoria Izumi, Bin Fang, Eric A. Welsh, and Hitendra S. Solanki
- Abstract
Supplementary Fig. S1. Differential response (2D and 3D culture) of KRASG12C specific covalent inhibitors (ARS-1620 and AMG510) in KRASG12C lung cancer cell lines and signaling rebound following KRASG12C inhibition. Supplementary Fig. S2. MetaCore literature network after ARS-1620 treatment in H358 cells. Supplementary Fig. S3. TMT design for expression and pY comparison in 8 KRASG12C lung cancer cell lines and two group comparison of phosphosites Supplementary Fig. S4. Analysis of CCLE data set for EMT markers and ERBB3 gene expression; and PCA representing 105 probeset TGFβ-EMT signatures (Gordian et al. 2019) translated into proteins expression to describe EMT activity in KRASG12C lung cancer cell lines. Supplementary Fig. S5. Impact of SHP2 inhibitors (SHP-099 and RMC-4550) on signaling in H358 cells and cell viability analysis following treatment with SHP2 inhibitors or in the combination with ARS-1620 in H358 and H1792 cells. Supplementary Fig. S6. Literature-based signaling network after KRASG12C inhibition in H1792 cells and cell viability analysis following treatment with ARS-1620 (1µM), AZD-4547 (1µM) and their combination in NSCLCG12C mutant cell lines. Supplementary Fig. S7. Effect of TGFβ stimulation on cell viability in response to KRASG12C inhibition and different combination strategies. Supplementary Fig. S8. Heatmap showing mean-centered log abundance expression values for indicated phosphosites/protein expression in KRASG12C mutant cell lines and E vs M 2-group comparison performed for FGFR1 gene expression in CCLE data set. Supplementary Fig. S9. Literature-based signaling network after KRASG12C inhibition in Calu1 cells and impact on signaling following treatment with ARS-1620 plus drugs known to target AXL Receptor.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Methods from Cell Type–specific Adaptive Signaling Responses to KRASG12C Inhibition
- Author
-
Eric B. Haura, Uwe Rix, John M. Koomen, Denis Imbody, Fumi Kinose, Sandip Chavan, Ryan Franzese, Brandon Stone, Lancia Darville, Victoria Izumi, Bin Fang, Eric A. Welsh, and Hitendra S. Solanki
- Abstract
Detailed Supplementary Methods
- Published
- 2023
- Full Text
- View/download PDF
9. Legends to Supplementary Data from Cell Type–specific Adaptive Signaling Responses to KRASG12C Inhibition
- Author
-
Eric B. Haura, Uwe Rix, John M. Koomen, Denis Imbody, Fumi Kinose, Sandip Chavan, Ryan Franzese, Brandon Stone, Lancia Darville, Victoria Izumi, Bin Fang, Eric A. Welsh, and Hitendra S. Solanki
- Abstract
Legends to Supplementary Figures and Tables
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Cell Type–specific Adaptive Signaling Responses to KRASG12C Inhibition
- Author
-
Eric B. Haura, Uwe Rix, John M. Koomen, Denis Imbody, Fumi Kinose, Sandip Chavan, Ryan Franzese, Brandon Stone, Lancia Darville, Victoria Izumi, Bin Fang, Eric A. Welsh, and Hitendra S. Solanki
- Abstract
Purpose:Covalent inhibitors of KRASG12C specifically target tumors driven by this form of mutant KRAS, yet early studies show that bypass signaling drives adaptive resistance. Although several combination strategies have been shown to improve efficacy of KRASG12C inhibitors (KRASi), underlying mechanisms and predictive strategies for patient enrichment are less clear.Experimental Design:We performed mass spectrometry–based phosphoproteomics analysis in KRASG12C cell lines after short-term treatment with ARS-1620. To understand signaling diversity and cell type–specific markers, we compared proteome and phosphoproteomes of KRASG12C cells. Gene expression patterns of KRASG12C cell lines and lung tumor tissues were examined.Results:Our analysis suggests cell type–specific perturbation to ERBB2/3 signaling compensates for repressed ERK and AKT signaling following ARS-1620 treatment in epithelial cell type, and this subtype was also more responsive to coinhibition of SHP2 and SOS1. Conversely, both high basal and feedback activation of FGFR or AXL signaling were identified in mesenchymal cells. Inhibition of FGFR signaling suppressed feedback activation of ERK and mTOR, while AXL inhibition suppressed PI3K pathway. In both cell lines and human lung cancer tissues with KRASG12C, we observed high basal ERBB2/3 associated with epithelial gene signatures, while higher basal FGFR1 and AXL were observed in cells/tumors with mesenchymal gene signatures.Conclusions:Our phosphoproteomic study identified cell type–adaptive responses to KRASi. Markers and targets associated with ERBB2/3 signaling in epithelial subtype and with FGFR1/AXL signaling in mesenchymal subtype should be considered in patient enrichment schemes with KRASi.
- Published
- 2023
- Full Text
- View/download PDF
11. Abstract B015: Wildtype RAS activity and PI3K signaling as new vulnerabilities in cells with acquired resistance to sotorasib
- Author
-
Denis Imbody, Hitendra S. Solanki, Bina Desai, Paul A. Stewart, Yaakov Stern, Ryoji Kato, Anurima Majumder, Liznair Bridenstine, Aobuli Xieraili, Bin Fang, Lancia Darville, Fumi Kinose, John M. Koomen, Uwe Rix, Andriy Marusyk, and Eric B. Haura
- Subjects
Cancer Research ,Oncology ,Molecular Biology - Abstract
Purpose: Sotorasib (AMG510) has demonstrated remarkable response in lung cancer patients with tumors driven by oncogenic KRASG12C mutation. However, recently published clinical data identified acquired mutations in RAS and other genomic alterations as potential mechanisms of resistance. The cause of resistance in more than half of the patient cohort was undetermined with genomic sequencing. We hypothesize that rewiring of signaling networks could be the newly acquired vulnerabilities in cells progressing after sotorasib treatment. Experimental Design: We performed whole exome sequencing, transcriptomic profiling, and mass spectrometry-based proteomics/phosphoproteomics of the parental and sotorasib resistant isogenic line of LU65 (LU65-AMGR). The omics analyses were integrated with functional screens to prioritize a set of druggable targets/pathways. Results: We did not observe obvious acquired mutations that drive resistance to sotorasib in LU65-AMGR cells. To investigate compensatory signaling critically important for the survival/growth of LU65-AMGR cells, we studied signaling perturbations using mass spectrometry-based phosphoproteomics approach. We identified 430 and 1,574 phosphosites differentially expressed in resistant cells (± 1.5-fold & p < 0.05) with phosphotyrosine (pY) and global phosphoproteome (pS/T/Y) enrichment, respectively. Analysis of phosphoproteomics data identified increased phosphorylation of multiple RTKs including EGFR, HER2, HER3, IGF1R, AXL and PDGFRA, including others. RAS-GTP pull-down show increased levels of RAS-GTP and NRAS-GTP in the resistant cells. RAS isoform quantification using parallel reaction monitoring (MRM) mass spectrometry further confirmed increased activity of NRAS in pull-down samples. Transcriptomic analysis revealed elevated genes responsible for anti-apoptosis pathways mediated by PI3K/AKT signaling. Enrichment of transcription factors such as AP1 and AP-2A could drive enhanced EGF expression and EGFR phosphorylation in sotorasib resistant cells. Consistent with enhanced RTK and RAS activity, Western blots confirmed higher phosphorylation of ERK and AKT in LU65-AMGR cells. We show higher EGFR phosphorylation in resistant cells when compared to the parental counterpart. While LU65 cells viability is reduced markedly by sotorasib combination with afatinib, an irreversible HER family inhibitor, resistant cells showed lesser effect on cell viability with afatinib combination. Consistent with our in vitro observations, the sotorasib and afatinib combination treatment significantly regressed tumor growth only in LU65 xenografts, but not in LU65-AMGR xenografts. Conclusions: Our data suggest that activation of multiple RTKs maintains RAS activity and PI3K signaling in LU65-AMGR cells. Thus, dual pan-RAS and PI3K inhibition could serve as promising strategy of treatment in relapsed tumors identified with WT RAS and PI3K signaling activation. Citation Format: Denis Imbody, Hitendra S. Solanki, Bina Desai, Paul A. Stewart, Yaakov Stern, Ryoji Kato, Anurima Majumder, Liznair Bridenstine, Aobuli Xieraili, Bin Fang, Lancia Darville, Fumi Kinose, John M. Koomen, Uwe Rix, Andriy Marusyk, Eric B. Haura. Wildtype RAS activity and PI3K signaling as new vulnerabilities in cells with acquired resistance to sotorasib [abstract]. In: Proceedings of the AACR Special Conference: Targeting RAS; 2023 Mar 5-8; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Res 2023;21(5_Suppl):Abstract nr B015.
- Published
- 2023
- Full Text
- View/download PDF
12. Plasma complement activation mechanisms differ in ornate (Terrapene ornata ornata) and eastern box turtles (Terrapene carolina carolina)
- Author
-
Lancia Darville, Mark Merchant, Laura Adamovicz, Matthew C. Allender, and Sarah J. Baker
- Subjects
Male ,0106 biological sciences ,0301 basic medicine ,Erythrocytes ,Physiology ,Zoology ,Terrapene ornata ,Hemolysis ,010603 evolutionary biology ,01 natural sciences ,Chromatography, Affinity ,03 medical and health sciences ,Genetics ,medicine ,Eastern box turtle ,Animals ,Complement Activation ,Molecular Biology ,Polyacrylamide gel electrophoresis ,Ecology, Evolution, Behavior and Systematics ,Sheep ,biology ,Temperature ,medicine.disease ,biology.organism_classification ,Turtles ,Complement system ,030104 developmental biology ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Lectin pathway ,Alternative complement pathway ,Electrophoresis, Polyacrylamide Gel ,Female ,Animal Science and Zoology ,Box turtle - Abstract
Eastern (Terrapene carolina carolina) and ornate (Terrapene ornata ornata) box turtles have robust plasma antibacterial activity, however, the mechanism behind this activity is unknown. We used sheep red blood cell (SRBC) hemolysis assays, mannan-affinity chromatography, sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), and matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) to explore the mechanisms of complement activity in box turtles. Plasma from both species demonstrated volume, time, and temperature-dependent SRBC hemolysis, with significantly greater hemolytic activity in ornate box turtle plasma. Hemolytic activity was highly attenuated following treatment with heat, EDTA, and salicylaldoxime in both species, but was unchanged after treatment with methylamine and ammonium hydroxide. Two abundant mannan-binding proteins (presumed C-type lectins) were identified in eastern box turtle plasma using SDS-PAGE and MALDI-TOF, but ornate box turtles did not express either protein. Eastern box turtles appear to rely on the lectin pathway of complement activation while ornate box turtles utilize the alternative pathway. This study provides further evidence that mechanisms underlying immune function are not always conserved between closely related species. This finding may have important implications for explaining species differences in susceptibility to emerging threats such as disease, toxicants, and climate change.
- Published
- 2020
- Full Text
- View/download PDF
13. Fucosylated Proteome Profiling Identifies a Fucosylated, Non-Ribosomal, Stress-Responsive Species of Ribosomal Protein S3
- Author
-
Eric Lau, Hui Ren, Lancia Darville, Connor M Forsyth, Elliot Medina, Ling Cen, John M. Koomen, Gregory W. Watson, Vince Luca, Jiqiang Yao, David Gonzalez Perez, and Daniel K. Lester
- Subjects
0301 basic medicine ,Ribosomal Proteins ,Glycosylation ,QH301-705.5 ,Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Ribosomal protein ,Cell Line, Tumor ,Neoplasms ,ribosomal protein S3 ,melanoma ,Animals ,Humans ,Biology (General) ,Gene ,Fucosylation ,Secretory pathway ,RNA ,General Medicine ,Ribosomal RNA ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Secretory protein ,030220 oncology & carcinogenesis ,Cancer cell ,fucosylation - Abstract
Simple Summary Dysregulated fucosylation has been characterized as an underlying cause or a contributor to the pathogenesis of several disease states. However, to date, there is not a clear understanding of how and what proteins, signaling pathways, and cellular processes are impacted by fucosylation. Here, we characterized the proteins recognized by a fucose-binding lectin and unexpectedly discovered that many intracellular proteins are putatively subject to posttranslational fucosylation. We further found that fucosylation on intracellular ribosomal protein S3 responds to stimulus, and that it appears to be independent of the currently characterized fucosylation pathway. This work suggests a to-date-underappreciated role for fucosylation on intracellular proteins and supports the existence of fucosylation capabilities within cells that is not fully known. Abstract Alterations in genes encoding for proteins that control fucosylation are known to play causative roles in several developmental disorders, such as Dowling-Degos disease 2 and congenital disorder of glycosylation type IIc (CDGIIc). Recent studies have provided evidence that changes in fucosylation can contribute to the development and progression of several different types of cancers. It is therefore important to gain a detailed understanding of how fucosylation is altered in disease states so that interventions may be developed for therapeutic purposes. In this report, we find that fucosylation occurs on many intracellular proteins. This is an interesting finding, as the fucosylation machinery is restricted to the secretory pathway and is thought to predominately affect cell-membrane-bound and secreted proteins. We find that Ribosomal protein S3 (RPS3) is fucosylated in normal tissues and in cancer cells, and that the extent of its fucosylation appears to respond to stress, including MAPK inhibitors, suggesting a new role in posttranslational protein function. Our data identify a new ribosome-independent species of fucosylated RPS3 that interacts with proteins involved in posttranscriptional regulation of RNA, such as Heterogeneous nuclear ribonucleoprotein U (HNRNPU), as well as with a predominance of non-coding RNAs. These data highlight a novel role for RPS3, which, given previously reported oncogenic roles for RPS3, might represent functions that are perturbed in pathologies such as cancer. Together, our findings suggest a previously unrecognized role for fucosylation in directly influencing intracellular protein functions.
- Published
- 2021
14. Cell-type Specific Adaptive Signaling Responses to KRAS(G12C) inhibition
- Author
-
Lancia Darville, Uwe Rix, Eric B. Haura, Eric A. Welsh, Denis Imbody, Hitendra S. Solanki, Brandon Stone, Victoria Izumi, Fumi Kinose, Ryan Franzese, Bin Fang, Sandip Chavan, and John M. Koomen
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Proteomics ,Cancer Research ,Epithelial-Mesenchymal Transition ,Receptor, ErbB-3 ,Receptor, ErbB-2 ,Cell ,Biology ,Article ,Piperazines ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Cell Line, Tumor ,Protein Interaction Mapping ,medicine ,Biomarkers, Tumor ,Humans ,Protein Interaction Maps ,Receptor, Fibroblast Growth Factor, Type 1 ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Alleles ,Mesenchymal stem cell ,Phosphoproteomics ,Computational Biology ,Phosphoproteins ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Amino Acid Substitution ,Cell culture ,030220 oncology & carcinogenesis ,Mutation ,SOS1 ,Cancer research ,Quinazolines ,Chromatography, Liquid ,Signal Transduction - Abstract
Purpose: Covalent inhibitors of KRASG12C specifically target tumors driven by this form of mutant KRAS, yet early studies show that bypass signaling drives adaptive resistance. Although several combination strategies have been shown to improve efficacy of KRASG12C inhibitors (KRASi), underlying mechanisms and predictive strategies for patient enrichment are less clear. Experimental Design: We performed mass spectrometry–based phosphoproteomics analysis in KRASG12C cell lines after short-term treatment with ARS-1620. To understand signaling diversity and cell type–specific markers, we compared proteome and phosphoproteomes of KRASG12C cells. Gene expression patterns of KRASG12C cell lines and lung tumor tissues were examined. Results: Our analysis suggests cell type–specific perturbation to ERBB2/3 signaling compensates for repressed ERK and AKT signaling following ARS-1620 treatment in epithelial cell type, and this subtype was also more responsive to coinhibition of SHP2 and SOS1. Conversely, both high basal and feedback activation of FGFR or AXL signaling were identified in mesenchymal cells. Inhibition of FGFR signaling suppressed feedback activation of ERK and mTOR, while AXL inhibition suppressed PI3K pathway. In both cell lines and human lung cancer tissues with KRASG12C, we observed high basal ERBB2/3 associated with epithelial gene signatures, while higher basal FGFR1 and AXL were observed in cells/tumors with mesenchymal gene signatures. Conclusions: Our phosphoproteomic study identified cell type–adaptive responses to KRASi. Markers and targets associated with ERBB2/3 signaling in epithelial subtype and with FGFR1/AXL signaling in mesenchymal subtype should be considered in patient enrichment schemes with KRASi.
- Published
- 2021
15. The impact of pre-treatment plasma sex hormones on the immune checkpoint inhibitors response to metastatic non-small cell lung cancer
- Author
-
Yumeng Zhang, Lancia Darville, Stephanie Hogue, Julie E. Hallanger-Johnson, Youngchul Kim, Jhanelle Elaine Gray, and Lary A. Robinson
- Subjects
Cancer Research ,Oncology - Abstract
e21088 Background: Immune checkpoint inhibitors (ICI) have changed the outcomes in metastatic non-small cell lung cancer (mNSCLC). The immune system partially determines the response to ICI. The impact of sex hormones on immune responses is well recognized in autoimmune disease and vaccine response. Little is known about the interplay between the sex hormones and the response to ICI in mNSCLC. Methods: This prospective, observational study included patients with mNSCLC at Moffitt Cancer Center who received ICI as monotherapy or combination with chemotherapy. Plasma was collected before the first ICI infusion. Extraction and detection of the hormones were optimized by modifying the previous method. Hormone levels were measured using ultra-high-performance liquid chromatography high-resolution mass spectrometry (UHPLC-HRMS). Estrogens included propyl pyrazole triol (PPT), 17β-estradiol (E2), and S-equol. Androgens included 5-androstenediol, 3β-androstenediol (3β-diol), and dehydroepiandrosterone (DHEA).Patients were divided based on clinical benefits, defined as a complete or partial response or stable disease for at least 12 months. Categorical variables were compared using the Chi-square test or Fischer exact test; continuous variables were compared using the Mann-Whitney U test. Results: Sixty-one patients were included and 50.8% were female. The median age was 67 years. Twenty-eight patients had clinical benefits from ICI. Patients in the clinical benefit group had higher BMI. The clinical benefit group had significantly lower androgen levels but no difference in estrogen levels. The median DHEA level was 3.7 ng/ml in the clinical benefit group and 9.6 ng/ml in the group without (p < 0.001). The 5-Androstenediol was also lower in the clinical benefits group (table I). When normalized to the age/sex-adjusted reference range, the difference of DHEA and 5-androstenediol was retained (p < 0.001 and 0.014, respectively). 3β-diol was below the limit of detection. Conclusions: The serum DHEA and 5-androstenediol levels were lower in mNSCLC patients who benefited from ICI, regardless of sex. Larger studies are needed to confirm the role of pre-treatment androgens levels as negative biomarkers for ICI response. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
16. Cereblon harnesses Myc-dependent bioenergetics and activity of CD8(+) T lymphocytes
- Author
-
Xue-Zhong Yu, John M. Koomen, Nicholas J. Lawrence, Mario R. Fernandez, William E. Goodheart, John L. Cleveland, Adam W. Mailloux, Ying Han, Anjali M. Rajadhyaksha, Muhammad Ayaz, Sang Y Yun, Matthew Beatty, Afua A. Akuffo, Jianing Fu, Timothy J. Garrett, Lancia Darville, Pearlie K. Epling-Burnette, Robert J. Gillies, Lanzhu Yue, Chunying Yang, Shiun Chang, Kun Jiang, Aileen Y. Alontaga, Rebecca S Hesterberg, Julia M.R. Billington, Jessica M. McDaniel, Jane L. Messina, Harshani R. Lawrence, Christelle M. Colin, Xiubao Ren, and Shonagh Russell
- Subjects
Immunobiology and Immunotherapy ,Immunology ,Melanoma, Experimental ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Biochemistry ,Ornithine decarboxylase ,Immunomodulation ,Proto-Oncogene Proteins c-myc ,Mice ,Animals ,Receptor ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Mice, Inbred BALB C ,biology ,Chemistry ,Effector ,Cereblon ,Biological activity ,Cell Biology ,Hematology ,Cell biology ,Ubiquitin ligase ,Mice, Inbred C57BL ,biology.protein ,Energy Metabolism ,Ex vivo ,CD8 - Abstract
Immunomodulatory drugs, such as thalidomide and related compounds, potentiate T-cell effector functions. Cereblon (CRBN), a substrate receptor of the DDB1-cullin-RING E3 ubiquitin ligase complex, is the only molecular target for this drug class, where drug-induced, ubiquitin-dependent degradation of known “neosubstrates,” such as IKAROS, AIOLOS, and CK1α, accounts for their biological activity. Far less clear is whether these CRBN E3 ligase-modulating compounds disrupt the endogenous functions of CRBN. We report that CRBN functions in a feedback loop that harnesses antigen-specific CD8+ T-cell effector responses. Specifically, Crbn deficiency in murine CD8+ T cells augments their central metabolism manifested as elevated bioenergetics, with supraphysiological levels of polyamines, secondary to enhanced glucose and amino acid transport, and with increased expression of metabolic enzymes, including the polyamine biosynthetic enzyme ornithine decarboxylase. Treatment with CRBN-modulating compounds similarly augments central metabolism of human CD8+ T cells. Notably, the metabolic control of CD8+ T cells by modulating compounds or Crbn deficiency is linked to increased and sustained expression of the master metabolic regulator MYC. Finally, Crbn-deficient T cells have augmented antigen-specific cytolytic activity vs melanoma tumor cells, ex vivo and in vivo, and drive accelerated and highly aggressive graft-versus-host disease. Therefore, CRBN functions to harness the activation of CD8+ T cells, and this phenotype can be exploited by treatment with drugs.
- Published
- 2020
17. LC-HRMS of derivatized urinary estrogens and estrogen metabolites in postmenopausal women
- Author
-
Steven A. Eschrich, Lusine Yaghjyan, Jayden K. Cline, Lancia Darville, Yessica C. Martinez, Shannan N Rich, Carrie M. Rozmeski, Kathleen M. Egan, and John M. Koomen
- Subjects
Male ,medicine.drug_class ,Urinary system ,Clinical Biochemistry ,Physiology ,Estrone ,Pilot Projects ,Urine ,030226 pharmacology & pharmacy ,01 natural sciences ,Biochemistry ,Sensitivity and Specificity ,Mass Spectrometry ,Article ,Analytical Chemistry ,Matrix (chemical analysis) ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,medicine ,Humans ,Derivatization ,Chromatography, High Pressure Liquid ,Postmenopausal women ,Chromatography ,Chemistry ,010401 analytical chemistry ,Reproducibility of Results ,Estrogens ,Cell Biology ,General Medicine ,medicine.disease ,0104 chemical sciences ,Postmenopause ,Estrogen ,Linear Models ,Female - Abstract
In order to undertake an epidemiologic study relating levels of parent estrogens (estrone and estradiol) and estrogen metabolites (EMs) to other breast cancer risk factors, we have optimized methods for EM quantification with ultra high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). A two-step approach was adopted; the first step comprised method development and evaluation of the method performance. The second step consisted of applying this method to quantify estrogens in postmenopausal women and determine if the observed patterns are consistent with the existing literature and prior knowledge of estrogen metabolism. First, 1-methylimidazole-2-sulfonyl chloride (MIS) was used to derivatize endogenous estrogens and estrogen metabolites in urine from study participants. Since C18 reversed phase columns have not been able to separate all the structurally related EMs, we used a C18-pentafluorophenyl (PFP) column. The parent estrogens and EMs were baseline resolved with distinct retention times on this C18-PFP column using a 30 min gradient. This method was used to quantify the parent estrogens and 13 EMs in urine samples collected in an initial pilot study involving males as well as pre- and peri-menopausal females to assess a range of EM levels in urine samples and enable comparison to the previous literature for assay evaluation. Detection limits ranged from 1 − 20 pg/mL depending on the EM. We evaluated matrix effects and interference as well as the intra- and inter-batch reproducibility including hydrolysis, extraction, derivatization and LC-MS analysis using charcoal-stripped human urine as a matrix. Methods were then applied to the measurement of estrogens in urine samples from 169 postmenopausal women enrolled in an epidemiological study to examine relationships between breast cancer risk, the intestinal microbiome, and urinary EMs. The results from our cohort are comparable to previous reports on urinary EMs in postmenopausal women and enabled thorough evaluation of the method.
- Published
- 2019
18. An immunoproteomic approach to characterize the CAR interactome and signalosome
- Author
-
Lancia Darville, John M. Koomen, Mibel Pabón-Saldaña, Sean Yoder, Maria C. Ramello, Wendy M. Kandell, Daniel N. Santiago, Maritza Lienlaf-Moreno, Daniel Abate-Daga, Uwe Rix, Eric B. Haura, Brent M. Kuenzi, Anders Berglund, Bin Fang, and Benzaid Ismahéne
- Subjects
Proteomics ,Adoptive cell transfer ,Proteome ,T-Lymphocytes ,T cell ,Mice, SCID ,Immunotherapy, Adoptive ,Biochemistry ,Interactome ,Article ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Receptor ,Molecular Biology ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Binding Sites ,Receptors, Chimeric Antigen ,Chemistry ,Cell Biology ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Second messenger system ,Phosphorylation ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
Adoptive transfer of T cells that express a chimeric antigen receptor (CAR) is an approved immunotherapy that may be curative for some hematological cancers. To better understand the therapeutic mechanism of action, we systematically analyzed prostate cancer-specific CAR signaling in human primary T cells by mass spectrometry. When we compared the interactomes and the signaling pathways activated by distinct CAR-T cells that shared the same antigen-binding domain but differed in their intracellular domains and their in vivo anti-tumor efficacy, we found that only second-generation CARs induced the expression of a constitutively phosphorylated form of CD3ζ that resembled the endogenous species. This phenomenon was independent of the choice of co-stimulatory domains, or the hinge/transmembrane region. Rather, it was dependent on the size of the intracellular domains. Moreover, the second-generation design was also associated with stronger phosphorylation of downstream secondary messengers, as evidenced by global phosphoproteome analysis. These results suggest that second-generation CARs can activate additional sources of CD3ζ signaling, and this may contribute to more intense signaling and superior antitumor efficacy that they display compared to third-generation CARs. Moreover, our results provide a deeper understanding of how CARs interact physically and/or functionally with endogenous T cell molecules, which will inform the development novel optimized immune receptors.
- Published
- 2019
- Full Text
- View/download PDF
19. HDAC11 deficiency disrupts oncogene-induced hematopoiesis in myeloproliferative neoplasms
- Author
-
Ross L. Levine, Chiara Conti, Lanzhu Yue, Eva Sahakian, Nathan P. Horvat, Amit Verma, John Powers, Narmin E. Amin, Afua A. Akuffo, Kenneth L. Wright, C. Gary Marshall, Pui Yee Ng, Ling Zhang, Javier Pinilla-Ibarz, Xiaozhang Zheng, John M. Koomen, Julia M.R. Billington, Zonghong Shao, Matthew Beatty, Lancia Darville, Christelle M. Colin, Pearlie K. Epling-Burnette, Eduardo M. Sotomayor, Que T. Lambert-Showers, Jennifer Y. Lee, Matthew W. Martin, Gary W. Reuther, Cem Murdun, H. Leighton Grimes, William E. Goodheart, and Vasundhara Sharma
- Subjects
Immunology ,Population ,Apoptosis ,Biology ,Biochemistry ,Histone Deacetylases ,Mice ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Progenitor cell ,education ,Cell Proliferation ,Mice, Knockout ,education.field_of_study ,Myeloproliferative Disorders ,Myeloid Neoplasia ,HDAC11 ,Cell Cycle ,food and beverages ,Cell Biology ,Hematology ,Janus Kinase 1 ,Oncogenes ,Hematopoiesis ,Transplantation ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,Haematopoiesis ,STAT Transcription Factors ,medicine.anatomical_structure ,Mutation ,Cancer research ,Histone deacetylase ,Bone marrow ,Stem cell - Abstract
Protein acetylation is an important contributor to cancer initiation. Histone deacetylase 6 (HDAC6) controls JAK2 translation and protein stability and has been implicated in JAK2-driven diseases best exemplified by myeloproliferative neoplasms (MPNs). By using novel classes of highly selective HDAC inhibitors and genetically deficient mouse models, we discovered that HDAC11 rather than HDAC6 is necessary for the proliferation and survival of oncogenic JAK2-driven MPN cells and patient samples. Notably, HDAC11 is variably expressed in primitive stem cells and is expressed largely upon lineage commitment. Although Hdac11is dispensable for normal homeostatic hematopoietic stem and progenitor cell differentiation based on chimeric bone marrow reconstitution, Hdac11 deficiency significantly reduced the abnormal megakaryocyte population, improved splenic architecture, reduced fibrosis, and increased survival in the MPLW515L-MPN mouse model during primary and secondary transplantation. Therefore, inhibitors of HDAC11 are an attractive therapy for treating patients with MPN. Although JAK2 inhibitor therapy provides substantial clinical benefit in MPN patients, the identification of alternative therapeutic targets is needed to reverse MPN pathogenesis and control malignant hematopoiesis. This study establishes HDAC11 as a unique type of target molecule that has therapeutic potential in MPN.
- Published
- 2019
20. Quantification of Apoptosis-Regulating Proteins to Guide Combination Therapy in Multiple Myeloma
- Author
-
L��pez-Ca��izares, Ashley, Lancia Darville, Magaletti, Dario, Meads, Mark, Fang, Bin, Shain, Kenneth, and Koomen, John
- Published
- 2019
- Full Text
- View/download PDF
21. CTEP 9557: A dose-escalation trial of combination dabrafenib, trametinib, and AT13387 in patients with BRAF mutant solid tumors
- Author
-
Lancia Darville, Meghan J. Mooradian, Anita Giobbie-Hurder, Harold N. Keer, Geoffrey I. Shapiro, Keiran S.M. Smalley, S. Percy Ivy, Elizabeth I. Buchbinder, James M. Cleary, Ryan J. Sullivan, John M. Koomen, Justine V. Cohen, Donald P. Lawrence, Amber Newton, Helen X. Chen, and Aparna Raj Parikh
- Subjects
Trametinib ,Cancer Research ,Oncology ,business.industry ,MEK inhibitor ,Mutant ,Cancer research ,Dose escalation ,Medicine ,Dabrafenib ,In patient ,business ,medicine.drug - Abstract
3609 Background: Combination BRAF and MEK inhibitor therapy is associated with response in patients (pts) with BRAF mutant (mut) solid tumors; however critical limitations for the durable activity of these agents remains. Preclinically, the addition of heat shock protein 90 (HSP90) inhibitors improves the efficacy of BRAF inhibitor (BRAFi) therapy in both BRAFi -sensitive and resistant mutant cell lines. Methods: CTEP study 9557 (NCT02097225) is a phase I study designed to determine the safety and efficacy of the small molecule HSP90inhibitor, AT13387, in combination with dabrafenib (dab) and trametinib (tram) in patients with BRAF V600E/K mut solid tumors. Prior chemotherapy, immunotherapy, BRAF and/or MEK exposure was permitted. The primary objective was to determine the maximum tolerated dose (MTD). Results: From July 2015 to June 2018, 22 patients with previously treated, metastatic BRAF V600E/K mut solid tumors were enrolled using a 3 + 3 design at four dose levels (DL) (Table). Pts were predominantly female (59%) with a median age of 57.5yrs (37 -75). The most common tumor type was BRAF V600Emut colon cancer (N=12). Dose limiting toxicities (DLTs) occurred in one patient in DL3 and one in DL4, specifically grade 3 myelosuppression and fatigue, respectively. The MTD was Dab 150mg [BID/PO], Tram 2mg [QD/PO] and AT1187 260mg/m2 [D1,8,15/IV]. Twenty-one of 22 pts were eligible for efficacy assessment. Best response, per RECIST 1.1, was partial response (PR) in 2 pts – one with colon ca (TKI-naïve), one with melanoma (TKI-resistant) - stable disease (SD) in 8 pts, and disease progression (PD) in 11 with a disease control rate (PR + SD) of 47.6% (90% CI: 29% - 67%). Median time to progression was significantly longer in DL3 (3.9 mths; 1.8-9.2) compared to DL1 (1.6mths; 0.9-1.7) or DL2 (1.5; 0.6-3.6). Median PFS and OS were 1.8mths (90% CI: 1.6 – 3.7mths) and 5.1 mths (90% CI: 2.5 -10.6mths), respectively. Median OS was not reached in DL3/4. Correlative data on the expression of the key signaling proteins relating to response will be presented at the meeting. Conclusions: HSP90 inhibition combined with BRAF/MEK inhibition was determined to be safe with evidence of disease control in a heavily pre-treated population of pts with BRAF V600E/K mut solid tumors. Clinical trial information: NCT02097225 . [Table: see text]
- Published
- 2020
- Full Text
- View/download PDF
22. Macrophage Metabolism of Apoptotic Cell-Derived Arginine Promotes Continual Efferocytosis and Resolution of Injury
- Author
-
Ze Zheng, Olga Ilkayeva, Lancia Darville, Arif Yurdagul, Christopher G. Kevil, Manikandan Subramanian, Brennan D. Gerlach, Deborah M. Muoio, Gopi K. Kolluru, Xiaobo Wang, John L. Cleveland, Christina C. Rymond, John M. Koomen, Scott B. Crown, Ira Tabas, and George Kuriakose
- Subjects
Male ,rac1 GTP-Binding Protein ,Physiology ,RNA Stability ,media_common.quotation_subject ,Apoptosis ,Arginine ,Ornithine Decarboxylase ,ELAV-Like Protein 1 ,Ornithine decarboxylase ,Jurkat Cells ,chemistry.chemical_compound ,Phagocytosis ,Putrescine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Myeloid Cells ,RNA, Messenger ,ARG1 ,Internalization ,Efferocytosis ,Molecular Biology ,media_common ,Arginase ,Macrophages ,Cell Biology ,Ornithine ,Cell biology ,Mice, Inbred C57BL ,chemistry ,Gene Deletion - Abstract
Continual efferocytic clearance of apoptotic cells (ACs) by macrophages prevents necrosis and promotes injury resolution. How continual efferocytosis is promoted is not clear. Here, we show that the process is optimized by linking the metabolism of engulfed cargo from initial efferocytic events to subsequent rounds. We found that continual efferocytosis is enhanced by the metabolism of AC-derived arginine and ornithine to putrescine by macrophage arginase 1 (Arg1) and ornithine decarboxylase (ODC). Putrescine augments HuR-mediated stabilization of the mRNA encoding the GTP-exchange factor Dbl, which activates actin-regulating Rac1 to facilitate subsequent rounds of AC internalization. Inhibition of any step along this pathway after first-AC uptake suppresses second-AC internalization, whereas putrescine addition rescues this defect. Mice lacking myeloid Arg1 or ODC have defects in efferocytosis in vivo and in atherosclerosis regression, while treatment with putrescine promotes atherosclerosis resolution. Thus, macrophage metabolism of AC-derived metabolites allows for optimal continual efferocytosis and resolution of injury.
- Published
- 2020
- Full Text
- View/download PDF
23. Label-free quantitative mass spectrometry analysis of differential protein expression in the developing cochlear sensory epithelium
- Author
-
Lancia Darville and Bernd H. A. Sokolowski
- Subjects
0301 basic medicine ,Proteomics ,lcsh:Cytology ,Immunoprecipitation ,Quantitative mass spectrometry ,Research ,Biology ,Development ,Hair cells ,Biochemistry ,Transmembrane protein ,Cell biology ,Cochlea ,Blot ,03 medical and health sciences ,030104 developmental biology ,Proteome ,lcsh:QH573-671 ,Networks ,Protein kinase A ,Receptor ,Molecular Biology ,Sensory epithelium - Abstract
Background The sensory epithelium of the inner ear converts the mechanical energy of sound to electro-chemical energy recognized by the central nervous system. This process is mediated by receptor cells known as hair cells that express proteins in a timely fashion with the onset of hearing. Methods The proteomes of 3, 14, and 30 day-old mice cochlear sensory epithelia were revealed, using label-free quantitative mass spectrometry (LTQ-Orbitrap). Statistical analysis using a one-way ANOVA followed by Bonferroni’s post-hoc test was used to show significant differences in protein expression. Ingenuity Pathway Analysis was used to observe networks of differentially expressed proteins, their biological processes, and associated diseases, while Cytoscape software was used to determine putative interactions with select biomarker proteins. These candidate biomarkers were further verified using Western blotting, while coimmunoprecipitation was used to verify putative partners determined using bioinformatics. Results We show that a comparison across all three proteomes shows that there are 447 differentially expressed proteins, with 387 differentially expressed between postnatal day 3 and 30. Ingenuity Pathway Analysis revealed ~ 62% of postnatal day 3 downregulated proteins are involved in neurological diseases. Several proteins are expressed exclusively on P3, including Parvin α, Drebrin1 (Drb1), Secreted protein acidic and cysteine rich (SPARC), Transmembrane emp24 domain-containing protein 10 (Tmed10). Coimmunoprecipitations showed that Parvin and SPARC interact with integrin-linked protein kinase and the large conductance calcium-activated potassium channel, respectively. Conclusions Quantitative mass spectrometry revealed the identification of numerous differentially regulated proteins over three days of postnatal development. These data provide insights into functional pathways regulating normal sensory and supporting cell development in the cochlea that include potential biomarkers. Interacting partners of two of these markers suggest the importance of these complexes in regulating cellular structure and synapse development. Electronic supplementary material The online version of this article (10.1186/s12953-018-0144-6) contains supplementary material, which is available to authorized users.
- Published
- 2018
24. Unification of de novo and acquired ibrutinib resistance in mantle cell lymphoma
- Author
-
Eduardo M. Sotomayor, Bijal D. Shah, J Tao, Kenneth H. Shain, Tint Lwin, Ying Han, Kai Fu, Lynn C. Moscinski, Huijuan Jiang, Ling Zhang, John M. Koomen, Allison Distler, Xiaohong Zhao, William S. Dalton, Dmitri Rebatchouk, Liang Zhang, Bin Fang, Timothy Jacobson, Chengfeng Bi, Mark B. Meads, Lancia Darville, Jiannong Li, Jianguo Tao, M. E. R. Silva, Michael Wang, Ariosto S. Silva, and Maurizio Di Liberto
- Subjects
0301 basic medicine ,Proteome ,Cell ,General Physics and Astronomy ,Lymphoma, Mantle-Cell ,Drug resistance ,Tyrosine-kinase inhibitor ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Piperidines ,hemic and lymphatic diseases ,Tumor Microenvironment ,Kinome ,Multidisciplinary ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Ibrutinib ,Reprogramming ,Signal Transduction ,Cell Survival ,medicine.drug_class ,Science ,Receptors, Antigen, B-Cell ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cell Proliferation ,Adenine ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,Lymphoma ,Pyrimidines ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,Immunology ,Cancer research ,Pyrazoles ,Mantle cell lymphoma ,Protein Kinases - Abstract
The novel Bruton's tyrosine kinase inhibitor ibrutinib has demonstrated high response rates in B-cell lymphomas; however, a growing number of ibrutinib-treated patients relapse with resistance and fulminant progression. Using chemical proteomics and an organotypic cell-based drug screening assay, we determine the functional role of the tumour microenvironment (TME) in ibrutinib activity and acquired ibrutinib resistance. We demonstrate that MCL cells develop ibrutinib resistance through evolutionary processes driven by dynamic feedback between MCL cells and TME, leading to kinome adaptive reprogramming, bypassing the effect of ibrutinib and reciprocal activation of PI3K-AKT-mTOR and integrin-β1 signalling. Combinatorial disruption of B-cell receptor signalling and PI3K-AKT-mTOR axis leads to release of MCL cells from TME, reversal of drug resistance and enhanced anti-MCL activity in MCL patient samples and patient-derived xenograft models. This study unifies TME-mediated de novo and acquired drug resistance mechanisms and provides a novel combination therapeutic strategy against MCL and other B-cell malignancies., Ibrutinib has demonstrated high response rates in B-cell lymphomas but a lot of ibrutinib-treated patients relapse with resistance. This study unified TME-mediated de novo and acquired drug resistance through B-cell receptor signalling and PI3K-AKT-mTOR axis and provides a combination therapeutic strategy against B-cell malignancies.
- Published
- 2017
- Full Text
- View/download PDF
25. Selective Targeting of Histone Deacetylase 11 Disables Metabolism of Myeloproliferative Neoplasms
- Author
-
Eduardo M. Sotomayor, Ross L. Levine, Narmin E. Amin, Julia M.R. Billington, Amit Verma, John Powers, Nathan P. Horvat, Zonghong Shao, Lancia Darville, Pearlie K. Epling-Burnette, Afua A. Akuffo, Kenneth P. Wright, H. Leighton Grimes, Cem Murdun, Gary C. Marshall, Vasundhara Sharma, Gary W. Reuther, Ling Zhang, Xiaozhang Zheng, Que T. Lambert-Showers, Mathew W. Martin, Jennifer Lee, Pui Yee Ng, Mathew Beatty, Lanzhu Yue, John M. Koomen, Eva Sahakian, Agni Christodoulidou, William E. Goodheart, and Javier Pinilla Ibarz
- Subjects
biology ,business.industry ,HDAC11 ,Immunology ,Cell Biology ,Hematology ,HDAC6 ,medicine.disease ,Biochemistry ,Transplantation ,Leukemia ,Histone ,Cancer research ,biology.protein ,Medicine ,Electrophoretic mobility shift assay ,Histone deacetylase ,business ,K562 cells - Abstract
Introduction: Acetylated histone and non-histone proteins are pharmacologic targets for both solid and hematological cancers including myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies driven by aberrant JAK2/STAT signaling. MPNs are characterized by epigenetic alterations, including aberrant acetylation, which makes this disease particularly interesting for targeting with HDAC inhibitors. Four classes of histone deacetylases (Class I-IV HDACs) regulate gene transcription and modulate cellular processes that drive the initiation and progression of cancer. Pan-HDAC and class I-selective HDAC inhibitors have gained traction in clinical settings, yet we reasoned that specific targeting of the 18 distinct HDAC proteins may establish roles for select HDACs as therapeutic vulnerabilities in MPNs. Methods: To explore the roles of individual HDACs in MPN, we first conducted an inhibitor screen of compounds having distinct HDAC selectivity based on electrophoretic mobility shift assays with full-length human HDAC proteins expressed in baculovirus and unique peptide substrates. Ultra-specific HDAC6 compounds were initially targeted for analysis based on its previously defined role in HSP90-mediated JAK2 stabilization and translation. Survival of MPN cell line models, MPN patient samples, leukemia cell lines, and MPN disease progression in mice transplanted with Hdac6-/-, and Hdac11-/- hematopoietic stem cells (HSCs) transduced with the MPLW515L oncogene, as well as Tg-Hdac11-eGfp mice were used to show the role of HDAC6 and HDAC11 in oncogene-driven and homeostatic hematopoiesis. As further proof of specificity, HDAC6 and HDAC11 were genetically ablated in MPN model cell lines using either RNA interference or inducible shRNA. For HDAC11 substrate identification, a combination of RNA-seq, acetylated proteome (SILAC), global metabolomics (LC-MS), Seahorse metabolic assays (Agilent Technologies), enzymatic assays, and acetylation-specific immunoblotting and mutation profiling were performed (Fig. 1). Results: Despite the established interplay between HDAC6, HSP90 and JAK2, neither a highly selective HDAC6 inhibitor, HDAC6 silencing, nor the Hdac6 deficiency suppressed MPN pathogenesis, although there were clear effects on the acetylation of α-tubulin, a well characterized HDAC6-selective substrate. Intriguingly, both inhibition of HDAC11 activity with highly-specific HDAC11 inhibitors and silencing HDAC11 using an inducible validated shRNA, identified HDAC11 as a therapeutic vulnerability for multiple human MPN cell lines. The Tg-Hdac11-eGFP reporter mice showed that HDAC11 is expressed in several hematopoietic cell types, including myeloid cells, erythroblasts, and megakaryocytes. Thus, Hdac11-/- and Hdac11+/+MPLWT bone marrow were examined for steady-state hematopoiesis and transplantation chimerism. These studies demonstrated that HDAC11 does not contribute to homeostatic or transplantated bone marrow reconstitution. However, in the oncogenic MPL model, recipient mice transplanted withoncogenic MPLW515L-expressing Hdac11-deficient HSCs displayed markedly impaired cytokine-independent colony-formation, had less fibrosis, and displayed improved survival in primary and secondary MPN hematopoietic stem cell transplantation; thus HDAC11 contributes to MPN pathogenesis (Fig. 1). Studies in additional leukemia cell lines, including THP-1, HL-60, and mantle lymphoma cell lines, but not in Ramos or K562 cells, established that HDAC11 contributes to oncogene-driven events in other cell types. Mechanistically, RNA-seq, SILAC proteomics, and metabolic profiling revealed that HDAC11 controls aerobic glycolysis by deacetylating Lys343 of the glycolytic enzyme enolase-1 (ENO1), functionally inactivating ENO1. Finally, the effects of targeting HDAC11 on metabolism were augmented by blocking compensatory pathways of oxidative phosphorylation that are induced via JAK2V617Fand MPLW515L oncogenic signaling. Conclusions: Our comprehensive screens of HDAC inhibitors, coupled with our biological, in vivo and molecular studies, indicate that HDAC11 is an attractive and potent target for disabling MPN metabolism and pathogenesis. These finding support the rationale for further development of clinical HDAC11 inhibitors for the treatment of metabolically-active cancers such as MPNs. Disclosures Pinilla Ibarz: Teva: Consultancy; TG Therapeutics: Consultancy; Sanofi: Speakers Bureau; Bayer: Speakers Bureau; Novartis: Consultancy; Bristol-Myers Squibb: Consultancy; Abbvie: Consultancy, Speakers Bureau; Takeda: Consultancy, Speakers Bureau; Janssen: Consultancy, Speakers Bureau. Reuther:Incyte Corporation: Research Funding. Levine:Loxo: Membership on an entity's Board of Directors or advisory committees; Roche: Consultancy, Research Funding; Lilly: Honoraria; C4 Therapeutics: Membership on an entity's Board of Directors or advisory committees; Isoplexis: Membership on an entity's Board of Directors or advisory committees; Imago Biosciences: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy; Gilead: Consultancy; Celgene: Consultancy, Research Funding; Qiagen: Membership on an entity's Board of Directors or advisory committees; Prelude Therapeutics: Research Funding; Amgen: Honoraria. Verma:BMS: Research Funding; Janssen: Research Funding; Stelexis: Equity Ownership, Honoraria; Acceleron: Honoraria; Celgene: Honoraria. Epling-Burnette:Incyte Corporation: Research Funding; Celgene Corporation: Patents & Royalties, Research Funding; Forma Therapeutics: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
26. Correction: Different mechanisms of serum complement activation in the plasma of common (Chelydra serpentina) and alligator (Macrochelys temminckii) snapping turtles
- Author
-
Ethan J. Kessler, Sarah J. Baker, Mark Merchant, and Lancia Darville
- Subjects
Multidisciplinary ,Innate immune system ,biology ,Science ,Alligator ,Common snapping turtle ,Alligator snapping turtle ,biology.organism_classification ,food.food ,Complement system ,law.invention ,Immune system ,food ,Biochemistry ,law ,biology.animal ,Medicine ,Turtle (robot) ,Chelydra - Abstract
Reptiles are declining worldwide yet our understanding of their immune function lags far behind other taxa. The innate immune system is the primary mode of defense in reptiles, and the serum complement cascade is its major component. We assessed serum complement activity of plasma in two closely related aquatic turtle species, the common snapping turtle (CST; Chelydra serpentina) and alligator snapping turtle (AST; Macrochelys temminckii). We used a sheep red blood cell (SRBC) hemolysis assay to assess serum complement activity. Although the antibacterial activities of the plasma of these turtle species are similar, the hemolytic activity was much stronger in CST than AST. Treatment with inhibitors of the serum complement cascade indicated differences in the mechanisms of complement activation between the turtle species. We subjected plasma from both turtle species to mannan affinity chromatography and analyzed the eluate with SDS-PAGE, which revealed that plasma from the CSTs contained only small amounts of one C-type lectin protein while the AST plasma contained high concentrations of two C-type lectins (31.0 and 35.9 kDa). Edman degradation analyses confirmed that the two AST proteins contained identical N-terminal sequences. Thus, the CST appears to rely more heavily on the alternative mechanism of serum complement activation, while the AST appears to rely more on the lectin-mediated pathway, which is a pattern recognition response to prokaryotes not activated by the SRBCs. These results are unique in that the use of serum complement pathways are generally assumed to be conserved within clades.
- Published
- 2019
- Full Text
- View/download PDF
27. The Lack of Cardiomyocyte Complex and Hybrid N‐glycans Result in Arrhythmic Dilated Cardiomyopathy, Heart Failure, and Premature Death
- Author
-
Lancia Darville, Andrew R. Ednie, Wei Deng, and Eric S. Bennett
- Subjects
Glycan ,medicine.medical_specialty ,biology ,business.industry ,Dilated cardiomyopathy ,medicine.disease ,Biochemistry ,Premature death ,Internal medicine ,Heart failure ,Genetics ,biology.protein ,medicine ,Cardiology ,business ,Molecular Biology ,Biotechnology - Published
- 2016
- Full Text
- View/download PDF
28. Protein Quantitation of the Developing Cochlea Using Mass Spectrometry
- Author
-
Bernd H. A. Sokolowski and Lancia Darville
- Subjects
0301 basic medicine ,Sensory epithelia ,03 medical and health sciences ,Label-free quantification ,030104 developmental biology ,Chromatography ,Chemistry ,Quantitative proteomics ,Lc ms ms ,Mass spectrometry ,Proteomics ,Cochlea ,Protein expression - Abstract
Mass spectrometry-based proteomics allows for the measurement of hundreds to thousands of proteins in a biological system. Additionally, mass spectrometry can also be used to quantify proteins and peptides. However, observing quantitative differences between biological systems using mass spectrometry-based proteomics can be challenging because it is critical to have a method that is fast, reproducible, and accurate. Therefore, to study differential protein expression in biological samples labeling or label-free quantitative methods can be used. Labeling methods have been widely used in quantitative proteomics, however label-free methods have become equally as popular and more preferred because they produce faster, cleaner, and simpler results. Here, we describe the methods by which proteins are isolated and identified from cochlear sensory epithelia tissues at different ages and quantitatively differentiated using label-free mass spectrometry.
- Published
- 2016
- Full Text
- View/download PDF
29. Proteome analysis of the leukocytes from the American alligator (Alligator mississippiensis) using mass spectrometry
- Author
-
Lancia Darville, Mark Merchant, Azeem Hasan, and Kermit K. Murray
- Subjects
Cell Extracts ,Proteomics ,Databases, Factual ,Proteome ,Physiology ,Alligator ,Peptide ,Peptide Mapping ,Biochemistry ,Mass Spectrometry ,Homology (biology) ,biology.animal ,Leukocytes ,Genetics ,Animals ,Electrophoresis, Gel, Two-Dimensional ,Amino Acid Sequence ,Databases, Protein ,American alligator ,Molecular Biology ,Gel electrophoresis ,chemistry.chemical_classification ,Alligators and Crocodiles ,Sequence Homology, Amino Acid ,biology ,Proteins ,biology.organism_classification ,Molecular biology ,chemistry ,Bottom-up proteomics ,Peptides ,Software ,Chromatography, Liquid - Abstract
Mass spectrometry was used in conjunction with gel electrophoresis and liquid chromatography, to determine peptide sequences from American alligator (Alligator mississippiensis) leukocytes and to identify similar proteins based on homology. The goal of the study was to generate an initial database of proteins related to the alligator immune system. We have adopted a typical proteomics approach for this study. Proteins from leukocyte extracts were separated using two-dimensional gel electrophoresis and the major bands were excised, digested and analyzed by on-line nano-LC MS/MS to generate peptide sequences. The sequences generated were used to identify proteins and characterize their functions. The protein identity and characterization of the protein function were based on matching two or more peptides to the same protein by searching against the NCBI database using MASCOT and Basic Local Alignment Search Tool (BLAST). For those proteins with only one peptide matching, the phylum of the matched protein was considered. Forty-three proteins were identified that exhibit sequence similarities to proteins from other vertebrates. Proteins related to the cytoskeletal system were the most abundant proteins identified. These proteins are known to regulate cell mobility and phagocytosis. Several other peptides were matched to proteins that potentially have immune-related function.
- Published
- 2010
- Full Text
- View/download PDF
30. A mass spectrometry approach for the study of deglycosylated proteins
- Author
-
Mark Merchant, Lancia Darville, and Kermit K. Murray
- Subjects
Chromatography ,biology ,Chemistry ,Lectin ,Proteomics ,Mass spectrometry ,Fetuin ,Sample preparation in mass spectrometry ,Analytical Chemistry ,Matrix-assisted laser desorption/ionization ,biology.protein ,Sample preparation ,Dialysis (biochemistry) ,Spectroscopy - Abstract
Mass spectrometry (MS) analysis, after enzymatic or chemical deglycosylation, requires preparatory steps to remove salts and buffers. In this work, the glycosylated protein fetuin and a lectin protein isolated from the serum of Alligator mississippiensis were used to evaluate methods for desalting samples after an enzymatic or chemical deglycosylation. Precipitation and dialysis were used to prepare the deglycosylated samples for MS analysis. Both the precipitation and dialysis methods were suitable for sample preparation prior to analysis by matrix assisted laser desorption ionization (MALDI) MS.
- Published
- 2011
- Full Text
- View/download PDF
31. Bottom-up and Shotgun Proteomics to Identify a Comprehensive Cochlear Proteome
- Author
-
Lancia Darville and Bernd H. A. Sokolowski
- Subjects
Electrophoresis ,Proteomics ,Antibody microarray ,Proteome ,General Immunology and Microbiology ,Difference gel electrophoresis ,General Chemical Engineering ,General Neuroscience ,Quantitative proteomics ,Computational biology ,Biology ,Bioinformatics ,Tandem mass spectrometry ,Chromatography, Ion Exchange ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Cochlea ,Mice ,Tandem Mass Spectrometry ,Mice, Inbred CBA ,Animals ,Bottom-up proteomics ,Shotgun proteomics - Abstract
Proteomics is a commonly used approach that can provide insights into complex biological systems. The cochlear sensory epithelium contains receptors that transduce the mechanical energy of sound into an electro-chemical energy processed by the peripheral and central nervous systems. Several proteomic techniques have been developed to study the cochlear inner ear, such as two-dimensional difference gel electrophoresis (2D-DIGE), antibody microarray, and mass spectrometry (MS). MS is the most comprehensive and versatile tool in proteomics and in conjunction with separation methods can provide an in-depth proteome of biological samples. Separation methods combined with MS has the ability to enrich protein samples, detect low molecular weight and hydrophobic proteins, and identify low abundant proteins by reducing the proteome dynamic range. Different digestion strategies can be applied to whole lysate or to fractionated protein lysate to enhance peptide and protein sequence coverage. Utilization of different separation techniques, including strong cation exchange (SCX), reversed-phase (RP), and gel-eluted liquid fraction entrapment electrophoresis (GELFrEE) can be applied to reduce sample complexity prior to MS analysis for protein identification.
- Published
- 2014
- Full Text
- View/download PDF
32. In-depth Proteomic Analysis of Mouse Cochlear Sensory Epithelium by Mass Spectrometry
- Author
-
Lancia Darville and Bernd H. A. Sokolowski
- Subjects
Proteome ,Proteolysis ,Biology ,Chemical Fractionation ,Tandem mass spectrometry ,Proteomics ,Mass spectrometry ,Biochemistry ,Article ,Epithelium ,Mice ,Tandem Mass Spectrometry ,medicine ,Animals ,medicine.diagnostic_test ,Cell Membrane ,Membrane Proteins ,Molecular Sequence Annotation ,General Chemistry ,Trypsin ,Molecular biology ,Peptide Fragments ,Cochlea ,Membrane protein ,Mice, Inbred CBA ,Digestion ,Hydrophobic and Hydrophilic Interactions ,medicine.drug - Abstract
Proteomic analysis of sensory organs such as the cochlea is challenging due to its small size and difficulties with membrane protein isolation. Mass spectrometry in conjunction with separation methods can provide a more comprehensive proteome, because of the ability to enrich protein samples, detect hydrophobic proteins, and identify low abundant proteins by reducing the proteome dynamic range. GELFrEE as well as different separation and digestion techniques were combined with FASP and nanoLC-MS/MS to obtain an in-depth proteome analysis of cochlear sensory epithelium from 30-day-old mice. Digestion with LysC/trypsin followed by SCX fractionation and multiple nanoLC-MS/MS analyses identified 3773 proteins with a 1% FDR. Of these, 694 protein IDs were in the plasmalemma. Protein IDs obtained by combining outcomes from GELFrEE/LysC/trypsin with GELFrEE/trypsin/trypsin generated 2779 proteins, of which 606 additional proteins were identified using the GELFrEE/LysC/trypsin approach. Combining results from the different techniques resulted in a total of 4620 IDs, including a number of previously unreported proteins. GO analyses showed high expression of binding and catalytic proteins as well as proteins associated with metabolism. The results show that the application of multiple techniques is needed to provide an exhaustive proteome of the cochlear sensory epithelium that includes many membrane proteins. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium with the data set identifier PXD000231.
- Published
- 2013
33. Differential Protein Analysis of Developing Mouse Cochlea using Label‐Free Quantitative Mass Spectrometry
- Author
-
Bernd H. A. Sokolowski and Lancia Darville
- Subjects
Chemistry ,Genetics ,Biophysics ,Mouse Cochlea ,Mass spectrometry ,Molecular Biology ,Biochemistry ,Differential (mathematics) ,Biotechnology ,Label free - Published
- 2013
- Full Text
- View/download PDF
34. Isolation and determination of the primary structure of a lectin protein from the serum of the American alligator (Alligator mississippiensis)
- Author
-
Kermit K. Murray, Vivekananda Reddy Siddavarapu, Lancia Darville, Mark Merchant, Venkata Maccha, and Azeem Hasan
- Subjects
Physiology ,Alligator ,Molecular Sequence Data ,Mannose ,Reptilian Proteins ,Biology ,Biochemistry ,Chromatography, Affinity ,chemistry.chemical_compound ,Protein sequencing ,Sequence Analysis, Protein ,Tandem Mass Spectrometry ,biology.animal ,Animals ,Amino Acid Sequence ,American alligator ,Molecular Biology ,Peptide sequence ,Mannan ,Alligators and Crocodiles ,Edman degradation ,Sequence Homology, Amino Acid ,Protein primary structure ,biology.organism_classification ,Molecular biology ,Peptide Fragments ,Mannose-Binding Lectins ,chemistry ,Sequence Alignment ,Protein Binding - Abstract
Mass spectrometry in conjunction with de novo sequencing was used to determine the amino acid sequence of a 35 kDa lectin protein isolated from the serum of the American alligator that exhibits binding to mannose. The protein N-terminal sequence was determined using Edman degradation and enzymatic digestion with different proteases was used to generate peptide fragments for analysis by liquid chromatography tandem mass spectrometry (LC MS/MS). Separate analysis of the protein digests with multiple enzymes enhanced the protein sequence coverage. De novo sequencing was accomplished using MASCOT Distiller and PEAKS software and the sequences were searched against the NCBI database using MASCOT and BLAST to identify homologous peptides. MS analysis of the intact protein indicated that it is present primarily as monomer and dimer in vitro . The isolated 35 kDa protein was ~ 98% sequenced and found to have 313 amino acids and nine cysteine residues and was identified as an alligator lectin. The alligator lectin sequence was aligned with other lectin sequences using DIALIGN and ClustalW software and was found to exhibit 58% and 59% similarity to both human and mouse intelectin-1. The alligator lectin exhibited strong binding affinities toward mannan and mannose as compared to other tested carbohydrates.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.