Network


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

Hotspot


Dive into the research topics where Deborah A. Raynes is active.

Publication


Featured researches published by Deborah A. Raynes.


FEBS Letters | 2002

HspBP1, a homologue of the yeast Fes1 and Sls1 proteins, is an Hsc70 nucleotide exchange factor

Mehdi Kabani; Catherine McLellan; Deborah A. Raynes; Vince Guerriero; Jeffrey L. Brodsky

The yeast FES1 and SLS1 genes encode conserved nucleotide exchange factors that act on the cytoplasmic and endoplasmic reticulum luminal Hsp70s, Ssa1p and BiP, respectively. We report here that mammalian HspBP1 is homologous to Fes1p and that HspBP1 promotes nucleotide dissociation from both Ssa1p and mammalian Hsc70. In contrast, Fes1p inefficiently strips nucleotide from mammalian Hsc70, and unlike HspBP1 does not inhibit chaperone‐mediated protein refolding in vitro. Together, our data indicate that HspBP1 is a member of this new class of nucleotide exchange factors that exhibit varying degrees of compartment and species specificity.


Cancer Science | 2009

Heat shock protein 70‐binding protein 1 is highly expressed in high‐grade gliomas, interacts with multiple heat shock protein 70 family members, and specifically binds brain tumor cell surfaces

Michael W. Graner; Deborah A. Raynes; Darell D. Bigner; Vince Guerriero

Chaperone proteins and heat shock proteins (HSP) are essential components of cellular protein folding systems under normal conditions; their expression and activities are upregulated during stress. Chronically stressed tumors frequently exhibit high chaperone protein levels, exploiting their anti‐apoptotic mechanisms and general proteome homeostasis amidst a background of genetic instability. Co‐chaperones interact with chaperones as malleable regulatory components of protein folding activity and may represent a conduit for modification of chaperone activity to the detriment of the tumor. We have initially characterized one such co‐chaperone, heat shock protein 70‐binding protein (HspBP) 1 from human brain tumors, their xenografts grown in immune‐compromised mice, and in syngeneic murine models in immune‐competent mice. Immunohistochemical analyses show HspBP1 overexpression (with unusual subcellular localizations) in patient brain tumors relative to normal brain tissue. This holds true for the xenograft and syngeneic murine tumor models. In biochemical affinity chromatography assays, HspBP1 interacts with members of the HSP70 family from brain tumor lysates and from surface‐derived samples, including HSP70, glucose regulated protein (GRP)75, GRP78, and HSP110. From normal brain lysates, only heat shock cognate (HSC)70, GRP75, and HSP110 bind to HspBP1. FACS analyses indicate that HspBP1 binds to brain tumor cell surfaces, possibly via HSP70 family members, and internalizes into cells. This has implications for HspBP1 biology as well as its utility as a tumor‐targeting agent. Our results suggest that HspBP1 may play a role in tumor (dys)regulation of chaperone proteins, and that HspBP1 may have extracellular roles with therapeutic implications. (Cancer Sci 2009; 100: 1870–1879)


Cell Stress & Chaperones | 2003

Expression of a unique drug-resistant Hsp90 ortholog by the nematode Caenorhabditis elegans

Cynthia L. David; Harold E. Smith; Deborah A. Raynes; Elizabeth J. Pulcini; Luke Whitesell

Abstract In all species studied to date, the function of heat shock protein 90 (Hsp90), a ubiquitous and evolutionarily conserved molecular chaperone, is inhibited selectively by the natural product drugs geldanamycin (GA) and radicicol. Crystal structures of the N-terminal region of yeast and human Hsp90 have revealed that these compounds interact with the chaperone in a Bergerat-type adenine nucleotide–binding fold shared throughout the gyrase, Hsp90, histidine kinase mutL (GHKL) superfamily of adenosine triphosphatases. To better understand the consequences of disrupting Hsp90 function in a genetically tractable multicellular organism, we exposed the soil-dwelling nematode Caenorhabditis elegans to GA under a variety of conditions designed to optimize drug uptake. Mutations in the gene encoding C elegans Hsp90 affect larval viability, dauer development, fertility, and life span. However, exposure of worms to GA produced no discernable phenotypes, although the amino acid sequence of worm Hsp90 is 85% homologous to that of human Hsp90. Consistent with this observation, we found that solid phase–immobilized GA failed to bind worm Hsp90 from worm protein extracts or when translated in a rabbit reticulocyte lysate system. Further, affinity precipitation studies using chimeric worm-vertebrate fusion proteins or worm C-terminal truncations expressed in reticulocyte lysate revealed that the conserved nucleotide-binding fold of worm Hsp90 exhibits the novel ability to bind adenosine triphosphate but not GA. Despite its unusual GA resistance, worm Hsp90 appeared fully functional when expressed in a vertebrate background. It heterodimerized with its vertebrate counterpart and showed no evidence of compromising its essential cellular functions. Heterologous expression of worm Hsp90 in tumor cells, however, did not render them GA resistant. These findings provide new insights into the nature of unusual N-terminal nucleotide-binding fold of Hsp90 and suggest that target-related drug resistance is unlikely to emerge in patients receiving GA-like chemotherapeutic agents.


Tumor Biology | 2003

Increased expression of the Hsp70 cochaperone HspBP1 in tumors

Deborah A. Raynes; Michael W. Graner; Rochelle Bagatell; Catherine McLellan; Vince Guerriero

Hsp70 levels are elevated in a number of different tumors. The Hsp70 cochaperone heat shock protein-binding protein 1 (HspBP1) has been shown to bind to Hsp70, inhibit its activity and promote dissociation of nucleotide from the Hsp70 ATPase domain. The purpose of this study was to determine if the levels of HspBP1 are altered in tumor cells. In this report, we show that HspBP1 levels are elevated in two mouse tumor models, 3LL cells (Lewis Lung carcinoma) and neuroblastoma tumors. The amounts of HspBP1 and Hsp70 in selected tissues, tumors and a rabbit reticulocyte lysate were determined using Western blots. It was found that the molar ratio of these two proteins was within a small range (0.21–0.42) in the normal and tumor tissues examined. This ratio was considerably below the HspBP1 to Hsp70 ratio of 4.0 needed for 50% inhibition of Hsp70-mediated refolding of a partially denatured protein in rabbit reticulocyte lysate. The ratio of HspBP1 to Hsp70 in these tissues is too low to inhibit Hsp70 globally in the cell, but is high enough to provide a pool of HspBP1 that could inhibit Hsp70 in a localized fashion. These studies have shown that HspBP1 is elevated in the tumors examined and therefore could be a new cancer marker.


Journal of Biological Chemistry | 2011

The heat shock binding protein (HspBP1) protects cells against the cytotoxic action of the Tag7-Hsp70 complex

Denis V. Yashin; Elena A. Dukhanina; Olga D. Kabanova; Elena A. Romanova; Tamara I. Lukyanova; Alexsander G. Tonevitskii; Deborah A. Raynes; N. V. Gnuchev; Vince Guerriero; Georgii P. Georgiev; Lidia P. Sashchenko

Heat shock-binding protein HspBP1 is a member of the Hsp70 co-chaperone family. The interaction between HspBP1 and the ATPase domain of the major heat shock protein Hsp70 up-regulates nucleotide exchange and reduces the affinity between Hsp70 and the peptide in its peptide-binding site. Previously we have shown that Tag7 (also known as peptidoglycan recognition protein PGRP-S), an innate immunity protein, interacts with Hsp70 to form a stable Tag7-Hsp70 complex with cytotoxic activity against some tumor cell lines. This complex can be produced in cytotoxic lymphocytes and released during interaction with tumor cells. Here the effect of HspBP1 on the cytotoxic activity of the Tag7-Hsp70 complex was examined. HspBP1 could bind not only to Hsp70, but also to Tag7. This interaction eliminated the cytotoxic activity of Tag7-Hsp70 complex and decreased the ATP concentration required to dissociate Tag7 from the peptide-binding site of Hsp70. Moreover, HspBP1 inhibited the cytotoxic activity of the Tag7-Hsp70 complex secreted by lymphocytes. HspBP1 was detected in cytotoxic CD8+ lymphocytes. This protein was released simultaneously with Tag7-Hsp70 during interaction of these lymphocytes with tumor cells. The simultaneous secretion of the cytotoxic complex with its inhibitor could be a mechanism protecting normal cells from the cytotoxic effect of this complex.


Scandinavian Journal of Rheumatology | 2011

Heat shock protein gene expression profile may differentiate between rheumatoid arthritis, osteoarthritis, and healthy controls

Lucie Sedlackova; Antonin Sosna; Pavla Vavrincova; J Frýdl; Vince Guerriero; Deborah A. Raynes; Ilona Hromadnikova

Objective: Heat shock proteins (Hsps) have been repeatedly implicated in the pathogenesis of rheumatoid arthritis (RA). The aim of this work was to study Hsp mRNA and protein levels to determine whether they can be used to differentiate between RA, osteoarthritis (OA), and healthy controls. Methods: Hsp27, Hsp60, Hsp70, Hsp90α, and HspBP1 mRNA expression was analysed using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) in 24 RA, 11 OA, and 21 healthy controls. Hsp70 and HspBP1 protein levels were measured in serum using an enzyme-linked immunosorbent assay (ELISA). Results: Hsp gene expression profiles differ significantly between inflammatory (RA) and non-inflammatory (OA) joint diseases, showing significantly increased Hsp27 and Hsp90α mRNA levels in RA synovial tissues. Up-regulated Hsp60 and Hsp90α together with down-regulated Hsp70 and elevated HspBP1/Hsp70 mRNA ratios can be used to differentiate between RA patients and healthy individuals through analysis of peripheral blood samples. Despite increased HspBP1 levels in RA sera, Hsp70 levels and the HspBP1/Hsp70 protein ratio remained identical in the RA patients and healthy individuals, which may contribute to the inhibition of Hsp70 anti-apoptotic activity. Conclusion: Hsp gene expression analysis can be implemented as a new diagnostic approach to facilitate differentiation between RA, OA, and healthy controls.


Biology of the Cell | 2009

Extracellular HspBP1 and Hsp72 synergistically activate epidermal growth factor receptor

A. L. Evdonin; Alexander Kinev; Natalia Tsupkina; Vince Guerriero; Deborah A. Raynes; N. D. Medvedeva

Background information. Heat‐inducible Hsp72 is the founding member of the Hsp70 (heat shock proteins of 70 kDa) family of molecular chaperones. It is localized primarily in cytoplasm and nucleus but is also found extracellularly. The source of e‐Hsp72 (extracellular Hsp72) is not precisely identified and may not be the same in every situation. A number of studies demonstrated that e‐Hsp72 plays an important role in cell survival, tumour rejection and immune response. However, currently little is known about regulation of e‐Hsp72 function. In cells, Hsp72 is controlled by co‐chaperones. An abundant co‐chaperone, HspBP1 (Hsp72‐binding protein 1) was found extracellularly in the serum. In the present study we analysed the secretion and function of e‐HspBP1 (extracellular HspBP1).


Cell Biology International | 2006

Expression of the cochaperone HspBP1 is not coordinately regulated with Hsp70 expression

Eric Gottwald; Mechthild Herschbach; Brigitte Lahni; Roger L. Miesfeld; Susan Kunz; Deborah A. Raynes; Vince Guerriero

Intracellular levels of the heat stress protein Hsp70 are elevated following exposure to elevated temperature. The cochaperone HspBP1 is an intracellular protein that is known to bind to and regulate Hsp70 activity. The purpose of this study was to determine if HspBP1 levels changed when Hsp70 levels were altered. Heat stress resulted in an increase in Hsp70 levels but no change in HspBP1 levels. Treatment of cells with the apoptosis inducing drug camptothecin lowered Hsp70 levels but again had no effect on HspBP1 levels. Cells treated with camptothecin plus heat stress did not exhibit an increase in Hsp70 levels. Over‐expression in cells stably transfected with HspBP1 cDNA resulted in a 290% increase in HspBP1 levels without a similar change in Hsp70 levels. These results demonstrate that Hsp70 and HspBP1 are not coordinately regulated but provide evidence that an increase in the ratio of HspBP1 to Hsp70 correlates with apoptosis, in a similar way to reducing the amount of Hsp70.


Journal of Immunoassay & Immunochemistry | 2006

Human Serum Contains Detectable Levels of the Hsp70 Cochaperone HspBP1 and Antibodies Bound to HspBP1

Deborah A. Raynes; Cynthia A. Thomson; John A. Stroster; Tara R. Newton; Peder Cuneo; Vince Guerriero

Abstract The identification of the proteins that comprise the serum proteome is a current major research goal that will provide useful information for the diagnosis and treatment of various diseases. It is well established that Hsp70 and Hsp70 antibodies are present in human serum. This study reports on the development of an ELISA assay for the Hsp70 co‐chaperone, HspBP1. HspBP1 is present in human serum at concentrations ranging between 0.74 to 3.98 ng/mL. No gender or age differences in the HspBP1 levels were identified. It was also found that human serum contained antibodies to HspBP1, and there were no gender or age differences in these levels. In addition, there was no correlation between the level of HspBP1 in a sample and the antibody titer. Finally, we found that HspBP1 in serum is complexed to anti‐HspBP1 antibodies. This report provides initial baseline data on HspBP1 in human serum and provides the methods for future studies to determine if these levels are altered in response to disease.


Biochimica et Biophysica Acta | 2000

Isolation and characterization of isoforms of HspBP1, inhibitors of Hsp70.

Deborah A. Raynes; Vince Guerriero

The protein sequences derived from cDNA sequences for Hsp70 binding proteins from human (HspBP2) and rat tissues (HspBPR) are presented in this paper. The derived amino acid sequences of these proteins differ from human HspBP1 in the number of consecutive glycines near the amino-terminus. These differences, however, do not alter the inhibitory activity.

Collaboration


Dive into the Deborah A. Raynes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael W. Graner

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge