- Browse by Author
Browsing by Author "Fehrenbacher, Jill"
Now showing 1 - 10 of 10
Results Per Page
Sort Options
Item APE1/REF-1 redox signaling regulates HIF1A-mediated CA9 expression in hypoxic pancreatic cancer cells : combination treatment in patient-derived pancreatic tumor model(2017-12-14) Logsdon, Derek Paul; Kelly, Mark; Fishel, Melissa; Jerde, Travis; Vasko, Michael; Fehrenbacher, JillPancreatic ductal adenocarcinoma (PDAC) is an extremely deadly disease characterized by aggressive metastasis and therapeutic resistance. Reactive stroma in pancreatic tumors contributes to tumor signaling, fibrosis, inflammation, and hypoxia. Hypoxia signaling creates a more aggressive phenotype with increased potential for metastasis and decreased therapeutic efficacy. Carbonic anhydrase IX (CA9) functions as part of the cellular response to hypoxia by regulating intracellular pH to promote cell survival. Apurinic/Apyrimidinic Endonuclease-1-Reduction/oxidation Effector Factor 1 (APE1/Ref-1) is a multi-functional protein with two major activities: endonuclease activity in DNA base excision repair and a redox signaling activity that reduces oxidized transcription factors, enabling them to bind target sequences in DNA. APE1/Ref-1 is a central node in redox signaling, contributing to the activation of transcription factors involved in tumor survival, growth, and hypoxia signaling. This work evaluates the mechanisms underlying PDAC cell responses to hypoxia and APE1/Ref-1 redox signaling control of hypoxia inducible factor 1 alpha (HIF1a), a critical factor in hypoxia-induced CA9 transcription. We hypothesized that obstructing the HIF-CA9 axis at two points via APE1/Ref-1 inhibition and CA9 inhibition results in enhanced PDAC cell killing under hypoxic conditions. We found that HIF1a-mediated induction of CA9 is significantly attenuated following APE1/Ref-1 knock-down or redox signaling inhibition in patient-derived PDAC cells and pancreatic cancer-associated fibroblast cells. Additionally, dual-targeting of APE1/Ref-1 redox signaling activity and CA9 activity results in enhanced acidification and cytotoxicity of PDAC cells under hypoxic conditions as well as decreased tumor growth in an ex-vivo 3-dimensional tumor co-culture model. Further experiments characterized novel analogs of clinically relevant drugs targeting the key enzymes in this pathway, resulting in improved potency. These results underscore the notion that combination therapy is essential and demonstrate the potential clinical utility of blocking APE1/Ref-1 and CA9 function for novel PDAC therapeutic treatment.Item APE1/Ref-1 – One Target with Multiple Indications: Emerging Aspects and New Directions(Scientific Archives, 2021) Mijit, Mahmut; Caston, Rachel; Gampala, Silpa; Fishel, Melissa L.; Fehrenbacher, Jill; Kelley, Mark R.; Pediatrics, School of MedicineIn the realm of DNA repair, base excision repair (BER) protein, APE1/Ref-1 (Apurinic/Apyrimidinic Endonuclease 1/Redox Effector - 1, also called APE1) has been studied for decades. However, over the past decade, APE1 has been established as a key player in reduction-oxidation (redox) signaling. In the review by Caston et al. (The multifunctional APE1 DNA repair-redox signaling protein as a drug target in human disease), multiple roles of APE1 in cancer and other diseases are summarized. In this Review, we aim to expand on the contributions of APE1 to various diseases and its effect on disease progression. In the scope of cancer, more recent roles for APE1 have been identified in cancer cell metabolism, as well as chemotherapy-induced peripheral neuropathy (CIPN) and inflammation. Outside of cancer, APE1 signaling may be a critical factor in inflammatory bowel disease (IBD) and is also an emergent area of investigation in retinal ocular diseases. The ability of APE1 to regulate multiple transcription factors (TFs) and therefore multiple pathways that have implications outside of cancer, makes it a particularly unique and enticing target. We discuss APE1 redox inhibitors as a means of studying and potentially combating these diseases. Lastly, we examine the role of APE1 in RNA metabolism. Overall, this article builds on our previous review to elaborate on the roles and conceivable regulation of important pathways by APE1 in multiple diseases.Item Characterization of a Novel Hunk Inhibitor in HER2+ Breast Cancer(2024-07) Dilday, Tinslee Y.; Yeh, Elizabeth; Fehrenbacher, Jill; Brustovetsy, Nickolay; Safa, Ahmad; Sankar, UmaHuman Epidermal Growth Factor Receptor 2 (HER2)-targeted agents have proven to be effective, however, the development of resistance to these agents has become an obstacle in treating HER2+ breast cancer. Prior evidence implicates Hormonally Upregulated Neu-associated Kinase (HUNK) as an anti-cancer target for primary and resistant HER2+ breast cancers. An inhibitor Staurosporine (STU) has been identified as a HUNK inhibitor in HER2+ breast cancer. While STU was determined as a promising tool for inhibiting HUNK, it is a broad-spectrum kinase inhibitor and has not moved forward clinically. Therefore, identifying a more selective inhibitor of HUNK could be critical for targeting HUNK in HER2+ breast and understanding mechanisms by which HUNK promotes resistance to HER2-inhibitors. Specifically, HUNK has been implicated in promoting autophagy as a resistance mechanism in HER2+ breast cancer. Previously, we have identified that HUNK binds and phosphorylates an autophagy inhibitory protein, Rubicon, at Serine (S) 92 in 293T cells. This phosphorylation event causes Rubicon to switch to being an autophagy promoter. However, the role that Rubicon S92 plays in HER2+ breast cancer has yet to be examined. In this study, a novel inhibitor of HUNK is characterized alongside Rubicon S92 phosphorylation. This study establishes that HUNK-mediated phosphorylation of Rubicon at S92 promotes tumorigenesis in HER2/neu+ breast cancer. HUNK inhibition prevents S92 Rubicon phosphorylation in HER2/neu+ breast cancer models and inhibits both autophagy and tumorigenesis. This study characterizes a downstream phosphorylation event as a measure of HUNK activity and identifies a novel HUNK inhibitor that has meaningful efficacy toward HER2+ breast cancer.Item FASN Negatively Regulates NF-kB/P65 Expression in Breast Cancer Cells by Disrupting Its Stability(2020-02) Barlow, Lincoln James; Lu, Tao; Zhang, Jian-Ting; Fehrenbacher, Jill; Herbert, Brittney-Shea; Safa, AhmadThe overexpression of the multi-domain enzyme fatty acid synthase (FASN) has long been associated with poor clinical prognosis and treatment outcome in various cancers. Previous research in the Zhang lab has determined a role for FASN in mediating increases in non-homologous end-joining (NHEJ) DNA double-strand break repair activity allowing for increased cancer cell survival, and this mechanism was found to involve inhibition of NF-kB/p65. The mechanism responsible for the regulation of NF-kB/p65 by FASN in cancer cells, however, remains unknown. To this end, I was able to determine that FASN negatively regulates both the expression and activity of NF-kB/p65 in breast cancer cells, and that this effect was likely mediated by the 16-carbon saturated fatty acid palmitate, the end product of FASN catalytic activity. Specifically, FASN was found to negatively regulate p65 expression by disrupting its protein stability as a result of an increase in poly-ubiquitination of p65 protein and subsequent proteasomal degradation. Further, I found that the phosphorylation site Thr254 of p65 is involved in the regulation of p65 protein stability by FASN, in that mutation of this residue resulted in a disruption in p65 stability. Finally, I was able to determine that FASN likely inhibits the ability of the peptidyl-prolyl cis/trans isomerase Pin1 to assist in maintaining p65 stability, in that both siRNA knockdown and pharmacological inhibition of Pin1 resulted in a reduction of p65 expression in FASN shRNA knockdown cells. The determination of this signaling mechanism serves to expand our understanding of the role of FASN in breast cancer cells and has the potential to assist in uncovering more effective ways to target the oncogenic FASN pathway to kill breast tumor cells and to overcome resistance to drug treatment.Item Function of a Unique Dually Localized EF-Hand Domain Containing Protein, TgEFP1, During the Lytic Cycle of the Human Parasite Toxoplasma Gondii(2022-08) Dave, Noopur Kirti; Arrizabalaga, Gustavo; Absalon, Sabrina; Fehrenbacher, Jill; Gilk, Stacey; Jerde, Travis; Mastracci, TeresaThe pathogenesis associated with toxoplasmosis is attributed to repeated rounds of the parasite lytic cycle, which has been shown to be regulated by calcium fluxes. However, little is known about the calcium homeostatic mechanisms utilized by T. gondii. Recently, our lab has identified a novel protein-TgEFP1 (TGGT1_255660), which is predicted to bind Ca2+ through its two EF-hand domains. Interestingly, TgEFP1 showed a unique dual localization at the PLV/ELC and the PV of the parasite. Previous work showed that the PLV/ELC harbors other ion binding and conducting proteins that are important for parasite survival and propagation. However, the function of this compartment in the parasite is unknown. Therefore, I hypothesize that the PLV/ELC, through the function of TgEFP1, plays a key role in calcium homeostasis of T. gondii. To test this hypothesis, we sought to characterize the function of TgEFP1 during the parasite lytic cycle and determine TgEFP1 interacting proteins that also localize to the PLV/ELC. Partial permeabilization and ultrastructure expansion microscopy techniques confirmed the dual localization of TgEFP1 at the PLV/ELC and the PV. TgEFP1 knockout parasites exhibited several phenotypic defects including a faster lytic rate, shorter intracellular cycle, and were more sensitive to calcium ionophore treatment. Signal peptide deletion led to a mislocalization of TgEFP1 as cytosolic puncta, while mutations at key calcium coordinating residues lead to exclusive localization of TgEFP1 at the PV. Lastly, immunoprecipitation assays followed by LC-MS/MS identified a novel lectin-like protein- TgLectin (TGGT1_258950) as a direct interactor of TgEFP1-HA. Collectively, these findings support that through the function of TgEFP1, the PLV/ELC, plays a key role in calcium-dependent processes during the lytic cycle of the parasite.Item HUNK as an Immune Regulator of Triple Negative Breast Cancer(2024-05) Ramos Solis, Nicole; Yeh, Elizabeth; Arrizabalaga, Gustavo; Fehrenbacher, Jill; Cook-Mills, Joan; Jerde, Travis J.Triple-negative breast cancer (TNBC) is a subtype of breast cancer characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Unlike other breast cancer types, TNBC tumors do not respond to endocrine therapy, and standardized treatment protocols for TNBC are currently unavailable. TNBC is recognized as a more metastatic, aggressive, and immunogenic subtype of breast cancer, rendering it to be more receptive to immunotherapy. Among the immune cell populations abundant in TNBC tumors, tumor-associated macrophages (TAMs) are particularly more prevalent and are particularly known to play a role in cancer metastasis. This work focuses on and investigates the involvement of the protein kinase HUNK in tumor immunity. With the use of gene expression analysis, such as NanoString's nCounter PanCancer Immune Profiling panel, we found that targeting HUNK is associated with alterations in the IL-4/IL-4R cytokine signaling pathway. Experimental analysis and work demonstrated that HUNK kinase activity regulates IL-4 production in mammary tumor cells, and this regulation is dependent on STAT3. Furthermore, in vivo, analysis shows that HUNK-dependent control of IL-4 secretion from tumor cells leads to the polarization of macrophages into an M2-like phenotype, and consequently, IL-4 induction promotes cancer metastasis and prompts macrophage's metastatic capacities. These findings underscore HUNK as a potential therapeutic target for mitigating TNBC metastasis by modulating the TAM population.Item Investigation of the Knockout of LMF1 on the Transcriptome of Toxoplasma gondii(2024-01) Thibodeau, Katherine E.; Arrizabalaga, Gustavo; Absalon, Sabrina; Fehrenbacher, Jill; Flak, Jonathan; Schmidt, NathanToxoplasma gondii is an obligate intracellular apicomplexan parasite that infects one third of the global population. There are limited treatments for Toxoplasmosis, however a potential drug target for Toxoplasma is its mitochondrion. While much is known about the function of this organelle in Toxoplasma, little is known about the mechanisms that regulate mitochondrial structure and division. The shape of the mitochondrion changes throughout the life cycle of the parasite. When inside a host cell, the mitochondrion is in a lasso shape, stretching around the periphery of the parasite, while in extracellular parasites it is collapsed towards the apical end of the parasite. While in a lasso shape the mitochondrion shows areas of contact with the parasite pellicle. We have determined that the proteins LMF1 (associated with the outer mitochondrial membrane) and IMC10 (inner membrane complex) interact and form a reversible tether that maintains the lasso shape of the mitochondrion. When either of these proteins are knocked out, the mitochondrion collapses. To elucidate the biological relevance of the interaction between the mitochondrion and the pellicle we explored the consequence of disrupting the interaction on the transcriptome of the parasite. RNA sequencing of the LMF1 knockout strain showed a disruption in the expression of genes involved in nucleotide metabolism and Coenzyme A biosynthesis, which might be an adaptation mechanism to the disruption of mitochondrial morphology. Current work focuses on investigating the connection between mitochondrial tethering and these pathways as well as a potential role for the mitochondrion/pellicle connection in metabolite transport.Item The role of Protein Kinase C in the modulation of neuronal sensitivity and neurite morphology following treatment with paclitaxel in cultured sensory neurons(2017-07-13) Darby, Lisa Monique; Fehrenbacher, Jill; Cummins, Theodore; Jerde, Travis; Obukhov, Alexander; Oxford, GerryPaclitaxel is a chemotherapeutic drug that is used in the treatment of solid tumors including breast and ovarian cancer. However, a debilitating and severe side effect associated with paclitaxel treatment is peripheral neuropathy. Clinically, peripheral neuropathy is characterized by a gain and loss of sensory neuronal function with symptoms including burning pain, tingling and numbness, respectively. In addition, paclitaxel also elicits a reduction in the length of intraepidermal nerve fibers. Currently, there are no effective therapies to prevent or alleviate the symptoms of peripheral neuropathy. Further studies are needed to elucidate the mechanisms underlying the changes in neuronal function and neurite morphology induced by paclitaxel in order to develop therapeutics to address the symptoms of peripheral neuropathy. Our laboratory previously demonstrated that paclitaxel altered capsaicin stimulated release of calcitonin gene-related peptide (CGRP) from cultured sensory neurons, indicating that paclitaxel altered the function of the transient receptor potential vanilloid I (TRPV1) channels. Because protein kinase C (PKC) modulates the function of TRPV1, we questioned whether PKC mediated changes in neuronal sensitivity induced by chronic treatment with paclitaxel. We used the release of CGRP as an index of neuronal sensitivity. Our data show that paclitaxel decreased the activity and membrane localization of the conventional PKC isozymes, PKCα and PKCβI/II, to elicit a reduction in the release of CGRP from cultured sensory neurons. For our neurite morphology studies, we focused on the importance of the novel PKC isozyme, PKCε, in mediating the changes in neurite morphology induced by treatment with paclitaxel because studies have demonstrated that PKCε is important for enhancing neurite outgrowth. Since our preliminary data showed a correlative reduction in PKCε protein expression and neurite length and branching following treatment with paclitaxel, we questioned whether loss of PKCε mediated altered neurite morphology induced by paclitaxel. Unexpectedly, we found that downregulation of PKCε did not exacerbate the reduction in neurite length and branching induced by paclitaxel. Our work highlights the significance of PKCα and PKCβI/II as critical mediators of changes in neuronal sensitivity induced by paclitaxel and illuminates our understanding of the mechanisms underlying the neurotoxic effects of paclitaxel on sensory neuronal function.Item The Role of T Cells in Toxoplasma gondii-Induced Prostatic Hyperplasia(2024-08) Schmidt, Tara D.; Jerde, Travis; Arrizabalaga, Gustavo; Fehrenbacher, Jill; Relich, Ryan; Schmidt, NathanChronic inflammation is the most common histological feature in Benign Prostatic Hyperplasia (BPH), and T cells are a key component of immune infiltrate. Advanced BPH is commonly associated with the formation of nodules, but it remains unclear whether a link exists among T cell infiltration, nodular development, and BPH progression. Using a Toxoplasma gondii (T. gondii) model and human specimens, we characterize the subtypes of T cells present during prostatic hyperplasia and their association with nodular development of the prostate. Using flow cytometry, we found that infecting male mice with T. gondii resulted in an increase of both CD4+ and CD8+ T cells in the prostate that was most prominent at 14 days post-infection. Next, we established the presence of microglandular hyperplasia (MGH) and glandular nodule formation at this timepoint through hematoxylin and eosin (H&E) staining. Immunofluorescence revealed that CD8+ cells were found proximal to forming glandular nodules relative to non-nodular glands. We also found that more CD8+ cells localized to non-nodular glands in nodular BPH tissue versus non-nodular BPH tissue. Finally, we discovered a higher prevalence of CD8+ cells in T. gondii IgG+ patients than in IgG- patients. All T. gondii IgG+ patients exhibited nodular BPH, whereas all but one IgG- patient exhibited non-nodular BPH. This study is the first to identify the subsets of T cells in T. gondii-infected mouse prostates. Additionally, the locality of CD4+ and CD8+ T cells to nodular and non-nodular glands within our mouse model and human BPH prostate tissue has never been analyzed. Translationally, CD8+ T cells may enhance nodular BPH progression, and T. gondii infection may promote this CD8+ T cell-mediated response. Future work will focus on dissecting the molecular pathways induced by secreted factors from these CD8+ T cells that may contribute to epithelial cell proliferation and re-activation of glandular patterning in BPH.Item Therapeutic Targeting of BET Proteins in Osteosarcoma(2025-03) Riyahi, Niknam; Pollok, Karen; Angus, Steve; Fehrenbacher, Jill; Jerde, Travis; Yeh, ElizabethOsteosarcoma (OS) is an aggressive bone cancer in pediatric, adolescent, and young adult patients with an exceedingly poor prognosis. An area of therapeutic opportunity is that OS exhibits increased oncogenic replication stress (RS), augmenting genome instability and tumor progression. Exploiting RS provides a treatment intervention. Bromodomain and extra-terminal (BET) proteins regulate DNA transcription, replication, and repair. BET inhibitors create an imbalance between transcription and replication kinetics, exacerbating oncogenic RS and increasing cell death. We hypothesized that BET inhibition would decrease OS tumor growth via modulation of gene expression and increased RS. Bivalent BET inhibitor AZD5153 monotherapy significantly suppressed the growth of patient-derived xenografts (PDXs) established from treatment-naïve male and female OS patients. AZD5153-mediated anti-tumor effect correlated with increased γ-H2AX levels, indicative of elevated RS. RNA-sequencing and kinome profiling revealed intra- and inter-tumoral molecular response mechanisms attributable to therapeutic response and potential resistance in BET inhibitor-treated PDX and OS cell lines. Reprogramming of signaling networks involved in DNA damage response, apoptosis, invasion, and Wnt signaling were identified. Combining AZD5153 with salvage agents demonstrated additive-to-synergistic cell growth inhibition in vitro with drug combinations inducing apoptosis. AZD5153 increased DNA-strand breaks that was further enhanced by salvage therapy. In a PDX established from a metastatic lesion of a patient treated with frontline and salvage therapy, a combination of AZD5153 and topotecan was well tolerated and increased survival probability compared to each agent alone. These data suggest that AZD5153 alone or in combination with low-dose salvage therapy holds promise as an efficacious treatment strategy in OS.