Network


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

Hotspot


Dive into the research topics where Kathryn A. Skelding is active.

Publication


Featured researches published by Kathryn A. Skelding.


Cell Cycle | 2011

Controlling the cell cycle: the role of calcium/calmodulin-stimulated protein kinases I and II

Kathryn A. Skelding; John A.P. Rostas; Nicole M. Verrills

Many studies have implicated Ca2+ and calmodulin (CaM) as regulators of the cell cycle. Ca2+/CaM-stimulated proteins, including the family of multifunctional Ca2+/CaM-stimulated protein kinases (CaMK), have also been identified as mediators of cell cycle progression. CaMKII is the best-characterized member of this family, and is regulated by multi-site phosphorylation and targeting. Using pharmacological inhibitors that were believed to be specific for CaMKII, CaMKII has been implicated in every phase of the cell cycle. However, these ‘specific’ inhibitors also produce effects on other CaMKs. These additional effects are usually ignored, and the effects of the inhibitors are normally attributed to CaMKII without further investigation. Using new specific molecular techniques, it has become clear that CaMKI is an important regulator of G1, whereas CaMKII is essential for regulating G2/M and the metaphase-anaphase transition. If the mechanisms controlling these events can be fully elucidated, new targets for controlling proliferative diseases may be identified.


Neurochemical Research | 2009

Regulation of CaMKII In vivo: The Importance of Targeting and the Intracellular Microenvironment

Kathryn A. Skelding; John A.P. Rostas

CaMKII (calcium/calmodulin-stimulated protein kinase II) is a multifunctional protein kinase that regulates normal neuronal function. CaMKII is regulated by multi-site phosphorylation, which can alter enzyme activity, and targeting to cellular microdomains through interactions with binding proteins. These proteins integrate CaMKII into multiple signalling pathways, which lead to varied functional outcomes following CaMKII phosphorylation, depending on the identity and location of the binding partner. A new phosphorylation site on CaMKII (Thr253) has been identified in vivo. Thr253 phosphorylation controls CaMKII purely by targeting, does not effect enzyme activity, and occurs in response to physiological and pathological stimuli in vivo, but only in CaMKII molecules present in specific cellular locations. This new phosphorylation site offers a potentially novel regulatory mechanism for controlling functional responses elicited by CaMKII that are restricted to specific subcellular locations and/or certain cell types, by controlling interactions with proteins that are expressed in the cell at that location.


Journal of Cerebral Blood Flow and Metabolism | 2012

αCaMKII is Differentially Regulated in Brain Regions that Exhibit Differing Sensitivities to Ischemia and Excitotoxicity

Kathryn A. Skelding; Neil J. Spratt; Phillip W. Dickson; John A.P. Rostas

Different brain regions exhibit differing sensitivities to ischemia/excitotoxicity. Whether these differences are due to perfusion or intrinsic factors has not been established. Herein, we found no apparent association between sensitivity to ischemia/excitotoxicity and the level of expression or basal phosphorylation of calcium/calmodulin-stimulated protein kinase II (αCaMKII) or glutamate receptors. However, we demonstrated significant differences in CaMKII-mediated responses after ischemia/excitotoxic stimulation in striatum and cortex. In vivo ischemia and in vitro excitotoxic stimulation produced more rapid phosphorylation of Thr253-αCaMKII in striatum compared with cortex, but equal rates of Thr286-αCaMKII phosphorylation. Phosphorylation by CaMKII of Ser831-GluA1 and Ser1303-GluN2B occurred more rapidly in striatum than in cortex after either stimulus. The differences between brain regions in CaMKII activation and its effects were not accounted for by differences in the expression of αCaMKII, glutamate receptors, or density of synapses. These results implicate intrinsic tissue differences in Thr253-αCaMKII phosphorylation in the differential sensitivities of brain regions to ischemia/excitotoxicity.


Cellular Signalling | 2010

Regulation of CaMKII by phospho-Thr253 or phospho-Thr286 sensitive targeting alters cellular function

Kathryn A. Skelding; Tatsuo Suzuki; Sarah L. Gordon; Jing Xue; Nicole M. Verrills; Phillip W. Dickson; John A.P. Rostas

Calcium/calmodulin-stimulated protein kinase II (CaMKII) is an important mediator of synaptic function that is regulated by multi-site phosphorylation and targeting through interactions with proteins. A new phosphorylation site at Thr253 has been identified in vivo, that does not alter CaMKII activity, but does alter CaMKII function through interactions with binding proteins. To identify these proteins, as well as to examine the specific effects following Thr253 or Thr286 phosphorylation on these interactions, we developed an in vitro overlay binding assay. We demonstrated that the interaction between CaMKII and its binding proteins was altered by the phosphorylation state of both the CaMKII and the partner, and identified a CaMKII-specific sequence that was responsible for the interaction between CaMKII and two interacting proteins. By comparing CaMKII binding profiles in tissue and cell extracts, we demonstrated that the CaMKII binding profiles varied with cell type, and also showed that overexpression of a CaMKII Thr253 phospho-mimic mutant in human neuroblastoma and breast cancer cells dramatically altered the morphology and growth rates when compared to overexpression of non-phosphorylated CaMKII. This data highlights the importance of the microenvironment in regulating CaMKII function, and describes a potentially new mechanism by which the functions of CaMKII can be regulated.


Advances in Experimental Medicine and Biology | 2012

The Role of Molecular Regulation and Targeting in Regulating Calcium/Calmodulin Stimulated Protein Kinases

Kathryn A. Skelding; John A.P. Rostas

Calcium/calmodulin-stimulated protein kinases can be classified as one of two types - restricted or multifunctional. This family of kinases contains several structural similarities: all possess a calmodulin binding motif and an autoinhibitory region. In addition, all of the calcium/calmodulin-stimulated protein kinases examined in this chapter are regulated by phosphorylation, which either activates or inhibits their kinase activity. However, as the multifunctional calcium/calmodulin-stimulated protein kinases are ubiquitously expressed, yet regulate a broad range of cellular functions, additional levels of regulation that control these cell-specific functions must exist. These additional layers of control include gene expression, signaling pathways, and expression of binding proteins and molecular targeting. All of the multifunctional calcium/calmodulin-stimulated protein kinases examined in this chapter appear to be regulated by these additional layers of control, however, this does not appear to be the case for the restricted kinases.


Oncotarget | 2016

Activation of protein phosphatase 2A in FLT3+ acute myeloid leukemia cells enhances the cytotoxicity of FLT3 tyrosine kinase inhibitors

Amanda M. Smith; Matthew D. Dun; Erwin M. Lee; Celeste L. Harrison; Richard G. S. Kahl; Hayley M. Flanagan; Nikita Panicker; Baratali Mashkani; Anthony S. Don; Jonathan C. Morris; Hamish D. Toop; Richard B. Lock; Jason A. Powell; Daniel Thomas; Mark A. Guthridge; Andrew S. Moore; Leonie K. Ashman; Kathryn A. Skelding; Anoop K. Enjeti; Nicole M. Verrills

Constitutive activation of the receptor tyrosine kinase Fms-like tyrosine kinase 3 (FLT3), via co-expression of its ligand or by genetic mutation, is common in acute myeloid leukemia (AML). In this study we show that FLT3 activation inhibits the activity of the tumor suppressor, protein phosphatase 2A (PP2A). Using BaF3 cells transduced with wildtype or mutant FLT3, we show that FLT3-induced PP2A inhibition sensitizes cells to the pharmacological PP2A activators, FTY720 and AAL(S). FTY720 and AAL(S) induced cell death and inhibited colony formation of FLT3 activated cells. Furthermore, PP2A activators reduced the phosphorylation of ERK and AKT, downstream targets shared by both FLT3 and PP2A, in FLT3/ITD+ BaF3 and MV4-11 cell lines. PP2A activity was lower in primary human bone marrow derived AML blasts compared to normal bone marrow, with blasts from FLT3-ITD patients displaying lower PP2A activity than WT-FLT3 blasts. Reduced PP2A activity was associated with hyperphosphorylation of the PP2A catalytic subunit, and reduced expression of PP2A structural and regulatory subunits. AML patient blasts were also sensitive to cell death induced by FTY720 and AAL(S), but these compounds had minimal effect on normal CD34+ bone marrow derived monocytes. Finally, PP2A activating compounds displayed synergistic effects when used in combination with tyrosine kinase inhibitors in FLT3-ITD+ cells. A combination of Sorafenib and FTY720 was also synergistic in the presence of a protective stromal microenvironment. Thus combining a PP2A activating compound and a FLT3 inhibitor may be a novel therapeutic approach for treating AML.


Scientific Reports | 2016

Phosphorylation of calcium/calmodulin-stimulated protein kinase II at T286 enhances invasion and migration of human breast cancer cells

Mengna Chi; Hamish Evans; Jackson Gilchrist; Jackson Mayhew; Alexander Hoffman; Elizabeth Pearsall; Helen Jankowski; Joshua S. Brzozowski; Kathryn A. Skelding

Calcium/calmodulin-stimulated protein kinase II (CaMKII) is a multi-functional kinase that controls a range of cellular functions, including proliferation, differentiation and apoptosis. The biological properties of CaMKII are regulated by multi-site phosphorylation. However, the role that CaMKII phosphorylation plays in cancer cell metastasis has not been examined. We demonstrate herein that CaMKII expression and phosphorylation at T286 is increased in breast cancer when compared to normal breast tissue, and that increased CAMK2 mRNA is associated with poor breast cancer patient prognosis (worse overall and distant metastasis free survival). Additionally, we show that overexpression of WT, T286D and T286V forms of CaMKII in MDA-MB-231 and MCF-7 breast cancer cells increases invasion, migration and anchorage independent growth, and that overexpression of the T286D phosphomimic leads to a further increase in the invasive, migratory and anchorage independent growth capacity of these cells. Pharmacological inhibition of CaMKII decreases MDA-MB-231 migration and invasion. Furthermore, we demonstrate that overexpression of T286D, but not WT or T286V-CaMKII, leads to phosphorylation of FAK, STAT5a, and Akt. These results demonstrate a novel function for phosphorylation of CaMKII at T286 in the control of breast cancer metastasis, offering a promising target for the development of therapeutics to prevent breast cancer metastasis.


Cellular Signalling | 2014

Dephosphorylation of CaMKII at T253 controls the metaphase-anaphase transition.

Alexander Hoffman; Helen Carpenter; Richard G. S. Kahl; Lauren F. Watt; Phillip W. Dickson; John A.P. Rostas; Nicole M. Verrills; Kathryn A. Skelding

Calcium/calmodulin-stimulated protein kinase II (CaMKII) is a multi-functional serine/threonine protein kinase that controls a range of cellular functions, including proliferation. The biological properties of CaMKII are regulated by multi-site phosphorylation and targeting via interactions with specific proteins. To investigate the role specific CaMKII phosphorylation sites play in controlling cell proliferation and cell cycle progression, we examined phosphorylation of CaMKII at two sites (T253 and T286) at various stages of the cell cycle, and also examined the effects of overexpression of wild-type (WT), T286D phosphomimic, T253D phosphomimic and T253V phosphonull forms of CaMKIIα in MDA-MB-231 breast cancer and SHSY5Y neuroblastoma cells on cellular proliferation and cell cycle progression. We demonstrate herein that whilst there is no change in total CaMKII expression or T286 phosphorylation throughout the cell cycle, a marked dephosphorylation of CaMKII at T253 occurs during the G2 and/or M phases. Additionally, we show by molecular inhibition, as well as pharmacological activation, that protein phosphatase 2A (PP2A) is the phosphatase responsible for this dephosphorylation. Furthermore, we show that inducible overexpression of WT, T286D and T253V forms of CaMKIIα in MDA-MB-231 and SHSY5Y cells increases cellular proliferation, with no alteration in cell cycle profiles. By contrast, overexpression of a T253D phosphomimic form of CaMKIIα significantly decreases proliferation, and cells accumulate in mitosis, specifically in metaphase. Taken together, these results strongly suggest that the dephosphorylation of CaMKII at T253 is involved in controlling the cell cycle, specifically the metaphase-anaphase transition.


Neurochemistry International | 2017

The role of Ca2+-calmodulin stimulated protein kinase II in ischaemic stroke – A potential target for neuroprotective therapies

John A.P. Rostas; Neil J. Spratt; Phillip W. Dickson; Kathryn A. Skelding

Studies in multiple experimental systems show that Ca2+-calmodulin stimulated protein kinase II (CaMKII) is a major mediator of ischaemia-induced cell death and suggest that CaMKII would be a good target for neuroprotective therapies in acute treatment of stroke. However, as CaMKII regulates many cellular processes in many tissues any clinical treatment involving the inhibition of CaMKII would need to be able to specifically target the functions of ischaemia-activated CaMKII. In this review we summarise new developments in our understanding of the molecular mechanisms involved in ischaemia-induced CaMKII-mediated cell death that have identified ways in which such specificity of CaMKII inhibition after stroke could be achieved. We also review the mechanisms and phases of tissue damage in ischaemic stroke to identify where and when CaMKII-mediated mechanisms may be involved.


Neurochemistry International | 2017

Ischaemia- and excitotoxicity-induced CaMKII-Mediated neuronal cell death: The relative roles of CaMKII autophosphorylation at T286 and T253

John A.P. Rostas; Alexander Hoffman; Lucy A. Murtha; Debbie Pepperall; Damian McLeod; Phillip W. Dickson; Neil J. Spratt; Kathryn A. Skelding

ABSTRACT Ischaemia/excitotoxicity produces persistent activation of CaMKII (Ca2+‐calmodulin stimulated protein kinase II) that initiates cell death. This study investigated the involvement of CaMKII phosphorylation at T286 and T253 in producing this persistent activation. In T286A‐&agr;CaMKII transgenic mice that lack the ability to phosphorylate &agr;CaMKII at T286, transient occlusion of the middle cerebral artery for 90 min resulted in no significant difference in infarct size compared to normal littermate controls. Overexpression of the phospho‐mimic mutant T286D‐&agr;CaMKII in differentiated neuroblastoma cell lines did not enhance excitotoxicity‐induced cell death compared to overexpression of wild type &agr;CaMKII. By contrast, overexpression of the phospho‐mimic mutant T253D‐&agr;CaMKII significantly enhanced excitotoxicity‐induced cell death whereas overexpression of the phospho‐null mutant T253V‐&agr;CaMKII produced no enhancement. These results indicate that T286 phosphorylation does not play a significant role in ischaemia/excitotoxicity induced CaMKII‐mediated cell death and suggest that T253 phosphorylation is required to produce the persistent activation of CaMKII involved in ischaemia/excitotoxicity induced cell death. HighlightsThe mechanism of ischaemia‐and excitotoxicity‐induced CaMKII‐mediated neuronal cell death is investigated.CaMKII phosphorylation at T286 does not play a major role in ischaemia‐ or excitotoxicity‐induced cell death.T253D‐&agr;CaMKII, but not T253V‐&agr;CaMKII, enhances excitotoxic cell death.Interaction of pT253‐CaMKII with a binding protein is proposed to activate CaMKII.Ischemic/excitotoxic cell death is proposed to require CaMKII phosphorylation at T253.

Collaboration


Dive into the Kathryn A. Skelding'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth W. Beagley

Queensland University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge