Network


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

Hotspot


Dive into the research topics where Carol A. Otey is active.

Publication


Featured researches published by Carol A. Otey.


PLOS Medicine | 2006

Palladin Mutation Causes Familial Pancreatic Cancer and Suggests a New Cancer Mechanism

Kay Pogue-Geile; Ru Chen; Mary P. Bronner; Tatjana Crnogorac-Jurcevic; Kara White Moyes; Sally E. Dowen; Carol A. Otey; David A. Crispin; Ryan George; David C. Whitcomb; Teresa A. Brentnall

Background Pancreatic cancer is a deadly disease. Discovery of the mutated genes that cause the inherited form(s) of the disease may shed light on the mechanism(s) of oncogenesis. Previously we isolated a susceptibility locus for familial pancreatic cancer to chromosome location 4q32–34. In this study, our goal was to discover the identity of the familial pancreatic cancer gene on 4q32 and determine the function of that gene. Methods and Findings A customized microarray of the candidate chromosomal region affecting pancreatic cancer susceptibility revealed the greatest expression change in palladin (PALLD), a gene that encodes a component of the cytoskeleton that controls cell shape and motility. A mutation causing a proline (hydrophobic) to serine (hydrophilic) amino acid change (P239S) in a highly conserved region tracked with all affected family members and was absent in the non-affected members. The mutational change is not a known single nucleotide polymorphism. Palladin RNA, measured by quantitative RT-PCR, was overexpressed in the tissues from precancerous dysplasia and pancreatic adenocarcinoma in both familial and sporadic disease. Transfection of wild-type and P239S mutant palladin gene constructs into HeLa cells revealed a clear phenotypic effect: cells expressing P239S palladin exhibited cytoskeletal changes, abnormal actin bundle assembly, and an increased ability to migrate. Conclusions These observations suggest that the presence of an abnormal palladin gene in familial pancreatic cancer and the overexpression of palladin protein in sporadic pancreatic cancer cause cytoskeletal changes in pancreatic cancer and may be responsible for or contribute to the tumors strong invasive and migratory abilities.


Nature Cell Biology | 2002

Dissecting the link between stress fibres and focal adhesions by CALI with EGFP fusion proteins

Zenon Rajfur; Partha Roy; Carol A. Otey; Lewis H. Romer; Ken Jacobson

Chromophore-assisted laser inactivation (CALI) is a light-mediated technique used to selectively inactivate proteins within cells. Here, we demonstrate that GFP can be used as a CALI reagent to locally inactivate proteins in living cells. We show that focused laser irradiation of EGFP–α-actinin expressed in Swiss 3T3 fibroblasts results in the detachment of stress fibres from focal adhesions (FAs), whereas the integrity of FAs, as determined by interference reflection microscopy (IRM), is preserved. Moreover, consistent with a function for focal adhesion kinase (FAK) in FA signalling and not FA structure, laser irradiation of EGFP–FAK did not cause either visible FA damage or stress fibre detachment, although in vitro CALI of isolated EGFP–FAK decreased its kinase activity, but not its binding to paxillin. These data indicate that CALI of specific FA components may be used to precisely dissect the functional significance of individual proteins required for the maintenance of this cytoskeletal structure. In vitro CALI experiments also demonstrated a reduction of EGFP–α-actinin binding to the cytoplasmic domain of the β1 integrin subunit, but not to actin. Thus, α-actinin is essential for the binding of microfilaments to integrins in the FA. CALI-induced changes in α-actinin result in the breakage of that link and the subsequent retraction of the stress fibre.


Cytoskeleton | 2001

Dynamics of α-actinin in focal adhesions and stress fibers visualized with α-actinin-green fluorescent protein

Magnus Edlund; Marc A. Lotano; Carol A. Otey

Motile cells undergo changes in cell adhesion, behavior, and shape that are mediated by small-scale cytoskeletal rearrangements. These rearrangements have proven difficult to follow quantitatively in living cells, without disrupting the very structures and delicate protein balances under study. We have expressed a prominent cytoskeletal protein, α-actinin, as a fusion with green fluorescent protein (αAGFP), and have followed this constructs movements within transfected mouse Swiss 3T3 and BALB/c fibroblasts. αAGFP was expressed at low levels to avoid overexpression artifacts. αAGFP localized to cellular structures, including stress fibers, focal adhesions, microspikes, and lamellipodia. High-resolution video-microscopy revealed that the αAGFP construct could be seen relocating to focal adhesions early in their formation and shortly thereafter to stress-fiber dense bodies. By Fluorescent Recovery After Photo-bleaching (FRAP) techniques, αAGFP was found to have similar exchange rates and protein stability in focal adhesions and stress fibers (despite the known differences in protein composition in these two structures). This raises the possibility that the two structures share common key regulatory factors and may not be as affected by protein-protein binding interactions as previously suggested. Additionally, the exchange rates revealed by video-microscopy and FRAP analysis of αAGFP are more rapid than those reported previously, which were obtained using microinjection of large excesses of fluorescently-tagged protein. Cell Motil. Cytoskeleton 48:190–200, 2001.


Mechanisms of Development | 1990

Actin-membrane interaction in focal adhesions

Keith Burridge; Glen Nuckolls; Carol A. Otey; Fredrick M. Pavalko; Keiko O. Simon; Christopher E. Turner

Focal adhesions are regions of the plasma membrane where cells in tissue culture adhere strongly to the underlying extracellular matrix, and which at their cytoplasmic face serve to anchor bundles of actin microfilaments. They provide an experimental model for studying the links between the cytoskeleton and the extracellular matrix. Members of the integrin family of extracellular matrix receptors are prominent components, spanning the membrane in focal adhesions, but there is evidence that other membrane components are also needed for these structures to form. A number of proteins are concentrated at the cytoplasmic face of focal adhesions. Recent efforts have sought to determine the links between actin and the integrin cytoplasmic domains. Using in vitro binding assays, two potential bridges between actin and integrin have been identified. One involves talin, which has recently been shown to bind actin directly. The other involves the actin-binding protein, alpha-actinin, which has been found to interact with several integrins. The physiological significance of these two potential bridges between actin and integrin remains to be determined in vivo.


Journal of Cell Science | 2006

Identification of palladin isoforms and characterization of an isoform-specific interaction between Lasp-1 and palladin

Andrew Rachlin; Carol A. Otey

Palladin is a recently described phosphoprotein with an important role in cytoskeletal organization. The major palladin isoform (90-92 kDa) binds to three actin-associated proteins (ezrin, VASP and α-actinin), suggesting that palladin functions as a cytoskeletal scaffold. Here, we describe the organization of the palladin gene, which encodes multiple isoforms, including one (140 kDa) with a similar localization pattern to 90 kDa palladin. Overexpression of the 90 kDa or 140 kDa isoforms in COS-7 cells results in rearrangements of the actin cytoskeleton into super-robust bundles and star-like arrays, respectively. Sequence analysis of 140 kDa palladin revealed a conserved binding site for SH3 domains, suggesting that it binds directly to the SH3-domain protein Lasp-1. Binding of 140 kDa palladin, but not 90 kDa palladin, to Lasp-1 was confirmed by yeast two-hybrid and GST-pull-down assays. Isoform-specific siRNA experiments suggested that 140 kDa palladin plays a role in recruiting Lasp-1 to stress fibers. These results add Lasp-1, an actin-binding protein with a crucial role in cell motility, to the growing list of palladins binding partners, and suggest that 140 kDa palladin has a specialized function in organizing the actin arrays that participate in cell migration and/or cellular contractility.


Journal of Biological Chemistry | 2008

Palladin is an actin cross-linking protein that uses immunoglobulin-like domains to bind filamentous actin

Richard D. S. Dixon; Daniel K. Arneman; Andrew Rachlin; Naresh R. Sundaresan; M. Joseph Costello; Sharon L. Campbell; Carol A. Otey

Palladin is a recently described phosphoprotein that plays an important role in cell adhesion and motility. Previous studies have shown that palladin overexpression results in profound changes in actin organization in cultured cells. Palladin binds to the actin-associated proteins α-actinin, vasodilator-stimulated phosphoprotein, profilin, Eps8, and ezrin, suggesting that it may affect actin organization indirectly. To determine its molecular function in generating actin arrays, we purified palladin and asked if it is also capable of binding to F-actin directly. In co-sedimentation and differential sedimentation assays, palladin was found to both bind and cross-link actin filaments. This bundling activity was confirmed by fluorescence and electron microscopy. Palladin fragments were then purified and used to determine the sequences necessary to bind and bundle F-actin. The Ig3 domain of palladin bound to F-actin, and a palladin fragment containing Ig3, Ig4, and the region linking these domains was identified as a fragment that was able to bundle F-actin. Because palladin has multiple Ig domains, and only one of them binds to F-actin, this suggests that different Ig domains may be specialized for distinct biological functions. In addition, our results suggest a potential role for palladin in generating specialized, actin-based cell morphologies via both direct actin cross-linking activity and indirect scaffolding activity.


International Review of Cytology-a Survey of Cell Biology | 2005

The palladin/myotilin/myopalladin family of actin-associated scaffolds.

Carol A. Otey; Andrew Rachlin; Monica Moza; Daniel K. Arneman; Olli Carpén

The dynamic remodeling of the actin cytoskeleton plays a critical role in cellular morphogenesis and cell motility. Actin-associated scaffolds are key to this process, as they recruit cohorts of actin-binding proteins and associated signaling complexes to subcellular sites where remodeling is required. This review is focused on a recently discovered family of three proteins, myotilin, palladin, and myopalladin, all of which function as scaffolds that regulate actin organization. While myotilin and myopalladin are most abundant in skeletal and cardiac muscle, palladin is ubiquitously expressed in the organs of developing vertebrates. Palladins function has been investigated primarily in the central nervous system and in tissue culture, where it appears to play a key role in cellular morphogenesis. The three family members each interact with specific molecular partners: all three bind to alpha-actinin; in addition, palladin also binds to vasodilator-stimulated phosphoprotein (VASP) and ezrin, myotilin binds to filamin and actin, and myopalladin also binds to nebulin and cardiac ankyrin repeat protein (CARP). Since mutations in myotilin result in two forms of muscle disease, an essential role for this family member in organizing the skeletal muscle sarcomere is implied.


FEBS Letters | 2004

Molecular analysis of the interaction between palladin and α-actinin

Mikko Rönty; Anu Taivainen; Monica Moza; Carol A. Otey; Olli Carpén

Palladin is a novel component of stress fiber dense regions. Antisense and transient overexpression studies have indicated an important role for palladin in the regulation of actin cytoskeleton. Palladin colocalizes and coimmunoprecipitates with α‐actinin, a dense region component, but the molecular details and functional significance of the interaction have not been studied. We show here a direct association between the two proteins and have mapped the binding site within a short sequence of palladin and in the carboxy‐terminal calmodulin domain of α‐actinin. Using transfection‐based targeting assays, we show that palladin is involved in targeting of α‐actinin to specific subcellular foci indicating a functional interplay between the two actin‐associated proteins.


Oncogene | 2014

Palladin promotes invasion of pancreatic cancer cells by enhancing invadopodia formation in cancer-associated fibroblasts

Silvia M. Goicoechea; Rafael Garcia-Mata; J. Staub; A. Valdivia; Lisa Sharek; C. G. Mcculloch; Rosa F. Hwang; Raul Urrutia; Jen Jen Yeh; Hong Jin Kim; Carol A. Otey

The stromal compartment surrounding epithelial-derived pancreatic tumors is thought to have a key role in the aggressive phenotype of this malignancy. Emerging evidence suggests that cancer-associated fibroblasts (CAFs), the most abundant cells in the stroma of pancreatic tumors, contribute to the tumor’s invasion, metastasis and resistance to therapy, but the precise molecular mechanisms that regulate CAFs behavior are poorly understood. In this study, we utilized immortalized human pancreatic CAFs to investigate molecular pathways that control the matrix-remodeling and invasion-promoting activity of CAFs. We showed previously that palladin, an actin-associated protein, is expressed at high levels in CAFs of pancreatic tumors and other solid tumors, and also in an immortalized line of human CAFs. In this study, we found that short-term exposure of CAFs to phorbol esters reduced the number of stress fibers and triggered the appearance of individual invadopodia and invadopodial rosettes in CAFs. Molecular analysis of invadopodia revealed that their composition resembled that of similar structures (that is, invadopodia and podosomes) described in other cell types. Pharmacological inhibition and small interfering RNA knockdown experiments demonstrated that protein kinase C, the small GTPase Cdc42 and palladin were necessary for the efficient assembly of invadopodia by CAFs. In addition, GTPase activity assays showed that palladin contributes to the activation of Cdc42. In mouse xenograft experiments using a mixture of CAFs and tumor cells, palladin expression in CAFs promoted the rapid growth and metastasis of human pancreatic tumor cells. Overall, these results indicate that high levels of palladin expression in CAFs enhance their ability to remodel the extracellular matrix by regulating the activity of Cdc42, which in turn promotes the assembly of matrix-degrading invadopodia in CAFs and tumor cell invasion. Together, these results identify a novel molecular signaling pathway that may provide new molecular targets for the inhibition of pancreatic cancer metastasis.


Oncogene | 2009

Palladin contributes to invasive motility in human breast cancer cells.

Silvia M. Goicoechea; Brian Keith Bednarski; Rafael Garcia-Mata; H. Prentice-Dunn; H.J. Kim; Carol A. Otey

Cancer metastasis involves multiple steps including detachment of the metastatic cells from neighboring cells, the acquisition of motility and invasion to other tissue. All of these steps require the reorganization of the actin cytoskeleton. In this study, we found that the protein palladin, a molecular scaffold with an important function in actin organization, is expressed at higher overall levels in tumors compared with benign breast tissue, and also expressed significantly higher in four invasive breast cancer cell lines when compared with four non-invasive cell lines. In addition, we found that palladin plays a key role in the formation of podosomes. Podosomes are actin-rich structures that function in adhesion and matrix degradation, and have been found in many invasive cell types. Our results show that phorbol ester treatment stimulated the formation of palladin-containing podosomes in invasive, but not in non-invasive cell lines. More importantly, palladin knockdown resulted in decreased podosome formation and a significant reduction in transwell migration and invasive motility. Palladin overexpression induced podosome formation in the non-invasive MCF7 cells, which are otherwise unable to form podosomes, suggesting that palladin plays a critical role in the assembly of podosomes. Overall, these results indicate that palladin overexpression contributes to the invasive behavior of metastatic cells.

Collaboration


Dive into the Carol A. Otey's collaboration.

Top Co-Authors

Avatar

Silvia M. Goicoechea

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Keith Burridge

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Andrew Rachlin

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Jin Kim

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Rosa F. Hwang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Fredrick M. Pavalko

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Malika Boukhelifa

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aldo Rustioni

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge