Publications

Published in , 1900

Proteolysis-targeting chimera against BCL-XL destroys tumor-infiltrating regulatory T cells.

Published in Nature Communications, 2021

Regulatory T cells (Tregs) play an important role in maintaining immune homeostasis and, within tumors, their upregulation is common and promotes an immunosuppressive microenvironment. Therapeutic strategies that can eliminate Tregs in the tumor (i.e., therapies that do not run the risk of affecting normal tissues), are urgently needed for the development of cancer immunotherapies. Here we report our discovery of B-cell lymphoma extra-large (BCL-XL) as a potential molecular target of tumor-infiltrating (TI) Tregs. We show that pharmacological degradation of BCL-XL using a newly developed platelet-sparing BCL-XL Proteolysis-targeting chimera (PROTAC) induces the apoptosis of TI-Tregs and the activation of TI-CD8+ T cells. Moreover, these activities result in an effective suppression of syngeneic tumor growth in immunocompetent, but not in immunodeficient or CD8+ T cell-depleted mice. Notably, treatment with BCL-XL PROTAC does not cause detectable damage within several normal tissues or thrombocytopenia. These findings identify BCL-XL as a target in the elimination of TI-Tregs as a component of cancer immunotherapies, and that the BCL-XL-specific PROTAC has the potential to be developed as a therapeutic for cancer immunotherapy.

Recommended citation: Kolb, R., De, U., Khan, S., Luo, Y., Kim, M., Yu, H., Wu, C., Zhang, X., Zhang, X., Borcherding, N., Zheng, G., Zhou, D., & Zhang, W. Proteolysis-targeting chimera against BCL-XL destroys tumor-infiltrating regulatory T cells. Nature Communications 2020.

Understanding and Targeting Human Cancer Regulatory T Cells to Improve Therapy.

Published in T Regulatory Cells in Human Health and Diseases, 2021

Regulatory T cells (Tregs) are critical in maintaining immune homeostasis under various pathophysiological conditions. A growing body of evidence demonstrates that Tregs play an important role in cancer progression and that they do so by suppressing cancer-directed immune responses. Tregs have been targeted for destruction by exploiting antibodies against and small-molecule inhibitors of several molecules that are highly expressed in Tregs—including immune checkpoint molecules, chemokine receptors, and metabolites. To date, these strategies have had only limited antitumor efficacy, yet they have also created significant risk of autoimmunity because most of them do not differentiate Tregs in tumors from those in normal tissues. Currently, immune checkpoint inhibitor (ICI)-based cancer immunotherapies have revolutionized cancer treatment, but the resistance to ICI is common and the elevation of Tregs is one of the most important mechanisms. Therapeutic strategies that can selectively eliminate Tregs in the tumor (i.e. therapies that do not run the risk of causing autoimmunity by affecting normal tissue), are urgently needed for the development of cancer immunotherapies. This chapter discusses specific properties of human Tregs under the context of cancer and the various ways to target Treg for cancer immunotherapy.

Recommended citation: Kolb, R., Borcherding, N., & Zhang, W. Understanding and Targeting Human Cancer Regulatory T Cells to Improve Therapy. T Regulatory Cells in Human Health and Diseases, 2021.

Mapping the immune environment in clear cell renal carcinoma by single-cell genomics.

Published in Communications Biology, 2021

Clear cell renal cell carcinoma (ccRCC) is one of the most immunologically distinct tumor types due to high response rate to immunotherapies, despite low tumor mutational burden. To characterize the tumor immune microenvironment of ccRCC, we applied single-cell-RNA sequencing (SCRS) along with T-cell-receptor (TCR) sequencing to map the transcriptomic heterogeneity of 25,688 individual CD45+ lymphoid and myeloid cells in matched tumor and blood from three patients with ccRCC. We also included 11,367 immune cells from four other individuals derived from the kidney and peripheral blood to facilitate the identification and assessment of ccRCC-specific differences. There is an overall increase in CD8+ T-cell and macrophage populations in tumor-infiltrated immune cells compared to normal renal tissue. We further demonstrate the divergent cell transcriptional states for tumor-infiltrating CD8+ T cells and identify a MKI67+ proliferative subpopulation being a potential culprit for the progression of ccRCC. Using the SCRS gene expression, we found preferential prediction of clinical outcomes and pathological diseases by subcluster assignment. With further characterization and functional validation, our findings may reveal certain subpopulations of immune cells amenable to therapeutic intervention.

Recommended citation: Borcherding, N., Vishwakarma, A., Voigt, A.P., Nepple, K., Salem, A., Jenkins, R.W., Zakharia, Y., & Zhang, W. Mapping the immune environment in clear cell renal carcinoma by single-cell genomics. Communications Biology 2021

The anti-tumor effects of cetuximab in combination with VTX-2337 are T cell dependent

Published in Scientific Reports, 2021

The Toll-like receptor 8 (TLR8) agonist VTX-2337 (motolimod) is an anti-cancer immunotherapeutic agent that is believed to augment natural killer (NK) and dendritic cell (DC) activity. The goal of this work is to examine the role of TLR8 expression/activity in head and neck squamous cell carcinoma (HNSCC) to facilitate the prediction of responders to VTX-2337-based therapy. The prognostic role of TLR8 expression in HNSCC patients was assessed by TCGA and tissue microarray analyses. The anti-tumor effect of VTX-2337 was determined in SCCVII/C3H, mEERL/C57Bl/6 and TUBO-human EGFR/BALB/c syngeneic mouse models. The effect of combined VTX-2337 and cetuximab treatment on tumor growth, survival and immune cell recruitment was assessed. TLR8 expression was associated with CD8+ T cell infiltration and favorable survival outcomes. VTX-2337 delayed tumor growth in all 3 syngeneic mouse models and significantly increased the survival of cetuximab-treated mice. The anti-tumor effects of VTX-2337+ cetuximab were accompanied by increased splenic lymphoid DCs and IFNγ+ CD4+ and tumor-specific CD8+ T cells. Depletion of CD4+ T cells, CD8+ T cells and NK cells were all able to abolish the anti-tumor effect of VTX-2337+ cetuximab. Altogether, VTX-2337 remains promising as an adjuvant for cetuximab-based therapy however patients with high TLR8 expression may be more likely to derive benefit from this drug combination compared to patients with low TLR8 expression.

Recommended citation: Cheng, Y., Borcherding, N., Ogunsakin, A., Lemke-Miller, C.D., Gibson-Corley, K.N., Rajan, A., Wongpattaraworakul, W., Chan, C.H., Salem, A.K., Weiner, G.J., Simons, A.L. The anti-tumor effects of cetuximab in combination with motolimod are T cell dependent. Scientific Reports.

AP-2γ Is Required for Maintenance of Multipotent Mammary Stem Cells

Published in Stem Cell Reports, 2021

Mammary gland ductal morphogenesis depends on the differentiation of mammary stem cells (MaSCs) into basal and luminal lineages. The AP-2γ transcription factor, encoded by Tfap2c, has a central role in mammary gland development but its effect in mammary lineages and specifically MaSCs is largely unknown. Here, we utilized an inducible, conditional knockout of Tfap2c to elucidate the role of AP-2γ in maintenance and differentiation of MaSCs. Loss of AP-2γ in the basal epithelium profoundly altered the transcriptomes and decreased the number of cells within several clusters of mammary epithelial cells, including adult MaSCs and luminal progenitors. AP-2γ regulated the expression of genes known to be required for mammary development, including Cebpb, Nfkbia, and Rspo1. As a result, AP-2γ-deficient mice exhibited repressed mammary gland ductal outgrowth and inhibition of regenerative capacity. The findings demonstrate that AP-2γ can regulate development of mammary gland structures potentially regulating maintenance and differentiation of multipotent MaSCs.

Recommended citation: Gu, V.W., Cho, E., Thompson, D.T., Cassady, V., Borcherding, N. , Koch, K.E., Wu, V.T., Lorenzen, A.W., Kulak, M.V., Williams, T., Zhang, W., Weigel, R.J. AP-2γ is Required for Maintenance of Pluripotent Mammary Stem Cells. Stem Cell Reports.

Repurposing anti-cancer drugs for COVID-19

Published in Drug Design, Development and Therapy, 2020

The novel coronavirus disease 2019 (COVID-19) pandemic has caused catastrophic damage to human life across the globe along with social and financial hardships. According to the Johns Hopkins University Coronavirus Resource Center, more than 41.3 million people worldwide have been infected, and more than 1,133,000 people have died as of October 22, 2020. At present, there is no available vaccine and a scarcity of efficacious therapies. However, there is tremendous ongoing effort towards identifying effective drugs and developing novel vaccines. Early data from Adaptive COVID-19 Treatment Trials (ACTT) sponsored by the National Institute of Allergy and Infectious Diseases (NIAID) and compassionate use study have shown promise for remdesivir, leading to emergency authorization by the Food and Drug Administration (FDA) for treatment of hospitalized COVID-19 patients. However, several randomized studies have now shown no benefit or increased adverse events associated with remdesivir treatment. Drug development is a time-intensive process and requires extensive safety and efficacy evaluations. In contrast, drug repurposing is a time-saving and cost-effective drug discovery strategy geared towards using existing drugs instead of de novo drug discovery. Treatments for cancer and COVID-19 often have similar goals of controlling inflammation, inhibiting cell division, and modulating the host microenvironment to control the disease. In this review, we focus on anti-cancer drugs that can potentially be repurposed for COVID-19 and are currently being tested in clinical trials.

Recommended citation: Borcherding, N., Jethava, Y., Vikas, P. (2020). Repurposing anti-cancer drugs for COVID-19. Drug Design, Development and Therapy 2020.

IL-12-Induced Immune Suppressive Deficit During CD8+ T-Cell Differentiation

Published in Frontiers in Immunology, 2020

Autoimmune diseases are characterized by regulatory deficit in both the CD4+ and CD8+ T-cell compartments. We have shown that CD8+ T-cells associated with acute relapse of multiple sclerosis are significantly deficient in their immune suppressive ability. We hypothesized that distinct CD8+ cytotoxic T-cell (Tc) lineages, determined by cytokine milieu during naïve T-cell differentiation, may harbor differential ability to suppress effector CD4+ T-cells. We differentiated purified human naïve CD8+ T-cells in vitro toward Tc0 (media control), Tc1 and Tc17 lineages. Using in vitro flow cytometric suppression assays, we observed that Tc0 and Tc17 cells had similar suppressive ability. In contrast, Tc1 cells showed significant loss of suppressive ability against ex vivo CD4+ T-cells and in vitro-differentiated Th0, Th1 and Th17 cells. Of note, Tc1 cells were also suboptimal in suppressing CD4-induced acute xenogeneic graft versus host disease (xGVHD) in vivo. Tc subtypes derived under various cytokine combinations revealed that IL-12-containing conditions resulted in less suppressive cells exhibiting dysregulated cytotoxic degranulation. RNA sequencing transcriptome analyses indicated differential regulation of inflammatory genes and enrichment in GM-CSF-associated pathways. These studies provide insights into the role of T-cell differentiation in CD8 suppressive biology and may reveal therapeutically targetable pathways to reverse suppressive deficit during immune-mediated disease.

Recommended citation: Renavikar, P.S., Sinha, S., Brate, A.A., Borcherding, N., Crawford, M.P., Steward-Tharp, S., & Karandikar, N. IL-12-mediated induction of immune suppressive deficit during CD8+ differentiation. Frontiers in Immunology 2020.

Multivariable Analysis of 169 Cases of Advanced Cutaneous Melanoma to Evaluate Antibiotic Exposure as Predictor of Survival to Anti-PD-1 Based Immunotherapies

Published in Antibiotics, 2020

Recently antibiotic exposure has been associated with worse outcomes in patients undergoing treatment with antibodies directed against programmed cell death protein-1 (PD-1). We reviewed data of 1264 patients enrolled at Melanoma Skin and Ocular Tissue Repositories at University of Iowa Hospitals and Clinic. Reviewed data included patient demographics, prior medical history, baseline hematologic and disease parameters and outcomes including progression-free survival (PFS) and overall survival (OS). Cox regression models were used to determine predictive markers. Overall, 169 patients with advanced cutaneous melanoma received anti-PD-1 based therapies. Median follow up was 18.46 (range 0.89 to 62.52) months. On multivariable analysis brain metastasis, higher absolute neutrophil count (ANC) and lower absolute lymphocyte count were associated with poorer PFS while brain and liver metastasis and lower albumin were associated with poorer OS. Prior antibiotics, radiation as well as age, gender, basal metabolic index (BMI), smoking status, BRAF mutation, line of therapy (first or latter), prior treatments (ipilimumab or BRAF inhibitors), hemoglobin, neutrophil-to-lymphocyte ratio, white blood cell, platelet and eosinophil counts were not associated with PFS or OS in multivariable analysis. Contrary to some prior studies BMI, radiation, and antibiotics were not associated with PFS or OS.

Recommended citation: Swami, U., Chennamadhavuni, A., Borcherding, N., Bossler, A.D., Mott, S.L., Garje, R., Zakharia, Y., & Milhem, M.M. Multivariate analysis of 169 cases of advanced cutaneous melanoma to evaluate antibiotic exposure as a predictor of survival to anti-PD-1 based immunotherapies. Antibiotics

CD4 T cell-intrinsic role for the T helper 17 signature cytokine IL-17: Effector resistance to immune suppression.

Published in PNAS, 2020

Untoward effector CD4+ T cell responses are kept in check by immune regulatory mechanisms mediated by CD4+ and CD8+ T cells. CD4+ T helper 17 (Th17) cells, characterized by IL-17 production, play important roles in the pathogenesis of autoimmune diseases (such as arthritis, multiple sclerosis, psoriasis, inflammatory bowel disease, among others) and in the host response to infection and cancer. Here, we demonstrate that human CD4+ T cells cells exposed to a Th17-differentiating milieu are significantly more resistant to immune suppression by CD8+ T cells compared to control Th0 cells. This resistance is mediated, in part, through the action of IL-17A, IL-17F, and IL-17AF heterodimer through their receptors (IL-17RA and IL-17RC) on CD4+ T cells themselves, but not through their action on CD8+ T cells or APC. We further show that IL-17 can directly act on non-Th17 effector CD4+ T cells to induce suppressive resistance, and this resistance can be reversed by blockade of IL-1β, IL-6, or STAT3. These studies reveal a role for IL-17 cytokines in mediating CD4-intrinsic immune resistance. The pathways induced in this process may serve as a critical target for future investigation and immunotherapeutic intervention.

Recommended citation: Crawford, M.P., Sinha, S., Renavikar, P.S., Borcherding, N., & Karandikar, N. Novel role for the T-helper 17 signature cytokine IL-17: CD4 resistance to immune suppression. PNAS 2020.

A transcriptomic map of murine and human alopecia areata.

Published in JCI Insights, 2020

Alopecia areata (AA) is one of the most common autoimmune conditions, presenting initially with loss of hair without other overt skin changes. The unremarkable appearance of the skin surface contrasts with the complex immune activity occurring at the hair follicle. AA pathogenesis is due to the loss of immune privilege of the hair follicle leading to autoimmune attack. Although the literature has focused on CD8+ T cells, vital roles for CD4+ T cells and antigen-presenting cells have been suggested. Here, we use single-cell sequencing to reveal distinct expression profiles of immune cells in murine AA. We found clonal expansions of both CD4+ and CD8+ T cells, with shared clonotypes across varied transcriptional states. The murine AA data were used to generate highly predictive models of human AA disease. Finally, single-cell sequencing of T cells in human AA recapitulated the clonotypic findings and the gene expression of the predictive models.

Recommended citation: Borcherding, N., Crotts, S.B., Ortolan, L.S., Henderson, N., Bormann, N.L., & Jabbari, A. A transcriptomic map of murine and human alopecia areata. JCI Insight 2020.

IL-1 Signaling in Tumor Microenvironment.

Published in Tumor Microenvironment, 2020

Interleukin 1 (IL-1) has long been known for its pleiotropic effects on inflammation that plays a complex, and sometimes contrasting, role in different stages of cancer development. As a major proinflammatory cytokine, IL-1β is mainly expressed by innate immune cells. IL-1α, however, is expressed by various cell types under physiological and pathological conditions. IL-1R1 is the main receptor for both ligands and is expressed by various cell types, including innate and adaptive immune cell types, epithelial cells, endothelial cells, adipocytes, chondrocytes, fibroblasts, etc. IL-1 and IL-1R1 receptor interaction leads to a set of common signaling pathways, mainly the NF-kB and MAP kinase pathways, as a result of complex positive and negative regulations. The variety of cell types with IL-1R1 expression dictates the role of IL-1 signaling at different stages of cancer, which under certain circumstances leads to contrasting roles in tumor development. Recent availability of IL-1R1 conditional knockout mouse model has made it possible to dissect the role of IL-1/IL-1R1 signaling transduction in different cell types within the tumor microenvironment. This chapter will focus on the role of IL-1/IL-1R1 in different cell types within the tumor microenvironment and discuss the potential of targeting this pathway in cancer therapy.

Recommended citation: Zhang, W., Borcherding, N., & Kolb, R. IL-1 Signaling in Tumor Microenvironment. Tumor Microenvironment 2020.

Cancer cell-intrinsic function of CD177 in attenuating β-catenin signaling.

Published in Oncogene, 2020

Aiming to identify immune molecules with a novel function in cancer pathogenesis, we found the cluster of differentiation 177 (CD177), a known neutrophil antigen, to be positively correlated with relapse-free, metastasis-free, or overall survival in breast cancer. In addition, CD177 expression is correlated with good prognosis in several other solid cancers including prostate, cervical, and lung. Focusing on breast cancer, we found that CD177 is expressed in normal breast epithelial cells and is significantly reduced in invasive cancers. Loss of CD177 leads to hyperproliferative mammary epithelium and contributes to breast cancer pathogenesis. Mechanistically, we found that CD177-deficiency is associated with an increase in β-catenin signaling. Here we identified CD177 as a novel regulator of mammary epithelial proliferation and breast cancer pathogenesis likely via the modulation of Wnt/β-catenin signaling pathway, a key signaling pathway involved in multiple cancer types.

Recommended citation: Kluz, P.N., Kolb, R., Xie, Q., Borcherding, N., Liu, Q., Wang, L., Zhang, Y., Li, W., Stipp, C., Gibson-Corley, K.N., Zhao, C., Qi, H.H., Bellizzi, A., Tao, A.W., Sugg, S., Weigel, R.J., Shen, X., Zhang, W. Cancer cell-intrinsic function of CD177 in attenuating β-Catenin signaling. Oncogene 2020.

scRepertoire: An R-based toolkit for single-cell immune receptor analysis.

Published in F1000Research, 2020

Single-cell sequencing is an emerging technology in the field of immunology and oncology that allows researchers to couple RNA quantification and other modalities, like immune cell receptor profiling at the level of an individual cell. A number of workflows and software packages have been created to process and analyze single-cell transcriptomic data. These packages allow users to take the vast dimensionality of the data generated in single-cell-based experiments and distill the data into novel insights. Unlike the transcriptomic field, there is a lack of options for software that allow for single-cell immune receptor profiling. Enabling users to easily combine mRNA and immune profiling, scRepertoire was built to process data derived from 10x Genomics Chromium Immune Profiling for both T-cell receptor (TCR) and immunoglobulin (Ig) enrichment workflows and subsequently interacts with the popular Seurat R package. The scRepertoire R package and processed data are open source and available on GitHub and provides in-depth tutorials on the capability of the package.

Recommended citation: Borcherding, N. & Bormann, N. scRepertoire: A R-based toolkit for single-cell immune receptor analysis. F1000Research 2020.

Coactivation of NF-κB and Notch signaling is sufficient to induce B-cell transformation and enables B-myeloid conversion.

Published in Blood, 2020

NF-κB and Notch signaling can be simultaneously activated in a variety of B-cell lymphomas. Patients with B-cell lymphoma occasionally develop clonally related myeloid tumors with poor prognosis. Whether concurrent activation of both pathways is sufficient to induce B-cell transformation and whether the signaling initiates B-myeloid conversion in a pathological context are largely unknown. Here, we provide genetic evidence that concurrent activation of NF-κB and Notch signaling in committed B cells is sufficient to induce B-cell lymphomatous transformation and primes common progenitor cells to convert to myeloid lineage through dedifferentiation, not transdifferentiation. Intriguingly, the converted myeloid cells can further transform, albeit at low frequency, into myeloid leukemia. Mechanistically, coactivation of NF-κB and Notch signaling endows committed B cells with the ability to self renew. Downregulation of BACH2, a lymphoma and myeloid gene suppressor, but not upregulation of CEBPα and/or downregulation of B-cell transcription factors, is an early event in both B-cell transformation and myeloid conversion. Interestingly, a DNA hypomethylating drug not only effectively eliminated the converted myeloid leukemia cells, but also restored the expression of green fluorescent protein, which had been lost in converted myeloid leukemia cells. Collectively, our results suggest that targeting NF-κB and Notch signaling will not only improve lymphoma treatment, but also prevent the lymphoma-to-myeloid tumor conversion. Importantly, DNA hypomethylating drugs might efficiently treat these converted myeloid neoplasms.

Recommended citation: Xiu, Y., Qianze Dong, Q., Fu, L., Bossler, A., Tang, X., Boyce, B., Borcherding, N., Byrd, J., Leidinger, M., Sardina, J.L., Xue, H., Li, Q., Feldman, A., Aifantis, I., Boccalatte, F., Khoury, J., Wang, W., Hu, S., Yuan, Y., Wang, E., Yuan, J., Colgan, J., Habelhah, H., Waldschmidt, T., Muschen, M., & Zhao, C. Concurrent activation of NF-κB and Notch signaling is sufficient to induce B cell transformation and enables B-myeloid conversion. Blood 2020.

A TFAP2C Gene Signature Is Predictive of Outcome in HER2-Positive Breast Cancer.

Published in Molecular Cancer Research, 2020

The AP-2γ transcription factor, encoded by the TFAP2C gene, regulates the expression of estrogen receptor-alpha (ERα) and other genes associated with hormone response in luminal breast cancer. Little is known about the role of AP-2γ in other breast cancer subtypes. A subset of HER2+ breast cancers with amplification of the TFAP2C gene locus becomes addicted to AP-2γ. Herein, we sought to define AP-2γ gene targets in HER2+ breast cancer and identify genes accounting for physiologic effects of growth and invasiveness regulated by AP-2γ. Comparing HER2+ cell lines that demonstrated differential response to growth and invasiveness with knockdown of TFAP2C, we identified a set of 68 differentially expressed target genes. CDH5 and CDKN1A were among the genes differentially regulated by AP-2γ and that contributed to growth and invasiveness. Pathway analysis implicated the MAPK13/p38δ and retinoic acid regulatory nodes, which were confirmed to display divergent responses in different HER2+ cancer lines. To confirm the clinical relevance of the genes identified, the AP-2γ gene signature was found to be highly predictive of outcome in patients with HER2+ breast cancer. We conclude that AP-2γ regulates a set of genes in HER2+ breast cancer that drive cancer growth and invasiveness. The AP-2γ gene signature predicts outcome of patients with HER2+ breast cancer and pathway analysis predicts that subsets of patients will respond to drugs that target the MAPK or retinoic acid pathways.

Recommended citation: Wu, V., Kiriazov, B., Koch, K., Gu, V., Beck A., Borcherding, N., Li, T., Addo, P., Wehrspan, Z., Zhang, W., Braun, T., Brown, B., Band, V., Band, B., Kulak, M., & Weigel, R. A TFAP2C Gene Signature is Predictive of Outcome in HER2-positive Breast Cancer. Molecular Cancer Research 2020.

Contribution of synergism between PHF8 and HER2 signalling to breast cancer development and drug resistance.

Published in EBioMedicine, 2020

Background HER2 plays a critical role in tumourigenesis and is associated with poor prognosis of patients with HER2-positive breast cancers. Although anti-HER2 drugs are beneficial for treating breast cancer, de novo, or acquired resistance often develops. Epigenetic factors are increasingly targeted for therapy; however, such mechanisms that interact with HER2 signalling are poorly understood.

Recommended citation: Liu, Q., Borcherding, N., Shao, P., Maina, P., Zhang, W., & Qi, H. Contribution of synergism between PHF8 and HER2 signaling to breast cancer development and drug resistance. EBioMedicine 2020.

Identification of novel TGF-β regulated genes with pro-migratory roles.

Published in Molecular Basis of Disease, 2019

Transforming growth factor-β (TGF-β) signaling plays fundamental roles in the development and homeostasis of somatic cells. Dysregulated TGF-β signaling contributes to cancer progression and relapse to therapies by inducing epithelial-to-mesenchymal transition (EMT), enriching cancer stem cells, and promoting immunosuppression. Although many TGF-β-regulated genes have been identified, only a few datasets were obtained by next-generation sequencing. In this study, we performed RNA-sequencing analysis of MCF10A cells and identified 1166 genes that were upregulated and 861 genes that were downregulated by TGF-β. Gene set enrichment analysis revealed that focal adhesion and metabolic pathways were the top enriched pathways of the up- and downregulated genes, respectively. Genes in these pathways also possess significant predictive value for renal cancers. Moreover, we confirmed that TGF-β induced expression of MICAL1 and 2, and the histone demethylase, KDM7A, and revealed their regulatory roles on TGF-β-induced cell migration. We also show a critical effect of KDM7A in regulating the acetylation of H3K27 on TGF-β-induced genes. In sum, this study identified novel effectors that mediate the pro-migratory role of TGF-β signaling, paving the way for future studies that investigate the function of MICAL family members in cancer and the novel epigenetic mechanisms downstream TGF-β signaling.

Recommended citation: Liu, Q., Borcherding, N., Shao, P., Cao, H., Zhang, W., & Qi, H. Novel effectors mediating the pro-migratory role of TGF-beta signaling. Molecular Basis of Disease 2019.

Metadherin enhances vulnerability of cancer cells to ferroptosis.

Published in Cell Death and Disease, 2019

Ferroptosis is an iron-dependent, non-apoptotic form of regulated cell death driven by lipid hydroperoxides within biological membranes. Although therapy-resistant mesenchymal-high cancers are particularly vulnerable to ferroptosis inducers, especially phospholipid glutathione peroxidase 4 (GPx4) inhibitors, the underlying mechanism is yet to be deciphered. As such, the full application of GPx4 inhibitors in cancer therapy remains challenging. Here we demonstrate that metadherin (MTDH) confers a therapy-resistant mesenchymal-high cell state and enhanced sensitivity to inducers of ferroptosis. Mechanistically, MTDH inhibited GPx4, as well as the solute carrier family 3 member 2 (SLC3A2, a system Xc− heterodimerization partner), at both the messenger RNA and protein levels. Our metabolomic studies demonstrated that MTDH reduced intracellular cysteine, but increased glutamate levels, ultimately decreasing levels of glutathione and setting the stage for increased vulnerability to ferroptosis. Finally, we observed an enhanced antitumor effect when we combined various ferroptosis inducers both in vitro and in vivo; the level of MTDH correlated with the ferroptotic effect. We have demonstrated for the first time that MTDH enhances the vulnerability of cancer cells to ferroptosis and may serve as a therapeutic biomarker for future ferroptosis-centered cancer therapy.

Recommended citation: Bi, J., Yang, S., Li, L., Dai, Q., Borcherding, N., Wagner, B.A., Buettner, G.R., Spitz, D.R., Leslie, K.K., Zhang, J., & Meng, X. Metadherin (MTDH) Enhances Vulnerability of Cancer Cells to Ferroptosis. Cell Death and Disease 2019.

ROR1 Potentiates FGFR Signaling in Basal-Like Breast Cancer.

Published in Cancers, 2019

Among all breast cancer types, basal-like breast cancer (BLBC) represents an aggressive subtype that lacks targeted therapy. We and others have found that receptor tyrosine kinase-like orphan receptor 1 (ROR1) is overexpressed in BLBC and other types of cancer and that ROR1 is significantly correlated with patient prognosis. In addition, using primary patient-derived xenografts (PDXs) and ROR1-knockout BLBC cells, we found that ROR1+ cells form tumors in immunodeficient mice. We developed an anti-ROR1 immunotoxin and found that targeting ROR1 significantly kills ROR1+ cancer cells and slows down tumor growth in ROR1+ xenografts. Our bioinformatics analysis revealed that ROR1 expression is commonly associated with the activation of FGFR-mediated signaling pathway. Further biochemical analysis confirmed that ROR1 stabilized FGFR expression at the posttranslational level by preventing its degradation. CRISPR/Cas9-mediated ROR1 knockout significantly reduced cancer cell invasion at cellular levels by lowering FGFR protein and consequent inactivation of AKT. Our results identified a novel signaling regulation from ROR1 to FGFR and further confirm that ROR1 is a potential therapeutic target for ROR1+ BLBC cells.

Recommended citation: Pandey, G., Borcherding, N., Kolb, R., Kluz, P., Li, W., Sugg, S., Zhang, J., Lai, D., & Zhang W. ROR1 potentiates FGFR signaling in basal-like breast cancer. Cancers 2019.

Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression.

Published in Clinical Cancer Research, 2019

Purpose: Cutaneous T-cell lymphomas (CTCL), encompassing a spectrum of T-cell lymphoproliferative disorders involving the skin, have collectively increased in incidence over the last 40 years. Sézary syndrome is an aggressive form of CTCL characterized by significant presence of malignant cells in both the blood and skin. The guarded prognosis for Sézary syndrome reflects a lack of reliably effective therapy, due, in part, to an incomplete understanding of disease pathogenesis.

Recommended citation: Borcherding, N., Voigt, A., Liu, V., Link, B.K., Zhang, W., & Jabbari, A. Single-cell profiling of cutaneous T-cell lymphoma reveals underlying heterogeneity associated with disease progression. Clinical Cancer Research 2019.

Application of flipped classroom pedagogy to the human gross anatomy laboratory: Student preferences and learning outcomes.

Published in Anatomical Science Education, 2018

To improve student preparedness for anatomy laboratory dissection, the dental gross anatomy laboratory was transformed using flipped classroom pedagogy. Instead of spending class time explaining the procedures and anatomical structures for each laboratory, students were provided online materials to prepare for laboratory on their own. Eliminating in‐class preparation provided the opportunity to end each period with integrative group activities that connected laboratory and lecture material and explored clinical correlations. Materials provided for prelaboratory preparation included: custom‐made, three‐dimensional (3D) anatomy videos, abbreviated dissection instructions, key atlas figures, and dissection videos. Data from three years of the course (n = 241 students) allowed for analysis of students’ preferences for these materials and detailed tracking of usage of 3D anatomy videos. Students reported spending an average of 27:22 (±17:56) minutes preparing for laboratory, similar to the 30 minutes previously allocated for in‐class dissection preparation. The 3D anatomy videos and key atlas figures were rated the most helpful resources. Scores on laboratory examinations were compared for the three years before the curriculum change (2011–2013; n = 242) and three years after (2014–2016; n = 241). There was no change in average grades on the first and second laboratory examinations. However, on the final semi‐cumulative laboratory examination, scores were significantly higher in the post‐flip classes (P = 0.04). These results demonstrate an effective model for applying flipped classroom pedagogy to the gross anatomy laboratory and illustrate a meaningful role for 3D anatomy visualizations in a dissection‐based course.

Recommended citation: Fleagle, T., Borcherding, N., & Hoffmann, D. A series of Custom 3D Anatomy Videos to Improve Preparation for Gross Anatomy Lab Dissection. Anatomical Science Education 2018.

The clinical promise of immunotherapy in triple-negative breast cancer.

Published in Cancer management and research, 2018

Triple-negative breast cancer (TNBC) is a heterogeneous disease with poorer outcomes compared to other breast cancer subtypes. Contributing to the worse prognosis in TNBC is the higher rates of relapse and rapid progression after relapse. Advances in targeted therapeutics and conventional chemotherapy for TNBC have been stymied due to the lack of specific targets. Moreover, the responses to chemotherapy in TNBC lack durability, partially accounting for the higher rates of relapse. Immunotherapy, notably immune-checkpoint blockade, has shown to improve survival and maintain robust antitumor responses in both hematologic and solid malignancies. Unlike lung cancer, melanoma, and bladder cancer, most breast cancers are not inherently immunogenic and typically have low T cell infiltration. However, among breast cancer subtypes, TNBC is characterized by greater tumor immune infiltrate and higher degree of stromal and intratumoral tumor-infiltrating lymphocytes (TILs), a predictive marker for responses to immunotherapy. Moreover, in TNBC, the high number of stromal TILs is predictive of more favorable survival outcomes and response to chemotherapy. Immunotherapy is being extensively explored in TNBC and clinical trials are showing some promising results. This article focuses on the rationale for immunotherapy in TNBC, to explore and discuss preclinical data, results from early clinical trials, and to summarize some ongoing trials. We will also discuss the potential application of immunotherapy in TNBC from a clinician’s perspective.

Recommended citation: Vikas, P., Borcherding, N., & Zhang, W. The Clinical Promise of Immunotherapy in Triple-Negative Breast Cancer. Clinical Management and Research 2018.

Obesity-associated inflammation promotes angiogenesis and breast cancer via angiopoietin-like 4.

Published in Oncogene, 2018

Interleukin 1 (IL-1) has long been known for its pleiotropic effects on inflammation that plays a complex, and sometimes contrasting, role in different stages of cancer development. As a major proinflammatory cytokine, IL-1β is mainly expressed by innate immune cells. IL-1α, however, is expressed by various cell types under physiological and pathological conditions. IL-1R1 is the main receptor for both ligands and is expressed by various cell types, including innate and adaptive immune cell types, epithelial cells, endothelial cells, adipocytes, chondrocytes, fibroblasts, etc. IL-1 and IL-1R1 receptor interaction leads to a set of common signaling pathways, mainly the NF-kB and MAP kinase pathways, as a result of complex positive and negative regulations. The variety of cell types with IL-1R1 expression dictates the role of IL-1 signaling at different stages of cancer, which under certain circumstances leads to contrasting roles in tumor development. Recent availability of IL-1R1 conditional knockout mouse model has made it possible to dissect the role of IL-1/IL-1R1 signaling transduction in different cell types within the tumor microenvironment. This chapter will focus on the role of IL-1/IL-1R1 in different cell types within the tumor microenvironment and discuss the potential of targeting this pathway in cancer therapy.

Recommended citation: Kolb, R., Kluz, P., Wei, T.Z., Borcherding, N., Bormann, N., Balcziak, L., Zhu, P., Davies, B., Gourronc, F., Gibson-Corley, K. Klingelhutz, A., Tan, N.S., Sutterwala, F., Vishwakarma, A., Liu, L., Ge, X., Jiang, B., Zhu, Y., Shen, X., & Zhang, W. Obesity-associated inflammation promotes angiogenesis and breast cancer via angiopoietin-like 4. Oncogene 2018.

TRGAted: A web tool for survival analysis using protein data in the Cancer Genome Atlas.

Published in F1000Research, 2018

Reverse-phase protein arrays (RPPAs) are a highthroughput approach to protein quantification utilizing an antibody-based micro-to-nano scale dot blot. Within the Cancer Genome Atlas (TCGA), RPPAs were used to quantify over 200 proteins in 8,167 tumor or metastatic samples. This protein-level data has particular advantages in assessing putative prognostic or therapeutic targets in tumors. However, many of the available pipelines do not allow for the partitioning of clinical and RPPA information to make meaningful conclusions. We developed a cloud-based application, TRGAted to enable researchers to better examine survival based on single or multiple proteins across 31 cancer types in the TCGA. TRGAted contains up-to-date overall survival, disease-specific survival, disease-free interval and progression-free interval information. Furthermore, survival information for primary tumor samples can be stratified based on gender, age, tumor stage, histological type, and subtype, allowing for highly adaptive and intuitive user experience. The code and processed data is open sourced and available on github and with a tutorial built into the application for assisting users.

Recommended citation: Borcherding, N., Bormann, N., Voigt, A., & Zhang, W. TRGAted: A web tool for survival analysis using protein data in the Cancer Genome Atlas. F1000Research 2018.

Re-Evaluating E-Cadherin and β-Catenin: A Pan-Cancer Proteomic Approach with an Emphasis on Breast Cancer.

Published in The American Journal of Pathology, 2018

E-cadherin is conventionally considered to be a good prognostic marker in cancer. The loss of E-cadherin is one of the key hallmarks of epithelial-to-mesenchymal transition, a biological process that promotes cancer cell invasiveness and metastasis. Recent evidence has cast doubt on the importance of epithelial-to-mesenchymal transition in metastasis. The availability of protein-level data in the Cancer Genome Atlas allows for the quantitative analysis of protein and prognosis. The prognostic values of E-cadherin and β-catenin were revisited across 19 cancer types, and high E-cadherin was found to correlate with good prognosis in most cancers. Conversely, higher E-cadherin and β-catenin correlated with shorter survival in invasive breast carcinoma. Stratifying breast cancers by histologic subtype revealed that the poor prognosis of E-cadherin and β-catenin proteins was characteristic of infiltrating ductal, but not lobular, carcinomas. To further corroborate the protein findings and examine cellular localization, immunohistochemistry was used for E-cadherin and β-catenin in 163 breast patient samples from the Iowa cohort. Most previous studies showing that reduced or absent E-cadherin and β-catenin was inversely associated with tumor stages in ductal carcinomas were confirmed. Taken together, these results lead us to question the prognostic values of E-cadherin and β-catenin in ductal carcinomas and indicate a complicated role of E-cadherin and β-catenin in breast cancer progression.

Recommended citation: Borcherding, N., Cole, K., Kluz, P., Pandey, G., Jorgensen, M., Kolb, R., Bellizzi, A., & Zhang, W. Reevaluating E-Cadherin and β-Catenin: A pan-cancer proteomic approach with emphasis on breast cancer. American Journal of Pathology 2018.

Keeping Tumors in Check: A Mechanistic Review of Clinical Response and Resistance to Immune Checkpoint Blockade in Cancer.

Published in Journal of Molecular Biology, 2018

Immune checkpoints are a diverse set of inhibitory signals to the immune system that play a functional role in adaptive immune response and self-tolerance. Dysregulation of these pathways is a vital mechanism in the avoidance of immune destruction by tumor cells. Immune checkpoint blockade (ICB) refers to targeted strategies to disrupt the tumor co-opted immune suppression to enhance anti-tumor immunity. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) are two immune checkpoints that have the widest range of antibody-based therapies. These therapies have gone from promising approaches to Food and Drug Administration-approved first- and second-line agents for a number of immunogenic cancers. The burgeoning investigations of ICB efficacy in blood and solid cancers have underscored the importance of identifying the predictors of response and resistance to ICB. Identification of response correlates is made complicated by the observations of mixed reactions, or different responses in multiple lesions from the same patient, and delayed responses that can occur over a year after the induction therapy. Factors that can influence response and resistance in ICB can illuminate underlying molecular mechanisms of immune activation and suppression. These same response predictors can guide the identification of patients who would benefit from ICB, reduce off-target immune-relate adverse events, and facilitate the use of combinatorial therapies to increase efficacy. Here we review the underlying principles of immune checkpoint therapy and results of single-agent ICB clinical trials, and summarize the predictors of response and resistance.

Recommended citation: Borcherding, N., Kolb, R., Gullicksrud, J., Vikas, P. Zhu, Y., & Zhang, W. Keeping tumors in check: A mechanistic review of clinical response and resistance to immune checkpoint blockade in cancer. Journal of Molecular Biology 2018.

Stabilization of NF-κB-inducing kinase suppresses MLL-AF9-induced acute myeloid leukemia.

Published in Cell Reports, 2018

Canonical NF-κB signaling is constitutively activated in acute myeloid leukemia (AML) stem cells and is required for maintenance of the self-renewal of leukemia stem cells (LSCs). However, any potential role for NF-κB non-canonical signaling in AML has been largely overlooked. Here, we report that stabilization of NF-κB-inducing kinase (NIK) suppresses AML. Mechanistically, stabilization of NIK activates NF-κB non-canonical signaling and represses NF-κB canonical signaling. In addition, stabilization of NIK-induced activation of NF-κB non-canonical signaling upregulates Dnmt3a and downregulates Mef2c, which suppresses and promotes AML development, respectively. Importantly, by querying the connectivity MAP using up- and downregulated genes that are present exclusively in NIK-stabilized LSCs, we discovered that verteporfin has anti-AML effects, suggesting that repurposing verteporfin to target myeloid leukemia is worth testing clinically. Our data provide a scientific rationale for developing small molecules to stabilize NIK specifically in myeloid leukemias as an attractive therapeutic option.

Recommended citation: Xiu, Y., Li, Q., Li, F., Boyce, B., Borcherding, N., Xue, X., & Zhao, C. Stabilization of NF-kB-inducing kinase suppresses MLL-AF9–induced acute myeloid leukemia. Cell Reports 2018.

A novel HER2 gene body enhancer contributes to HER2 expression.

Published in Oncogene, 2017

The transcriptional regulation of the human epidermal growth factor receptor-2 (HER2) contributes to an enhanced HER2 expression in HER2-positive breast cancers with HER2 gene amplification and HER2-low or HER2-negative breast cancers following radiotherapy or endocrine therapy, and this drives tumorigenesis and the resistance to therapy. Epigenetic mechanisms are critical for transcription regulation, however, such mechanisms in the transcription regulation of HER2 are limited to the involvement of tri-methylated histone 3 lysine 4 (H3K4me3) and acetylated histone 3 lysine 9 (H3K9ac) at the HER2 promoter region. Here, we report the identification of a novel enhancer in the HER2 3’ gene body, which we have termed HER2 gene body enhancer (HGE). The HGE starts from the 3’ end of intron 19 and extends into intron 22, possesses enhancer histone modification marks in specific cells and enhances the transcriptional activity of the HER2 promoters. We also found that TFAP2C, a known regulator of HER2, binds to HGE and is required for its enhancer function and that DNA methylation in the HGE region inhibits the histone modifications characterizing enhancer and is inversely correlated with HER2 expression in breast cancer samples. The identification of this novel enhancer sheds a light on the roles of epigenetic mechanisms in HER2 transcription, in both HER2-positive breast cancer samples and individuals with HER2-low or HER2-negative breast cancers undergoing radiotherapy or endocrine therapy.

Recommended citation: Liu, Q., Kulak, M., Borcherding, N., Maina, P., Zhang, W., Weigel, R., & Qi, H. A novel HER2 gene-body enhancer contributes to HER2 expression. Oncogene 2018.

Content Evaluation, Usage Profile, and Student Preferences Related to a Medical Student Wiki for Student-Authored Learning Resources (Carverpedia)

Published in Medical Science Educator, 2017

We identified a lack of continuity in student-generated learning aids and the sharing of advice and resources between medical graduating classes. Carverpedia, built in the UIowa Wiki platform, is a novel collaboration between pre-clinical medical students and faculty to produce a centralized location for learning aids at the Carver College of Medicine. Carverpedia is designed to organize files and links in a centralized location accessible to all students. Previously, items were shared through various modalities (e.g., Facebook, email, etc.) without equal access for every student or well-developed archiving. After one academic year of use, there were 217 resources posted to the wiki site by 34 authors. Content posted on Carverpedia differed from Facebook content, with increased content summaries and active-learning. Site utilization, collected by Google Analytics, found 2979 sessions on Carverpedia and a total of 10,253 page views. Site usage and posting of resources was highly correlated with basic science exam schedule. This corresponded with the students surveyed on site usage, with the majority of students using Carverpedia for basic science courses, like Gross Anatomy and Foundations of Cellular Life. Of note, surveyed students found faculty-authored content, practice questions, content summaries, and active-learning resources the most useful of the resources posted. Overall, we found the Wiki format is effective as a means of creating an online community of support and collaboration for student-authored and faculty-authored learning resources.

Recommended citation: Borcherding, N., Rudolph, M., Tierney, A., Vyn, & R., Hoffmann. Content evaluation, usage profile and student preferences related to a medical student wiki for student-authored learning resources. Medical Science Educator 2017.

Obesity-associated NLRC4 inflammasome activation drives breast cancer progression.

Published in Nature Communications, 2016

Obesity is associated with an increased risk of developing breast cancer and is also associated with worse clinical prognosis. The mechanistic link between obesity and breast cancer progression remains unclear, and there has been no development of specific treatments to improve the outcome of obese cancer patients. Here we show that obesity-associated NLRC4 inflammasome activation/ interleukin (IL)-1 signalling promotes breast cancer progression. The tumour microenvironment in the context of obesity induces an increase in tumour-infiltrating myeloid cells with an activated NLRC4 inflammasome that in turn activates IL-1β, which drives disease progression through adipocyte-mediated vascular endothelial growth factor A (VEGFA) expression and angiogenesis. Further studies show that treatment of mice with metformin inhibits obesity-associated tumour progression associated with a marked decrease in angiogenesis. This report provides a causal mechanism by which obesity promotes breast cancer progression and lays out a foundation to block NLRC4 inflammasome activation or IL-1β signalling transduction that may be useful for the treatment of obese cancer patients.

Recommended citation: Kolb, R., Phan L., Borcherding, N., Liu, Y., Yuan, F., Janowski, A.M., Xie, Q., Markan, K.R., Li, W., Potthoff, M.J., Fuentes-Mattei, E., Knudsen, M., Lee, M., Yeung, S., Cassel, S., Sutterwala, F., & Zhang, W. Obesity-associated NLRC4 inflammasome activation drives breast cancer progression. Nature Communications 2016.

Calpain-5 Expression in the Retina Localizes to Photoreceptor Synapses.

Published in Investigative Ophthalmology and Visual Science, 2016

Purpose: We characterize calpain-5 (CAPN5) expression in retinal and neuronal subcellular compartments.

Recommended citation: Schaefer, K., Toral, M., Velez, G., Cox, A., Baker, S., Borcherding, N. , Colgan, D., Bondada, V., Mashburn, C., Yu, C., Geddes, J., Tsang, S., Bassuk, A., & Mahajan, V. Calpain-5 expression in the retina localizes to photoreceptor synapses. Investigative Ophthalmology and Visual Science 2016.

Paracrine WNT5A signaling in the mammary healthy and neoplastic mammary tissue.

Published in Molecular and Cellular Oncology, 2015

Paracrine signaling between mammary epithelial cells has long been appreciated. Recently, we found that Wnt5a, a novel noncanonical Wnt ligand of luminal origin, counteracts canonical Wnt signaling in basal mammary epithelial cells through a paracrine pathway, inhibits the expansion of Erbb2-induced tumor-initiating cells, and suppresses tumor incidence and metastasis.

Recommended citation: Kusner, D., Borcherding, N., & Zhang, W. Paracrine WNT5A signaling in the mammary healthy and neoplastic mammary tissue. Molecular and Cellular Oncology 2016.

Transcriptome analysis of basal and luminal tumor-initiating cells in ErbB2-driven breast cancer

Published in Genomic Data, 2015

Breast cancer is the leading cause of cancer-related mortality for females worldwide. Improving early screening strategies and understanding the events that lead to tumor initiation have led to demonstrable improvements in clinical outcome. Our previous work revealed a variance in the tumorigenic capacity between different mammary epithelial cell populations in an MMTV-ErbB2 mouse model. In order to greater understand how different mammary epithelial cells influence the tumorigenic capacity in ErbB2-induced breast cancer, we transplanted different cell populations from pre-neoplastic MMTV-ErbB2 female mice into recipient mice for tumorigenic study. We found that different mammary epithelial cells bear different tumorigenic potentials even when induced by the same ErbB2 proto-oncogene. To understand the difference in tumors formed from different epithelial cells, we performed gene expression profiling using these tumors (GSE64487). Several genes were further validated using real-time reverse transcription polymerase chain reaction (RT-PCR). Here we provide further details on the experimental methods and microarray analysis. This data provides a resource to further understanding how different mammary cell populations can initiate ErbB2-driven tumors and the role of these cell populations as putative tumor-initiating cells (TICs).

Recommended citation: Borcherding, N., Bormann, N., Kusner, D., Kolb R., & Zhang, W. Transcriptome analysis of basal and luminal tumor-initiating cells in ErbB2-driven breast cancer. Genomic Data 2015.

Paracrine WNT5A signaling inhibits expansion of tumor-initiating cells.

Published in Cancer Research, 2015

It is not well understood how paracrine communication between basal and luminal cell populations in the mammary gland affects tumorigenesis. During ErbB2-induced mammary tumorigenesis, enriched mammary stem cells that represent a subpopulation of basal cells exhibit enhanced tumorigenic capacity compared with the corresponding luminal progenitors. Transcript profiling of tumors derived from basal and luminal tumor-initiating cells (TIC) revealed preferential loss of the noncanonical Wnt ligand WNT5A in basal TIC-derived tumors. Heterozygous loss of WNT5A was correlated with shorter survival of breast cancer patients. In a mouse model of ErbB2-induced breast cancer, Wnt5a heterozygosity promoted tumor multiplicity and pulmonary metastasis. As a TGFβ substrate, luminal cell-produced WNT5A induced a feed-forward loop to activate SMAD2 in a RYK and TGFβR1-dependent manner to limit the expansion of basal TIC in a paracrine fashion, a potential explanation for the suppressive effect of WNT5A in mammary tumorigenesis. Our results identify the WNT5A/RYK module as a spatial regulator of the TGFβ–SMAD signaling pathway in the context of mammary gland development and carcinogenesis, offering a new perspective on tumor suppression provided by basal–luminal cross-talk in normal mammary tissue

Recommended citation: Borcherding, N., Kusner, D., Kolb, R., Xie, Q., Li, W., Yuan, F., Velez, G., Askeland, R., Weigel, R.J., & Zhang, W. Paracrine WNT5A Signaling Inhibits Expansion of Tumor-Initiating Cells. Cancer Research 2015.

Bone Marrow Stromal Antigen 2 (BST-2) DNA Is Demethylated in Breast Tumors and Breast Cancer Cells

Published in PLoS ONE, 2015

Background Bone marrow stromal antigen 2 (BST-2) is a known anti-viral gene that has been recently identified to be overexpressed in many cancers, including breast cancer. BST-2 is critical for the invasiveness of breast cancer cells and the formation of metastasis in vivo. Although the regulation of BST-2 in immune cells is unraveling, it is unknown how BST-2 expression is regulated in breast cancer. We hypothesized that meta-analyses of BST-2 gene expression and BST-2 DNA methylation profiles would illuminate mechanisms regulating elevated BST-2 expression in breast tumor tissues and cells.

Recommended citation: Mahauad-Fernandez, W.D., Borcherding, N. C. , Zhang, W., & Okeoma, C.M. Bone Marrow Stromal Antigen 2 (BST-2) DNA Is Demethylated in Breast Tumors and Breast Cancer Cells. PLoS One 2015.

Characterization of a novel mouse model with genetic deletion of CD177.

Published in PLoS ONE, 2015

Neutrophils play an essential role in the innate immune response to infection. Neutrophils migrate from the vasculature into the tissue in response to infection. Recently, a neutrophil cell surface receptor, CD177, was shown to help mediate neutrophil migration across the endothelium through interactions with PECAM1. We examined a publicly available gene array dataset of CD177 expression from human neutrophils following pulmonary endotoxin instillation. Among all 22,214 genes examined, CD177 mRNA was the most upregulated following endotoxin exposure. The high level of CD177 expression is also maintained in airspace neutrophils, suggesting a potential involvement of CD177 in neutrophil infiltration under infectious diseases. To determine the role of CD177 in neutrophils in vivo, we constructed a CD177-genetic knockout mouse model. The mice with homozygous deletion of CD177 have no discernible phenotype and no significant change in immune cells, other than decreased neutrophil counts in peripheral blood. We examined the role of CD177 in neutrophil accumulation using a skin infection model with Staphylococcus aureus. CD177 deletion reduced neutrophil counts in inflammatory skin caused by S. aureus. Mechanistically we found that CD177 deletion in mouse neutrophils has no significant impact in CXCL1/KC- or fMLP-induced migration, but led to significant cell death. Herein we established a novel genetic mouse model to study the role of CD177 and found that CD177 plays an important role in neutrophils.

Recommended citation: Xie, Q., Klesney-Tait, J., Keck, K., Parlet, C., Borcherding, N. , Kolb, R., Li, W., Tygrett, L., Waldschmidt, T., Olivier, A., Chen, S., Liu, G. H., Li, X., & Zhang, W. Characterization of a novel mouse model with genetic deletion of CD177. Protein & Cell 2015.

NIAM-deficient mice are predisposed to the development of proliferative lesions including B-cell lymphomas.

Published in PLoS One, 2014

Nuclear Interactor of ARF and Mdm2 (NIAM, gene designation Tbrg1) is a largely unstudied inhibitor of cell proliferation that helps maintain chromosomal stability. It is a novel activator of the ARF-Mdm2-Tip60-p53 tumor suppressor pathway as well as other undefined pathways important for genome maintenance. To examine its predicted role as a tumor suppressor, we generated NIAM mutant (NIAMm/m) mice homozygous for a β-galactosidase expressing gene-trap cassette in the endogenous gene. The mutant mice expressed significantly lower levels of NIAM protein in tissues compared to wild-type animals. Fifty percent of aged NIAM deficient mice (14 to 21 months) developed proliferative lesions, including a uterine hemangioma, pulmonary papillary adenoma, and a Harderian gland adenoma. No age-matched wild-type or NIAM+/m heterozygous animals developed lesions. In the spleen, NIAMm/m mice had prominent white pulp expansion which correlated with enhanced increased reactive lymphoid hyperplasia and evidence of systemic inflammation. Notably, 17% of NIAM mutant mice had splenic white pulp features indicating early B-cell lymphoma. This correlated with selective expansion of marginal zone B cells in the spleens of younger, tumor-free NIAM-deficient mice. Unexpectedly, basal p53 expression and activity was largely unaffected by NIAM loss in isolated splenic B cells. In sum, NIAM down-regulation in vivo results in a significant predisposition to developing benign tumors or early stage cancers. These mice represent an outstanding platform for dissecting NIAM’s role in tumorigenesis and various anti-cancer pathways, including p53 signaling.

Recommended citation: Reed, S.M., Hagen, J., Muniz, V.P., Rosean, T.R., Borcherding, N. , Sciegienka, S., Goeken, J.A., Naumann, P.W., Zhang, W., Tompkins, V.S., Janz, S., Meyerholz, D.K., & Quelle, D.E. NIAM-deficient mice are predisposed to the development of proliferative lesions including B-cell lymphomas. PLoS One 2014.

Ubiquitin-conjugating enzyme Ubc13 controls breast cancer metastasis through a TAK1-p38 MAP kinase cascade.

Published in PNAS, 2014

Metastatic spread is the leading cause of cancer mortality. Breast cancer (BCa) metastatic recurrence can happen years after removal of the primary tumor. Here we show that Ubc13, an E2 enzyme that catalyzes K63-linked protein polyubiquitination, is largely dispensable for primary mammary tumor growth but is required for metastatic spread and lung colonization by BCa cells. Loss of Ubc13 inhibited BCa growth and survival only at metastatic sites. Ubc13 was dispensable for transforming growth factor β (TGFβ)-induced SMAD activation but was required for activation of non-SMAD signaling via TGFβ-activating kinase 1 (TAK1) and p38, whose activity controls expression of numerous metastasis promoting genes. p38 activation restored metastatic activity to Ubc13-deficient cells, and its pharmacological inhibition attenuated BCa metastasis in mice, suggesting it is a therapeutic option for metastatic BCa.

Recommended citation: Wu, X., Zhang, W., Font-Burgada, J., Palmer, T., Hamil, A. S., Biswas, S. K., Poidinger, M., Borcherding, N., Xie, Q., Ellies, L. G., Ltyle, N. K., Wu, L. W., Fox, R. G., Yang, J., Dowdy, S. F., Reya, T., & Karin, M. Ubiquitin-conjugating enzyme Ubc13 controls breast cancer metastasis through a TAK1-p38 MAP kinase cascade. Proceedings of the National Academy of Science 2014.

ROR1, an embryonic protein with an emerging role in cancer biology.

Published in Protein & Cell, 2014

Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a member of the ROR family consisting of ROR1 and ROR2. RORs contain two distinct extracellular cysteine-rich domains and one transmembrane domain. Within the intracellular portion, ROR1 possesses a tyrosine kinase domain, two serine/threonine-rich domains and a proline-rich domain. RORs have been studied in the context of embryonic patterning and neurogenesis through a variety of homologs. These physiologic functions are dichotomous based on the requirement of the kinase domain. A growing literature has established ROR1 as a marker for cancer, such as in CLL and other blood malignancies. In addition, ROR1 is critically involved in progression of a number of blood and solid malignancies. ROR1 has been shown to inhibit apoptosis, potentiate EGFR signaling, and induce epithelial-mesenchymal transition (EMT). Importantly, ROR1 is only detectable in embryonic tissue and generally absent in adult tissue, making the protein an ideal drug target for cancer therapy.

Recommended citation: Borcherding, N., Kusner, D., Liu, G. H., & Zhang, W. ROR1, an embryonic protein with an emerging role in cancer biology. Protein Cell 2014.

Dietary resistant starch prevents urinary excretion of 25-hydroxycholecalciferol and vitamin D-binding protein in type 1 diabetic rats.

Published in Journal of Nutrition, 2013

Diabetes is a rapidly growing epidemic affecting millions of Americans and has been implicated in a number of devastating secondary complications. We previously demonstrated that type 2 diabetic rats exhibit vitamin D deficiency due to aberrant megalin-mediated endocytosis and excessive urinary excretion of 25-hydroxycholecalciferol (25D3) and vitamin D-binding protein (DBP). Here, we examined whether a model of type 1 diabetes [T1D; streptozotocin (STZ)-treated Sprague-Dawley rats] would similarly excrete abnormally high concentrations of 25D3 and DBP due to renal damage and compromised expression of megalin and its endocytic partner, disabled-2 (Dab2). Moreover, we tested whether feeding diabetic rats starch that is resistant to digestion could alleviate these abnormalities. Control (n = 12) rats were fed a standard, semipurified diet (AIN-93G) containing 55% total dietary starch and STZ-treated rats were fed the AIN-93G diet (n = 12) or a diet containing 55% high-amylose maize that is partially resistant to digestion [20% total dietary resistant starch (RS); n = 12] for 2 and 5 wk. The RS diet attenuated weight loss and polyuria in STZ-treated rats. Histology and immunohistochemistry revealed that dietary RS also attenuated the loss of Dab2 expression in renal proximal tubules. Moreover, urinary concentrations of both 25D3 and DBP were elevated ∼10-fold in STZ-treated rats (5 wk post STZ injection), which was virtually prevented by the RS. We also observed a ∼1.5-fold increase in megalin mRNA expression in STZ-treated rats, which was attenuated by feeding rats the RS diet for 2 wk. Taken together, these studies indicate that consumption of low-glycemic carbohydrates can attenuate disruption of vitamin D homeostasis in T1D through the rescue of megalin-mediated endocytosis in the kidney.

Recommended citation: Smazal, A. L., Borcherding, N. C., Anderegg, A. S., Schalinske, K. L., Whitley, E. M., & Rowling, M. J. (2013). Dietary resistant starch prevents urinary excretion of 25-hydroxycholecalciferol and vitamin D-binding protein in type 1 diabetic rats. Journal of Nutrition 2013.

A NIK-IKKα Module Expands ErbB2-Induced Tumor-Initiating Cells by Stimulating Nuclear Export of p27/Kip1.

Published in Cancer Cell, 2013

IκB kinase α (IKKα) activity is required for ErbB2-induced mammary tumorigenesis. Here, we show that IKKα and its activator, NF-κB-inducing kinase (NIK), support the expansion of tumor-initiating cells (TICs) that copurify with a CD24medCD49fhi population from premalignant ErbB2-expressing mammary glands. Upon activation, IKKα enters the nucleus, phosphorylates the cyclin-dependent kinase (CDK) inhibitor p27/Kip1, and stimulates its nuclear export or exclusion. Reduced p27 expression rescues mammary tumorigenesis in mice deficient in IKKα kinase activity and restores TIC self-renewal. IKKα is also likely to be involved in human breast cancer, where its expression shows an inverse correlation with metastasis-free survival, and its presence in the nucleus of invasive ductal carcinomas (IDCs) is associated with decreased nuclear p27 abundance.

Recommended citation: Zhang, W., Tan, W., Wu, X., Poustovoitov, M., Strasner, A., Li, W., Borcherding, N. , Ghassemian, M., & Karin, M. A NIK-IKKalpha module expands ErbB2-induced tumor-initiating cells by stimulating nuclear export of p27/Kip1. Cancer Cell 2013.