Network


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

Hotspot


Dive into the research topics where Susan B. Hopkinson is active.

Publication


Featured researches published by Susan B. Hopkinson.


BioEssays | 1998

STRUCTURE AND ASSEMBLY OF HEMIDESMOSOMES

Jonathan C. R. Jones; Susan B. Hopkinson; Lawrence E. Goldfinger

The hemidesmosome is a complex junction containing many proteins. The keratin cytoskeleton attaches to its cytoplasmic plaque, while its transmembrane elements interact with components of the extracellular matrix. Hemidesmosome assembly involves recruitment of α6β4 integrin heterodimers, as well as cytoskeletal elements and cytoskeleton‐associated proteins to the cell surface. In our cell culture models, these phenomena appear to be triggered by laminin‐5 in the extracellular matrix. Cell interaction with laminin‐5 apparently induces both phosphorylation and dephosphorylation of subunits of α6β4 integrin. There is emerging evidence that such events are necessary for subsequent cytoskeleton anchorage to the hemidesmosome cytoplasmic plaque. Once assembled, the hemidesmosome plays an essential role in maintaining firm epithelial adhesion to the basement membrane, with hemidesmosome disruption being a hallmark of certain devastating blistering diseases. However, the hemidesmosome is more than just a stable anchor, as it may also be the site of signal transduction, mediated by its α6β4 integrin component. This review discusses our current knowledge of the structure and assembly of the hemidesmosome. BioEssays 20:488–494, 1998.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Complex interactions between the laminin α4 subunit and integrins regulate endothelial cell behavior in vitro and angiogenesis in vivo

Annette M. Gonzalez; Meredith Gonzales; G. Scott Herron; Usha Nagavarapu; Susan B. Hopkinson; Daisuke Tsuruta; Jonathan C. R. Jones

The α4 laminin subunit is a component of the basement membrane of blood vessels where it codistributes with the integrins αvβ3, α3β1, and α6β1. An antibody against the G domain (residues 919-1207; G919–1207) of the α4 laminin subunit inhibits angiogenesis in a mouse–human chimeric model, indicating the functional importance of this domain. Additional support for the latter derives from the ability of recombinant G919–1207 to support endothelial cell adhesion. In particular, endothelial cell adhesion to G919–1207 is half-maximal at 1.4 nM, whereas residues 919-1018 and 1016–1207 of the G domain are poor cellular ligands. Function blocking antibodies against integrins αvβ3 and β1 and a combination of antibodies against α3 and α6 integrin subunits inhibit endothelial cell attachment to G919–1207. Moreover, both αvβ3 and α3β1 integrin bind with high affinity to G919–1207. Together, our studies demonstrate that the G domain of laminin α4 chain is a specific, high affinity ligand for the αvβ3 and α3β1 integrin heterodimers and that these integrins, together with α6β1, function cooperatively to mediate endothelial cell–α4 laminin interaction and hence blood vessel development. We propose a model based on these data that reconcile apparent discrepancies in the recent literature with regard to the role of the αvβ3 integrin in angiogenesis.


Journal of Biological Chemistry | 2006

Integrin β4 Regulates Migratory Behavior of Keratinocytes by Determining Laminin-332 Organization

Bernd U. Sehgal; Phillip J. DeBiase; Sumio Matzno; Teng-Leong Chew; Jessica N. Claiborne; Susan B. Hopkinson; Alan Russell; M. Peter Marinkovich; Jonathan C. R. Jones

Whether α6β4 integrin regulates migration remains controversial. β4 integrin-deficient (JEB) keratinocytes display aberrant migration in that they move in circles, a behavior that mirrors the circular arrays of laminin (LM)-332 in their matrix. In contrast, wild-type keratinocytes and JEB keratinocytes, induced to express β4 integrin, assemble laminin-332 in linear tracks over which they migrate. Moreover, laminin-332-dependent migration of JEB keratinocytes along linear tracks is restored when cells are plated on wild-type keratinocyte matrix, whereas wild-type keratinocytes show rotation over circular arrays of laminn-332 in JEB keratinocyte matrix. The activities of Rac1 and the actin cytoskeleton-severing protein cofilin are low in JEB keratinocytes compared with wild-type cells but are rescued following expression of wild-type β4 integrin in JEB cells. Additionally, in wild-type keratinocytes Rac1 is complexed with α6β4 integrin. Moreover, Rac1 or cofilin inactivation induces wild-type keratinocytes to move in circles over rings of laminin-332 in their matrix. Together these data indicate that laminin-332 matrix organization is determined by the α6β4 integrin/actin cytoskeleton via Rac1/cofilin signaling. Furthermore, our results imply that the organizational state of laminin-332 is a key determinant of the motility behavior of keratinocytes, an essential element of skin wound healing and the successful invasion of epidermal-derived tumor cells.


Journal of Biological Chemistry | 2007

The Slingshot Family of Phosphatases Mediates Rac1 Regulation of Cofilin Phosphorylation, Laminin-332 Organization, and Motility Behavior of Keratinocytes

Kristina Kligys; Jessica N. Claiborne; Phillip J. DeBiase; Susan B. Hopkinson; Yvonne Y. Wu; Kensaku Mizuno; Jonathan C. R. Jones

The motility of keratinocytes is an essential component of wound closure and the development of epidermal tumors. In vitro, the specific motile behavior of keratinocytes is dictated by the assembly of laminin-332 tracks, a process that is dependent upon α6β4 integrin signaling to Rac1 and the actin-severing protein cofilin. Here we have analyzed how cofilin phosphorylation is regulated by phosphatases (slingshot (SSH) or chronophin (CIN)) downstream of signaling by α6β4 integrin/Rac1 in human keratinocytes. Keratinocytes express all members of the SSH family (SSH1, SSH2, and SSH3) and CIN. However, expression of phosphatase-dead versions of all three SSH proteins, but not dominant inactive CIN, results in phosphorylation/inactivation of cofilin, changes in actin cytoskeleton organization, loss of cell polarity, and assembly of aberrant arrays of laminin-332 in human keratinocytes. SSH activity is regulated by 14-3-3 protein binding, and intriguingly, 14-3-3/α6β4 integrin protein interaction is required for keratinocyte migration. We wondered whether 14-3-3 proteins function as regulators of Rac1-mediated keratinocyte migration patterns. In support of this hypothesis, inhibition of Rac1 results in an increase in 14-3-3 protein association with SSH. Thus, we propose a novel mechanism in which α6β4 integrin signaling via Rac1, 14-3-3 proteins, and SSH family members regulates cofilin activation, cell polarity, and matrix assembly, leading to specific epidermal cell migration behavior.


Journal of Cell Science | 2009

Laminin deposition in the extracellular matrix: a complex picture emerges.

Kevin J. Hamill; Kristina Kligys; Susan B. Hopkinson; Jonathan C. R. Jones

Laminins are structural components of basement membranes. In addition, they are key extracellular-matrix regulators of cell adhesion, migration, differentiation and proliferation. This Commentary focuses on a relatively understudied aspect of laminin biology: how is laminin deposited into the extracellular matrix? This topic has fascinated researchers for some time, particularly considering the diversity of patterns of laminin that can be visualized in the matrix of cultured cells. We discuss current ideas of how laminin matrices are assembled, the role of matrix receptors in this process and how laminin-associated proteins modulate matrix deposition. We speculate on the role of signaling pathways that are involved in laminin-matrix deposition and on how laminin patterns might play an important role in specifying cell behaviors, especially directed migration. We conclude with a description of new developments in the way that laminin deposition is being studied, including the use of tagged laminin subunits that should allow the visualization of laminin-matrix deposition and assembly by living cells.


The FASEB Journal | 2002

Microfilament-dependent movement of the beta3 integrin subunit within focal contacts of endothelial cells.

Daisuke Tsuruta; Meredith Gonzales; Susan B. Hopkinson; Carol Otey; Satya Khuon; Robert D. Goldman; Jonathan C. R. Jones

To gain insight into the dynamic properties of focal contacts, we induced expression of green fluorescent protein‐tagged β3 integrin (GFP‐β3) and actinin‐1 (GFP‐actinin‐1) in endothelial cells. Both tagged proteins localize with αvβ3 integrin in focal contacts distributed towards the periphery of transfected cells. Labeled focal contacts migrate at about 0.1 μm/min in stationary live endothelial cells. We compared β3 integrin and actinin‐1 dynamics in focal contacts by using fluorescence recovery after photobleaching. Recovery of signal in bleached focal contacts that have incorporated actinin‐1 is rapid and occurs within less than 4 min. This recovery is energy‐dependent. In contrast, recovery of bleached focal contacts that contain GFP‐β3 integrin takes longer than 30 min. Yet, when a narrow stripe of fluorescence is bleached across a β3 integrinlabeled focal contact, recovery is complete within 16 min. The latter recovery is energy‐dependent and is blocked not only by actin‐filament disrupting drugs but also by a myosin light chain kinase inhibitor. Thus, integrins are not immobile when incorporated into focal contacts, as some have suggested. We propose that integrins are mobile within the confines of focal contacts and that this mobility is supported by an actin‐associated molecular motor.


Journal of Cell Science | 2005

Laminin-6 assembles into multimolecular fibrillar complexes with perlecan and participates in mechanical-signal transduction via a dystroglycan-dependent, integrin-independent mechanism

Jonathan C. R. Jones; Kimberly Lane; Susan B. Hopkinson; Emilia Lecuona; Robert Christopher Geiger; David A. Dean; Eduardo Correa-Meyer; Meredith Gonzales; Kevin P. Campbell; Jacob I. Sznajder; Scott Budinger

Mechanical ventilation is a valuable treatment regimen for respiratory failure. However, mechanical ventilation (especially with high tidal volumes) is implicated in the initiation and/or exacerbation of lung injury. Hence, it is important to understand how the cells that line the inner surface of the lung [alveolar epithelial cells (AECs)] sense cyclic stretching. Here, we tested the hypothesis that matrix molecules, via their interaction with surface receptors, transduce mechanical signals in AECs. We first determined that rat AECs secrete an extracellular matrix (ECM) rich in anastamosing fibers composed of the α3 laminin subunit, complexed with β1 and γ1 laminin subunits (i.e. laminin-6), and perlecan by a combination of immunofluorescence microscopy and immunoblotting analyses. The fibrous network exhibits isotropic expansion when exposed to cyclic stretching (30 cycles per minute, 10% strain). Moreover, this same stretching regimen activates mitogen-activated-protein kinase (MAPK) in AECs. Stretch-induced MAPK activation is not inhibited in AECs treated with antagonists to α3 or β1 integrin. However, MAPK activation is significantly reduced in cells treated with function-inhibiting antibodies against the α3 laminin subunit and dystroglycan, and when dystroglycan is knocked down in AECs using short hairpin RNA. In summary, our results support a novel mechanism by which laminin-6, via interaction with dystroglycan, transduces a mechanical signal initiated by stretching that subsequently activates the MAPK pathway in rat AECs. These results are the first to indicate a function for laminin-6. They also provide novel insight into the role of the pericellular environment in dictating the response of epithelial cells to mechanical stimulation and have broad implications for the pathophysiology of lung injury.


Journal of Biological Chemistry | 2000

Spatial regulation and activity modulation of plasmin by high affinity binding to the G domain of the alpha 3 subunit of laminin-5

Lawrence E. Goldfinger; Luohua Jiang; Susan B. Hopkinson; M. Sharon Stack; Jonathan C. R. Jones

Cells in complex tissues contact extracellular matrix that interacts with integrin receptors to influence gene expression, proliferation, apoptosis, adhesion, and motility. During development, tissue remodeling, and tumorigenesis, matrix components are modified by enzymatic digestion with subsequent effects on integrin binding and signaling. We are interested in understanding the mechanisms by which broad spectrum proteinases such as plasmin are targeted to their extracellular matrix protein substrates. We have utilized plasmin-mediated cleavage of the epithelial basement membrane glycoprotein laminin-5 as a model to evaluate molecular events that direct plasmin activity to specific structural domains. We report that plasminogen and tissue plasminogen activator (tPA) exhibit high affinity, specific binding to the G1 subdomain of the N terminus of the laminin-5 α3 subunit, with equilibrium dissociation constants of 50 nm for plasminogen and 80 nm for tPA. No high affinity binding to the G2, G3, and G4 subdomains was observed. As a result of binding to the G1 subdomain, the catalytic efficiency of tPA-catalyzed plasminogen activation is enhanced 32-fold, leading to increased matrix-associated plasmin that is positioned favorably for cleavage within the G4 subdomain as we have reported previously (Goldfinger, L. E., Stack, M. S., and Jones, J. C. R. (1998) J. Cell Biol. 141, 255–265). Thus, physical constraints dictated by interaction of proteinase and matrix macromolecule control not only enzymatic activity but may regulate substrate targeting of proteinases.


Molecular Biology of the Cell | 2009

BPAG1e Maintains Keratinocyte Polarity through β4 Integrin–mediated Modulation of Rac 1 and Cofilin Activities

Kevin J. Hamill; Susan B. Hopkinson; Philip Debiase; Jonathan C. R. Jones

alpha6beta4 integrin, a component of hemidesmosomes, also plays a role in keratinocyte migration via signaling through Rac1 to the actin-severing protein cofilin. Here, we tested the hypothesis that the beta4 integrin-associated plakin protein, bullous pemphigoid antigen 1e (BPAG1e) functions as a scaffold for Rac1/cofilin signal transduction. We generated keratinocyte lines exhibiting a stable knockdown in BPAG1e expression. Knockdown of BPAG1e does not affect expression levels of other hemidesmosomal proteins, nor the amount of beta4 integrin expressed at the cell surface. However, the amount of Rac1 associating with beta4 integrin and the activity of both Rac1 and cofilin are significantly lower in BPAG1e-deficient cells compared with wild-type keratinocytes. In addition, keratinocytes deficient in BPAG1e exhibit loss of front-to-rear polarity and display aberrant motility. These defects are rescued by inducing expression of constitutively active Rac1 or active cofilin. These data indicate that the BPAG1e is required for efficient regulation of keratinocyte polarity and migration by determining the activation of Rac1.


Journal of Biological Chemistry | 2012

α6β4 Integrin, a Master Regulator of Expression of Integrins in Human Keratinocytes

Kristina Kligys; Yvonne Wu; Susan B. Hopkinson; Surinder Kaur; Leonidas C. Platanias; Jonathan C. R. Jones

Background: Keratinocyte migration involves the coordinated expression of various integrin heterodimers. Results: Loss of α6β4 integrin expression impairs cell migration and decreases α2 and α3 integrin subunit expression via transcriptional and translational mechanisms. Conclusion: Migration of human keratinocytes requires α6β4 integrin-dependent regulation of integrin subunit expression. Significance: α6β4 integrin controls integrin expression profiles and thereby regulates migration. Three major laminin and collagen-binding integrins in skin (α6β4, α3β1, and α2β1) are involved in keratinocyte adhesion to the dermis and dissemination of skin cells during wound healing and/or tumorigenesis. Knockdown of α6 integrin in keratinocytes not only results in motility defects but also leads to decreased surface expression of the α2, α3, and β4 integrin subunits. Whereas α2 integrin mRNA levels are decreased in α6 integrin knockdown cells, α3 and β4 integrin mRNAs levels are unaffected. Expression of either α6 or α3 integrin in α6 integrin knockdown cells restores α2 integrin mRNA levels. Moreover, re-expression of α6 integrin increases β4 integrin protein at the cell surface, which results in an increase in α3 integrin expression via activation of initiation factor 4E-binding protein 1. Our data indicate that the α6β4 integrin is a master regulator of transcription and translation of other integrin subunits and underscore its pivotal role in wound healing and cancer.

Collaboration


Dive into the Susan B. Hopkinson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sho Hiroyasu

Washington State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge