402 results on '"Harikrishna, Nakshatri"'
Search Results
152. Additional file 3: of Dependence receptor UNC5A restricts luminal to basal breast cancer plasticity and metastasis
- Author
-
Padua, Maria, Poornima Bhat-Nakshatri, Manjushree Anjanappa, Mayuri Prasad, Yangyang Hao, Rao, Xi, Liu, Sheng, Wan, Jun, Yunlong Liu, McElyea, Kyle, Jacobsen, Max, Sandusky, George, Althouse, Sandra, Perkins, Susan, and Harikrishna Nakshatri
- Abstract
Description of patients and characteristics of their tumors (nâ =â 221). (DOCX 98Â kb)
- Published
- 2018
- Full Text
- View/download PDF
153. Pharmacological dual inhibition of tumor and tumor-induced functional limitations in transgenic model of breast cancer
- Author
-
George E. Sandusky, Peter A. Crooks, Harikrishna Nakshatri, Max Jacobson, Courtney Finnearty, Ruizhong Wang, Mayuri S. Prasad, Narsimha Reddy Penthala, Poornima Bhat-Nakshatri, Kyle McElyea, Jianguo Liu, Manjushree Anjanappa, Teresa A. Zimmers, Victoria Sefcsik, Maria B. Padua, and Rachael Redmond
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Breast Neoplasms ,Mammary Neoplasms, Animal ,Mice, Transgenic ,Biology ,Article ,Cachexia ,Metastasis ,03 medical and health sciences ,Mice ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Autocrine signalling ,Mammary tumor ,Myogenesis ,Skeletal muscle ,Cancer ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Oncology ,Sarcopenia ,Female - Abstract
Breast cancer progression is associated with systemic effects, including functional limitations and sarcopenia without the appearance of overt cachexia. Autocrine/paracrine actions of cytokines/chemokines produced by cancer cells mediate cancer progression and functional limitations. The cytokine-inducible transcription factor NF-κB could be central to this process, as it displays oncogenic functions and is integral to the Pax7:MyoD:Pgc-1β:miR-486 myogenesis axis. We tested this possibility using the MMTV-PyMT transgenic mammary tumor model and the NF-κB inhibitor dimethylaminoparthenolide (DMAPT). We observed deteriorating physical and functional conditions in PyMT+ mice with disease progression. Compared with wild-type mice, tumor-bearing PyMT+ mice showed decreased fat mass, impaired rotarod performance, and reduced grip strength as well as increased extracellular matrix (ECM) deposition in muscle. Contrary to acute cachexia models described in the literature, mammary tumor progression was associated with reduction in skeletal muscle stem/satellite-specific transcription factor Pax7. Additionally, we observed tumor-induced reduction in Pgc-1β in muscle, which controls mitochondrial biogenesis. DMAPT treatment starting at 6 to 8 weeks age prior to mammary tumor occurrence delayed mammary tumor onset and tumor growth rates without affecting metastasis. DMAPT overcame cancer-induced functional limitations and improved survival, which was accompanied with restoration of Pax7, Pgc-1β, and mitochondria levels and reduced ECM levels in skeletal muscles. In addition, DMAPT restored circulating levels of 6 out of 13 cancer-associated cytokines/chemokines changes to levels seen in healthy animals. These results reveal a pharmacological approach for overcoming cancer-induced functional limitations, and the above-noted cancer/drug-induced changes in muscle gene expression could be utilized as biomarkers of functional limitations. Mol Cancer Ther; 16(12); 2747–58. ©2017 AACR.
- Published
- 2017
154. A system for detecting high impact-low frequency mutations in primary tumors and metastases
- Author
-
Jennifer B. Nelson, Raghavendra G. Mirmira, Kennethe Nephew, Yangyang Hao, Manjushree Anjanappa, Fang Fang, Kathy D. Miller, Harikrishna Nakshatri, Mohammad Reza Saadatzadeh, Yunlong Liu, Lang Li, Aaron A. Cohen-Gadol, Sarah A. Tersey, Poornima Bhat-Nakshatri, and Edward Simpson
- Subjects
0301 basic medicine ,Cancer Research ,Somatic cell ,Biopsy ,DNA Mutational Analysis ,Primary Cell Culture ,Notch signaling pathway ,Genomics ,Biology ,medicine.disease_cause ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,FHIT ,Neoplasms ,Genetics ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Gene Regulatory Networks ,Epigenetics ,Molecular Biology ,Gene ,Mutation ,Whole Genome Sequencing ,Cancer ,High-Throughput Nucleotide Sequencing ,Epithelial Cells ,Fibroblasts ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cancer research ,Feasibility Studies ,Signal Transduction - Abstract
Tumor complexity and intratumor heterogeneity contribute to subclonal diversity. Despite advances in next-generation sequencing (NGS) and bioinformatics, detecting rare mutations in primary tumors and metastases contributing to subclonal diversity is a challenge for precision genomics. Here, in order to identify rare mutations, we adapted a recently described epithelial reprograming assay for short-term propagation of epithelial cells from primary and metastatic tumors. Using this approach, we expanded minor clones and obtained epithelial cell-specific DNA/RNA for quantitative NGS analysis. Comparative Ampliseq Comprehensive Cancer Panel sequence analyses were performed on DNA from unprocessed breast tumor and tumor cells propagated from the same tumor. We identified previously uncharacterized mutations present only in the cultured tumor cells, a subset of which has been reported in brain metastatic but not primary breast tumors. In addition, whole-genome sequencing identified mutations enriched in liver metastases of various cancers, including Notch pathway mutations/chromosomal inversions in 5/5 liver metastases, irrespective of cancer types. Mutations/rearrangements in FHIT, involved in purine metabolism, were detected in 4/5 liver metastases, and the same four liver metastases shared mutations in 32 genes, including mutations of different HLA-DR family members affecting OX40 signaling pathway, which could impact the immune response to metastatic cells. Pathway analyses of all mutated genes in liver metastases showed aberrant tumor necrosis factor and transforming growth factor signaling in metastatic cells. Epigenetic regulators including KMT2C/MLL3 and ARID1B, which are mutated in >50% of hepatocellular carcinomas, were also mutated in liver metastases. Thus, irrespective of cancer types, organ-specific metastases may share common genomic aberrations. Since recent studies show independent evolution of primary tumors and metastases and in most cases mutation burden is higher in metastases than primary tumors, the method described here may allow early detection of subclonal somatic alterations associated with metastatic progression and potentially identify therapeutically actionable, metastasis-specific genomic aberrations.
- Published
- 2017
155. Inflammation-associated microRNA changes in circulating exosomes of heart failure patients
- Author
-
Zeb Saeed, Ruizhong Wang, Srikant Devaraj, Harikrishna Nakshatri, Faheemullah Beg, and Kamalesh Masoor
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,lcsh:Medicine ,Inflammation ,Exosomes ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Internal medicine ,microRNA ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,lcsh:Science (General) ,lcsh:QH301-705.5 ,Aged ,Heart Failure ,business.industry ,lcsh:R ,MicroRNA ,General Medicine ,Middle Aged ,medicine.disease ,Control cell ,Microvesicles ,Rats ,MicroRNAs ,Research Note ,030104 developmental biology ,Endocrinology ,lcsh:Biology (General) ,Cell culture ,Heart failure ,Biomarker (medicine) ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,business ,Biomarkers ,lcsh:Q1-390 - Abstract
Objective MiR-486 and miR-146a are cardiomyocyte-enriched microRNAs that control cell survival and self-regulation of inflammation. These microRNAs are released into circulation and are detected in plasma or in circulating exosomes. Little is known whether heart failure affects their release into circulation, which this study investigated. Results Total and exosome-specific microRNAs in plasma of 40 heart failure patients and 20 controls were prepared using the miRVana Kit. We measured exosomal and total plasma microRNAs separately because exosomes serve as cargos that transfer biological materials and alter signaling in distant organs, whereas microRNAs in plasma indicate the level of tissue damage and are mostly derived from dead cells. qRT-PCR was used to quantify miR-486, miR-146a, and miR-16. Heart failure did not significantly affect plasma miR-486/miR-16 and miR-146a/miR-16 ratio, although miR-146a/miR-16 showed a trend of elevated expression (2.3 ± 0.79, p = 0.27). By contrast, circulating exosomal miR-146a/miR-16 ratio was higher in heart failure patients (2.46 ± 0.51, p = 0.05). miR-146a is induced in response to inflammation as a part of inflammation attenuation circuitry. Indeed, Tnfα and Gm-csf increased miR-146a but not miR-486 in the cardiomyocyte cell line H9C2. These results, if confirmed in a larger study, may help to develop circulating exosomal miR-146a as a biomarker of heart failure.
- Published
- 2017
156. RareVar: A Framework for Detecting Low-Frequency Single-Nucleotide Variants
- Author
-
Xiaoling Xuei, Yangyang Hao, Harikrishna Nakshatri, Yunlong Liu, Lang Li, and Howard J. Edenberg
- Subjects
0301 basic medicine ,Pooling ,Word error rate ,Computational biology ,Biology ,Polymorphism, Single Nucleotide ,DNA sequencing ,03 medical and health sciences ,Gene Frequency ,Neoplasms ,Genetics ,medicine ,Biomarkers, Tumor ,Humans ,Computer Simulation ,Molecular Biology ,Allele frequency ,Research Articles ,Protocol (science) ,Point mutation ,Computational Biology ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Primary tumor ,Computational Mathematics ,030104 developmental biology ,Computational Theory and Mathematics ,Modeling and Simulation ,Benchmark (computing) ,Algorithms - Abstract
Accurate identification of low-frequency somatic point mutations in tumor samples has important clinical utilities. Although high-throughput sequencing technology enables capturing such variants while sequencing primary tumor samples, our ability for accurate detection is compromised when the variant frequency is close to the sequencer error rate. Most current experimental and bioinformatic strategies target mutations with ≥5% allele frequency, which limits our ability to understand the cancer etiology and tumor evolution. We present an experimental and computational modeling framework, RareVar, to reliably identify low-frequency single-nucleotide variants from high-throughput sequencing data under standard experimental protocols. RareVar protocol includes a benchmark design by pooling DNAs from already sequenced individuals at various concentrations to target variants at desired frequencies, 0.5%-3% in our case. By applying a generalized, linear model-based, position-specific error model, followed by machine-learning-based variant calibration, our approach outperforms existing methods. Our method can be applied on most capture and sequencing platforms without modifying the experimental protocol.
- Published
- 2017
157. Systemic Actions of Breast Cancer Facilitate Functional Limitations
- Author
-
Harikrishna Nakshatri and Ruizhong Wang
- Subjects
0301 basic medicine ,Cancer Research ,microrna ,functional limitations ,Review ,Disease ,Bioinformatics ,MyoD ,cachexia ,lcsh:RC254-282 ,Cachexia ,Skeletal muscle fibrosis ,03 medical and health sciences ,breast cancer ,0302 clinical medicine ,Breast cancer ,microRNA ,medicine ,skeletal muscle ,business.industry ,Skeletal muscle ,Muscle weakness ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,medicine.symptom ,business - Abstract
Breast cancer is a disease of a specific organ, but its effects are felt throughout the body. The systemic effects of breast cancer can lead to functional limitations in patients who suffer from muscle weakness, fatigue, pain, fibromyalgia, or many other dysfunctions, which hasten cancer-associated death. Mechanistic studies have identified quite a few molecular defects in skeletal muscles that are associated with functional limitations in breast cancer. These include circulating cytokines such as TNF-α, IL-1, IL-6, and TGF-β altering the levels or function of myogenic molecules including PAX7, MyoD, and microRNAs through transcriptional regulators such as NF-κB, STAT3, and SMADs. Molecular defects in breast cancer may also include reduced muscle mitochondrial content and increased extracellular matrix deposition leading to energy imbalance and skeletal muscle fibrosis. This review highlights recent evidence that breast cancer-associated molecular defects mechanistically contribute to functional limitations and further provides insights into therapeutic interventions in managing functional limitations, which in turn may help to improve quality of life in breast cancer patients.
- Published
- 2020
- Full Text
- View/download PDF
158. Tonsoku-Like Protein Facilitates Chromatin Transcription Complex (TONSL-FACT): Illuminating Unsuspected Culprits in Breast Cancer Initiation
- Author
-
Fang Fang, Jacob K. Olson, Yunlong Liu, Rebecca C. Dirks, Xi Rao, Harikrishna Nakshatri, and Kenneth P. Nephew
- Subjects
Breast cancer ,business.industry ,Transcription preinitiation complex ,Cancer research ,medicine ,Surgery ,medicine.disease ,business ,Chromatin - Published
- 2019
- Full Text
- View/download PDF
159. Abstract 1006: Non-linear relationship between estradiol-regulated changes in chromatin architecture and gene expression
- Author
-
Yunlong Liu, Xiaona Chu, Yue Wang, Duojiao Chen, Poornima Bhat-Nakshatri, Harikrishna Nakshatri, and Taylor M. Parker
- Subjects
Regulation of gene expression ,Cancer Research ,Histone ,Oncology ,Cistrome ,biology ,Pioneer factor ,biology.protein ,FOXA1 ,Enhancer ,Chromatin remodeling ,Chromatin ,Cell biology - Abstract
Gene expression in estrogen receptor (ER)-positive breast cancer cells is predominantly driven by ER-estradiol (E2) signaling. However, the effects of ER-E2 signaling on the overall chromatin architecture and accessibility to transcription regulators are not well understood. Chromatin architecture is a key determinant to gene expression patterns and therefore a comprehensive notation is essential for the interpretation of the ER cistrome. To investigate the effects of E2 signaling on chromatin architecture, we employed Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq), a high-throughput method to map chromatin accessibility via a hyperactive transposase that inserts sequencing tags into open regions. ATAC-seq was performed on MCF-7 cells, an ERα+ breast cancer cell line, treated with E2 or vehicle control for 1 or 3 hours. Results from the ATAC-seq were compared to RNA-seq data of 3-hours E2-treated MCF-7 cells to determine which regions of E2-regulated open or closed chromatin coincide with increased or decreased respective gene expression. Interestingly, we observed greater chromatin accessibility changes in genes repressed by E2 compared to genes induced by E2. Integration of ATAC-seq and RNA-seq data revealed distinct categories of E2-induced chromatin remodeling and downstream gene expression including increased chromatin accessibility, but repression of the corresponding gene, as well as decreased chromatin accessibility, but induction of the corresponding gene. Validation of the chromatin configurations predicted by ATAC-seq of select corresponding genes is currently being performed by ChIP-qPCR against specific histone modifications, such as H3K27Ac, a marker of open chromatin. To mechanistically dissect the distinct modes of E2 chromatin remodelling, we performed motif enrichment analysis of chromatin regions affected by E2. This analysis showed expected enrichment of estrogen response element (ERE), and pioneer factor FOXA1 and GATA3 response elements in E2-inducible genes with enhanced chromatin accessibility after E2-treatment. Regions in which the corresponding mRNA showed decreased transcript levels but displayed open chromatin in their promoter or enhancer region were enriched for the PBX3 motif and the ERE motif, respectively. Our results reveal previously unrecognized modes of E2-mediated restructuring of chromatin architecture and its correlation with transcription dynamics. This study provides grounds for future work to study fundamental relationships between chromatin accessibility and gene regulation in breast cancer under E2 and anti-estrogen treated conditions as well as in metastatic breast cancers with ERα mutations. Citation Format: Taylor M. Parker, Duojiao Chen, Poornima Bhat-Nakshatri, Xiaona Chu, Yunlong Liu, Yue Wang, Harikrishna Nakshatri. Non-linear relationship between estradiol-regulated changes in chromatin architecture and gene expression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1006.
- Published
- 2019
- Full Text
- View/download PDF
160. Abstract P3-05-20: ESRP1 adds sp(l)ice to endocrine resistance
- Author
-
Harikrishna Nakshatri, Chirayu P. Goswami, Yesim Gökmen-Polar, Yaseswini Neelamraju, Sarath Chandra Janga, and Sunil Badve
- Subjects
Cancer Research ,medicine.diagnostic_test ,Fulvestrant ,Alternative splicing ,Estrogen receptor ,RNA-binding protein ,Biology ,medicine.disease ,Bioinformatics ,Breast cancer ,Oncology ,RNA splicing ,medicine ,Cancer research ,Oncotype DX ,Gene ,medicine.drug - Abstract
Introduction: De novo or acquired resistance to endocrine therapy limits its utility in a significant number of estrogen receptor (ER) breast cancers. An increasing number of molecular assays predict the likelihood of distant recurrence in tamoxifen-treated patients with node-negative, ER+ breast cancer. However, these do not provide the mechanistic basis for endocrine resistance. It is crucial to identify novel targets and improve the success of endocrine therapies. We previously shown that epithelial splicing regulatory proteins 1 and 2 (ESRP1 and ESRP2), RNA binding proteins that promote splicing, are significantly elevated in cases with high Oncotype DX scores (innate resistance) and in ERα-positive cells with acquired tamoxifen resistance (MCF-7 LCC2 cells) and fulvestrant and tamoxifen resistance (MCF-7/LCC9 cells). The aim of this study was to investigate the ESRP1/ESRP2-regulated alternative splicing events leading to innate and acquired endocrine resistance. Methods: A combinatorial bioinformatics approach was employed to identify genes altered through alternative splicing by ESRP1/ESRP2 in endocrine resistance. Briefly, genes with ESRP1/ESRP2 motifs were screened in the human genome. This data was integrated with tamoxifen-treated datasets, and filtered using protein-protein interactions (PPI; BIOGRID) and clinical outcome (KM plotter. Potential target gene transcripts were further narrowed down using an algorithm based on motif binding location and the Cancer Genome Atlas (TCGA) breast cancer datasets with low and high ESRP1 cases. The alternative transcripts were further validated using splice variant specific-custom qRT-PCR in low and high RS Oncotype cases as surrogate for endocrine sensitivity and resistance. Results: Motif analysis in combination with tamoxifen-treated datasets identified 2212 differentially expressed genes. Further collective analysis of number of PPI (>50) and survival data from KM plotter (P Conclusion: ESRP1/ESRP2 by inducing alternative splicing play an important role in tamoxifen resistance and recurrence of ER+ breast cancer. Targeting alternative splicing may offer novel avenues for combating endocrine-resistance in breast cancer. Citation Format: Yesim Gokmen-Polar, Yaseswini Neelamraju, Chirayu Pankaj Goswami, Harikrishna Nakshatri, Sarath Chandra Janga, Sunil Badve. ESRP1 adds sp(l)ice to endocrine resistance [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P3-05-20.
- Published
- 2015
- Full Text
- View/download PDF
161. Abstract P3-05-15: Divergent activation of AKT1 and AKT2 isoforms downstream of PI3K mutation impacts response of breast cancer cells to estradiol and PI3K inhibitors
- Author
-
Chirayu P. Goswami, Mathieu Lupien, Luca Magnani, Poornima Bhat-Nakshatri, Harikrishna Nakshatri, and Sunil Badve
- Subjects
Cancer Research ,Estrogen receptor ,AKT1 ,Cancer ,AKT2 ,Biology ,medicine.disease ,Breast cancer ,Oncology ,Cancer research ,medicine ,FOXA1 ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Background: PI3K mutations are observed in 30% of breast cancers, which are more common in estrogen receptor (ERα) positive breast cancers compared to ERα-negative breast cancers. AKT, a the major kinase downstream of PI3K, modulates ERα activity. It is unknown whether PI3K mutation leads to preferential activation of specific AKT isoform with ability to modulate ERα function. Results: We report elevated AKT1 but not AKT2 mRNA in breast cancers with PI3K mutation. Breast epithelial cells with targeted substitution of PI3K with PI3K-E545K or PI3K-H1047R demonstrated elevated AKT1_pS473 compared to AKT2_pS474, distinct AKT substrate phosphorylation, and enhanced Estrogen Receptor (ERα) activity. AKT1 had a dominant role in ERα:estradiol (E2)-dependent gene expression and proliferation. We have identified an unique gene expression signature in ERα-positive breast cancers that is dependent on ERα, estradiol (E2), AKT1, and the pioneer factor FOXA1. Elevated expression of this signature in ERα-positive tumor was associated with better response to endocrine therapy and outcome. In addition, AKT1 determined the sensitivity to PI3α-specific inhibitor BYL719 and pan-PI3K inhibitor BKM120. In contrast, AKT2 controlled global gene expression and elevated levels of an AKT2-directed protein signature predicted poor outcome in breast cancer patients. Conclusions: PI3K mutation favors AKT1 activation leading to imbalance in AKT isoform-specific kinome/proteome and an effect on specific signaling pathways, particularly ERα:E2 signaling network. Knowledge gained from this functional dissection of AKT isoform activity downstream of PI3K mutation can be exploited for therapeutic stratification, particularly for anti-estrogen and PI3K inhibitor-based therapies. Citation Format: Harikrishna Nakshatri, Chirayu Goswami, Sunil Badve, Luca Magnani, Mathieu Lupien, Poornima Bhat-Nakshatri. Divergent activation of AKT1 and AKT2 isoforms downstream of PI3K mutation impacts response of breast cancer cells to estradiol and PI3K inhibitors [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P3-05-15.
- Published
- 2015
- Full Text
- View/download PDF
162. Angiopoietin-2 mediates blood-brain barrier impairment and colonization of triple-negative breast cancer cells in brain
- Author
-
Haim Ovadia, Harikrishna Nakshatri, Shalom Avraham, Yigong Fu, Shuxian Jiang, and Hava Avraham
- Subjects
Pathology ,medicine.medical_specialty ,Tight junction ,business.industry ,Angiopoietin 2 ,medicine.disease ,Blood–brain barrier ,Pathology and Forensic Medicine ,medicine.anatomical_structure ,Breast cancer ,In vivo ,cardiovascular system ,medicine ,business ,Trebananib ,Triple-negative breast cancer ,Brain metastasis - Abstract
Although the incidence of breast cancer metastasis (BCM) in brain has increased significantly in triple-negative breast cancer (TNBC), the mechanisms remain elusive. Using in vivo mouse models for BCM in brain, we observed that TNBC cells crossed the blood-brain barrier (BBB), lodged in the brain microvasculature and remained adjacent to brain microvascular endothelial cells (BMECs). Breaching of the BBB in vivo by TNBCs resulted in increased BBB permeability and changes in ZO-1 and claudin-5 tight junction (TJ) protein structures. Angiopoietin-2 expression was elevated in BMECs and was correlated with BBB disruption. Secreted Ang-2 impaired TJ structures and increased BBB permeability. Treatment of mice with the neutralizing Ang-2 peptibody trebananib prevented changes in the BBB integrity and BMEC destabilization, resulting in inhibition of TNBC colonization in brain. Thus, Ang-2 is involved in initial steps of brain metastasis cascade, and inhibitors for Ang-2 may serve as potential therapeutics for brain metastasis.
- Published
- 2014
- Full Text
- View/download PDF
163. Abstract P1-05-05: Epithelial cell reprogramming assay reveals inter-individual heterogeneity in stem/basal and progenitor cell profile of the normal breast
- Author
-
Harikrishna Nakshatri, Manjushree Anjanappa, and Poornima Bhat-Nakshatri
- Subjects
Cancer Research ,Cell type ,education.field_of_study ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,Cell ,Population ,Biology ,Hyperplasia ,medicine.disease ,Molecular biology ,Flow cytometry ,medicine.anatomical_structure ,Oncology ,Cancer stem cell ,medicine ,Progenitor cell ,education ,Reprogramming - Abstract
Objectives: The majority of our understanding of mammary epithelial cell hierarchy is from mouse models. In addition, due to limited replication potential, primary cells of either mouse or human origin have rarely been used for functional studies. In this study, we took advantage of the recently developed epithelial cell-reprogramming assay that utilizes irradiated fibroblasts and ROCK inhibitor to characterize breast epithelial cells from individuals with varying degree of breast cancer risk. Methods: Primary breast tissues were collected at the time of surgery and cultured either immediately or at convenient time after freezing in liquid nitrogen with ROCK inhibitor. Flow cytometry sorting was employed to fractionate and propagate stem-like/basal (CD49f+/EpCAM-), luminal progenitor (CD49f+/EpCAM+), and mature (CD49f-/EpCAM+) cell enriched fractions. Similar fractionation/characterization was performed using CD133, CD10, CD271, EGFR and ALDEFLUOR. Results: Inter-individual variability was observed in proportion of CD49f+/EpCAM+, CD49-/EpCAM+ and CD133+ cells. Epithelial cells from a patient considered high risk based on the history of hyperplasia as well as cells from BRCA1 and BRCA2 patients displayed unique CD49f and EpCAM staining pattern compared to cells isolated from adjoining “normal” tissue of two cancer patients. In addition, CD133+ population was higher in BRCA1 and BRCA2 mutant patients. Morphologically, cells isolated from BRCA1 patient displayed higher levels of cells with mesenchymal phenotype. Upon short-term culturing of cells under regular 2D culture without reprogramming conditions, cell surface marker profile remained unchanged suggesting that most of the cell surface marker profile is cell intrinsic. CD49f+/EpCAM-, CD49f+/EpCAMhigh, CD49f+/EpCAMmedium, CD49f-/EpCAM+, CD133+/EpCAM+, and CD10+/EpCAM+ cells were amicable for further propagation in culture. CD49f+/EpCAMmedium cells cultured for two weeks remained CD49f+/EpCAMmedium. Additional marker profiling including cancer stem cell markers is underway to further refine stem, progenitor, and mature cells. Conclusion: Ability to propagate distinct subpopulation of normal cells from patients with different breast cancer risk would allow investigation of the impact of cell type origin of cancer on the course of the disease including response to therapy and the development of in vitro assay for breast cancer risk assessment. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P1-05-05.
- Published
- 2013
- Full Text
- View/download PDF
164. Abstract P5-09-09: ESRP1 and ESRP2 expression in tamoxifen resistance
- Author
-
Xiaoping Gu, Harikrishna Nakshatri, Yesim Gökmen-Polar, Chirayu Goswami, and Sunil Badve
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,In silico ,Estrogen receptor ,Cancer ,medicine.disease ,Endocrinology ,Breast cancer ,Internal medicine ,Gene expression ,medicine ,Hormonal therapy ,skin and connective tissue diseases ,Oncotype DX ,business ,Tamoxifen ,medicine.drug - Abstract
Background: Current standard of care therapies with antiestrogens targeting estrogen receptor α (ER) signaling improve disease-free survival (DFS) in early-stage of breast cancer. However a significant number of cases exhibit de novo or acquired endocrine resistance and recur. It is thus important to identify novel targets of resistance and select these patients for additional therapeutic options which might include continued/extended hormonal therapy. The Oncotype Dx recurrence score (RS) in current practice predicts the likelihood of distant recurrence in tamoxifen-treated patients with node-negative, ER+ breast cancer. However, Oncotype Dx does not provide the mechanistic basis for endocrine resistance. In this study, we aimed to investigate the impact of two epithelial splicing regulatory proteins 1 and 2 (ESRP1 and ESRP2) expression on recurrence and endocrine resistance such as tamoxifen of ER+ breast cancer. Methods: Publicly available gene expression datasets were analyzed for the overexpression of ESRP1 and ESRP2 in breast cancer. To validate the in silico findings for the ESRP1 and ESRP2 expression, we further performed quantitative real-time RT-PCR (qRT-PCR) in a cohort of 60 paraffin-embedded ER-positive node-negative breast carcinomas with low, intermediate, and high (19, 21, and 20 cases, respectively) Oncotype DX scores. To further determine the correlation between endocrine resistance and ESRP1/ESRP2 expression, we evaluated their expression levels in an established in vitro model of ER+/tamoxifen-resistance (MCF7/LCC2: an acquired tamoxifen resistant model of human breast cancer). Results: Using publicly available breast cancer gene expression datasets, we have identified that overexpression of the epithelial splicing regulatory proteins 1 and 2 (ESRP1 and ESRP2) correlates with worse prognosis in ER+ significantly, but not in ER- breast cancers. qRT-PCR analysis further revealed that ESRP1 and ESRP2 expressions are positively correlated with high Oncotype Dx scores (P Conclusion: These findings suggest a role for the elevated expression of ESRP1 and ESRP2 in tamoxifen resistance and recurrence of ER+ breast cancer. Further studies are ongoing to determine their mechanistic role in ER+ cancer and methods of targeting ESRP1 & 2. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P5-09-09.
- Published
- 2013
- Full Text
- View/download PDF
165. ANTXR1, a Stem Cell-Enriched Functional Biomarker, Connects Collagen Signaling to Cancer Stem-like Cells and Metastasis in Breast Cancer
- Author
-
Chirayu P. Goswami, Poornima Bhat-Nakshatri, Harikrishna Nakshatri, Sunil Badve, and Daohong Chen
- Subjects
Cancer Research ,Lung Neoplasms ,Blotting, Western ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Receptors, Cell Surface ,Collagen Type VI ,Real-Time Polymerase Chain Reaction ,Article ,Metastasis ,Mice ,Cancer stem cell ,Medullary breast carcinoma ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Breast ,RNA, Messenger ,Cells, Cultured ,Cell Proliferation ,Fluorescent Dyes ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Microfilament Proteins ,CD44 ,Wnt signaling pathway ,Cancer ,Flow Cytometry ,medicine.disease ,Neoplasm Proteins ,Survival Rate ,Wnt Proteins ,Phenotype ,Oncology ,Low Density Lipoprotein Receptor-Related Protein-6 ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Female ,Breast disease ,Stem cell - Abstract
Cancer stem-like cells are thought to contribute to tumor recurrence. The anthrax toxin receptor 1 (ANTXR1) has been identified as a functional biomarker of normal stem cells and breast cancer stem-like cells. Primary stem cell-enriched basal cells (CD49f+/EpCAM−/Lin−) expressed higher levels of ANTXR1 compared with mature luminal cells. CD49f+/EpCAM−, CD44+/EpCAM−, CD44+/CD24−, or ALDEFLUOR-positive subpopulations of breast cancer cells were enriched for ANTXR1 expression. CD44+/CD24−/ANTXR1+ cells displayed enhanced self-renewal as measured by mammosphere assay compared with CD44+/CD24−/ANTXR1− cells. Activation of ANTXR1 by its natural ligand C5A, a fragment of collagen VI α3, increased stem cell self-renewal in mammosphere assays and Wnt signaling including the expression of the Wnt receptor–lipoprotein receptor-related protein 6 (LRP6), phosphorylation of GSK3α/β, and elevated expression of Wnt target genes. RNAi-mediated silencing of ANTXR1 enhanced the expression of luminal-enriched genes but diminished Wnt signaling including reduced LRP6 and ZEB1 expression, self-renewal, invasion, tumorigenicity, and metastasis. ANTXR1 silencing also reduced the expression of HSPA1A, which is overexpressed in metastatic breast cancer stem cells. Analysis of public databases revealed ANTXR1 amplification in medullary breast carcinoma and overexpression in estrogen receptor-negative breast cancers with the worst outcome. Furthermore, ANTXR1 is among the 10% most overexpressed genes in breast cancer and is coexpressed with collagen VI. Thus, ANTXR1:C5A interactions bridge a network of collagen cleavage and remodeling in the tumor microenvironment, linking it to a stemness signaling network that drives metastatic progression. Cancer Res; 73(18); 5821–33. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
166. HOXB13 Mediates Tamoxifen Resistance and Invasiveness in Human Breast Cancer by Suppressing ERα and Inducing IL-6 Expression
- Author
-
Sunju Park, Soonweng Cho, Howard Y. Chang, Chirayu P. Goswami, Saraswati Sukumar, Zhe Zhang, Leslie Cope, Helen Sadik, Christopher B. Umbricht, Harikrishna Nakshatri, Nilay Shah, Kideok Jin, Rajnish A. Gupta, Ashley Cimino-Mathews, and Leigh Ann Cruz
- Subjects
Cancer Research ,Estrogen receptor ,Apoptosis ,Mice ,Breast ,Neoplasm Metastasis ,Phosphorylation ,Luciferases ,Promoter Regions, Genetic ,skin and connective tissue diseases ,STAT3 ,Cells, Cultured ,Regulation of gene expression ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,TOR Serine-Threonine Kinases ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Oncology ,Female ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,medicine.drug ,STAT3 Transcription Factor ,Chromatin Immunoprecipitation ,Antineoplastic Agents, Hormonal ,Blotting, Western ,Down-Regulation ,Mice, Nude ,Breast Neoplasms ,Real-Time Polymerase Chain Reaction ,Article ,Breast cancer ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,Transcription factor ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Homeodomain Proteins ,Interleukin-6 ,Cell growth ,Estrogen Receptor alpha ,medicine.disease ,Tamoxifen ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein - Abstract
Most breast cancers expressing the estrogen receptor α (ERα) are treated successfully with the receptor antagonist tamoxifen (TAM), but many of these tumors recur. Elevated expression of the homeodomain transcription factor HOXB13 correlates with TAM-resistance in ERα-positive (ER+) breast cancer, but little is known regarding the underlying mechanism. Our comprehensive evaluation of HOX gene expression using tiling microarrays, with validation, showed that distant metastases from TAM-resistant patients also displayed high HOXB13 expression, suggesting a role for HOXB13 in tumor dissemination and survival. Here we show that HOXB13 confers TAM resistance by directly downregulating ERα transcription and protein expression. HOXB13 elevation promoted cell proliferation in vitro and growth of tumor xenografts in vivo. Mechanistic investigations showed that HOXB13 transcriptionally upregulated interleukin (IL)-6, activating the mTOR pathway via STAT3 phosphorylation to promote cell proliferation and fibroblast recruitment. Accordingly, mTOR inhibition suppressed fibroblast recruitment and proliferation of HOXB13-expressing ER+ breast cancer cells and tumor xenografts, alone or in combination with TAM. Taken together, our results establish a function for HOXB13 in TAM resistance through direct suppression of ERα and they identify the IL-6 pathways as mediator of disease progression and recurrence. Cancer Res; 73(17); 5449–58. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
167. Individualized Breast Cancer Characterization through Single-Cell Analysis of Tumor and Adjacent Normal Cells
- Author
-
Manjushree Anjanappa, Harikrishna Nakshatri, George E. Sandusky, Angelo A. Cardoso, Safa F. Mohamad, Edward F. Srour, Andrea M. Gunawan, Lijun Cheng, Susan Rice, Yan Dong, and Lang Li
- Subjects
0301 basic medicine ,Cancer Research ,Breast Neoplasms ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Flow cytometry ,03 medical and health sciences ,Single-cell analysis ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Cluster Analysis ,Humans ,Gene Regulatory Networks ,Progenitor cell ,Precision Medicine ,Clonogenic assay ,medicine.diagnostic_test ,Gene Expression Profiling ,Epithelial Cells ,Genomics ,Flow Cytometry ,Phenotype ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Cell culture ,Immunology ,Cancer research ,Female ,Single-Cell Analysis ,Reprogramming - Abstract
There is a need to individualize assays for tumor molecular phenotyping, given variations in the differentiation status of tumor and normal tissues in different patients. To address this, we performed single-cell genomics of breast tumors and adjacent normal cells propagated for a short duration under growth conditions that enable epithelial reprogramming. Cells analyzed were either unselected for a specific subpopulation or phenotypically defined as undifferentiated and highly clonogenic ALDH+/CD49f+/EpCAM+ luminal progenitors, which express both basal cell and luminal cell–enriched genes. We analyzed 420 tumor cells and 284 adjacent normal cells for expression of 93 genes that included a PAM50-intrinsic subtype classifier and stemness-related genes. ALDH+/CD49f+/EpCAM+ tumor and normal cells clustered differently compared with unselected tumor and normal cells. PAM50 gene-set analyses of ALDH+/CD49f+/EpCAM+ populations efficiently identified major and minor clones of tumor cells, with the major clone resembling clinical parameters of the tumor. Similarly, a stemness-associated gene set identified clones with divergent stemness pathway activation within the same tumor. This refined expression profiling technique distinguished genes truly deregulated in cancer from genes that identify cellular precursors of tumors. Collectively, the assays presented here enable more precise identification of cancer-deregulated genes, allow for early identification of therapeutically targetable tumor cell subpopulations, and ultimately provide a refinement of precision therapeutics for cancer treatment. Cancer Res; 77(10); 2759–69. ©2017 AACR.
- Published
- 2016
168. Statistical modeling for sensitive detection of low-frequency single nucleotide variants
- Author
-
Yangyang Hao, Yunlong Liu, Howard J. Edenberg, Harikrishna Nakshatri, Xiaoling Xuei, Pengyue Zhang, and Lang Li
- Subjects
0301 basic medicine ,Systematic error ,Sequence analysis ,Pooling ,Sequencing data ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Genetics ,Humans ,Models, Statistical ,Base Sequence ,Research ,Chromosome Mapping ,High-Throughput Nucleotide Sequencing ,Statistical model ,Sequence Analysis, DNA ,Cancer Early Detection ,3. Good health ,030104 developmental biology ,DNA microarray ,Algorithms ,Software ,Biotechnology ,Count data - Abstract
Background Sensitive detection of low-frequency single nucleotide variants carries great significance in many applications. In cancer genetics research, tumor biopsies are a mixture of normal and tumor cells from various subpopulations due to tumor heterogeneity. Thus the frequencies of somatic variants from a subpopulation tend to be low. Liquid biopsies, which monitor circulating tumor DNA in blood to detect metastatic potential, also face the challenge of detecting low-frequency variants due to the small percentage of the circulating tumor DNA in blood. Moreover, in population genetics research, although pooled sequencing of a large number of individuals is cost-effective, pooling dilutes the signals of variants from any individual. Detection of low frequency variants is difficult and can be cofounded by sequencing artifacts. Existing methods are limited in sensitivity and mainly focus on frequencies around 2 % to 5 %; most fail to consider differential sequencing artifacts. Results We aimed to push down the frequency detection limit close to the position specific sequencing error rates by modeling the observed erroneous read counts with respect to genomic sequence contexts. 4 distributions suitable for count data modeling (using generalized linear models) were extensively characterized in terms of their goodness-of-fit as well as the performances on real sequencing data benchmarks, which were specifically designed for testing detection of low-frequency variants; two sequencing technologies with significantly different chemistry mechanisms were used to explore systematic errors. We found the zero-inflated negative binomial distribution generalized linear mode is superior to the other models tested, and the advantage is most evident at 0.5 % to 1 % range. This method is also generalizable to different sequencing technologies. Under standard sequencing protocols and depth given in the testing benchmarks, 95.3 % recall and 79.9 % precision for Ion Proton data, 95.6 % recall and 97.0 % precision for Illumina MiSeq data were achieved for SNVs with frequency > = 1 %, while the detection limit is around 0.5 %. Conclusions Our method enables sensitive detection of low-frequency single nucleotide variants across different sequencing platforms and will facilitate research and clinical applications such as pooled sequencing, cancer early detection, prognostic assessment, metastatic monitoring, and relapses or acquired resistance identification. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2905-x) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
169. Molecular insights of pathways resulting from two common PIK3CA mutations in breast cancer
- Author
-
Luca Magnani, Harikrishna Nakshatri, Chirayu P. Goswami, Sunil Badve, Mathieu Lupien, Poornima Bhat-Nakshatri, and Imperial College London
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,AKT1 ,Breast Neoplasms ,P110α ,medicine.disease_cause ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,Breast cancer ,Internal medicine ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,Gene Regulatory Networks ,RNA, Messenger ,Oncology & Carcinogenesis ,Protein kinase B ,neoplasms ,PI3K/AKT/mTOR pathway ,Mutation ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Estrogen Receptor alpha ,Cancer ,medicine.disease ,030104 developmental biology ,Cancer research ,Female ,business ,Estrogen receptor alpha ,Proto-Oncogene Proteins c-akt ,1112 Oncology And Carcinogenesis ,Signal Transduction - Abstract
The PI3K pathway is activated in approximately 70% of breast cancers. PIK3CA gene mutations or amplifications that affect the PI3K p110α subunit account for activation of this pathway in 20% to 40% of cases, particularly in estrogen receptor alpha (ERα)-positive breast cancers. AKT family of kinases, AKT1–3, are the downstream targets of PI3K and these kinases activate ERα. Although several inhibitors of PI3K have been developed, none has proven effective in the clinic, partly due to an incomplete understanding of the selective routing of PI3K signaling to specific AKT isoforms. Accordingly, we investigated in this study the contribution of specific AKT isoforms in connecting PI3K activation to ERα signaling, and we also assessed the utility of using the components of PI3K–AKT isoform–ERα signaling axis as predictive biomarkers of response to PI3K inhibitors. Using a variety of physiologically relevant model systems with defined natural or knock-in PIK3CA mutations and/or PI3K hyperactivation, we show that PIK3CA-E545K mutations (found in ∼20% of PIK3CA-mutant breast cancers), but not PIK3CA-H1047R mutations (found in 55% of PIK3CA-mutant breast cancers), preferentially activate AKT1. Our findings argue that AKT1 signaling is needed to respond to estrogen and PI3K inhibitors in breast cancer cells with PIK3CA-E545K mutation, but not in breast cancer cells with other PIK3CA mutations. This study offers evidence that personalizing treatment of ER-positive breast cancers to PI3K inhibitor therapy may benefit from an analysis of PIK3CA–E545K–AKT1–estrogen signaling pathways. Cancer Res; 76(13); 3989–4001. ©2016 AACR.
- Published
- 2016
170. Abstract P6-07-08: Dual TGFβ/BMP inhibition allows in vitro expansion of multiple cell types from normal and cancerous breast
- Author
-
Kathy D. Miller, Poornima Bhat-Nakshatri, Amv Storniolo, Harikrishna Nakshatri, M Ananappa, Brijesh Kumar, Prasad, and Yunlong Liu
- Subjects
Cancer Research ,Stromal cell ,biology ,Chemistry ,CD44 ,Mesenchymal stem cell ,Myoepithelial cell ,Oncology ,biology.protein ,Cancer research ,CD90 ,Stem cell ,ALCAM ,Adult stem cell - Abstract
Functional modeling of breast epithelial hierarchy and stromal-epithelial cell interactions has been difficult due to inability to obtain sufficient stem-progenitor-mature epithelial cells and stromal cells. The recently developed epithelial reprogramming assay has partially overcome this limitation, allowing propagation of epithelial cells with stem, luminal progenitor and mature cell features. However, characterizing stromal cells using this assay is difficult because irradiated fibroblasts which can be difficult to distinguish from stromal cells are needed as feeder layer. A recent study demonstrated expansion of airway basal stem cells without a feeder layer through pharmacologic inhibition of TGFβ/BMP/SMAD signaling. We sought to develop this method for culture and expansion of cells from normal and cancerous breast samples. With appropriate modifications to growth media, we were able to obtain normal and stromal cells from breast biopsies of healthy women. The expanded cell population included CD10+/EpCAM- basal/myoepithelial cells, CD49f+/EpCAM+ luminal progenitor cells, CD49f-/EpCAM+ mature luminal cells, CD73+/EpCAM+/CD90- rare endogenous pluripotent somatic stem cells, CD73+/CD90+/EpCAM- mesenchymal stem cells, ALCAM (CD166)+/EpCAM+ cells, CD44+/CD24- cells, CD44+/CD24+ cells and ALDFLUOR+ stem/luminal progenitor cells. Epithelial cells were KRT14+, KRT19+ or both further documenting heterogeneity within epithelial cell population. We have extended this technique to grow breast epithelial cells from high-risk patients including BRCA1 mutant-carriers, tumor-adjacent normal and tumor cells from the same patient, pleural effusions and liver metastasis from breast cancer patients. Phenotypic characterization showed differences in the differentiation state of adjacent-normal and tumor cells. Tumor cells from pleural effusions showed remarkable phenotypic heterogeneity with a fraction of these cells expressing estrogen receptor. The assay described here, therefore, is versatile and provides resources to model epithelial-stromal interactions under normal and cancerous conditions as well as for genomics and screening of drugs to target metastasis on an individual level. Susan G. Komen for the Cure and Department of Defense supported this work. Citation Format: Nakshatri H, Ananappa M, Prasad MS, Kumar B, Liu Y, Storniolo AM, Miller KD, Bhat-Nakshatri P. Dual TGFβ/BMP inhibition allows in vitro expansion of multiple cell types from normal and cancerous breast [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P6-07-08.
- Published
- 2018
- Full Text
- View/download PDF
171. FOXA1 is an independent prognostic marker for ER-positive breast cancer
- Author
-
Rutika Mehta, David G. Huntsman, Torsten O. Nielsen, Harikrishna Nakshatri, Rohit Jain, Jennifer R. Choo, Samuel Leung, and Sunil Badve
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Antineoplastic Agents, Hormonal ,Estrogen receptor ,Breast Neoplasms ,Biology ,Basal (phylogenetics) ,Breast cancer ,Recurrence ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Survival analysis ,Tissue microarray ,Prognosis ,medicine.disease ,Survival Analysis ,Tamoxifen ,Receptors, Estrogen ,Immunohistochemistry ,Female ,FOXA1 ,medicine.drug - Abstract
Forkhead box protein A1 (FOXA1) is a "pioneer factor" that plays a role in controlling nearly 50% of estrogen receptor target genes. FOXA1 expression correlates with estrogen receptor (ER)-positivity especially in luminal subtype A breast cancers. The aim of this study was to investigate the precise role of FOXA1 in breast cancer using a large population-based cohort. Nuclear expression of FOXA1 was analyzed in a tissue microarray of 4,444 invasive breast cancer cases using immunohistochemistry and correlated with clinicopathologic variables using previously described methods and cutoff points. The entire cohort was equally divided into a training and validation set. All survival analyses were performed using a previously defined cutoff (3) for validation. Additional X-tile analysis performed to analyze prognostic effects of low and high FOXA1 levels identified 24 as a cutoff. Bonferroni-Holmes test was used as appropriate. FOXA1 expression significantly correlated positively with markers of good prognosis or ER-positivity, and negatively with tumor size, tumor grade, nodal status, Ki67, HER2 expression, and basal subtype (each P value
- Published
- 2011
- Full Text
- View/download PDF
172. High-level expression of forkhead-box protein A1 in metastatic prostate cancer
- Author
-
Harikrishna Nakshatri, Rohit Jain, Muhammad T. Idrees, Sunil Badve, and Rutika Mehta
- Subjects
PCA3 ,Oncology ,Intraepithelial neoplasia ,medicine.medical_specialty ,Histology ,business.industry ,GATA3 ,Perineural invasion ,General Medicine ,medicine.disease ,Pathology and Forensic Medicine ,Prostate cancer ,medicine.anatomical_structure ,Prostate ,Internal medicine ,Medicine ,FOXA1 ,business ,Lymph node - Abstract
Jain R K, Mehta R J, Nakshatri H, Idrees M T & Badve S S (2011) Histopathology58, 766–772 High-level expression of forkhead-box protein A1 in metastatic prostate cancer Aims: Expression of forkhead-box protein A1 (FOXA1), a transcription factor important for normal development of the prostate gland, is thought to be controlled by steroid hormones and GATA3. The aim of this study was to investigate the expression and potential role of FOXA1 and GATA3 transcription factors as prognostic factors in prostate cancer. Methods and results: Expression of FOXA1, GATA3 and androgen receptor (AR) was retrospectively analysed by immunohistochemistry in a series of 80 primary tumours and 28 metastatic prostate cancers, including 15 matched paired samples. Nuclear AR expression did not significantly differ between primary and metastatic tumours. High-level nuclear FOXA1 expression was seen in 19% of primary and 89% of metastatic tumours (P
- Published
- 2011
- Full Text
- View/download PDF
173. Control of EVI-1 oncogene expression in metastatic breast cancer cells through microRNA miR-22
- Author
-
J B Patel, Guohua Wang, Michael J. Thomson, Harikrishna Nakshatri, Hitesh Nidumanda Appaiah, Scott M. Hammond, Rutika Mehta, Sunil Badve, P. S. Steeg, Yunlong Liu, Poornima Bhat-Nakshatri, and Riesa M. Burnett
- Subjects
CA15-3 ,Cancer Research ,Mice, Nude ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Metastasis ,Mice ,Breast cancer ,Cancer stem cell ,Cell Line, Tumor ,Proto-Oncogenes ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Cancer ,medicine.disease ,Metastatic breast cancer ,MDS1 and EVI1 Complex Locus Protein ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Receptors, Estrogen ,Cancer cell ,Cancer research ,Female ,Carcinogenesis ,Neoplasm Transplantation ,Transcription Factors - Abstract
Metastasis in breast cancer carries a disproportionately worse prognosis than localized primary disease. To identify microRNAs (miRNA) involved in metastasis, the expression of 254 miRNAs was measured across the following cell lines using microarray analysis: MDA-MB-231 breast cancer cells, cells that grew as a tumor in the mammary fat pad of nude mice (TMD-231), metastatic disease to the lungs (LMD-231), bone (BMD-231) and adrenal gland (ADMD-231). A brain-seeking variant of this cell line (231-BR) was used additionally in validation studies. Twenty miRNAs were upregulated and seven were downregulated in metastatic cancer cells compared with TMD-231 cells. The expression of the tumor suppressor miRNAs let-7 and miR-22 was consistently downregulated in metastatic cancer cells. These metastatic cells expressed higher levels of putative/proven miR-22 target oncogenes ERBB3, CDC25C and EVI-1. Introduction of miR-22 into cancer cells reduced the levels of ERBB3 and EVI-1 as well as phospho-AKT, an EVI-1 downstream target. The miR-22 primary transcript is located in the 5'-untranslated region of an open reading frame C17orf91, and the promoter/enhancer of C17orf91 drives miR-22 expression. We observed elevated C17orf91 expression in non-basal subtype compared with basal subtype breast cancers. In contrast, elevated expression of EVI-1 was observed in basal subtype and was associated with poor outcome in estrogen receptor-negative breast cancer patients. These results suggest that metastatic cancer cells increase specific oncogenic signaling proteins through downregulation of miRNAs. Identifying such metastasis-specific oncogenic pathways may help to manipulate tumor behavior and aid in the design of more effective targeted therapies.
- Published
- 2010
- Full Text
- View/download PDF
174. ITF2 is a target of CXCR4 in MDA-MB-231 breast cancer cells and is associated with reduced survival in estrogen receptor-negative breast cancer
- Author
-
Poornima Bhat-Nakshatri, Rutika Mehta, Harikrishna Nakshatri, Hitesh Nidumanda Appaiah, Sunil Badve, and Mangesh A. Thorat
- Subjects
Receptors, CXCR4 ,Cancer Research ,Transplantation, Heterologous ,Mice, Nude ,Estrogen receptor ,Breast Neoplasms ,Kaplan-Meier Estimate ,Biology ,CXCR4 ,Cell Line ,Metastasis ,Mice ,Chemokine receptor ,Transcription Factor 4 ,Breast cancer ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Transcription factor ,Oligonucleotide Array Sequence Analysis ,Pharmacology ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Reverse Transcriptase Polymerase Chain Reaction ,Microarray analysis techniques ,Gene Expression Profiling ,Mammary Neoplasms, Experimental ,Flow Cytometry ,medicine.disease ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Receptors, Estrogen ,Oncology ,Cancer research ,Molecular Medicine ,Female ,Transcription Factors - Abstract
CXCR4, a chemokine receptor, plays an important role in breast cancer growth, invasion, and metastasis. The transcriptional targets of CXCR4 signaling are not known. Microarray analysis of CXCR4-enriched and CXCR4-low subpopulations of the MDA-MB-231 breast cancer cell line, which has a constitutively active CXCR4 signaling network, revealed differential expression of ∼ 200 genes in the CXCR4-enriched subpopulation. ITF2, upregulated in CXCR4-enriched cells, was investigated further. Expression array datasets of primary breast tumors revealed higher ITF2 expression in estrogen receptor negative tumors, which correlated with reduced progression free and overall survival and suggested its relevance in breast cancer progression. CXCL12, a CXCR4 ligand, increased ITF2 expression in MDA-MB-231 cells. ITF2 is a basic helix-loop-helix transcription factor that controls the epithelial-to-mesenchymal transition and the function of the ID family (inhibitor-of-differentiation) of transcription factors, such as ID2. ID2 promotes differentiation of breast epithelial cells and its reduced expression in breast cancer is associated with an unfavorable prognosis. Both CXCR4 and ITF2 repressed ID2 expression. In xenograft studies, CXCR4-enriched cells formed large tumors and exhibited significantly elevated lung metastasis. Short interfering RNA against ITF2 reduced invasion of the CXCR4-enriched MDA-MB-231 subpopulation, whereas ITF2 overexpression restored the invasive capacity of MDA-MB-231 cells expressing CXCR4shRNA. Furthermore, overexpression of ITF2 in these cells enhanced tumor growth. We propose that ITF2 is one of the CXCR4 targets, which is involved in CXCR4-dependent tumor growth and invasion of breast cancer cells.
- Published
- 2010
- Full Text
- View/download PDF
175. Abstract 4993: Breast epithelial cell lines from normal breast with luminal and intrinsic subtypes -enriched gene expression document inter-individual differences in differentiation cascade
- Author
-
Harikrishna Nakshatri, Jun Wan, Yunlong Liu, Sheng Liu, Mayuri S. Prasad, Anna Maria Storniolo, Natascia Marino, Brijesh Kumar, Manjushree Anjanappa, Xi Rao, and Poornima Nakshatri
- Subjects
Cancer Research ,Matrigel ,education.field_of_study ,Cell signaling ,Cell ,Population ,Cancer ,Biology ,medicine.disease ,medicine.anatomical_structure ,Breast cancer ,Oncology ,Cell culture ,Cancer research ,medicine ,education ,Immortalised cell line - Abstract
Breast cancers are classified into five intrinsic subtypes based on gene expression profile. It is suggested that these intrinsic subtypes originate from specific developmental stage of breast epithelial cell hierarchy, stem-progenitor-mature cell. However, normal breast epithelial cell lines representing these intrinsic subtypes are yet to be created. Using normal breast tissues of ancestry-mapped Caucasian, African American, and Hispanic women and a primary cell culturing system that allows growth of normal epithelial cells of different developmental stages including estrogen receptor-positive mature luminal cells, we created 15 human telomerase-immortalized breast epithelial cell lines. These cells formed acini on a matrigel and ductal structures on 3-dimensional collagen or hydrogel, indicating that these cell lines have retained characteristics of normal breast epithelial cells. RNA sequencing and PAM50 intrinsic subtype clustering algorithms were used to identify the intrinsic subtypes of the immortalized cell lines together with two well characterized “normal” breast epithelial cell lines MCF10A and HMEC as well as luminal breast cancer cell line MCF-7. Unlike MCF10A and HMEC, which are enriched for basal-like gene expression pattern, our cell lines classified into luminal A, basal, and normal-like subtypes. This was also reflected in the immunofluorescence staining with basal marker KRT14 and luminal marker KRT19. Few of these cell lines were dual positive for KRT14 and KRT19, but in varying proportions. Cell lines representing claudin-low subtypes were also created, which are phenotypically (CD201+/EpCAM-) different from the above cell lines. Cell lines showed inter-individual differences in stemness/differentiation capabilities and variable basal activity of signaling molecules such as NF-kB, AP-1 and pERK, which is consistent with, possibly reflecting, recent discoveries of genetic variations in gene regulatory regions among general population that contribute to widespread differences in gene expression/signaling under “normal” state. As majority of breast cancers are believed to originate from luminal progenitor cells, which are well represented our cell lines, these cell lines are ideal to delineate the impact of inter-individual and ethnic differences in normal breast biology on breast cancer initiation and progression as well as to determine whether cell-type-origin instead of genomic aberration drives intrinsic subtype-enriched gene expression patterns in breast tumors. Citation Format: Brijesh Kumar, Mayuri Prasad, Manjushree Anjanappa, Poornima Nakshatri, Natascia Marino, Anna Maria Storniolo, Xi Rao, Sheng Liu, Jun Wan, Yunlong Liu, Harikrishna Nakshatri. Breast epithelial cell lines from normal breast with luminal and intrinsic subtypes -enriched gene expression document inter-individual differences in differentiation cascade [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4993.
- Published
- 2018
- Full Text
- View/download PDF
176. A water soluble parthenolide analog suppresses in vivo tumor growth of two tobacco‐associated cancers, lung and bladder cancer, by targeting NF‐κB and generating reactive oxygen species
- Author
-
James E. Klaunig, Praveen Kusumanchi, Peter A. Crooks, Rajasubramaniam Shanmugam, Hitesh Appaiah, Tyler Peat, Harikrishna Nakshatri, Christopher Sweeney, Sundar Neelakantan, William Matthews, and Liang Cheng
- Subjects
Male ,Cancer Research ,Programmed cell death ,Lung Neoplasms ,Nitrosamines ,Cell ,Mice, Nude ,Electrophoretic Mobility Shift Assay ,Biology ,Article ,Mice ,chemistry.chemical_compound ,Cyclin D1 ,Tobacco ,medicine ,Animals ,Parthenolide ,RNA, Small Interfering ,Carcinogen ,DNA Primers ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,NF-kappa B ,Cell cycle ,Flow Cytometry ,XIAP ,medicine.anatomical_structure ,Urinary Bladder Neoplasms ,Oncology ,chemistry ,Apoptosis ,Immunology ,Carcinogens ,Cancer research ,Reactive Oxygen Species ,Sesquiterpenes ,Cell Division - Abstract
Dimethylaminoparthenolide (DMAPT) is a water soluble parthenolide analog with preclinical activity in hematologic malignancies. Using non-small lung cancer (NSCLC) cell lines (A549 and H522) and an immortalized human bronchial epithelial cell line (BEAS2B) and TCC cell lines (UMUC-3, HT-1197 and HT-1376) and a bladder papilloma (RT-4), we aimed to characterize DMAPT's anticancer activity in tobacco-associated neoplasms. Flow cytometric, electrophoretic mobility gel shift assays (EMSA), and Western blot studies measured generation of reactive oxygen species (ROS), inhibition of NFκB DNA binding, and changes in cell cycle distribution and apoptotic proteins. DMAPT generated ROS with subsequent JNK activation and also decreased NFκB DNA binding and antiapoptotic proteins, TRAF-2 and XIAP. DMAPT-induced apoptotic cell death and altered cell cycle distribution with upregulation of p21 and p73 levels in a cell type-dependent manner. DMAPT suppressed cyclin D1 in BEAS2B. DMAPT retained NFκB and cell cycle inhibitory activity in the presence of the tobacco carcinogen nitrosamine ketone, 4(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Using a BrdU accumulation assay, 5-20 μM of DMAPT was shown to inhibit cellular proliferation of all cell lines by more than 95%. Oral dosing of DMAPT suppressed in vivo A549 and UMUC-3 subcutaneous xenograft growth by 54% (p = 0.015) and 63% (p < 0.01), respectively, and A549 lung metastatic volume by 28% (p = 0.043). In total, this data demonstrates DMAPT's novel anticancer properties in both early and late stage tobacco-associated neoplasms as well as its significant in vivo activity. The data provides support for the conduct of clinical trials in TCC and NSCLC.
- Published
- 2010
- Full Text
- View/download PDF
177. Subcellular Localization of Activated AKT in Estrogen Receptor- and Progesterone Receptor-Expressing Breast Cancers
- Author
-
Mangesh A. Thorat, Shikha Bose, Dmitry Turbin, Sandra E. Dunn, Jason S. Carroll, Samuel Leung, Tim R. Geistlinger, Harikrishna Nakshatri, Poornima Bhat-Nakshatri, Sunil Badve, Myles Brown, Nikail Collins, and Michael A. Teitell
- Subjects
Estrogen receptor ,Cancer ,AKT1 ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,Progesterone receptor ,Cancer research ,medicine ,Breast disease ,FOXA1 ,Protein kinase B ,Tamoxifen ,medicine.drug - Abstract
Activated v-AKT murine thymoma viral oncogene homolog 1 (AKT)/protein kinase B (PKB) kinase (pAKT) is localized to the plasma membrane, cytoplasm, and/or nucleus in 50% of cancers. The clinical importance of pAKT localization and the mechanism(s) controlling this compartmentalization are unknown. In this study, we examined nuclear and cytoplasmic phospho-AKT (pAKT) expression by immunohistochemistry in a breast cancer tissue microarray (n = 377) with ≈15 years follow-up and integrated these data with the expression of estrogen receptor (ER)α, progesterone receptor (PR), and FOXA1. Nuclear localization of pAKT (nuclear-pAKT) was associated with long-term survival (P = 0.004). Within the ERα+/PR+ subgroup, patients with nuclear-pAKT positivity had better survival than nuclear-pAKT–negative patients (P ≤ 0.05). The association of nuclear-pAKT with the ERα+/PR+ subgroup was validated in an independent cohort (n = 145). TCL1 family proteins regulate nuclear transport and/or activation of AKT. TCL1B is overexpressed in ERα-positive compared with ERα-negative breast cancers and in lung metastasis–free breast cancers. Therefore, we examined the possible control of TCL1 family member(s) expression by the estrogen:ERα pathway. Estradiol increased TCL1B expression and increased nuclear-pAKT levels in breast cancer cells; short- interfering RNA against TCL1B reduced nuclear-pAKT. Overexpression of nuclear-targeted AKT1 in MCF-7 cells increased cell proliferation without compromising sensitivity to the anti-estrogen, tamoxifen. These results suggest that subcellular localization of activated AKT plays a significant role in determining its function in breast cancer, which in part is dependent on TCL1B expression.
- Published
- 2010
- Full Text
- View/download PDF
178. A water-soluble parthenolide analogue suppresses in vivo prostate cancer growth by targeting NFκB and generating reactive oxygen species
- Author
-
Harikrishna Nakshatri, Sundar Neelakantan, Rajasubramaniam Shanmugam, Praveen Kusumanchi, Peter A. Crooks, Liang Cheng, William Matthews, and Christopher Sweeney
- Subjects
chemistry.chemical_classification ,medicine.medical_specialty ,Reactive oxygen species ,Programmed cell death ,medicine.diagnostic_test ,Urology ,Biology ,Flow cytometry ,Small hairpin RNA ,chemistry.chemical_compound ,Endocrinology ,Oncology ,chemistry ,Apoptosis ,In vivo ,Internal medicine ,medicine ,Cancer research ,Parthenolide ,Viability assay - Abstract
BACKGROUND To characterize the molecular changes associated with DMAPT-induced prostate cancer cell death and its in vivo activity. METHODS CWR22Rv1 and PC-3 were subjected to flow cytometry, electrophoretic mobility shift assays, and Western blot studies to measure DMAPT's ability to generate reactive oxygen species (ROS), inhibit NFκB DNA binding, and cause changes in anti-apoptotic proteins. N-acetyl cysteine (NAC) and short hairpin RNA (shRNA) were used to determine the contribution of ROS and JNK2 activation, respectively. The BrdU incorporation assay was used to measure proliferation and trypan blue studies assessed cell viability after DMAPT treatment. The in vivo activity of DMAPT as a single agent and in combination with bicalutamide or docetaxel was assessed in a subcutaneous xenograft model with athymic nude female mice. RESULTS DMAPT generated ROS with subsequent JNK activation and inhibited NFκB DNA binding and expression of NFκB-regulated anti-apoptotic proteins. DMAPT increased necrotic and apoptotic cell death in a cell-type-dependent manner and both types of cell death were blocked by NAC. Additionally, shRNA JNK2 partially blocked the anti-proliferative activity of DMAPT. DMAPT inhibited CWR22Rv1 and PC-3 cellular proliferation by 100% with 10 and 20 µM respectively and in vivo, DMAPT was more effective at inhibiting growth than biclutamide (CWR22v1) and docetaxel (PC-3). CONCLUSIONS DMAPT promotes cell death by both generating ROS and inhibition of NFκB. Its in vivo activity supports the conduct of clinical trials in patients with castrate-resistant disease. Prostate 70: 1074–1086, 2010. © 2010 Wiley-Liss, Inc.
- Published
- 2010
- Full Text
- View/download PDF
179. Expression of Forkhead-box protein A1, a marker of luminal A type breast cancer, parallels low Oncotype DX 21-gene recurrence scores
- Author
-
Mangesh A. Thorat, Foluso O. Ademuyiwa, Harikrishna Nakshatri, Sunil Badve, and Rohit Jain
- Subjects
Adult ,Hepatocyte Nuclear Factor 3-alpha ,Pathology ,medicine.medical_specialty ,Gene Expression ,Estrogen receptor ,Breast Neoplasms ,GATA3 Transcription Factor ,Pathology and Forensic Medicine ,Breast cancer ,Progesterone receptor ,Biomarkers, Tumor ,medicine ,Humans ,Pathology, Molecular ,Aged ,medicine.diagnostic_test ,business.industry ,Gene Expression Profiling ,GATA3 ,Middle Aged ,Progesterone Receptor Status ,Prognosis ,medicine.disease ,Immunohistochemistry ,Cohort ,Female ,Reagent Kits, Diagnostic ,Neoplasm Recurrence, Local ,FOXA1 ,business ,Oncotype DX - Abstract
The Oncotype DX assay is one of the molecular tests that provide predictive and prognostic information to breast cancer patients with estrogen receptor (ER)-positive and node-negative disease. This study evaluates the association of Forkhead-box protein A1 (FOXA1) and GATA-binding protein 3 (GATA3) expressions with Oncotype DX recurrences scores in 77 cases of patients with ER-positive node-negative breast carcinomas diagnosed at Indiana University. The data were correlated with patient age, tumor size, histologic type, Scarff-Bloom-Richardson score, histologic grade, and progesterone receptor status. The median FOXA1 and GATA3 scores were 240 and 200, respectively. The Oncotype DX recurrence scores were low in 57%, intermediate in 30%, and high in 13% of cases. FOXA1 expression correlated negatively with Oncotype DX recurrence scores (P=0.004), and histologic type (P=0.0004). Oncotype DX recurrences score also correlated negatively with progesterone receptor (P=0.035) with 100% of progesterone receptor-negative cases having high or intermediate Oncotype DX scores. FOXA1 and GATA3 expressions correlated positively (P=0.014). The correlation between FOXA1 expression and Oncotype DX recurrence scores remained significant after adjusting for multiple comparisons and controlling for confounders such as histological type, grade, and progesterone receptor. A statistically significant correlation between the Oncotype DX recurrence scores and FOXA1 expression in our diverse cohort of ER-positive breast cancer patients was observed. We propose that this may represent a more cost-effective strategy to further risk stratify patients with good prognosis in whom chemotherapy may be omitted. To confirm these findings, further studies in a larger cohort of patients are warranted.
- Published
- 2010
- Full Text
- View/download PDF
180. NF-κB inhibition in human hepatocellular carcinoma and its potential as adjunct to sorafenib based therapy
- Author
-
Menggang Yu, Jian Min Wu, Mary A. Maluccio, Harikrishna Nakshatri, Hongmiao Sheng, Romil Saxena, Poornima Bhat-Nakshatri, and Nicholas J. Skill
- Subjects
Niacinamide ,Sorafenib ,Cancer Research ,Programmed cell death ,Carcinoma, Hepatocellular ,MAP Kinase Signaling System ,Pyridines ,Antineoplastic Agents ,Apoptosis ,chemistry.chemical_compound ,NF-KappaB Inhibitor alpha ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Neoplasm Invasiveness ,neoplasms ,Cell Proliferation ,Cell growth ,business.industry ,Phenylurea Compounds ,Benzenesulfonates ,Liver Neoplasms ,NF-kappa B ,Transcription Factor RelA ,medicine.disease ,digestive system diseases ,Vascular endothelial growth factor ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,chemistry ,Cell culture ,Hepatocellular carcinoma ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,I-kappa B Proteins ,Signal transduction ,business ,Signal Transduction ,medicine.drug - Abstract
Nuclear factor-kappaB (NF-kappaB) has been shown to play an important role in the development and progression of cancer. In this study, we systematically examined NF-kappaBp65 signaling pathway in both human hepatocellular carcinoma (HCC) tissue and HCC cell lines. NF-kappaBp65 signaling pathway is aberrantly expressed and activated in both human HCC tissue and HCC Hep3B cells. Inhibition of NF-kappaB activity significantly reduced proliferation and invasion of Hep3B cells as well as down-regulated the expression of invasion-related molecules including matrix metalloproteinase (MMP)-2, MMP-9, membrane type-1 MMP (MT1-MMP), urokinase plasminogen activator (uPA) and vascular endothelial growth factor (VEGF). Hep3B cells exhibited a dose-dependent increase in apoptosis after receiving sorafenib treatment. Inhibition of NF-kappaB activity strongly sensitized Hep3B cells to sorafenib-induced cell death. Mechanistically, combined treatment of sorafenib and NF-kappaB inhibition enhanced inhibition of MAPK signaling and down-regulation of anti-apoptotic protein Mcl-1 expression. These observations indicate that inhibition of NF-kappaB may be a potential antineoplastic therapy for HCC, especially the combination of NF-kappaB inhibition and sorafenib provides a novel therapeutic strategy for patients with advanced-stage HCC.
- Published
- 2009
- Full Text
- View/download PDF
181. Breast Cancer Stem Cells and Intrinsic Subtypes: Controversies Rage On
- Author
-
Harikrishna Nakshatri, Sunil Badve, and Edward F. Srour
- Subjects
Cellular differentiation ,Medicine (miscellaneous) ,Breast Neoplasms ,Biology ,Aldehyde Dehydrogenase 1 Family ,Mice ,Breast cancer ,Cancer stem cell ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,CD24 ,Gene Expression Profiling ,Mesenchymal stem cell ,CD44 ,CD24 Antigen ,Retinal Dehydrogenase ,Cell Differentiation ,Mesenchymal Stem Cells ,General Medicine ,Aldehyde Dehydrogenase ,Genes, erbB-2 ,medicine.disease ,Isoenzymes ,Hyaluronan Receptors ,Immunology ,Cancer cell ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Female ,Stem cell - Abstract
Heterogeneity is a well-documented phenomenon in breast cancer; one of the explanations for this phenomenon is the presence of cancer stem cells (CSCs) with the capacity to differentiate along divergent pathways. These CSCs undergo asymmetric and symmetric division resulting in both expansion of the stem cell pool and the production of morphologically and functionally distinct differentiated daughter cells. Breast cancer cells that express the cell surface molecule CD44 but lack the expression of CD24 have been described as CSCs. Breast cancer cells expressing elevated levels of Aldehyde Dehydrogenase 1 (ALDH1) are also described as CSCs with ALDH1+/CD44+/CD24- subpopulation displaying highest tumorigenic potential in NOD/SCID models. The CSC hypothesis for tumor heterogeneity raises three important questions. First, in unrelated gene expression studies, breast cancers have been classified to five intrinsic subtypes; luminal type A, luminal type B, basal type, ErbB2/HER2-positive and normal-like. Therefore, do these intrinsic subtypes of breast cancer have distinct CSCs of their own or are ALDH1+ or CD44+/CD24- cells common CSCs for all intrinsic subtypes? Secondly, do ALDH1+ or CD44+/CD24- CSCs originate from normal cells of same phenotype or can differentiated cancer cells acquire ALDH1 or CD44+/CD24- status due to mutagenic events? Third, do ALDH1+, ALDH1-, CD44+/CD24- and non-CD44+/CD24- cancer cells differ in their ability to metastasize and respond to chemotherapy? In this review, we present our views on these questions based on studies conducted by several laboratories including ours and present evidence for a strong association of CD44+/CD24- phenotype with basal-like or mesenchymal-like cancer cells.
- Published
- 2009
- Full Text
- View/download PDF
182. Amplified in breast cancer 1 expression in breast cancer
- Author
-
Mangesh A. Thorat, Dmitry Turbin, David G. Huntsman, Akira Morimiya, Harikrishna Nakshatri, Jeng Mh, Zhang Q, Samuel Leung, and Sunil Badve
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Prognostic variable ,Histology ,Estrogen receptor ,Breast Neoplasms ,Biology ,Pathology and Forensic Medicine ,Nuclear Receptor Coactivator 3 ,Breast cancer ,Internal medicine ,Progesterone receptor ,medicine ,Humans ,Aged ,Histone Acetyltransferases ,Aged, 80 and over ,Tissue microarray ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Survival Analysis ,Endocrinology ,Trans-Activators ,Female ,Breast disease ,Follow-Up Studies - Abstract
Aims: The amplified in breast cancer 1 (AIB1), steroid receptor co-activator family member, acts as an oestrogen receptor (ER) co-activator. Acting with HER-2, it is thought to play a role in endocrine resistance by facilitating ER–growth factor crosstalk. The aim was to analyse AIB1 expression by immunohistochemistry and study its correlations with other prognostic variables in breast cancer and its effect on survival. Methods: A tissue microarray comprising tumours from 438 patients with 15.4 years’ median follow-up was used. Interpretable AIB1 expression obtained in 395 patients was analysed along with other prognostic factors in breast cancer. Results: AIB1 expression scores ranged from 0 to 30; positive AIB1 expression (score > 14) was seen in 146/395 breast cancers; it correlated negatively with ER (P = 0.003) and progesterone receptor (PR) (P = 0.007), and positively with HER-2 (P = 0.005) and tumour grade (P = 0.014). It did not correlate with nodal status (P = 0.437). Among ER+ patients, AIB1 expression showed a trend towards loss of PR expression (29% versus 20%; P = 0.14). AIB1 did not predict survival on univariate or multivariate analysis. Conclusions: AIB1 expression correlates with HER-2 expression in breast cancer and shows a trend of association with loss of PR expression in ER+ tumours. Our study supports the postulated role of AIB1 in ER–growth factor interactions.
- Published
- 2008
- Full Text
- View/download PDF
183. Oestrogen-receptor-positive breast cancer: towards bridging histopathological and molecular classifications
- Author
-
Harikrishna Nakshatri and Sunil Badve
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Pathology ,medicine.medical_specialty ,Estrogen receptor ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,Pathology and Forensic Medicine ,Growth factor receptor ,Biomarkers, Tumor ,medicine ,Humans ,medicine.diagnostic_test ,Cancer ,General Medicine ,Gene signature ,Prognosis ,medicine.disease ,Neoplasm Proteins ,Receptors, Estrogen ,Cancer research ,Immunohistochemistry ,Female ,Breast disease ,FOXA1 ,Receptors, Progesterone ,Oncotype DX - Abstract
The oestrogen receptor (ER) pathway is key for survival and progression in a significant proportion of breast cancers. The ER can be activated by oestrogen or activated due to "crosstalk" with growth factor receptor pathways. Activated ER signals through transcriptional and non-transcriptional mechanisms. Immunohistochemistry (IHC), in spite of the shortcomings, remains the method of choice as it provides for in situ assessment of ER expression within the tumour cells. This capability is lost in tissue grinding methods that assess oestrogen-binding activity or messenger RNAs in tumours. IHC is also not influenced by the presence of non-tumoural cells or low amounts of tumour cells within samples examined. It is clear that ER-positive tumours do not represent a single entity. Irrespective of the terminology used, low-grade ER-positive (also known as luminal A) tumours need to be differentiated from high-grade/highly proliferative ER-positive tumours. This can be done in a variety of ways including but not limited to analysis of FOXA1 and GATA-3 by IHC, and limited molecular profiling by Oncotype DX, MGH2-gene signature, intrinsic gene signature or MapQuant Dx. Several areas of ER biology are still poorly understood; these include: its function in the cytoplasm/plasma membrane, its role in the differentiation to proliferation switch, and pathways associated with resistance to hormonal therapy. A detailed understanding of these areas will permit better classification and a personalised approach to management of ER-positive breast cancers.
- Published
- 2008
- Full Text
- View/download PDF
184. Antimyeloma Effects of a Sesquiterpene Lactone Parthenolide
- Author
-
Colin D. Crean, Harikrishna Nakshatri, Attaya Suvannasankha, H. Scott Boswell, Rajasubramaniam Shanmugam, Rafat Abonour, and Sherif S. Farag
- Subjects
Cancer Research ,Stromal cell ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,Apoptosis ,Bone Marrow Cells ,Biology ,Lactones ,chemistry.chemical_compound ,Cell Line, Tumor ,Leukocytes ,medicine ,Humans ,Parthenolide ,Insulin-Like Growth Factor I ,Cell Proliferation ,Dose-Response Relationship, Drug ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Cell growth ,Growth factor ,NF-kappa B ,Coculture Techniques ,medicine.anatomical_structure ,Oncology ,chemistry ,Biochemistry ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,Bone marrow ,Growth inhibition ,Multiple Myeloma ,Sesquiterpenes - Abstract
Purpose: Nuclear factor-κB (NF-κB), activated in multiple myeloma (MM) cells by microenvironmental cues, confers resistance to apoptosis. The sesquiterpene lactone parthenolide targets NF-κB. However, its therapeutic potential in MM is not known.Experimental Designs: We explored the effects of parthenolide on MM cells in the context of the bone marrow microenvironment.Results: Parthenolide inhibited growth of MM cells lines, including drug-resistant cell lines, and primary cells in a dose-dependent manner. Parthenolide overcame the proliferative effects of cytokines interleukin-6 and insulin-like growth factor I, whereas the adhesion of MM cells to bone marrow stromal cells partially protected MM cells against parthenolide effect. In addition, parthenolide blocked interleukin-6 secretion from bone marrow stromal cells triggered by the adhesion of MM cells. Parthenolide cytotoxicity is both caspase-dependent and caspase-independent. Parthenolide rapidly induced caspase activation and cleavage of PARP, MCL-1, X-linked inhibitor of apoptosis protein, and BID. Parthenolide rapidly down-regulated cellular FADD-like IL-1β–converting enzyme inhibitory protein, and direct targeting of cellular FADD-like IL-1β–converting enzyme inhibitory protein using small interfering RNA oligonucleotides inhibited MM cell growth and lowered the parthenolide concentration required for growth inhibition. An additive effect and synergy were observed when parthenolide was combined with dexamethasone and TNF-related apoptosis-inducing ligand, respectively.Conclusion: Collectively, parthenolide has multifaceted antitumor effects toward both MM cells and the bone marrow microenvironment. Our data support the clinical development of parthenolide in MM therapy.
- Published
- 2008
- Full Text
- View/download PDF
185. Striatin-3γ inhibits estrogen receptor activity by recruiting a protein phosphatase
- Author
-
Kenneth P. Nephew, Robert M. Bigsby, Bailin Tan, Harikrishna Nakshatri, and Xinghua Long
- Subjects
Molecular Sequence Data ,Phosphatase ,DUSP6 ,Autoantigens ,Dephosphorylation ,Mice ,chemistry.chemical_compound ,Endocrinology ,Cell Line, Tumor ,Chlorocebus aethiops ,polycyclic compounds ,Animals ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Protein Phosphatase 2 ,Phosphorylation ,Estrogen receptor activity ,Molecular Biology ,Estrogen receptor beta ,biology ,Estrogen Antagonists ,Estrogen Receptor alpha ,Protein phosphatase 2 ,Okadaic acid ,Rats ,Cell biology ,Receptors, Estrogen ,chemistry ,COS Cells ,biology.protein ,Cancer research ,Calmodulin-Binding Proteins ,Female ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists ,HeLa Cells - Abstract
A splicing variant of rat striatin-3 (rSTRN3gamma) was found to associate with estrogen receptor-alpha (ERalpha) in a ligand-dependent manner. In two-hybrid and pull-down analyses, estradiol induced an interaction between rSTRN3gamma and ERalpha. STRN3gamma protein was found in nuclear extracts from rat uterus and human cell lines. Overexpression of rSTRN3gamma induced a decrease in ERalpha transcriptional activity but had no effect on ERbeta activity. Immunoprecipitation analyses showed that rSTRN3gamma interacts with both the ERalpha and the catalytic subunit of protein phosphatase 2A (PP2A(C)). The transrepressor action of rSTRN3gamma was overcome by okadaic acid, an inhibitor of PP2A(C), and by cotransfection of PP2A(C) siRNA. rSTRN3gamma caused dephosphorylation of ERalpha at serine 118 and this was abrogated by okadaic acid. ERalpha lacking phosphorylation sites at either serine 118 or 167 was insensitive to the corepressor action of rSTRN3gamma. These observations suggest that an rSTRN3gamma-PP2A(C) complex is recruited to agonist-activated ERalpha, thereby leading to its dephosphorylation and inhibiting transcription.
- Published
- 2008
- Full Text
- View/download PDF
186. Additional file 1: of Statistical modeling for sensitive detection of low-frequency single nucleotide variants
- Author
-
Yangyang Hao, Pengyue Zhang, Xiaoling Xuei, Harikrishna Nakshatri, Edenberg, Howard, Li, Lang, and Yunlong Liu
- Subjects
body regions ,nervous system ,viruses ,fungi - Abstract
Summary of major somatic callers, their methods of detecting SNV candidates and the limitations. (PDF 60Â kb)
- Published
- 2016
- Full Text
- View/download PDF
187. Additional file 5: of Statistical modeling for sensitive detection of low-frequency single nucleotide variants
- Author
-
Yangyang Hao, Pengyue Zhang, Xiaoling Xuei, Harikrishna Nakshatri, Edenberg, Howard, Li, Lang, and Yunlong Liu
- Abstract
Vuongâ s non-nested test on 4 distributions applied to Illumina MiSeq training data. (PDF 65Â kb)
- Published
- 2016
- Full Text
- View/download PDF
188. Additional file 4: of Statistical modeling for sensitive detection of low-frequency single nucleotide variants
- Author
-
Yangyang Hao, Pengyue Zhang, Xiaoling Xuei, Harikrishna Nakshatri, Edenberg, Howard, Li, Lang, and Yunlong Liu
- Abstract
Genomic sequence context features used in GLM. (PDF 71Â kb)
- Published
- 2016
- Full Text
- View/download PDF
189. Additional file 9: of Statistical modeling for sensitive detection of low-frequency single nucleotide variants
- Author
-
Yangyang Hao, Pengyue Zhang, Xiaoling Xuei, Harikrishna Nakshatri, Edenberg, Howard, Li, Lang, and Yunlong Liu
- Subjects
Quantitative Biology::Quantitative Methods ,Statistics::Applications ,Statistics::Methodology ,Quantitative Biology::Genomics ,Statistics::Computation - Abstract
Zero-inflated Poisson GLM coefficients for Illumina MiSeq training datasets. (PDF 71Â kb)
- Published
- 2016
- Full Text
- View/download PDF
190. Parthenolide Sensitizes Cells to X-Ray-Induced Cell Killing through Inhibition of NF-κB and Split-Dose Repair
- Author
-
Marc S. Mendonca, Helen Chin-Sinex, Nicholas Datzman, Jaime Gomez-Millan, Sachin Mehta, Laura V. Benjamin, Kathleen Comerford, Fatima Patino, Christopher Sweeney, Monica Nye, Michael Hardacre, and Harikrishna Nakshatri
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Biophysics ,Apoptosis ,Biology ,chemistry.chemical_compound ,Tubulin ,Cell Line, Tumor ,Humans ,Radiology, Nuclear Medicine and imaging ,Parthenolide ,Radiation ,Cell growth ,X-Rays ,NF-kappa B ,NF-κB ,Cell biology ,Cell killing ,Proto-Oncogene Proteins c-bcl-2 ,chemistry ,Cell culture ,Phosphorylation ,Tumor Suppressor Protein p53 ,Signal transduction ,Sesquiterpenes ,Protein Binding - Abstract
Human cancers have multiple alterations in cell signaling pathways that promote resistance to cytotoxic therapy such as X rays. Parthenolide is a sesquiterpene lactone that has been shown to inhibit several pro-survival cell signaling pathways, induce apoptosis, and enhance chemotherapy-induced cell killing. We investigated whether parthenolide would enhance X-ray-induced cell killing in radiation resistant, NF-kappaB-activated CGL1 cells. Treatment with 5 microM parthenolide for 48 to 72 h inhibited constitutive NF-kappaB binding and cell growth, reduced plating efficiency, and induced apoptosis through stabilization of p53 (TP53), induction of the pro-apoptosis protein BAX, and phosphorylation of BID. Parthenolide also enhanced radiation-induced cell killing, increasing the X-ray sensitivity of CGL1 cells by a dose modification factor of 1.6. Flow cytometry revealed that parthenolide reduced the percentage of X-ray-resistant S-phase cells due to induction of p21 waf1/cip1 (CDKN1A) and the onset of G1/S and G2/M blocks, but depletion of radioresistant S-phase cells does not explain the observed X-ray sensitization. Further studies demonstrated that the enhancement of X-ray-induced cell killing by parthenolide is due to inhibition of split-dose repair.
- Published
- 2007
- Full Text
- View/download PDF
191. MOZ and MOZ-CBP cooperate with NF-κB to activate transcription from NF-κB–dependent promoters
- Author
-
Zhenyun Yang, Theodore G. Gabig, Colin D. Crean, Elisha M. Comer, Edward M. Chan, H. Scott Boswell, Harikrishna Nakshatri, Rebecca J. Chan, Amy M. Fruehwald, Robert J. Goulet, and Zachary D. Brown
- Subjects
Cancer Research ,Oncogene Proteins, Fusion ,Transcription, Genetic ,Immunoprecipitation ,Protein subunit ,Biology ,Cell Line ,chemistry.chemical_compound ,Transcription (biology) ,Coactivator ,Genetics ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,DNA Primers ,Zinc finger ,Base Sequence ,Interleukin-8 ,NF-kappa B ,Promoter ,NF-κB ,Cell Biology ,Hematology ,Molecular biology ,chemistry - Abstract
Monocytic zinc finger (MOZ) maintains hematopoietic stem cells and, upon fusion to the coactivator CREB-binding protein (CBP), induces acute myeloid leukemia (AML). Leukemic stem cells in AML often exhibit excessive signal-dependent activity of the transcription factor nuclear factor (NF)-kappaB. Because aberrant interaction between NF-kappaB and coactivators represents an alternative mechanism for enhancing NF-kappaB activity, we evaluated whether MOZ and MOZ-CBP cooperate with NF-kappaB to activate transcription from NF-kappaB-dependent promoters.The ability of MOZ, MOZ mutants, and MOZ-CBP to enhance expression of NF-kappaB-dependent promoters was tested in reporter studies. The interaction between MOZ and NF-kappaB was evaluated by both coimmunoprecipitation and glutathione S-transferase pulldown assays.MOZ activates transcription from the NF-kappaB-dependent interleukin-8 promoter; interestingly, this effect is markedly enhanced by CBP. Although MOZ has less potent transcriptional activity than MOZ-CBP, both proteins cooperate with steroid receptor coactivator-1 to activate transcription. MOZ also induces multiple NF-kappaB-dependent viral promoters. Importantly, MOZ associates in a protein complex with the p65 subunit of NF-kappaB and interacts directly with p65 in vitro. Transcriptional activity of MOZ requires its C-terminal domain, which is absent from MOZ-CBP, indicating that the transcriptional activity of MOZ-CBP derives from its CBP sequence.MOZ interacts with the p65 subunit of NF-kappaB and enhances expression of NF-kappaB-dependent promoters. The more potent transcriptional activity of MOZ-CBP derives from its CBP sequence. Thus, interaction between NF-kappaB and MOZ-CBP may play an important role in the pathogenesis of certain acute myeloid leukemias.
- Published
- 2007
- Full Text
- View/download PDF
192. Effect of Celecoxib and Novel Agent LC-1 in a Hamster Model of Lung Cancer
- Author
-
Sundar Neelakantan, Christopher Sweeney, Matthew Ralstin, Harikrishna Nakshatri, Peter A. Crooks, Reid C. Vegeler, Michele T. Yip-Schneider, Huangbing Wu, and C. Max Schmidt
- Subjects
medicine.medical_specialty ,Lung Neoplasms ,Nitrosamines ,Hamster ,Adenocarcinoma ,Pharmacology ,Random Allocation ,Cricetinae ,Internal medicine ,medicine ,Animals ,Cyclooxygenase Inhibitors ,Prostaglandin E2 ,Lung cancer ,Lung ,Carcinogen ,Sulfonamides ,Dose-Response Relationship, Drug ,Mesocricetus ,biology ,business.industry ,NF-kappa B ,Cancer ,medicine.disease ,biology.organism_classification ,Disease Models, Animal ,Endocrinology ,Celecoxib ,Cyclooxygenase 2 ,Carcinogens ,Pyrazoles ,Female ,Surgery ,business ,Sesquiterpenes ,medicine.drug - Abstract
Lung cancer is the leading cause of cancer deaths in the United States. Inflammatory molecules, cyclooxygenase-2 (COX-2) and nuclear factor kappa B (NF-kappaB) have been implicated in lung carcinogenesis. The therapeutic potential of celecoxib, a COX-2 selective inhibitor, and LC-1, a pro-apoptotic drug with accompanying inhibition of NF-kappaB, were investigated.Syrian golden hamsters (n = 140) underwent N-nitroso-bis(2-oxopropyl)amine (BOP) injection weekly for 6 wk. Hamsters were randomized into seven groups: placebo and low/high doses of LC-1, celecoxib, and LC-1/celecoxib. Treatments were given via orogastric lavage for 32 wk. Immunohistochemistry was used to determine COX-2 expression and NF-kappaB activity. Ki-67 labeling was used as an index of proliferation. COX activity was measured by prostaglandin E(2) enzyme-linked immunosorbent assay.BOP successfully induced lung adenocarcinoma in 63% of placebo animals. Lung tumors strongly expressed COX-2 and NF-kappaB. Prostaglandin E(2) levels were decreased in celecoxib compared with placebo groups (P0.05) reflecting suppression of COX activity, but no decrease in NF-kappaB was seen as measured by immunohistochemistry in the tumors. There was no significant difference in tumor size, tumor incidence, or tumor proliferation index between placebo and treatment groups.Carcinogen exposure results in increased COX-2 and NF-kappaB expression and suggests a role in carcinogenesis. Celecoxib and LC-1 did not have any effect in preventing lung cancer development when co-administered with and continued after the carcinogen BOP. Higher doses that can result in suppression of NF-kappaB activity will need to be explored to determine the viability of this approach to prevent lung cancer development.
- Published
- 2007
- Full Text
- View/download PDF
193. Identity Profiling of Cell Surface Markers by Multiplex Gold Nanorod Probes
- Author
-
Chenxu Yu, Harikrishna Nakshatri, and Joseph Irudayaraj
- Subjects
Molecular Probe Techniques ,Nanoprobe ,Breast Neoplasms ,Bioengineering ,Nanotechnology ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,General Materials Science ,Multiplex ,Surface plasmon resonance ,Nanotubes ,Cluster of differentiation ,Chemistry ,Gene Expression Profiling ,Mechanical Engineering ,General Chemistry ,Surface Plasmon Resonance ,Condensed Matter Physics ,Molecular biology ,Dark field microscopy ,Neoplasm Proteins ,Antigens, Surface ,Cytokines ,Nanorod ,Gold ,Molecular probe - Abstract
Gold nanorod molecular probes (GNrMPs) were designed and fabricated for multiplex identification of cell surface markers in HBECs. Cells were probed directly using dark field microscopy integrated with a spectral imager for simultaneous detection of up to three surface markers. The immunophenotype composition of these cell lines indicative of their metastasis potential was assessed using the GNrMPs. The technique has the potential to become an important tool for diagnosis and prognosis of breast and other cancers.
- Published
- 2007
- Full Text
- View/download PDF
194. Suppression of pancreatic tumor growth by combination chemotherapy with sulindac and LC-1 is associated with cyclin D1 inhibitionin vivo
- Author
-
Constantin T. Yiannoutsos, C. Max Schmidt, Peter A. Crooks, Sundar Neelakantan, Matthew Ralstin, Christopher Sweeney, Harikrishna Nakshatri, Michele T. Yip-Schneider, Huangbing Wu, and Stephen Noble
- Subjects
Cancer Research ,Mice, Nude ,Pharmacology ,Mice ,chemistry.chemical_compound ,Sulindac ,Cyclin D1 ,Pancreatic tumor ,In vivo ,Cell Line, Tumor ,Pancreatic cancer ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Parthenolide ,biology ,Chemistry ,NF-kappa B ,Combination chemotherapy ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,Oncology ,Prostaglandin-Endoperoxide Synthases ,biology.protein ,Cyclooxygenase ,Sesquiterpenes ,medicine.drug - Abstract
The design of novel targeted or combination therapies may improve treatment options for pancreatic cancer. Two targets of recent interest are nuclear factor-κB (NF-κB) and cyclooxygenase (COX), known to be activated or overexpressed, respectively, in pancreatic cancer. We have previously shown that parthenolide, a proapoptotic drug associated with NF-κB inhibition, enhanced the growth suppression of pancreatic cancer cells by the COX inhibitor sulindac in vitro. In the present study, a bioavailable analogue of parthenolide, LC-1, and sulindac were evaluated in vivo using a xenograft model of human pancreatic cancer. Treatment groups included placebo, low-dose/high-dose LC-1 (20 and 40 mg/kg), low-dose/high-dose sulindac (20 and 60 mg/kg), and low-dose combination LC-1/sulindac (20 mg/kg each). In MiaPaCa-2 xenografts, tumor growth was inhibited by either high-dose sulindac or LC-1. In BxPC-3 xenografts, tumor size was significantly reduced by treatment with the low-dose LC-1/sulindac combination or high-dose sulindac alone (P < 0.05). Immunohistochemistry of BxPC-3 tumors revealed a significant decrease in Ki-67 and CD31 staining by high-dose sulindac, with no significant changes in COX-1/COX-2 levels or activity in any of the treatment groups. NF-κB DNA-binding activity was significantly decreased by high-dose LC-1. Cyclin D1 protein levels were reduced by the low-dose LC-1/sulindac combination or high-dose sulindac alone, correlating with BxPC-3 tumor suppression. These results suggest that LC-1 and sulindac may mediate their antitumor effects, in part, by altering cyclin D1 levels. Furthermore, this study provides preclinical evidence for the therapeutic efficacy of these agents. [Mol Cancer Ther 2007;6(6):1736–44]
- Published
- 2007
- Full Text
- View/download PDF
195. FOXA1 as a therapeutic target for breast cancer
- Author
-
Harikrishna Nakshatri and Sunil Badve
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Pharmacology ,Oncology ,medicine.medical_specialty ,business.industry ,Clinical Biochemistry ,Estrogen receptor ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,medicine.disease ,Gene expression profiling ,Breast cancer ,Text mining ,Internal medicine ,Drug Discovery ,medicine ,Humans ,Molecular Medicine ,Female ,Luminal type ,FOXA1 ,business ,Estrogen receptor beta - Abstract
Gene expression profiling studies have classified breast cancer into five intrinsic subtypes with distinct prognostic significance: luminal type A, luminal type B, normal-like, HER-2-positive and basal type. These studies have also uncovered novel diagnostic markers and molecular targets. FOXA1, a winged-helix transcription factor belonging to the forkhead family, is one among them as it is expressed predominantly in luminal type A breast cancer, which is characterized by the presence of estrogen receptor-alpha (ERalpha) with favorable prognosis. FOXA1 is a 'pioneer' factor that binds to chromatinized DNA, opens the chromatin and enhances binding of ERalpha to its target genes. It is essential for the expression of approximately 50% of ERalpha:estrogen-regulated genes. Thus, a network comprising FOXA1, ERalpha and estrogen constitutes a major proliferation and survival signal for luminal type A breast cancer. However, by controlling differentiation and by regulating the expression of cell cycle inhibitor p27kip1 and the cell adhesion molecule E-cadherin, FOXA1 may prevent metastatic progression of luminal type A breast cancer. This article reviews possible roles of FOXA family transcription factors in breast cancer initiation, hormone dependency and speculates on the potential of FOXA1 as a therapeutic target.
- Published
- 2007
- Full Text
- View/download PDF
196. 2-Methoxyestradiol Inhibits the Anaphase-Promoting Complex and Protein Translation in Human Breast Cancer Cells
- Author
-
Harikrishna Nakshatri, Yesim Gökmen-Polar, Christoph H. Borchers, Cheng Fan, David Ketelsen, Nancy Klauber-DeMore, Cam Patterson, Rajendra Bhati, George W. Sledge, and Michael J. Dial
- Subjects
Cancer Research ,Cell cycle checkpoint ,Apoptosis ,Breast Neoplasms ,Cell Growth Processes ,Biology ,Anaphase-Promoting Complex-Cyclosome ,Tubulin ,Cell Line, Tumor ,Gene expression ,Humans ,Cyclin B1 ,Oligonucleotide Array Sequence Analysis ,Estradiol ,Cell Cycle ,Ubiquitin-Protein Ligase Complexes ,Molecular biology ,Tubulin Modulators ,Genetic translation ,2-Methoxyestradiol ,Oncology ,Mitotic spindle assembly checkpoint ,Securin ,Protein Biosynthesis ,Mutation ,Cancer cell ,Anaphase-promoting complex - Abstract
2-Methoxyestradiol (2ME2), an estradiol metabolite with antiproliferative and antiangiogenic activities, is in phase I/II clinical trials for breast cancer. 2ME2 inhibits microtubule polymerization and causes cells to arrest in G2-M. The purpose of this study was to further elucidate the molecular mechanism of 2ME2. MDA-MB-435 breast cancer cells were treated with 2ME2 (2 μmol/L) or vehicle alone. RNA was extracted and genomic profiling was done using 22k Agilent microarrays. Expression Analysis Systematic Explorer was used to determine enrichment of Gene Ontology categories. Protein isolates were subjected to Western blot analysis. Protein synthesis was measured with a [35S]methionine pulse assay. An MDA-MB-435 cell line with two β-tubulin mutations (2ME2R) was used to determine whether novel mechanisms were tubulin-dependent. Gene Ontology categories enriched include genes that regulate the mitotic spindle assembly checkpoint, apoptosis, and the cytosolic ribosome. The target of the mitotic spindle assembly checkpoint is the anaphase-promoting complex (APC). APC inhibition was confirmed by measuring protein levels of its targets securin and cyclin B1, which were increased in 2ME2-treated cells. Because gene expression in the cytosolic ribosome category was decreased, we evaluated whether 2ME2 decreases protein translation. This was confirmed with a pulse assay, which showed decreased isotope incorporation in 2ME2-treated cells, which was maintained in the tubulin-resistant 2ME2R cells. APC inhibition was not maintained in 2ME2R cells. 2ME2 induces tubulin-dependent cell cycle arrest through regulation of genes involved in the mitotic spindle assembly checkpoint, which results in inhibition of the APC and tubulin-independent inhibition of protein translation. [Cancer Res 2007;67(2):702–8]
- Published
- 2007
- Full Text
- View/download PDF
197. The Platelet-derived Growth Factor Receptor α Is Destabilized by Geldanamycins in Cancer Cells
- Author
-
Yi Chun Lai, Liyun Cao, David B. Donner, Harikrishna Nakshatri, Daniela Matei, and M. Satpathy
- Subjects
Cell signaling ,Receptor, Platelet-Derived Growth Factor alpha ,Leupeptins ,Lactams, Macrocyclic ,Antineoplastic Agents ,Biochemistry ,Piperazines ,Receptor tyrosine kinase ,Mice ,Growth factor receptor ,Cell Line, Tumor ,Benzoquinones ,polycyclic compounds ,Animals ,Humans ,Receptor ,Molecular Biology ,Cell Proliferation ,Insulin-like growth factor 1 receptor ,Platelet-Derived Growth Factor ,Antibiotics, Antineoplastic ,biology ,3T3 Cells ,Cell Biology ,female genital diseases and pregnancy complications ,Pyrimidines ,Imatinib mesylate ,Benzamides ,Imatinib Mesylate ,biology.protein ,Cancer research ,Tyrosine kinase ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
The heat shock protein HSP90 serves as a chaperone for receptor protein kinases, steroid receptors, and other intracellular signaling molecules. Targeting HSP90 with ansamycin antibiotics disrupts the normal processing of clients of the HSP90 complex. The platelet-derived growth factor receptor alpha (PDGFRalpha) is a tyrosine kinase receptor up-regulated and activated in several malignancies. Here we show that the PDGFRalpha forms a complex with HSP90 and the co-chaperone cdc37 in ovarian, glioblastoma, and lung cancer cells. Treatment of cancer cell lines expressing the PDGFRalpha with the HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) promotes degradation of the receptor. Likewise, phospho-Akt, a downstream target, is degraded after treatment with 17-AAG. In contrast, PDGFRalpha expression is not affected by 17-AAG in normal human smooth muscle cells or 3T3 fibroblasts. PDGFRalpha degradation by 17-AAG is inhibited by the proteasome inhibitor MG132. High molecular weight, ubiquitinated forms of the receptor are detected in cells treated with 17-AAG and MG132. Degradation of the receptor is also inhibited by a specific neutralizing antibody to the PDGFRalpha but not by a neutralizing antibody to PDGF or by imatinib mesylate (Gleevec). Ultimately, PDGFRalpha-mediated cell proliferation is inhibited by 17-AAG. These results show that 17-AAG promotes PDGFRalpha degradation selectively in transformed cells. Thus, not only mutated tyrosine kinases but also overexpressed receptors in cancer cells can be targeted by 17-AAG.
- Published
- 2007
- Full Text
- View/download PDF
198. TFAP2C expression in breast cancer: correlation with overall survival beyond 10 years of initial diagnosis
- Author
-
George E. Sandusky, Kathy D. Miller, Harikrishna Nakshatri, Susan M. Perkins, Tudor Vladislav, Sandra Althouse, Casey Bales, and Sunil Badve
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Pathology ,Estrogen receptor ,Breast Neoplasms ,Disease-Free Survival ,Correlation ,Breast cancer ,Internal medicine ,medicine ,Biomarkers, Tumor ,Endocrine system ,Humans ,Aged ,Proportional Hazards Models ,Tissue microarray ,business.industry ,GATA3 ,Estrogen Receptor alpha ,Middle Aged ,medicine.disease ,Survival Analysis ,Transcription Factor AP-2 ,Tissue Array Analysis ,Immunohistochemistry ,Female ,FOXA1 ,business ,Follow-Up Studies - Abstract
Recurrence and death in a significant number of patients with ERα-positive breast cancer occurs 10–20 years after diagnosis. Prognostic markers for late events have been more elusive. TFAP2C (AP2γ) regulates the expression of ERα, the ERα pioneer factors FOXA1 and GATA3, and controls ERα-dependent transcription. The purpose of this investigation is to determine the long-term prognostic value of TFAP2C. A tissue microarray (TMA) consisting of breast tumors from 451 patients with median follow-up time of 10.3 years was created and tested for the expression of TFAP2C by immunohistochemistry. Wilcoxon Rank-Sum and Kruskal–Wallis tests were used to determine if TFAP2C H-scores correlate with other tumor markers. Cox proportional hazards regression models were used to determine whether TFAP2C H-scores and other tumor markers were related to overall and disease-free survival in univariate and multivariable models. TFPAC2 overexpression did not impact overall survival during the first 10 years after diagnosis, but was associated with a shorter survival after 10 years (HR 3.40, 95 % CI 1.58, 7.30; p value = 0.002). This late divergence persisted in ER-positive (HR 2.86, 95 % CI 1.29, 6.36; p value = 0.01) and endocrine therapy-positive subgroups (HR 4.19, 95 % CI 1.72, 10.23; p value = 0.002). For the ER+ and endocrine therapy subgroup, the HR was 3.82 (95 % CI 1.53, 9.50; p value = 0.004). TFAP2C H-scores were not correlated with other tumor markers or related to disease-free survival. In this hypothesis-generating study, we show that higher TFAP2C scores correlate with poor overall survival after 10 years of diagnosis in ERα-positive and endocrine therapy-treated subgroups.
- Published
- 2015
199. Ethnicity-Dependent and -Independent Heterogeneity in Healthy Normal Breast Hierarchy Impacts Tumor Characterization
- Author
-
Harikrishna Nakshatri, Poornima Bhat-Nakshatri, and Manjushree Anjanappa
- Subjects
Heterozygote ,Epithelial-Mesenchymal Transition ,Down-Regulation ,Breast Neoplasms ,Cell Count ,Article ,Transcriptome ,chemistry.chemical_compound ,Genetic Heterogeneity ,Antigens, CD ,Antigens, Neoplasm ,Cell Line, Tumor ,Genetic predisposition ,Biomarkers, Tumor ,Ethnicity ,Humans ,Progenitor cell ,Multidisciplinary ,biology ,Genetic heterogeneity ,CD24 ,BRCA1 Protein ,CD44 ,Epithelial cell adhesion molecule ,Epithelial Cells ,Epithelial Cell Adhesion Molecule ,Tissue Donors ,Extracellular Matrix ,Black or African American ,Gene Expression Regulation, Neoplastic ,Phenotype ,chemistry ,Immunology ,Mutation ,biology.protein ,Neoplastic Stem Cells ,Female ,Stem cell ,Cell Adhesion Molecules - Abstract
Recent reports of widespread genetic variation affecting regulation of gene expression raise the possibility of significant inter-individual differences in stem-progenitor-mature cell hierarchy in adult organs. This has not been explored because of paucity of methods to quantitatively assess subpopulation of normal epithelial cells on individual basis. We report the remarkable inter-individual differences in differentiation capabilities as documented by phenotypic heterogeneity in stem-progenitor-mature cell hierarchy of the normal breast. Ethnicity and genetic predisposition are partly responsible for this heterogeneity, evidenced by the finding that CD44+/CD24- and PROCR+/EpCAM- multi-potent stem cells were elevated significantly in African American women compared with Caucasians. ALDEFLUOR+ luminal stem/progenitor cells were lower in BRCA1-mutation carriers compared with cells from healthy donors (p = 0.0014). Moreover, tumor and adjoining-normal breast cells of the same patients showed distinct CD49f+/EpCAM+ progenitor, CD271+/EpCAM- basal and ALDEFLUOR+ cell profiles. These inter-individual differences in the rate of differentiation in the normal breast may contribute to a substantial proportion of transcriptome, epigenome and signaling pathway alterations and consequently has the potential to spuriously magnify the extent of documented tumor-specific gene expression. Therefore, comparative analysis of phenotypically defined subpopulations of normal and tumor cells on an individual basis may be required to identify cancer-specific aberrations.
- Published
- 2015
200. Restoring chemotherapy and hormone therapy sensitivity by parthenolide in a xenograft hormone refractory prostate cancer model
- Author
-
Michele T. Yip-Schneider, Qi Huang Zheng, Stephanie Kelich, Yesim Gökmen-Polar, Harikrishna Nakshatri, George W. Sledge, Poornima Bhat-Nakshatri, Christopher Sweeney, Timothy R. DeGrado, Gary D. Hutchins, Liang Cheng, Michael A. Miller, Rajasubramaniam Shanmugam, Vetrichelvan Jayaprakasan, and Kathy D. Miller
- Subjects
medicine.medical_specialty ,Bicalutamide ,Angiogenesis ,business.industry ,Urology ,medicine.disease ,Prostate cancer ,chemistry.chemical_compound ,Endocrinology ,Oncology ,Docetaxel ,chemistry ,In vivo ,Apoptosis ,Internal medicine ,medicine ,Cancer research ,Hormonal therapy ,Parthenolide ,business ,medicine.drug - Abstract
BACKGROUND Nuclear Factor kappa B (NFκB) is a eukaryotic transcription factor that is constitutively active in human cancers and can be inhibited by the naturally occurring sesquiterpene lactone, parthenolide (P). METHODS The in vitro effects of P were assessed using the androgen independent cell line, CWR22Rv1, and human umbilical endothelial cells (HUVECs). The in vivo activity of P as a single agent and its ability to augment the efficacy of docetaxel and the anti-androgen, bicalutamide, were determined using the CWR22Rv1 xenograft model. RESULTS Parthenolide at low micromolar concentration inhibited proliferation of CWR22Rv1 and HUVEC cells, promoted apoptosis and abrogated NFκB-DNA binding. Parthenolide downregulated anti-apoptotic genes under NFκB control, TRAF 1 and 2, and promoted sustained activation of c-jun-NH2 kinase (JNK). Parthenolide also augmented the in vivo efficacy of docetaxel and restored sensitivity to anti-androgen therapy. CONCLUSION These studies demonstrate parthenolide's anti-tumor and anti-angiogenic activity, and its potential to augment the efficacy of chemotherapy and hormonal therapy. Prostate 66: 1498–1511, 2006. © 2006 Wiley-Liss, Inc.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.