Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael E. Wright is active.

Publication


Featured researches published by Michael E. Wright.


The EMBO Journal | 1998

Caspase‐mediated activation and induction of apoptosis by the mammalian Ste20‐like kinase Mst1

Jonathan D. Graves; Yukiko Gotoh; Kevin E. Draves; Diane Ambrose; David K. M. Han; Michael E. Wright; Jonathan Chernoff; Edward A. Clark; Edwin G. Krebs

Mst1 is a ubiquitously expressed serine–threonine kinase, homologous to the budding yeast Ste20, whose physiological regulation and cellular function are unknown. In this paper we show that Mst1 is specifically cleaved by a caspase 3‐like activity during apoptosis induced by either cross‐linking CD95/Fas or by staurosporine treatment. CD95/Fas‐induced cleavage of Mst1 was blocked by the cysteine protease inhibitor ZVAD‐fmk, the more selective caspase inhibitor DEVD‐CHO and by the viral serpin CrmA. Caspase‐mediated cleavage of Mst1 removes the C‐terminal regulatory domain and correlates with an increase in Mst1 activity in vivo, consistent with caspase‐mediated cleavage activating Mst1. Overexpression of either wild‐type Mst1 or a truncated mutant induces morphological changes characteristic of apoptosis. Furthermore, exogenously expressed Mst1 is cleaved, indicating that Mst1 can activate caspases that result in its cleavage. Kinase‐dead Mst1 did not induce morphological alterations and was not cleaved upon overexpression, indicating that Mst1 must be catalytically active in order to mediate these effects. Mst1 activates MKK6, p38 MAPK, MKK7 and SAPK in co‐transfection assays, suggesting that Mst1 may activate these pathways. Our findings suggest the existence of a positive feedback loop involving Mst1, and possibly the SAPK and p38 MAPK pathways, which serves to amplify the apoptotic response.


Molecular & Cellular Proteomics | 2005

Mass Spectrometry-based Expression Profiling of Clinical Prostate Cancer

Michael E. Wright; David K. Han; Ruedi Aebersold

The maturation of MS technologies has provided a rich opportunity to interrogate protein expression patterns in normal and disease states by applying expression protein profiling methods. Major goals of this research strategy include the identification of protein biomarkers that demarcate normal and disease populations, and the identification of therapeutic biomarkers for the treatment of diseases such as cancer (Celis, J. E., and Gromov, P. (2003) Proteomics in translational cancer research: Toward an integrated approach. Cancer Cell 3, 9–151). Prostate cancer is one disease that would greatly benefit from implementing MS-based expression profiling methods because of the need to stratify the disease based on molecular markers. In this review, we will summarize the current MS-based methods to identify and validate biomarkers in human prostate cancer. Lastly, we propose a reverse proteomic approach implementing a quantitative MS research strategy to identify and quantify biomarkers implicated in prostate cancer development. With this approach, the absolute levels of prostate cancer biomarkers will be identified and quantified in normal and diseased samples by measuring the levels of native peptide biomarkers in relation to a chemically identical but isotopically labeled reference peptide. Ultimately, a centralized prostate cancer peptide biomarker expression database could function as a repository for the identification, quantification, and validation of protein biomarker(s) during prostate cancer progression in men.


Genome Biology | 2003

Identification of androgen-coregulated protein networks from the microsomes of human prostate cancer cells

Michael E. Wright; Jimmy K. Eng; James Sherman; David M. Hockenbery; Peter S. Nelson; Timothy Galitski; Ruedi Aebersold

BackgroundAndrogens play a critical role in the development of prostate cancer-dysregulation of androgen-regulated growth pathways can led to hormone-refractory prostate cancer. A comprehensive understanding of androgen-regulated cellular processes has not been achieved to date. To this end, we have applied a large-scale proteomic approach to define cellular processes that are responsive to androgen treatment in LNCaP prostate cancer cells.ResultsUsing isotope-coded affinity tags and mass spectrometry we identified and quantified the relative abundance levels of 1,064 proteins and found that distinct cellular processes were coregulated by androgen while others were essentially unaffected. Subsequent pharmacological perturbation of the cellular process for energy generation confirmed that androgen starvation had a profound effect on this pathway.ConclusionsOur results provide evidence for the role of androgenic hormones in coordinating the expression of critical components involved in distinct cellular processes and further establish a foundation for the comprehensive reconstruction of androgen-regulated protein networks and pathways in prostate cancer cells.


Molecular & Cellular Proteomics | 2007

Identification of Putative Androgen Receptor Interaction Protein Modules Cytoskeleton and Endosomes Modulate Androgen Receptor Signaling in Prostate Cancer Cells

Rohini J. Jasavala; Harryl D. Martinez; Jaykumar Thumar; Armann Andaya; Anne-Claude Gingras; Jimmy K. Eng; Ruedi Aebersold; David K. Han; Michael E. Wright

We have developed a novel androgen receptor (AR) expression system in the 293 human embryonic kidney cell line that recapitulates AR biochemical activity as a steroid hormone receptor in prostate cancer cells. We used this system to identify putative AR-binding proteins in the cytosolic and nuclear compartments of mammalian cells using a large scale co-immunoprecipitation strategy coupled to quantitative mass spectrometry. For example, the heat shock 70 and 90 chaperones, which are known regulators of steroid hormone receptor, were identified as AR-binding proteins. AR purification enriched for proteins involved in RNA processing, protein transport, and cytoskeletal organization, suggesting a functional link between AR and these protein modules in mammalian cells. For example, AR purification in the nuclear compartment led to the specific enrichment of α-actinin-4, clathrin heavy chain, and serine-threonine protein kinase C δ. Short interfering RNA knockdown studies and co-transcriptional reporter assays revealed that clathrin heavy chain possessed co-activator activity during AR-mediated transcription, whereas α-actinin-4 and protein kinase C δ displayed both co-activator and co-repressor activity during AR-mediated transcription that was dependent upon their relative expression levels. Lastly immunohistochemical staining of prostate tissue showed that α-actinin-4 levels decreased in the nucleus of high grade cancerous prostate samples, suggesting its possible deregulation in advanced prostate cancers as previously observed in late stage metastatic breast cancers. Taken together, these findings suggest AR binds to specific protein modules in mammalian cells and that these protein modules may provide a molecular framework for interrogating AR function in normal and cancerous prostate epithelial cells.


Current protocols in human genetics | 2009

Protein Identification Using Sorcerer 2 and SEQUEST

Deborah H. Lundgren; Harryl D. Martinez; Michael E. Wright; David K. Han

Sage‐Ns Sorcerer 2 provides an integrated data analysis system for comprehensive protein identification and characterization. It runs on a proprietary version of SEQUESTR, the most widely used search engine for identifying proteins in complex mixtures. The protocol presented here describes the basic steps performed to process mass spectrometric data with Sorcerer 2 and how to analyze results using TPP and Scaffold. The unit also provides an overview of the SEQUESTR algorithm, along with Sorcerer‐SEQUESTR enhancements, and a discussion of data filtering methods, important considerations in data interpretation, and additional resources that can be of assistance to users running Sorcerer and interpreting SEQUESTR results. Curr. Protoc. Bioinform. 28:13.3.1‐13.3.21.


BMC Cancer | 2015

Androgen receptor and chemokine receptors 4 and 7 form a signaling axis to regulate CXCL12-dependent cellular motility

Jordy J. Hsiao; Brandon H. Ng; Melinda M. Smits; Jiahui Wang; Rohini J. Jasavala; Harryl D. Martinez; Jinhee Lee; Jhullian J Alston; Hiroaki Misonou; James S. Trimmer; Michael E. Wright

BackgroundIdentifying cellular signaling pathways that become corrupted in the presence of androgens that increase the metastatic potential of organ-confined tumor cells is critical to devising strategies capable of attenuating the metastatic progression of hormone-naïve, organ-confined tumors. In localized prostate cancers, gene fusions that place ETS-family transcription factors under the control of androgens drive gene expression programs that increase the invasiveness of organ-confined tumor cells. C-X-C chemokine receptor type 4 (CXCR4) is a downstream target of ERG, whose upregulation in prostate-tumor cells contributes to their migration from the prostate gland. Recent evidence suggests that CXCR4-mediated proliferation and metastasis of tumor cells is regulated by CXCR7 through its scavenging of chemokine CXCL12. However, the role of androgens in regulating CXCR4-mediated motility with respect to CXCR7 function in prostate-cancer cells remains unclear.MethodsImmunocytochemistry, western blot, and affinity-purification analyses were used to study how androgens influenced the expression, subcellular localization, and function of CXCR7, CXCR4, and androgen receptor (AR) in LNCaP prostate-tumor cells. Moreover, luciferase assays and quantitative polymerase chain reaction (qPCR) were used to study how chemokines CXCL11 and CXCL12 regulate androgen-regulated genes (ARGs) in LNCaP prostate-tumor cells. Lastly, cell motility assays were carried out to determine how androgens influenced CXCR4-dependent motility through CXCL12.ResultsHere we show that, in the LNCaP prostate-tumor cell line, androgens coordinate the expression of CXCR4 and CXCR7, thereby promoting CXCL12/CXCR4-mediated cell motility. RNA interference experiments revealed functional interactions between AR and CXCR7 in these cells. Co-localization and affinity-purification experiments support a physical interaction between AR and CXCR7 in LNCaP cells. Unexpectedly, CXCR7 resided in the nuclear compartment and modulated AR-mediated transcription. Moreover, androgen-mediated cell motility correlated positively with the co-localization of CXCR4 and CXCR7 receptors, suggesting that cell migration may be linked to functional CXCR4/CXCR7 heterodimers. Lastly, CXCL12-mediated cell motility was CXCR7-dependent, with CXCR7 expression required for optimal expression of CXCR4 protein.ConclusionsOverall, our results suggest that inhibition of CXCR7 function might decrease the metastatic potential of organ-confined prostate cancers.


Molecular & Cellular Proteomics | 2003

PROTEOME-3D: An Interactive Bioinformatics Tool for Large-Scale Data Exploration and Knowledge Discovery

Deborah H. Lundgren; Jimmy K. Eng; Michael E. Wright; David K. Han

Comprehensive understanding of biological systems requires efficient and systematic assimilation of high-throughput datasets in the context of the existing knowledge base. A major limitation in the field of proteomics is the lack of an appropriate software platform that can synthesize a large number of experimental datasets in the context of the existing knowledge base. Here, we describe a software platform, termed PROTEOME-3D, that utilizes three essential features for systematic analysis of proteomics data: creation of a scalable, queryable, customized database for identified proteins from published literature; graphical tools for displaying proteome landscapes and trends from multiple large-scale experiments; and interactive data analysis that facilitates identification of crucial networks and pathways. Thus, PROTEOME-3D offers a standardized platform to analyze high-throughput experimental datasets for the identification of crucial players in co-regulated pathways and cellular processes.


Retina-the Journal of Retinal and Vitreous Diseases | 2012

Proteomic analysis of vitreous biopsy techniques.

Jessica M. Skeie; Eric N. Brown; Harryl D. Martinez; Stephen R. Russell; Emily S. Birkholz; James C. Folk; H. Culver Boldt; Karen M. Gehrs; Edwin M. Stone; Michael E. Wright; Vinit B. Mahajan

Purpose: To compare vitreous biopsy methods using analysis platforms used in proteomics biomarker discovery. Methods: Vitreous biopsies from 10 eyes were collected sequentially using a 23-gauge needle and a 23-gauge vitreous cutter instrument. Paired specimens were evaluated by UV absorbance spectroscopy, sodium dodecyl sulfate–polyacrylamide gel electrophoresis, and liquid chromatography tandem mass spectrometry (LC-MS/MS). Results: The total protein concentration obtained with a needle and vitrectomy instrument biopsy averaged 1.10 mg/mL (standard error of the mean = 0.35) and 1.13 mg/mL (standard error of the mean = 0.25), respectively. In eight eyes with low or medium viscidity, there was a very high correlation (R2 = 0.934) between the biopsy methods. When data from 2 eyes with high viscidity vitreous were included, the correlation was reduced (R2 = 0.704). The molecular weight protein sodium dodecyl sulfate–polyacrylamide gel electrophoresis profiles of paired needle and vitreous cutter samples were similar, except for a minority of pairs with single band intensity variance. Using LC-MS/MS, equivalent peptides were identified with similar frequencies (R2 ≥ 0.90) in paired samples. Conclusion: Proteins and peptides collected from vitreous needle biopsies are nearly equivalent to those obtained from a vitreous cutter instrument. This study suggests both techniques may be used for most proteomic and biomarker discovery studies of vitreoretinal diseases, although a minority of proteins and peptides may differ in concentration.


Journal of Biological Chemistry | 2016

Discovery Proteomics Identifies a Molecular Link between the Coatomer Protein Complex I and Androgen Receptor-dependent Transcription

Jordy J. Hsiao; Melinda M. Smits; Brandon H. Ng; Jinhee Lee; Michael E. Wright

Aberrant androgen receptor (AR)-dependent transcription is a hallmark of human prostate cancers. At the molecular level, ligand-mediated AR activation is coordinated through spatial and temporal protein-protein interactions involving AR-interacting proteins, which we designate the “AR-interactome.” Despite many years of research, the ligand-sensitive protein complexes involved in ligand-mediated AR activation in prostate tumor cells have not been clearly defined. Here, we describe the development, characterization, and utilization of a novel human LNCaP prostate tumor cell line, N-AR, which stably expresses wild-type AR tagged at its N terminus with the streptavidin-binding peptide epitope (streptavidin-binding peptide-tagged wild-type androgen receptor; SBP-AR). A bioanalytical workflow involving streptavidin chromatography and label-free quantitative mass spectrometry was used to identify SBP-AR and associated ligand-sensitive cytosolic proteins/protein complexes linked to AR activation in prostate tumor cells. Functional studies verified that ligand-sensitive proteins identified in the proteomic screen encoded modulators of AR-mediated transcription, suggesting that these novel proteins were putative SBP-AR-interacting proteins in N-AR cells. This was supported by biochemical associations between recombinant SBP-AR and the ligand-sensitive coatomer protein complex I (COPI) retrograde trafficking complex in vitro. Extensive biochemical and molecular experiments showed that the COPI retrograde complex regulates ligand-mediated AR transcriptional activation, which correlated with the mobilization of the Golgi-localized ARA160 coactivator to the nuclear compartment of prostate tumor cells. Collectively, this study provides a bioanalytical strategy to validate the AR-interactome and define novel AR-interacting proteins involved in ligand-mediated AR activation in prostate tumor cells. Moreover, we describe a cellular system to study how compartment-specific AR-interacting proteins influence AR activation and contribute to aberrant AR-dependent transcription that underlies the majority of human prostate cancers.


Proceedings of the National Academy of Sciences of the United States of America | 1997

MRIT, a novel death-effector domain-containing protein, interacts with caspases and BclXL and initiates cell death

David K. Han; Preet M. Chaudhary; Michael E. Wright; Cynthia Friedman; Barbara J. Trask; Rodney T. Riedel; Dale Baskin; Stephen M. Schwartz; Leroy Hood

Collaboration


Dive into the Michael E. Wright's collaboration.

Top Co-Authors

Avatar

David K. Han

University of Connecticut

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jimmy K. Eng

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Brandon H. Ng

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Deborah H. Lundgren

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emily S. Birkholz

University of Iowa Hospitals and Clinics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge