Network


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

Hotspot


Dive into the research topics where Amy J. Aldrich is active.

Publication


Featured researches published by Amy J. Aldrich.


Cancer Research | 2009

A Novel Splice Variant of GLI1 That Promotes Glioblastoma Cell Migration and Invasion

Hui-Wen Lo; Hu Zhu; Xinyu Cao; Amy J. Aldrich; Francis Ali-Osman

The family of GLI zinc finger transcription factors regulates the expression of genes involved in many important cellular processes, notably embryonal development and cellular differentiation. The glioma-associated oncogene homologue 1 (GLI1) isoform, in particular, has attracted much attention because of its frequent activation in many human cancers and its interactions with other signaling pathways, such as those mediated by K-RAS, transforming growth factor-beta, epidermal growth factor receptor, and protein kinase A. Here, we report the identification of a novel truncated GLI1 splice variant, tGLI1, with an in-frame deletion of 123 bases (41 codons) spanning the entire exon 3 and part of exon 4 of the GLI1 gene. Expression of tGLI1 is undetectable in normal cells but is high in glioblastoma multiforme (GBM) and other cancer cells. Although tGLI1 undergoes nuclear translocalization and transactivates GLI1-binding sites similar to GLI1, unlike GLI1, it is associated with increased motility and invasiveness of GBM cells. Using microarray analysis, we showed >100 genes to be differentially expressed in tGLI1-expressing compared with GLI1-expressing GBM cells, although both cell types expressed equal levels of known GLI1-regulated genes, such as PTCH1. We further showed one of the tGLI1 up-regulated genes, CD24, an invasion-associated gene, to be required for the migratory and invasive phenotype of GBM cells. These data provide conclusive evidence for a novel gain-of-function GLI1 splice variant that promotes migration and invasiveness of GBM cells and open up a new research paradigm on the role of the GLI1 pathway in malignancy.


Molecular Cancer Therapeutics | 2012

XIAP Inhibition and Generation of Reactive Oxygen Species Enhances TRAIL Sensitivity in Inflammatory Breast Cancer Cells

Jennifer L. Allensworth; Katherine M. Aird; Amy J. Aldrich; Ines Batinic-Haberle; Gayathri R. Devi

We recently identified superoxide dismutase (SOD) overexpression and decreased induction of reactive oxygen species (ROS)-mediated apoptosis in models of inflammatory breast cancer (IBC) cells with acquired therapeutic resistance. This population of cells has high expression of X-linked inhibitor of apoptosis protein (XIAP), which inhibits both extrinsic and intrinsic apoptosis pathways. We therefore wanted to evaluate the effect of classical apoptosis-inducing agent TRAIL, a proapoptotic receptor agonist that selectively triggers death receptor (DR)-mediated apoptosis in cancer cells, in the IBC acquired resistance model. XIAP levels and subsequent inhibition of caspase activity inversely correlated with TRAIL sensitivity in our models of IBC. These include SUM149, a basal-type cell line isolated from primary IBC tumors and isogenic SUM149-derived lines rSUM149 and SUM149 wtXIAP, models of acquired therapeutic resistance with endogenous and exogenous XIAP overexpression, respectively. Inhibition of XIAP function using embelin, a plant-derived cell permeable small molecule, in combination with TRAIL caused a synergistic decrease in cell viability. Embelin treatment resulted in activation of extracellular signal–regulated kinase (ERK)1/2 and ROS accumulation, which correlated with downregulation of antioxidant protein SOD1 and consumption of redox modulator reduced glutathione in the XIAP-overexpressing cells. Simultaneous treatment with an SOD mimic, which protects against ROS accumulation, reversed the decrease in cell viability caused by embelin + TRAIL treatment. Embelin primes IBC cells for TRAIL-mediated apoptosis by its direct action on the anti-caspase activity of XIAP and by shifting the cellular redox balance toward oxidative stress–mediated apoptosis. Thus, ROS modulators represent a novel approach to enhance efficacy of TRAIL-based treatment protocols in IBC. Mol Cancer Ther; 11(7); 1518–27. ©2012 AACR.


British Journal of Cancer | 2011

Targeting GLI1 expression in human inflammatory breast cancer cells enhances apoptosis and attenuates migration.

Zainab Thomas; W Gibson; Jonathan Z. Sexton; Katherine M. Aird; Shalonda M. Ingram; Amy J. Aldrich; Herbert Kim Lyerly; Gayathri R. Devi; Kevin P. Williams

Background:Inflammatory breast cancer (IBC) is an aggressive subtype of breast cancer with distinct molecular profiles. Gene expression profiling previously identified sonic hedgehog (SHH) as part of a gene signature that is differentially regulated in IBC patients.Methods:The effects of reducing GLI1 levels on protein expression, cell proliferation, apoptosis and migration were determined by immunoblots, MTT assay, Annexin-V/PI assay and conventional and automated cell migration assays.Results:Evaluation of a panel of breast cancer cell lines revealed elevated GLI1 expression, typically a marker for hedgehog-pathway activation, in a triple-negative, highly invasive IBC cell line, SUM149 and its isogenic-derived counterpart rSUM149 that has acquired resistance to ErbB1/2 targeting strategies. Downregulation of GLI1 expression in SUM149 and rSUM149 by small interfering RNA or a small molecule GLI1 inhibitor resulted in decreased proliferation and increased apoptosis. Further, GLI1 suppression in these cell lines significantly inhibited cell migration as assessed by a wound-healing assay compared with MCF-7, a non-invasive cell line with low GLI1 expression. A novel high-content migration assay allowed us to quantify multiple effects of GLI1 silencing including significant decreases in cell distance travelled and linearity of movement.Conclusion:Our data reveal a role for GLI1 in IBC cell proliferation, survival and migration, which supports the feasibility of targeting GLI1 as a novel therapeutic strategy for IBC patients.


Molecular Oncology | 2015

Disulfiram (DSF) acts as a copper ionophore to induce copper-dependent oxidative stress and mediate anti-tumor efficacy in inflammatory breast cancer

Jennifer L. Allensworth; Myron K. Evans; François Bertucci; Amy J. Aldrich; Richard A. Festa; Pascal Finetti; Naoto T. Ueno; Rachid Safi; Donald P. McDonnell; Dennis J. Thiele; Steven Van Laere; Gayathri R. Devi

Cancer cells often have increased levels of reactive oxygen species (ROS); however, acquisition of redox adaptive mechanisms allows for evasion of ROS‐mediated death. Inflammatory breast cancer (IBC) is a distinct, advanced BC subtype characterized by high rates of residual disease and recurrence despite advances in multimodality treatment. Using a cellular model of IBC, we identified an oxidative stress response (OSR) signature in surviving IBC cells after administration of an acute dose of an ROS inducer. Metagene analysis of patient samples revealed significantly higher OSR scores in IBC tumor samples compared to normal or non‐IBC tissues, which may contribute to the poor response of IBC tumors to common treatment strategies, which often rely heavily on ROS induction. To combat this adaptation, we utilized a potent redox modulator, the FDA‐approved small molecule Disulfiram (DSF), alone and in combination with copper. DSF forms a complex with copper (DSF‐Cu) increasing intracellular copper concentration both in vitro and in vivo, bypassing the need for membrane transporters. DSF‐Cu antagonized NFκB signaling, aldehyde dehydrogenase activity and antioxidant levels, inducing oxidative stress‐mediated apoptosis in multiple IBC cellular models. In vivo, DSF‐Cu significantly inhibited tumor growth without significant toxicity, causing apoptosis only in tumor cells. These results indicate that IBC tumors are highly redox adapted, which may render them resistant to ROS‐inducing therapies. DSF, through redox modulation, may be a useful approach to enhance chemo‐ and/or radio‐sensitivity for advanced BC subtypes where therapeutic resistance is an impediment to durable responses to current standard of care.


PLOS ONE | 2013

Immunologic Targeting of FOXP3 in Inflammatory Breast Cancer Cells

Smita K. Nair; Amy J. Aldrich; Eoin McDonnell; Qing Cheng; Anshu Aggarwal; Pujan Patel; Monique M. Williams; David Boczkowski; H. Kim Lyerly; Michael A. Morse; Gayathri R. Devi

The forkhead transcription factor FOXP3 is necessary for induction of regulatory T lymphocytes (Tregs) and their immunosuppressive function. We have previously demonstrated that targeting Tregs by vaccination of mice with murine FOXP3 mRNA-transfected dendritic cells (DCs) elicits FOXP3-specific T cell responses and enhances tumor immunity. It is clear that FOXP3 expression is not restricted to T-cell lineage and herein, using RT-PCR, flow cytometry, and western immunoblot we demonstrate for the first time that FOXP3 is expressed in inflammatory breast cancer (IBC) cells, SUM149 (triple negative, ErbB1-activated) and SUM190 (ErbB2-overexpressing). Importantly, FOXP3-specific T cells generated in vitro using human FOXP3 RNA-transfected DCs as stimulators efficiently lyse SUM149 cells. Interestingly, an isogenic model (rSUM149) derived from SUM149 with an enhanced anti-apoptotic phenotype was resistant to FOXP3-specific T cell mediated lysis. The MHC class I cellular processing mechanism was intact in both cell lines at the protein and transcription levels suggesting that the resistance to cytolysis by rSUM149 cells was not related to MHC class I expression or to the MHC class I antigen processing machinery in these cells. Our data suggest that FOXP3 may be an effective tumor target in IBC cells however increased anti-apoptotic signaling can lead to immune evasion.


Cancer Letters | 2013

Quantitative high-throughput efficacy profiling of approved oncology drugs in inflammatory breast cancer models of acquired drug resistance and re-sensitization.

Kevin P. Williams; Jennifer L. Allensworth; Shalonda M. Ingram; Ginger R. Smith; Amy J. Aldrich; Jonathan Z. Sexton; Gayathri R. Devi

Although there is no standard treatment protocol for inflammatory breast cancer (IBC), multi-modality treatment has improved survival. In this study we profiled the NCI approved oncology drug set in a qHTS format to identify those that are efficacious in basal type and ErbB2 overexpressing IBC models. Further, we characterized the sensitivity of an acquired therapeutic resistance model to the oncology drugs. We observed that lapatinib-induced acquired resistance in SUM149 cells led to cross-resistance to other targeted- and chemotherapeutic drugs. Removal of the primary drug to which the model was developed led to re-sensitization to multiple drugs to a degree comparable to the parental cell line; this coincided with the cells regaining the ability to accumulate ROS and reduced expression of anti-apoptotic factors and the antioxidant SOD2. We suggest that our findings provide a unique IBC model system for gaining an understanding of acquired therapeutic resistance and the effect of redox adaptation on anti-cancer drug efficacy.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2008

Myocyte Specific Overexpression of Myoglobin Impairs Angiogenesis After Hind-Limb Ischemia

Surovi Hazarika; Michael Angelo; Yongjun Li; Amy J. Aldrich; Shelley I. Odronic; Zhen Yan; Jonathan S. Stamler; Brian H. Annex

Objective—In preclinical models of peripheral arterial disease the angiogenic response is typically robust, though it can be impaired in conditions such as hypercholesterolemia and diabetes where the endothelium is dysfunctional. Myoglobin (Mb) is expressed exclusively in striated muscle cells. We hypothesized that myocyte specific overexpression of myoglobin attenuates ischemia-induced angiogenesis even in the presence of normal endothelium. Methods and Results—Mb overexpressing transgenic (MbTg, n=59) and wild-type (WT, n=56) C57Bl/6 mice underwent unilateral femoral artery ligation/excision. Perfusion recovery was monitored using Laser Doppler. Ischemia-induced changes in muscle were assessed by protein and immunohistochemistry assays. Nitrite/nitrate and protein-bound NO, and vasoreactivity was measured. Vasoreactivity was similar between MbTg and WT. In ischemic muscle, at d14 postligation, MbTg increased VEGF-A, and activated eNOS the same as WT mice but nitrate/nitrite were reduced whereas protein-bound NO was higher. MbTg had attenuated perfusion recovery at d21 (0.37±0.03 versus 0.47±0.02, P<0.05), d28 (0.40±0.03 versus 0.50±0.04, P<0.05), greater limb necrosis (65.2% versus 15%, P<0.001), a lower capillary density, and greater apoptosis versus WT. Conclusion—Increased Mb expression in myocytes attenuates angiogenesis after hind-limb ischemia by binding NO and reducing its bioavailability. Myoglobin can modulate the angiogenic response to ischemia even in the setting of normal endothelium.


Cancer Gene Therapy | 2012

Depleting regulatory T cells with arginine-rich, cell-penetrating, peptide-conjugated morpholino oligomer targeting FOXP3 inhibits regulatory T-cell function

Michael A. Morse; Amy Hobeika; Delila Serra; Katherine M. Aird; Matthew McKinney; Amy J. Aldrich; Clay Tm; D Mourich; Herbert Kim Lyerly; Patrick L. Iversen; Gayathri R. Devi

CD4+CD25+regulatory T cells (Treg) impair anti-tumor and anti-viral immunity. As there are higher Treg levels in cancer patients compared with healthy individuals, there is considerable interest in eliminating them or altering their function as part of cancer or viral immunotherapy strategies. The scurfin transcriptional regulator encoded by the member of the forkhead winged helix protein family (FOXP3) is critical for maintaining the functions of Treg. We hypothesized that targeting FOXP3 expression with a novel arginine-rich, cell-penetrating, peptide-conjugated phosphorodiamidate morpholino (PPMO) based antisense would eliminate Treg and enhance the induction of effector T-cell responses. We observed that the PPMO was taken up by activated T cells in vitro and could downregulate FOXP3 expression, which otherwise increases during antigen-specific T-cell activation. Generation of antigen-specific T cells in response to peptide stimulation was enhanced by pre-treatment of peripheral blood mononuclear cells with the FOXP3-targeted PPMO. In summary, modulation of Treg levels using the FOXP3 PPMO antisense-based genomic strategy has the potential to optimize immunotherapy strategies in cancer and viral immunotherapy.


Cancer Research | 2016

Abstract LB-015: XIAP induction by the MAPK-eIF4G1 pathway drives NFκB activation in inflammatory breast cancer growth and therapeutic resistance

Myron K. Evans; Joseph Geradts; Courtney M. Edwards; Arianna Price; Arjun J. Arora; Amy J. Aldrich; Adrian Ramirez; Timothy J. Robinson; Peter B. Vermeulen; Steven Van Laere; Gayathri R. Devi

Inflammatory breast cancer (IBC) is the most lethal, distinct form of breast cancer, however, the basis for its aggressiveness and rapid acquisition of drug resistance is not fully understood. Using immunohistochemical analysis, we identified a strong correlation between high grade, high stage, and triple-negative status and elevated expression of the X-linked inhibitor of apoptosis protein (XIAP) in IBC. Molecular profiling of multiple IBC cell lines revealed that modulating XIAP expression can significantly alter the expression pattern of a 79-gene, characteristic IBC profile that was previously obtained from clinical samples. Using specific antagonists and RNAi, we determined that the mitogen activated protein kinase (MAPK) pathway and its interaction with the protein synthesis initiation factor eIF4G1, both of which are elevated in IBC, cooperate to drive XIAP induction and resistance to therapeutic apoptosis. Further, we found that XIAP expression directly correlates with activation of the transcription factor NFκB, a molecular component defining IBC. Mutational analysis revealed that the BIR1 domain of XIAP is essential for subsequent TAB1:IKKβ-dependent NFκB activation. After determining this association between XIAP and TAB1, we tested a peptide-mediated strategy used to block the BIR1:TAB1 interaction antagonized NFκB activity which led to decreased anchorage independent growth and reversed resistance to an EGFR tyrosine kinase inhibitor. Most significantly, orthotopic implantation of XIAP silenced IBC cells revealed the necessity of expression for IBC tumor growth, while overexpression of XIAP enhanced tumor growth. Our findings establish that XIAP augments the malignant properties in IBC by enhancing NFκB function, identify a druggable pathway with multiple targets, and provide feasibility for the development of novel therapeutics targeting the BIR1 domain of XIAP in IBC. Grant Support: Supported by American Cancer Society, the Duke Cancer Institute as part of the P30 Cancer Center Support Grant NIH CA014236, the Duke Department of Surgery, and DOD grant W81XWH-13-1-0047. Citation Format: Myron K. Evans, Joseph Geradts, Courtney Edwards, Arianna Price, Arjun J. Arora, Amy J. Aldrich, Adrian Ramirez, Timothy J. Robinson, Peter B. Vermeulen, Steven van Laere, Gayathri R. Devi. XIAP induction by the MAPK-eIF4G1 pathway drives NFκB activation in inflammatory breast cancer growth and therapeutic resistance. [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 LB-015.


Cancer Research | 2015

Abstract P6-14-05: A novel link between anti-apoptotic signaling, NFκB, and SMAD7 in IBC pathobiology

Myron K. Evans; Scott J. Sauer; Amy J. Aldrich; Joseph Geradts; Peter B. Vermeulen; Luc Dirix; Steven Van Laere; Gayathri R. Devi

Background: Inflammatory breast cancer (IBC) has the highest lethality amongst all subtypes of breast cancer and develops rapid therapeutic resistance. High NFκB activation has been identified as a distinct molecular determinant in IBC pathobiology; however, the precise sequence of its activation and functional consequence in IBC remains unknown. Our previous work identified increased expression of the X-linked inhibitor of apoptosis protein (XIAP) due to altered translation in IBC, while other studies have noted a crosstalk between XIAP and NFκB. We hypothesized that XIAP drives NFκB activation in IBC promoting therapeutic resistance and tumorigenesis. Methods: NFκB phosphorylation, nuclear translocation, and target gene expression were evaluated in triple-negative SUM149 IBC cells with targeted overexpression or knockdown of XIAP. Using specific point mutants, we assessed the domain and mechanism of XIAP-mediated NFκB activation in IBC. We evaluated proliferation and viability in 2D and 3D culture of SUM149 cells treated with JSH-23, a small molecule inhibitor of NFκB nuclear translocation. We monitored the effects of XIAP overexpression or knockdown on in vivo tumorigenicity in IBC xenograft models by measuring tumor growth and NFκB signaling. IHC analysis of XIAP and NFκB was performed on tumor microarrays containing both non-IBC and IBC. Results: Knockdown of XIAP significantly decreased NFκB activation in IBC cells. Domain analysis revealed the necessity of the BIR1 domain of XIAP and TAB1:IKKβ complex formation in activating NFκB. NFκB antagonism inhibited proliferation of cells and sensitized therapy-resistant, XIAP overexpressing cells to targeted therapy. Loss of XIAP inhibited tumor growth of SUM149 tumor cells, correlating with decreased ALDH activity and varied epithelial-mesenchymal characteristics in these cells, while overexpression of XIAP significantly enhanced tumor growth of SUM149 cells. Further analysis revealed altered SMAD7 expression in XIAP knockdown cells, revealing crosstalk between XIAP, NFκB, and TGFβ signaling in IBC. IHC analysis of XIAP expression in invasive non-IBC tumors correlated with triple-negative status as well as increased grade and stage of tumors. In IBC tumors, XIAP expression associated with increased NFκB. Conclusions: In summary, our studies reveal that XIAP expression is necessary for NFκB activation in IBC and is critical for IBC development and progression. This study provides a novel insight into how an anti-apoptotic protein may regulate survival signaling and disease progression and may guide further research into innovative inhibitors of this interaction. Citation Format: Myron K Evans, Scott J Sauer, Amy J Aldrich, Joseph Geradts, Peter Vermeulen, Luc Dirix, Steven Van Laere, Gayathri R Devi. A novel link between anti-apoptotic signaling, NFκB, and SMAD7 in IBC pathobiology [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P6-14-05.

Collaboration


Dive into the Amy J. Aldrich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin P. Williams

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar

Jonathan Z. Sexton

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shalonda M. Ingram

North Carolina Central University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ginger R. Smith

North Carolina Central University

View shared research outputs
Researchain Logo
Decentralizing Knowledge