Network


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

Hotspot


Dive into the research topics where Nicholas J. Sullivan is active.

Publication


Featured researches published by Nicholas J. Sullivan.


Oncogene | 2009

Interleukin-6 induces an epithelial–mesenchymal transition phenotype in human breast cancer cells

Nicholas J. Sullivan; A K Sasser; A E Axel; F Vesuna; V Raman; N Ramirez; Tatiana M. Oberyszyn; B M Hall

Breast tumor interleukin-6 (IL-6) levels increase with tumor grade, and elevated serum IL-6 correlates with poor breast cancer patient survival. Epithelial–mesenchymal transition (EMT) phenotypes such as impaired E-cadherin expression or aberrant Vimentin induction are associated with enhanced metastasis and unfavorable clinical outcome in breast cancer. Despite this fact, few tumor microenvironment-derived extracellular signaling factors capable of provoking such a phenotypic transition have been identified. In this study, we showed that IL-6 promoted E-cadherin repression among a panel of estrogen receptor-α-positive human breast cancer cells. Furthermore, ectopic stable IL-6 expressing MCF-7 breast adenocarcinoma cells (MCF-7IL−6) exhibited an EMT phenotype characterized by impaired E-cadherin expression and induction of Vimentin, N-cadherin, Snail and Twist. MCF-7IL−6 cells formed xenograft tumors that displayed loss of E-cadherin, robust Vimentin induction, increased proliferative indices, advanced tumor grade and undifferentiated histology. Finally, we showed aberrant IL-6 production and STAT3 activation in MCF-7 cells that constitutively express Twist, a metastatic regulator and direct transcriptional repressor of E-cadherin. To our knowledge, this is the first study that shows IL-6 as an inducer of an EMT phenotype in breast cancer cells and implicates its potential to promote breast cancer metastasis.


The FASEB Journal | 2007

Interleukin-6 is a potent growth factor for ER-α-positive human breast cancer

A. Kate Sasser; Nicholas J. Sullivan; Adam W. Studebaker; Lindsay F. Hendey; Amy Axel; Brett Hall

Bone is the primary anatomical site of breast cancer metastasis, and bone metastasis is associated with increased morbidity and mortality. Mesenchy‐mal stem cells (MSC) are a predominant fibroblast cell population within the bone marrow, and metastatic breast cancer cells that seed within bone would predictably encounter MSC or their soluble factors. Therefore, we examined the impact of primary human MSC on a panel of estrogen receptor‐alpha (ERα)‐positive (MCF‐7, T47D, BT474, and ZR‐75–1) and ERα‐negative (MDA‐MB‐231 and MDA‐MB‐468) human breast tumor cell lines. All ERα‐positive breast tumor cell lines displayed low basal activation of signal transducer and activator of transcription 3 (STAT3) until exposed to MSC, which induced chronic phosphorylation of STAT3 on tyrosine‐705. Paracrine IL‐6 was found to be the principal mediator of STAT3 phosphorylation in coculture studies, and MSC induction of STAT3 phos‐phorylation was lost when IL‐6 was depleted from MSC conditioned media or the IL‐6 receptor was blocked on tumor cells. Enhanced tumor cell growth rates were observed in the ERα‐positive mammary tumor cell line MCF‐7 after paracrine and autocrine IL‐6 exposure, where MCF‐7 growth rates were enhanced by > 2‐fold when cocultured with MSC in vitro and even more pronounced in vivo with autocrine IL‐6 production.—Sasser, A. K., Sullivan, N. J., Studebaker, A. W., Hendey, L. F., Axel, A. E., Hall, B. M. Interleukin‐6 is a potent growth factor for ER‐α‐positive human breast cancer. FASEB J. 21, 3763–3770 (2007)


Journal of Investigative Dermatology | 2012

UV Light B–Mediated Inhibition of Skin Catalase Activity Promotes Gr-1+CD11b+ Myeloid Cell Expansion

Nicholas J. Sullivan; Kathleen L. Tober; Erin M. Burns; Jonathan Schick; Judith A. Riggenbach; Thomas A. Mace; Matthew A. Bill; Gregory S. Young; Tatiana M. Oberyszyn; Gregory B. Lesinski

Skin cancer incidence and mortality are higher in men compared to women, but the causes of this sex discrepancy remain largely unknown. Ultraviolet light exposure induces cutaneous inflammation and neutralizes cutaneous antioxidants. Gr-1+CD11b+ myeloid cells are heterogeneous bone marrow-derived cells that promote inflammation-associated carcinogenesis. Reduced activity of catalase, an antioxidant present within skin, has been associated with skin carcinogenesis. We utilized the outbred, immune competent Skh-1 hairless mouse model of ultraviolet light B (UVB)-induced inflammation and non-melanoma skin cancer to further define sex discrepancies in UVB-induced inflammation. Our results demonstrated that male skin had relatively lower baseline catalase activity, which was inhibited following acute UVB exposure in both sexes. Further analysis revealed that skin catalase activity inversely correlated with splenic Gr-1+CD11b+ myeloid cell percentage. Acute UVB exposure induced Gr-1+CD11b+ myeloid cell skin infiltration, which was inhibited to a greater extent in males by topical catalase treatment. In chronic UVB studies, we demonstrated that the percentage of splenic Gr-1+CD11b+ myeloid cells was 55% higher in male tumor-bearing mice compared to their female counterparts. Together, our findings indicate that lower skin catalase activity in male mice may at least in part contribute to increased UVB-induced Gr-1+CD11b+ myeloid cells and subsequent skin carcinogenesis.


Clinical Cancer Research | 2009

Prognostic significance of interleukin-6 single nucleotide polymorphism genotypes in neuroblastoma: rs1800795 (promoter) and rs8192284 (receptor).

Joanne P. Lagmay; Wendy B. London; Thomas G. Gross; Amanda M. Termuhlen; Nicholas J. Sullivan; Amy Axel; Bethany L. Mundy; Mark Ranalli; Jason Canner; Patrick McGrady; Brett Hall

Purpose: Neuroblastoma is a childhood cancer of the sympathetic nervous system and many patients present with high-risk disease. Risk stratification, based on pathology and tumor-derived biomarkers, has improved prediction of clinical outcomes, but overall survival (OS) rates remain unfavorable and new therapeutic targets are needed. Some studies suggest a link between interleukin (IL)-6 and more aggressive behavior in neuroblastoma tumor cells. Therefore, we examined the impact of two IL-6 single nucleotide polymorphisms (SNP) on neuroblastoma disease progression. Experimental Design: DNA samples from 96 high-risk neuroblastoma patients were screened for two SNP that are known to regulate the serum levels of IL-6 and the soluble IL-6 receptor, rs1800795 and rs8192284, respectively. The genotype for each SNP was determined in a blinded fashion and independent statistical analysis was done to determine SNP-related event-free survival (EFS) and OS rates. Results: The rs1800795 IL-6 promoter SNP is an independent prognostic factor for EFS and OS in high-risk neuroblastoma patients. In contrast, the rs8192284 IL-6 receptor SNP revealed no prognostic value. Conclusions: The rs1800795 SNP [−174 IL-6 (G > C)] represents a novel and independent prognostic marker for both EFS and OS in high-risk neuroblastoma. Because the rs1800795 SNP [−174 IL-6 (G > C)] has been shown to correlate with production of IL-6, this cytokine may represent a target for development of new therapies in neuroblastoma. (Clin Cancer Res 2009;15(16):5234–9)


Neoplasia | 2015

Function of Integrin-Linked Kinase in Modulating the Stemness of IL-6–Abundant Breast Cancer Cells by Regulating γ-Secretase–Mediated Notch1 Activation in Caveolae

En-Chi Hsu; Samuel K. Kulp; Han Li Huang; Huang Ju Tu; Santosh B. Salunke; Nicholas J. Sullivan; Duxin Sun; Max S. Wicha; Charles L. Shapiro; Ching-Shih Chen

Interleukin-6 (IL-6) and Notch signaling are important regulators of breast cancer stem cells (CSCs), which drive the malignant phenotype through self-renewal, differentiation, and development of therapeutic resistance. We investigated the role of integrin-linked kinase (ILK) in regulating IL-6–driven Notch1 activation and the ability to target breast CSCs through ILK inhibition. Ectopic expression/short hairpin RNA-mediated knockdown of ILK, pharmacological inhibition of ILK with the small molecule T315, Western blot analysis, immunofluorescence, and luciferase reporter assays were used to evaluate the regulation of IL-6–driven Notch1 activation by ILK in IL-6–producing triple-negative breast cancer cell lines (MDA-MB-231, SUM-159) and in MCF-7 and MCF-7IL-6 cells. The effects of ILK on γ-secretase complex assembly and cellular localization were determined by immunofluorescence, Western blots of membrane fractions, and immunoprecipitation. In vivo effects of T315-induced ILK inhibition on CSCs in SUM-159 xenograft models were assessed by mammosphere assays, flow cytometry, and tumorigenicity assays. Results show that the genetic knockdown or pharmacological inhibition of ILK suppressed Notch1 activation and the abundance of the γ-secretase components presenilin-1, nicastrin, and presenilin enhancer 2 at the posttranscriptional level via inhibition of caveolin-1-dependent membrane assembly of the γ-secretase complex. Accordingly, knockdown of ILK inhibited breast CSC-like properties in vitro and the breast CSC subpopulation in vivo in xenograft tumor models. Based on these findings, we propose a novel function of ILK in regulating γ-secretase–mediated Notch1 activation, which suggests the targeting of ILK as a therapeutic approach to suppress IL-6–induced breast CSCs.


Carcinogenesis | 2016

Integrin-linked kinase as a novel molecular switch of the IL-6-NF-κB signaling loop in breast cancer

En-Chi Hsu; Samuel K. Kulp; Han Li Huang; Huang Ju Tu; Min Wu Chao; Yu Chou Tseng; Ming Chen Yang; Santosh B. Salunke; Nicholas J. Sullivan; Wen Chung Chen; Jianying Zhang; Che-Ming Teng; Wen-Mei Fu; Duxin Sun; Max S. Wicha; Charles L. Shapiro; Ching-Shih Chen

Substantial evidence has clearly demonstrated the role of the IL-6-NF-κB signaling loop in promoting aggressive phenotypes in breast cancer. However, the exact mechanism by which this inflammatory loop is regulated remains to be defined. Here, we report that integrin-linked kinase (ILK) acts as a molecular switch for this feedback loop. Specifically, we show that IL-6 induces ILK expression via E2F1 upregulation, which, in turn, activates NF-κB signaling to facilitate IL-6 production. shRNA-mediated knockdown or pharmacological inhibition of ILK disrupted this IL-6-NF-κB signaling loop, and blocked IL-6-induced cancer stem cells in vitro and estrogen-independent tumor growth in vivo Together, these findings establish ILK as an intermediary effector of the IL-6-NF-κB feedback loop and a promising therapeutic target for breast cancer.


Journal of Investigative Dermatology | 2012

Ultraviolet Light B-Mediated Inhibition of Skin Catalase Activity Promotes Gr-1+CD11b+ Myeloid Cell Expansion

Nicholas J. Sullivan; Kathleen L. Tober; Erin M. Burns; Jonathan Schick; Judith A. Riggenbach; Thomas A. Mace; Matthew A. Bill; Gregory S. Young; Tatiana M. Oberyszyn; Gregory B. Lesinski

Skin cancer incidence and mortality are higher in men compared to women, but the causes of this sex discrepancy remain largely unknown. Ultraviolet light exposure induces cutaneous inflammation and neutralizes cutaneous antioxidants. Gr-1+CD11b+ myeloid cells are heterogeneous bone marrow-derived cells that promote inflammation-associated carcinogenesis. Reduced activity of catalase, an antioxidant present within skin, has been associated with skin carcinogenesis. We utilized the outbred, immune competent Skh-1 hairless mouse model of ultraviolet light B (UVB)-induced inflammation and non-melanoma skin cancer to further define sex discrepancies in UVB-induced inflammation. Our results demonstrated that male skin had relatively lower baseline catalase activity, which was inhibited following acute UVB exposure in both sexes. Further analysis revealed that skin catalase activity inversely correlated with splenic Gr-1+CD11b+ myeloid cell percentage. Acute UVB exposure induced Gr-1+CD11b+ myeloid cell skin infiltration, which was inhibited to a greater extent in males by topical catalase treatment. In chronic UVB studies, we demonstrated that the percentage of splenic Gr-1+CD11b+ myeloid cells was 55% higher in male tumor-bearing mice compared to their female counterparts. Together, our findings indicate that lower skin catalase activity in male mice may at least in part contribute to increased UVB-induced Gr-1+CD11b+ myeloid cells and subsequent skin carcinogenesis.


Journal of Investigative Dermatology | 2012

UV Light B|[ndash]|Mediated Inhibition of Skin Catalase Activity Promotes Gr-1|[plus]|CD11b|[plus]| Myeloid Cell Expansion

Nicholas J. Sullivan; Kathleen L. Tober; Erin M. Burns; Jonathan Schick; Judith A. Riggenbach; Thomas A. Mace; Matthew A. Bill; Gregory S. Young; Tatiana M. Oberyszyn; Gregory B. Lesinski

Skin cancer incidence and mortality are higher in men compared to women, but the causes of this sex discrepancy remain largely unknown. Ultraviolet light exposure induces cutaneous inflammation and neutralizes cutaneous antioxidants. Gr-1+CD11b+ myeloid cells are heterogeneous bone marrow-derived cells that promote inflammation-associated carcinogenesis. Reduced activity of catalase, an antioxidant present within skin, has been associated with skin carcinogenesis. We utilized the outbred, immune competent Skh-1 hairless mouse model of ultraviolet light B (UVB)-induced inflammation and non-melanoma skin cancer to further define sex discrepancies in UVB-induced inflammation. Our results demonstrated that male skin had relatively lower baseline catalase activity, which was inhibited following acute UVB exposure in both sexes. Further analysis revealed that skin catalase activity inversely correlated with splenic Gr-1+CD11b+ myeloid cell percentage. Acute UVB exposure induced Gr-1+CD11b+ myeloid cell skin infiltration, which was inhibited to a greater extent in males by topical catalase treatment. In chronic UVB studies, we demonstrated that the percentage of splenic Gr-1+CD11b+ myeloid cells was 55% higher in male tumor-bearing mice compared to their female counterparts. Together, our findings indicate that lower skin catalase activity in male mice may at least in part contribute to increased UVB-induced Gr-1+CD11b+ myeloid cells and subsequent skin carcinogenesis.


Cancer Research | 2010

Abstract 4202: Sex disparities in the cutaneous response to acute ultraviolet light exposure

Kathleen L. Tober; Judith A. Riggenbach; Jonathan Schick; Nicholas J. Sullivan; Lina Abbaoui; Tatiana M. Oberyszyn

Epidemiological evidence has identified a 3-fold increase in squamous cell skin cancer incidence in men compared to women. The historical explanation for this sex-bias is increased occupational exposure to ultraviolet (UV) light. Our studies demonstrated, however, that when chronically exposed to equivalent doses of UV light, male mice developed more skin tumors that were larger and more aggressive than those arising in female mice. Additionally, the skin of male mice acutely exposed to UV light had higher levels of oxidative DNA damage and decreased total antioxidant capacity compared to females (Thomas-Ahner, JM Cancer Research 2007). The hypothesis for the current study is that the topical application of anti-inflammatory and/or antioxidant compounds may differentially affect the acute UVB induced inflammatory response in the sexes. To test this hypothesis, female and male Skh-1 mice were exposed to 2240J/m2 UVB light followed immediately by topical application of either an anti-inflammatory compound (Solaraze), an antioxidant (Vitamin E), or a combination of the 2 compounds. Animals were sacrificed at 24 and 48 hrs following a single exposure and topical treatment. Skin was analyzed for myeloperoxidase (MPO) activity as a measure of neutrophil infiltration, catalase activity, and immunohistochemically for the presence of p53 protein in the epidermis. In response to UVB exposure, vehicle treated female mice had significantly increased MPO activity compared to male mice. Topical treatments with Solaraze, Vitamin E or the combination significantly reduced MPO levels in female mice but had no effects on the already low levels of UV-induced MPO in male skin. Catalase is a key enzyme used for detoxifying hydrogen peroxide in the skin. Baseline levels of catalase were lower in male skin compared to female skin and UVB exposure significantly reduced catalase in both sexes. Topical treatment with Solaraze significantly increased male catalase activity to levels similar to female unirradiated skin while vitamin E partially restored catalase activity in female mice but had no effect in male skin. The single exposure to UVB induced higher numbers of p53 positive epidermal cells in male skin, however at this time point topical treatment had no effect on UVB-induced p53 levels in either sex. Our data suggests that UVB differentially effects inflammation in the sexes and that topical application of anti-inflammatory or anti-oxidant compounds have differential effects on markers of an acute inflammatory response in the skin. Future studies will evaluate the effects of additional compounds and delivery vehicles following both acute and chronic UVB exposure. This work was supported by NIH NCI CA133629. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4202.


Cancer Research | 2010

Abstract 3378: E-cadherin suppresses breast cancer cell growth and interleukin-6 production

Nicholas J. Sullivan; Jonathan Schick; Judith A. Riggenbach; Kathleen L. Tober; Brett M. Hall; Tatiana M. Oberyszyn

Carcinomas arise from epithelial tissues and represent the most common type of human cancer. E-cadherin is an epithelial-specific intercellular adhesion molecule, which localizes to epithelial cell adherens junctions and is a critical mediator of carcinoma cell invasion and metastasis. Although impaired E-cadherin expression can be clinically utilized to predict breast cancer cell invasiveness, metastasis, and patient survival, E-cadherin has received less attention for its role as a putative regulator of epithelial cell growth. We have previously shown that interleukin-6 (IL-6), a pleiotropic inflammatory cytokine associated with poor clinical outcome in breast cancer patients, is a potent growth factor and repressor of E-cadherin in human breast cancer cells. Consistent with clinical reports and our previous findings, we hypothesized that E-cadherin repression may be responsible for IL-6-induced breast cancer cell growth. The current study demonstrated that IL-6 enhanced the growth rates of estrogen receptor-alpha (ERα)-positive breast cancer cells in a three-dimensional (3D) tumor growth assay (TGA) in a dose-dependent manner. We showed that IL-6 also repressed E-cadherin protein levels in ERα-positive breast cancer cells in a 3D TGA in a dose-dependent manner. We have previously shown that IL-6 treatment does not affect E-cadherin-negative MDA-MB-231 cell growth, and the current study showed similar results in MCF-7 cells which express Twist and therefore lack E-cadherin. MDA-MB-231 cells lack E-cadherin and express IL-6, thus allowing us to express stable ectopic E-cadherin to evaluate its impact on cell growth and IL-6 production. Ectopic E-cadherin expression significantly suppressed MDA-MB-231 cell growth (P-value Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3378.

Collaboration


Dive into the Nicholas J. Sullivan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge