Network


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

Hotspot


Dive into the research topics where Rosemary O’Connor is active.

Publication


Featured researches published by Rosemary O’Connor.


Cell Communication and Signaling | 2013

RACK(1) to the future – a historical perspective

Dorit Ron; David R. Adams; George S. Baillie; Aideen Long; Rosemary O’Connor; Patrick A. Kiely

This perspective summarises the first and long overdue RACK1 meeting held at the University of Limerick, Ireland, May 2013, in which RACK1’s role in the immune system, the heart and the brain were discussed and its contribution to disease states such as cancer, cardiac hypertrophy and addiction were described. RACK1 is a scaffolding protein and a member of the WD repeat family of proteins. These proteins have a unique architectural assembly that facilitates protein anchoring and the stabilisation of protein activity. A large body of evidence is accumulating which is helping to define the versatile role of RACK1 in assembling and dismantling complex signaling pathways from the cell membrane to the nucleus in health and disease. In this commentary, we first provide a historical perspective on RACK1. We also address many of the pertinent and topical questions about this protein such as its role in transcription, epigenetics and translation, its cytoskeletal contribution and the merits of targeting RACK1 in disease.


Immunology | 2011

Tim‐4 inhibition of T‐cell activation and T helper type 17 differentiation requires both the immunoglobulin V and mucin domains and occurs via the mitogen‐activated protein kinase pathway

Wei Cao; Michelle Ryan; Deirdre A. Buckley; Rosemary O’Connor; Michael R. Clarkson

Emerging experimental data suggest an important role for the T‐cell immunoglobulin mucin 1 (Tim‐1):Tim‐4 pathway in autoimmune and alloimmune responses in vivo. Using a Tim‐4 ectodomain human IgG Fc fusion protein we studied the role of Tim‐4 in T‐cell activation, signalling and differentiation responses in vitro. We demonstrate that Tim‐4Fc can inhibit naive and pre‐activated T‐cell activation, proliferation and cytokine secretion via a Tim‐1‐independent pathway. Tim‐4 contains immunoglobulin variable (IgV) and mucin domains; to identify which domain accounts for the inhibitory effect novel Tim‐4 fusion proteins containing either the IgV or mucin domain were generated. We demonstrate that both IgV and mucin domains are required for the inhibitory effects and that they are mediated at least in part by inhibition of extracellular signal‐regulated kinase pathway activity. Given the emerging interest in the role of the Tim family in T helper type 17 (Th17) cells, which play an important role in autoimmune disease and transplantation tolerance, our data show that Tim‐4Fc can prevent polarization of CD4+ T cells to the Th17 phenotype. Collectively, our results highlight an inhibitory role for Tim‐4Fc in vitro, which we propose is mediated by a receptor other than Tim‐1. In addition, this study provides new insights into the role of Tim‐4Fc in regulating Th17 immune responses and may open a new avenue for autoimmune therapy.


Oncotarget | 2016

IGF-1R inhibition sensitizes breast cancer cells to ATM-Related Kinase (ATR) inhibitor and cisplatin

Ciara O’Flanagan; Amy Lyons; Fionola M. Fogarty; Nuala McCabe; Richard D. Kennedy; Rosemary O’Connor

The complexity of the IGF-1 signalling axis is clearly a roadblock in targeting this receptor in cancer therapy. Here, we sought to identify mediators of resistance, and potential co-targets for IGF-1R inhibition. By using an siRNA functional screen with the IGF-1R tyrosine kinase inhibitor (TKI) BMS-754807 in MCF-7 cells we identified several genes encoding components of the DNA damage response (DDR) pathways as mediators of resistance to IGF-1R kinase inhibition. These included ATM and Ataxia Telangiectasia and RAD3-related kinase (ATR). We also observed a clear induction of DDR in cells that were exposed to IGF-1R TKIs (BMS-754807 and OSI-906) as indicated by accumulation of γ-H2AX, and phosphorylated Chk1. Combination of the IGF-1R/IR TKIs with an ATR kinase inhibitor VE-821 resulted in additive to synergistic cytotoxicity compared to either drug alone. In MCF-7 cells with stably acquired resistance to the IGF-1R TKI (MCF-7-R), DNA damage was also observed, and again, dual inhibition of the ATR kinase and IGF-1R/IR kinase resulted in synergistic cytotoxicity. Interestingly, dual inhibition of ATR and IGF-1R was more effective in MCF-7-R cells than parental cells. IGF-1R TKIs also potentiated the effects of cisplatin in a panel of breast cancer cell lines. Overall, our findings identify induction of DDR by IGF-1R kinase inhibition as a rationale for co-targeting the IGF-1R with ATR kinase inhibitors or cisplatin, particularly in cells with acquired resistance to TKIs.


Frontiers in Endocrinology | 2015

IGF-1 Receptor and Adhesion Signaling: An Important Axis in Determining Cancer Cell Phenotype and Therapy Resistance

Orla T. Cox; Emilie Tresse; Milan Bustamante-Garrido; Ravi Kiran-Deevi; Rosemary O’Connor

IGF-1R expression and activation levels generally cannot be correlated in cancer cells, suggesting that cellular proteins may modulate IGF-1R activity. Strong candidates for such modulation are found in cell-matrix and cell–cell adhesion signaling complexes. Activated IGF-1R is present at focal adhesions, where it can stabilize β1 integrin and participate in signaling complexes that promote invasiveness associated with epithelial mesenchymal transition (EMT) and resistance to therapy. Whether IGF-1R contributes to EMT or to non-invasive tumor growth may be strongly influenced by the degree of extracellular matrix engagement and the presence or absence of key proteins in IGF-1R-cell adhesion complexes. One such protein is PDLIM2, which promotes both cell polarization and EMT by regulating the stability of transcription factors including NFκB, STATs, and beta catenin. PDLIM2 exhibits tumor suppressor activity, but is also highly expressed in certain invasive cancers. It is likely that distinct adhesion complex proteins modulate IGF-1R signaling during cancer progression or adaptive responses to therapy. Thus, identifying the key modulators will be important for developing effective therapeutic strategies and predictive biomarkers.


Alzheimers & Dementia | 2006

O3-02-02

Cora O’Neill; Aileen M. Moloney; Suzanne Timmons; Mary Kelliher; Peter Dockery; Rosemary O’Connor; Janet A. Johnston; Rivka Ravid; Rebecca J. Griffin

tion of pro-death signaling. Recent studies of human postmortem brains linked the molecular and pathological lesions in AD to major impairments in: 1) insulin and insulin-like growth factor (IGF) gene expression; 2) expression and function of the insulin and IGF receptors; 3) neuronal survival signaling; and 4) acetylcholine homeostasis, and showed each of these abnormalities increases with progression of AD. The co-existence of insulin/IGF deficiency and insulin/IGF resistance suggests that AD represents a brain-specific form of diabetes, i.e. Type 3 diabetes. We generated an experimental animal model in which intracerebral Streptozotocin (icSTZ) was used to deplete brain and not pancreatic insulin/IGF, and produce neurodegeneration that is similar to human AD. The ic-STZ-injected rats did not have hyperglycemia, and pancreatic architecture and insulin immunoreactivity were similar to control, yet their brains were reduced in size and exhibited neurodegeneration with cell loss, gliosis, and increased immunoreactivity for p53, activated glycogen synthase kinase 3 , phospho-tau, ubiquitin, and amyloid. Real time quantitative RT-PCR studies demonstrated that the ic-STZ-treated brains had significantly reduced expression of genes corresponding to neurons, oligodendroglia, and choline acetyltransferase, and increased expression of genes encoding glial fibrillary acidic protein, microglia-specific proteins, acetylcholinesterase, tau, and amyloid precursor protein. These abnormalities were associated with reduced expression of genes encoding insulin, IGF-II, insulin receptor, IGF-I receptor, and insulin receptor substrate-1, and reduced ligand binding to the insulin and IGF-II receptors. Further studies showed that treatment with peroxisome-proliferator activated receptor agonists effectively prevented the ic-STZ-induced Type 3 diabetes and preserved learning and memory. These results demonstrate that many of the characteristic features of AD-type neurodegeneration can be produced experimentally by selectively impairing insulin/IGF functions together with increasing oxidative stress, and support our hypothesis that AD represents a neuro-endocrine disorder associated with brain-specific perturbations in insulin and IGF signaling mechanisms, i.e. Type 3 diabetes. The results also suggest that early treatment with insulin sensitizer agent may prevent or reduce the severity of AD.


Archive | 2004

The IGF-1 Receptor in Cell Survival: Signalling and Regulation

Patrick A. Kiely; Denise M. O’Gorman; Anthony Lyons; Rosemary O’Connor

The suppression of apoptosis by the IGF system is critical for normal cell development, proliferation, differentiation and motility. Aberrations in IGF signalling mechanisms contribute to cell transformation, tumour progression and metastasis. Many questions remain to be answered as to how exactly the IGF system mediates its effects both in normal and tumour cells and how the IGF-1R interacting proteins and downstream signalling cascades are regulated. The importance of the IGF system is underscored by the significant interest in the development of anti-IGF therapies for IGF sensitive cancers. Future developments in cancer therapy are likely to focus on methods to target these therapies to diseased but not normal cells.


Oncogene | 2018

FES-related tyrosine kinase activates the insulin-like growth factor-1 receptor at sites of cell adhesion

Joanna Stanicka; Leonie Rieger; Orla T. Cox; Michael Coleman; Ciara O’Flanagan; Barbara Addario; Nuala McCabe; Richard D. Kennedy; Rosemary O’Connor

IGF-1 receptor (IGF-1R) and integrin cooperative signaling promotes cancer cell survival, proliferation, and motility, but whether this influences cancer progression and therapy responses is largely unknown. Here we investigated the non-receptor tyrosine adhesion kinase FES-related (FER), following its identification as a potential mediator of sensitivity to IGF-1R kinase inhibition in a functional siRNA screen. We found that FER and the IGF-1R co-locate in cells and can be co-immunoprecipitated. Ectopic FER expression strongly enhanced IGF-1R expression and phosphorylation on tyrosines 950 and 1131. FER phosphorylated these sites in an IGF-1R kinase-independent manner and also enhanced IGF-1-mediated phosphorylation of SHC, and activation of either AKT or MAPK-signaling pathways in different cells. The IGF-1R, β1 Integrin, FER, and its substrate cortactin were all observed to co-locate in cell adhesion complexes, the disruption of which reduced IGF-1R expression and activity. High FER expression correlates with phosphorylation of SHC in breast cancer cell lines and with a poor prognosis in patient cohorts. FER and SHC phosphorylation and IGF-1R expression could be suppressed with a known anaplastic lymphoma kinase inhibitor (AP26113) that shows high specificity for FER kinase. Overall, we conclude that FER enhances IGF-1R expression, phosphorylation, and signaling to promote cooperative growth and adhesion signaling that may facilitate cancer progression.


Oncotarget | 2017

Rethinking the bile acid/gut microbiome axis in cancer

John P. Phelan; F. Jerry Reen; Jose A. Caparros-Martin; Rosemary O’Connor; Fergal O’Gara

Dietary factors, probiotic agents, aging and antibiotics/medicines impact on gut microbiome composition leading to disturbances in localised microbial populations. The impact can be profound and underlies a plethora of human disorders, including the focus of this review; cancer. Compromised microbiome populations can alter bile acid signalling and produce distinct pathophysiological bile acid profiles. These in turn have been associated with cancer development and progression. Exposure to high levels of bile acids, combined with localised molecular/genome instability leads to the acquisition of bile mediated neoplastic alterations, generating apoptotic resistant proliferation phenotypes. However, in recent years, several studies have emerged advocating the therapeutic benefits of bile acid signalling in suppressing molecular and phenotypic hallmarks of cancer progression. These studies suggest that in some instances, bile acids may reduce cancer phenotypic effects, thereby limiting metastatic potential. In this review, we contextualise the current state of the art to propose that the bile acid/gut microbiome axis can influence cancer progression to the extent that classical in vitro cancer hallmarks of malignancy (cell invasion, cell migration, clonogenicity, and cell adhesion) are significantly reduced. We readily acknowledge the existence of a bile acid/gut microbiome axis in cancer initiation, however, in light of recent advances, we focus exclusively on the role of bile acids as potentially beneficial molecules in suppressing cancer progression. Finally, we theorise that suppressing aggressive malignant phenotypes through bile acid/gut microbiome axis modulation could uncover new and innovative disease management strategies for managing cancers in vulnerable cohorts.


Cancer Research | 2016

Abstract 200: PDLIM2 phosphorylation in determining breast cancer phenotype

Orla T. Cox; Janina Berghoff; Deirdre A. Buckley; Rosemary O’Connor

Several genes that are involved in cancer progression are induced by Insulin-like Growth Factor-1 (IGF-1). One of them encodes the PDZ-LIM protein PDLIM2, which is localized at the cytoskeleton and nucleus in epithelial and hematopoietic cells. PDLIM2 is repressed in certain cancers including breast cancers, but it is highly expressed in invasive cancer cells in triple negative breast cancer (TNBC), where it has also been associated with poor survival. PDLIM2 regulates the stability and activity of transcription factors including beta catenin, STATs, NFκB and IRFs and has been associated with ubiquitination and destabilization of proteins such as NF-κB, E-cadherin, and p27 KIP1 . However, PDLIM2 itself is also regulated by posttranslational modifications, but how this controls PDLIM2 activity is still largely unclear. Therefore, this study aimed to investigate PDLIM2 phosphorylation and its effect on PDLIM2 sub-cellular location and function in transcription factor regulation. The PDLIM2 protein sequence includes multiple potential phosphorylation sites and it migrates as several distinct phosphatase-sensitive species in SDS PAGE and 2D electrophoresis, indicating phosphorylation at several residues. Using cellular fractionation and immunofluorescence we show that PDLIM2 accumulates in the nucleus in response to cell stimulation with IGF-1 or TGFβ. Moreover, we show that inhibitors of Rho Kinases suppress PDLIM2 phosphorylation in response to these stimuli. In addition, we demonstrate that mutation of the predicted Rho Kinase phosphorylation sites in PDLIM2 reduce protein stability, an effect that can be reversed by application of proteasomal inhibitor. Overall, our results indicate that phosphorylation by Rho kinases plays an important role in the subcellular location and regulation of PDLIM2 function, which opens a new avenue in targeting PDLIM2 activity in invasive cancer cells. Citation Format: Orla T. Cox, Janina Berghoff, Deirdre A. Buckley, Rosemary O’Connor. PDLIM2 phosphorylation in determining breast cancer phenotype. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 200.


Cancer Research | 2016

Abstract 204: Disruption of the RACK1/PP2A complex results in a decrease in the adhesion, proliferation, migration and invasion capabilities of breast cancer cells

Maeve Kiely; Rosemary O’Connor; David R. Adams; George S. Baillie; Patrick A. Kiely

Conflicting reports implicate the scaffolding protein RACK1 in the progression of breast cancer. PP2A has a well-established role as a tumour suppressor within signalling pathways but is also known to play a pro-carcinogenic role in certain circumstances [1]. RACK1 has been identified as a direct binding partner of PP2A to regulate cell migration and stabilize PP2A activity [2]. Our objective was to further characterise the interaction between PP2A and RACK1 in breast cancer cells. The PP2A holoenzyme is assembled in MCF-7 cells and we found that both the C subunit and A subunit of PP2A are assembled on the RACK1 scaffold. We used immobilized peptide arrays representing the entire PP2A-Catalytic protein to identify amino acids on the C subunit of PP2A that are required for the binding of RACK1. Once identified, these sites were mutated and expressed in the context of the full length PP2A C subunit protein and stable cell lines were generated. When the RACK1/PP2A interaction was disrupted, cells exhibited reduced PP2A phosphatase activity, confirming the role for RACK1 in stabilizing PP2A activity. We used the stable cell lines to determine that disruption of the RACK1/PP2A complex also reduces the adhesion, proliferation, migration and invasion of this breast cancer cell model. The work has significantly advanced our understanding of the RACK1/PP2A complex and indicates a pro-carcinogenic role for the RACK1/PP2A complex. This work has highlighted a novel mechanism to target PP2A activity and may provide a potential therapeutic target in the treatment of breast cancer. 1. Kiely, M. and P.A. Kiely, PP2A: The Wolf in Sheep9s Clothing? Cancers, 2015. 7(2): p. 648-669. 2. Kiely, P.A., et al., Tyrosine 302 in RACK1 is essential for insulin-like growth factor-I-mediated competitive binding of PP2A and β1 integrin and for tumor cell proliferation and migration. Journal of Biological Chemistry, 2008. 283(34): p. 22952-22961. Citation Format: Maeve Kiely, Rosemary O’Connor, David Adams, George S. Baillie, Patrick A. Kiely. Disruption of the RACK1/PP2A complex results in a decrease in the adhesion, proliferation, migration and invasion capabilities of breast cancer cells. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 204.

Collaboration


Dive into the Rosemary O’Connor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Orla T. Cox

University College Cork

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rivka Ravid

Netherlands Institute for Neuroscience

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge