Network


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

Hotspot


Dive into the research topics where Amy K. Roshak is active.

Publication


Featured researches published by Amy K. Roshak.


Journal of Biological Chemistry | 1996

Manipulation of Distinct NFκB Proteins Alters Interleukin-1β-induced Human Rheumatoid Synovial Fibroblast Prostaglandin E2 Formation

Amy K. Roshak; Jeffrey R. Jackson; Kevin McGough; Marie Chabot-Fletcher; Eugene Mochan; Lisa A. Marshall

Interleukin 1β (IL-1β) up-regulates human rheumatoid synovial fibroblast (RSF) 85-kDa phospholipase A2 (PLA2) and mitogen-inducible cyclooxygenase (COX) II. Promoter regions for these genes contain a motif that closely resembles the “classic” NFκB consensus site. Immunoblot analysis identified NFκB1 (p50), RelA (p65), and c-Rel in RSF. Upon IL-1β-stimulation, p65 and c-Rel but not p50 protein levels were reduced suggesting nuclear translocation. IL-1β-induced RSF nuclear extracts contained a p65-containing complex, which bound to the classical NFκB consensus motif. An NFκB classical oligonucleotide decoy produced a concentration-dependent decrease in IL-1-stimulated PGE2 production (IC50 = ∼2 μM), indicating a role of NFκB. Utilization of antisense technology showed that p65 but not p50 or c-Rel mediated IL-1β-stimulated PGE2 formation. Treated RSF could not transcribe COX II or 85-kDa PLA2 mRNA, which reduced their respective proteins. Interestingly, stimulated IL-8 production was not inhibited by the classical NFκB decoy but was reduced by treatment with antisense to both p65 and c-Rel supporting preferential binding of c-Rel-p65 to the “alternative” IL-8 κB motif. Taken together, these data provide the first direct evidence for a role of p65 in COX II and 85-kDa PLA2 gene induction and support the IL-1 activation and participation of distinct NFκB protein dimers in RSF prostanoid and IL-8 formation.


Cellular Signalling | 2000

The human polo-like kinase, PLK, regulates cdc2/cyclin B through phosphorylation and activation of the cdc25C phosphatase

Amy K. Roshak; Elizabeth A. Capper; Christina S. Imburgia; James Fornwald; Gilbert F. Scott; Lisa A. Marshall

Entry into mitosis by mammalian cells is triggered by the activation of the cdc2/cyclin B holoenzyme. This is accomplished by the specific dephosphorylation of key residues by the cdc25C phosphatase. The polo-like kinases are a family of serine/threonine kinases which are also implicated in the control of mitotic events, but their exact regulatory mechanism is not known. Recently, a Xenopus homologue, PLX1, was reported to phosphorylate and activate cdc25, leading to activation of cdc2/cyclin B. Jurkat T leukemia cells were chemically arrested and used to verify that PLK protein expression and its phosphorylation state is regulated with respect to cell cycle phase (i.e., protein is undetectable at G1/S, accumulates at S phase and is modified at G2/M). Herein, we show for the first time that endogenous human PLK protein immunoprecipitated from the G2/M-arrested Jurkat cells directly phosphorylates human cdc25C. In addition, we demonstrate that recombinant human (rh) PLK also phosphorylates rhcdc25C in a time- and concentration-dependent manner. Phosphorylation of endogenous cdc25C and recombinant cdc25C by PLK resulted in the activation of the phosphatase as assessed by dephosphorylation of cdc2/cyclin B. These data are the first to demonstrate that human PLK is capable of phosphorylating and positively regulating human cdc25C activity, allowing cdc25C to dephosphorylate inactive cdc2/cyclin B. As this event is required for cell cycle progression, we define at least one key regulatory mode of action for human PLK in the initiation of mitosis.


Inflammation Research | 2002

Scytonemin-a marine natural product inhibitor of kinases key in hyperproliferative inflammatory diseases

C. S. Stevenson; Elizabeth A. Capper; Amy K. Roshak; Brian L. Marquez; K. Grace; William H. Gerwick; Robert S. Jacobs; Lisa A. Marshall

Chronic inflammatory diseases are associated with the persistent production of proinflammatory mediators and tissue hyperplasia. Protein kinases play an important role in regulating the signaling events controlling mediator release and cell proliferation. For instance, in in vivo models, the tumor promoter, phorbol myristate acetate (PMA), is used as an initiator of inflammation, both acute and chronic. PMA produces these effects by activating protein kinase C (PKC), an enzyme involved in a number o f cellular activities, including growth, differentiation, and mediator formation [1]. In addition, cell cycle progression is highly regulated predominantly through kinase activity. One example is polo-like kinase 1 (PLK1), an enzyme important for the G2-M transition and mitotic spindle formation. Therefore, by targeting certain kinases involved in both processes, it may be possible to reduce the adverse nature o f chronic inflammatory disorders. Here we report the novel pharmacological properties o f scy-


Journal of Biological Chemistry | 2000

Human Calcium-independent Phospholipase A2 Mediates Lymphocyte Proliferation

Amy K. Roshak; Elizabeth A. Capper; Christopher Stevenson; Christopher Eichman; Lisa A. Marshall

Activation of lymphocytes induces blastogenesis and cell division which is accompanied by membrane lipid metabolism such as increased fatty acid turnover. To date little is known about the enzymatic mechanism(s) regulating this process. Release of fatty acids such as arachidonic acid requiressn-2-deacylation catalyzed by a class of enzymes known as phospholipases A2 (PLA2, EC3.1.1.4). Herein, we confirm that human peripheral blood B or T lymphocytes (PBL) do not possess measurable levels of 85-kDa PLA2 as assessed by Western immunoblot. Low levels of 14-kDa PLA2 protein and activity were detectable in the particulate fraction of PBL and Jurkat cells. Western immunoblot analysis indicates that PBLs possess the calcium-independent PLA2 (iPLA2) protein. Calcium-independentsn-2-acylhydrolytic activity was measurable in PBL cytosols and could be inhibited by the selective iPLA2inhibitor bromoenol lactone. Mitogen activation of PBLs resulted in maintenance of activity levels which remained constant over 72 h suggesting an important role for iPLA2 in this proliferative process. Indeed, evaluation of iPLA2 activity in cell cycle-arrested Jurkat T cell fractions revealed the highest iPLA2 levels occurring at the G2/M phase. Addition of the iPLA2 inhibitors, bromoenol lactone, or arachidonyl trifluoromethyl ketone (AAOCF3), inhibited both mitogen-induced PBL as well as Jurkat T cell proliferation. Moreover, specific depletion of iPLA2 protein by antisense treatment also resulted in marked suppression of cell division. Inhibition of Jurkat cell proliferation was not associated with arrest at a particular phase of the cell cycle nor was it associated with apoptosis as assessed by flow cytometry. These findings provide the first evidence that iPLA2 plays a key role in the lymphocyte proliferative response.


Journal of Biological Chemistry | 1997

Cytosolic 85-kDa Phospholipase A2-mediated Release of Arachidonic Acid Is Critical for Proliferation of Vascular Smooth Muscle Cells

Karen M. Anderson; Amy K. Roshak; James D. Winkler; Mark McCord; Lisa A. Marshall

Recent evidence suggests that arachidonic acid (AA) may be involved in regulating cellular proliferation. The predominant mechanism of AA release from cellular phospholipids is via phospholipase A2 (PLA2) hydrolysis. The purpose of this study was to examine the roles of the distinct 14-kDa and 85-kDa PLA2 enzymes in human coronary artery vascular smooth muscle cell (hCAVSMC) proliferation. Cultured hCAVSMCs proliferate in the presence of growth medium with a typical doubling time of 30–40 h, grow at a slower proliferative rate upon reaching confluency (day 8), and eventually undergo contact inhibition of growth (day 10). Neither Type II 14-kDa PLA2activity nor mass changed over a 10-day culture period. In contrast, 85-kDa PLA2 protein activity and mRNA decreased as time in culture progressed. This reduction in 85-kDa PLA2correlated with reductions in DNA synthesis and suggested a possible association between 85-kDa PLA2 and proliferation. To directly evaluate the role of the 85-kDa PLA2 in proliferation we examined the effects of an 85-kDa PLA2inhibitor (AACOCF3) and 85-kDa PLA2 antisense oligonucleotides on proliferation. Both reagents dose dependently inhibited proliferation, whereas a 14-kDa PLA2 inhibitor (SB203347), a calcium-independent PLA2 inhibitor (HELSS), an 85-kDa sense oligonucleotide, and a nonrelevant scrambled control oligonucleotide had no effect. The mechanism by which 85-kDa PLA2 influences cellular proliferation remains unclear. Inhibition of 85-kDa PLA2 activity produced neither phase-specific cell cycle arrest nor apoptosis (fluorescence-activated cell sorter analysis). Addition of AA (20 μm) attenuated the effects of both AACOCF3 and 85-kDa antisense oligonucleotides implicating AA as a key mediator in cellular proliferation. However, although prostaglandin E2(PGE2) was present in the culture medium, it peaked early (day 3) in culture, and indomethacin had no effect on cellular proliferation indicating that hCAVSMC proliferation was not mediated through PGE2. These data provide the first direct evidence that PLA2 is involved in control of VSMC proliferation and indicate that 85-kDa PLA2-mediated liberation of AA is critical for cellular proliferation.


Journal of Biological Chemistry | 1997

Depletion of Human Monocyte 85-kDa Phospholipase A2 Does Not Alter Leukotriene Formation

Lisa A. Marshall; Brian Bolognese; James D. Winkler; Amy K. Roshak

Human monocytes possess several acylhydrolase activities and are capable of producing both prostanoids (PG) and leukotriene (LT) products upon acute stimulation with calcium ionophore, A23187 or phagocytosis of zymosan particles. The cytosolic 85-kDa phospholipase (PLA) A2 co-exists with the 14-kDa PLA2 in the human monocyte, but their respective roles in LT production are not well understood. Reduction in 85-kDa PLA2 cellular protein levels by initiation site-directed antisense (SK 7111) or exposure to the 85-kDa PLA2 inhibitor, arachidonyl trifluoromethyl ketone (AACOCF3), prevented A23187 or zymosan-stimulated monocyte prostanoid formation. In contrast, neither treatment altered stimulated LTC4 production. This confirmed the important role of the 85-kDa PLA2 in prostanoid formation but suggests that it has less of a role in LT biosynthesis. Alternatively, treatment of monocytes with the selective, active site-directed 14-kDa PLA2 inhibitor, SB 203347, prior to stimulation had no effect on prostanoid formation at concentrations that totally inhibited LT formation. Addition of 20 μM exogenous arachidonic acid to monocytes exposed to SK 7111 or SB 203347 did not alter A23187-induced PGE2 or LTC4 generation, respectively, indicating that these agents had no effect on downstream arachidonic acid-metabolizing enzymes in this setting. Taken together, these results provide evidence that the 85-kDa PLA2 may play a more significant role in the formation of PG than LT. Further, utilization of SB 203347 provides intriguing data to form the hypothesis that a non-85-kDa PLA2 sn-2 acyl hydrolase, possibly the 14-kDa PLA2, may provide substrate for LT formation.


Current Opinion in Pharmacology | 2002

Small-molecule inhibitors of NF-κB for the treatment of inflammatory joint disease

Amy K. Roshak; James F. Callahan; Simon M. Blake

Abstract Recent advances in our understanding of the role of cytokine networks in inflammatory processes have led to the development of novel biological agents for the treatment of chronic inflammatory diseases. At the present time, significant efforts are focused on characterizing the complex signal transduction cascades that are activated by these cytokines and, in turn, regulate their expression. The transcription factor NF-κB is a pivotal regulator of the inducible expression of key proinflammatory mediators, and activated NF-κB has been observed in several debilitating inflammatory disorders, including rheumatoid arthritis and osteoarthritis. In light of its central role in inflammation, the identification of inhibitors of NF-κB should provide novel therapeutics for the treatment of chronic joint disease .


Journal of Leukocyte Biology | 1999

Anti-human RP105 sera induces lymphocyte proliferation.

Amy K. Roshak; Karen M. Anderson; Stephen D. Holmes; Zdenka L. Jonak; Brian Bolognese; Jonathan A. Terrett; Lisa A. Marshall

Cellular environment dictates whether antigen binding to the B lymphocyte receptor together with co‐stimulatory molecules will result in proliferation, anergy, or apoptosis. Murine RP105 is a member of the leucine‐rich repeat family of proteins, which is specifically expressed on mature B cells. Monoclonal antibodies to the murine RP105 induce proliferation and protect B cells from apoptosis, suggesting an important regulatory role in murine B lymphocyte function. We identified a human RP105 homolog and mapped the gene to chromosome 5q 12.3‐13.1. Tissue distribution analysis shows that the transcript is found predominately in lymphoid tissues including spleen, tonsils, appendix, and peripheral blood leukocytes. Polymerase chain reaction analysis of isolated primary human cell populations confirms that mRNA exists in spleen B lymphocytes and monocytes but not T lymphocytes. Western blot analysis demonstrates specific expression of human RP105 in human B lymphocytes. Murine anti‐human RP105 sera was generated using DNA immunization. The antisera contained antibodies that recognized and bound to human B lymphocytes from both spleen and peripheral blood as assessed by flow cytometry. Assessment of biological function showed that human peripheral blood leukocytes incubated with anti‐RP105 sera were induced to proliferate as measured by tritiated thymidine incorporation. Moreover, anti‐CD40 and interleukin‐4‐treated cells but not those exposed to anti‐RP105 sera produced soluble CD23, suggesting distinct functional roles. This is the first demonstration of both the existence of RP105 protein on human B lymphocytes and its role in the regulation of B lymphocyte activation. J. Leukoc. Biol. 65: 43–49; 1999.


Biochimica et Biophysica Acta | 1997

Evidence that 85 kDa phospholipase A2 is not linked to CoA-independent transacylase-mediated production of platelet-activating factor in human monocytes

James D. Winkler; Brian Bolognese; Amy K. Roshak; Chiu-Mei Sung; Lisa A. Marshall

Platelet-activating factor (PAF) production is carefully controlled in inflammatory cells. The specific removal of arachidonate (AA) from 1-O-alkyl-2-arachidonoyl-sn-glycero-3-phosphocholine (GPC), thought to be mediated by CoA-independent transacylase (CoA-IT), is required to generate the PAF precursor 1-O-alkyl-2-lyso-GPC in human neutrophils. Exposure of A23187-stimulated human monocytes to the CoA-IT inhibitors SK&F 98625 and SK&F 45905 inhibited PAF formation (IC50s of 10 and 12 microM, respectively), indicating that these cells also need CoA-IT activity for PAF production. Because CoA-IT activity transfers arachidonate to a 2-lyso phospholipid substrate, its activity is obligated to an sn-2 acyl hydrolase to form the 2-lyso phospholipid substrate. SB 203347, an inhibitor of 14 kDa phospholipase A2 (PLA2), and AACOCF3, an inhibitor of 85 kDa PLA2, both inhibited AA release from A23187-stimulated human monocytes. However, AACOCF3 had no effect on A23187-induced PAF formation at concentrations as high as 3 microM. Further, depletion of 85 kDa PLA2 using antisense (SB 7111, 1 microM) had no effect on PAF production, indicating a lack of a role of 85 kDa PLA2 in PAF biosynthesis. Both SB 203347 and the 14 kDa PLA2 inhibitor scalaradial blocked PAF synthesis in monocytes (IC50s of 2 and 0.5 microM, respectively), suggesting a key role of 14 kDa PLA2 in this process. Further, A23187-stimulated monocytes produced two forms of PAF: 80% 1-O-alkyl-2-acetyl-GPC and 20% 1-acyl-2-acetyl-GPC, which were both equally inhibited by SB 203347. In contrast, inhibition of CoA-IT using SK&F 45905 (20 microM) had a greater effect on the production of 1-O-alkyl (-80%) than of 1-acyl (-14%) acetylated material. Finally, treatment of U937 cell membranes with exogenous human recombinant (rh) type II 14 kDa PLA2, but not rh 85 kDa PLA2, induced PAF production. Elimination of membrane CoA-IT activity by heat treatment impaired the ability of 14 kDa PLA2 to induce PAF formation. Taken together, these results suggest that a 14 kDa PLA2-like activity, and not 85 kDa PLA2, is coupled to monocyte CoA-IT-induced PAF production.


Advances in Experimental Medicine and Biology | 1999

RESPECTIVE ROLES OF THE 14 KDA AND 85 KDA PHOSPHOLIPASE A2 ENZYMES IN HUMAN MONOCYTE EICOSANOID FORMATION

Lisa A. Marshall; Brian Bolognese; Amy K. Roshak

Human monocytes possess both the cytosolic 85 kDa phospholipase (PLA) A2 and a 14 kDa PLA2 and are capable of simultaneously producing prostanoids (PG), leukotrienes (LT) and platelet activating factor (PAF). As the exact roles of the two enzymes in monocyte lipid mediator formation was unclear, both selective PLA2 inhibitors and antisense were used to elucidate their respective roles. Reduction in 85 kDa PLA2 cellular protein levels by initiation site-directed antisense (SK 7111) or exposure to the 85 kDa PLA2 inhibitor, arachidonyl trifluormethyl ketone (AACOCF3), prevented A23187 or zymosan-stimulated monocytes prostanoid formation but not LTC4 or PAF production. This confirmed the important role of the 85 kDa PLA2 in prostanoid formation but indicated a less significant role in LT or PAF biosynthesis. Alternatively, treatment of monocytes with the selective, active-site-directed 14 kDa PLA2 inhibitor, SB 203347, totally inhibited LT and PAF formation, while prostanoid formation was not altered. Addition of 20 uM exogenous arachidonic acid (AA) to monocytes exposed to SB 203347 did not alter A23187-induced LTC4 generation, indicating that SB 203347 had no effect on downstream AA metabolizing enzymes in this setting. Taken together, these results provide evidence that the 14 kDa PLA2 provides substrate for monocyte LT and PAF formation, while the 85 kDa PLA2 plays a more significant role in the formation of PG.

Collaboration


Dive into the Amy K. Roshak's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James D. Winkler

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen M. Anderson

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge