Network


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

Hotspot


Dive into the research topics where Laura D. Nelson is active.

Publication


Featured researches published by Laura D. Nelson.


Bioscience Reports | 2011

Curcumin regulates miR-21 expression and inhibits invasion and metastasis in colorectal cancer

Giridhar Mudduluru; Jonahunnatha N. George-William; Santoshi Muppala; Irfan A. Asangani; Regalla Kumarswamy; Laura D. Nelson; Heike Allgayer

Curcumin has promising potential in cancer prevention and therapy by interacting with proteins and modifying their expression and activity, which includes transcription factors, inflammatory cytokines and factors of cell survival, proliferation and angiogenesis. miR-21 is overexpressed in many tumours, promoting progression and metastasis. In the present study, we examined the potential of curcumin to regulate miR-21, tumour growth, invasion and in vivo metastasis in colorectal cancer. In Rko and HCT116 cells, we identified two new transcriptional start sites of the miR-21 gene and delineated its promoter region. PMA stimulation induced miR-21 expression via motifs bound with AP-1 (activator protein 1) transcription factors. Curcumin treatment reduced miR-21 promoter activity and expression in a dose-dependent manner by inhibiting AP-1 binding to the promoter, and induced the expression of the tumour suppressor Pdcd4 (programmed cell death protein 4), which is a target of miR-21. Curcumin-treated Rko and HCT116 cells were arrested in the G2/M phase with increasing concentrations. Furthermore, curcumin inhibited tumour growth, invasion and in vivo metastasis in the chicken-embryo-metastasis assay [CAM (chorionallantoic membrane) assay]. Additionally, curcumin significantly inhibited miR-21 expression in primary tumours generated in vivo in the CAM assay by Rko and HCT116 cells (P<0.00006 and P<0.035 respectively). Taken together, this is the first paper to show that curcumin inhibits the transcriptional regulation of miR-21 via AP-1, suppresses cell proliferation, tumour growth, invasion and in vivo metastasis, and stabilizes the expression of the tumour suppressor Pdcd4 in colorectal cancer.


Journal of the American Chemical Society | 2015

Improving paclitaxel delivery: in vitro and in vivo characterization of PEGylated polyphosphoester-based nanocarriers.

Fuwu Zhang; Shiyi Zhang; Stephanie F. Pollack; Richen Li; Amelia M. Gonzalez; Jingwei Fan; Jiong Zou; Sarah E. Leininger; Adriana Pavía-Sanders; Rachel Johnson; Laura D. Nelson; Jeffery E. Raymond; Mahmoud Elsabahy; Dennis M. P. Hughes; Mark W. Lenox; Tiffany P. Gustafson; Karen L. Wooley

Nanomaterials have great potential to offer effective treatment against devastating diseases by providing sustained release of high concentrations of therapeutic agents locally, especially when the route of administration allows for direct access to the diseased tissues. Biodegradable polyphosphoester-based polymeric micelles and shell cross-linked knedel-like nanoparticles (SCKs) have been designed from amphiphilic block-graft terpolymers, PEBP-b-PBYP-g-PEG, which effectively incorporate high concentrations of paclitaxel (PTX). Well-dispersed nanoparticles physically loaded with PTX were prepared, exhibiting desirable physiochemical characteristics. Encapsulation of 10 wt% PTX, into either micelles or SCKs, allowed for aqueous suspension of PTX at concentrations up to 4.8 mg/mL, as compared to <2.0 μg/mL for the aqueous solubility of the drug alone. Drug release studies indicated that PTX released from these nanostructures was defined through a structure-function relationship, whereby the half-life of sustained PTX release was doubled through cross-linking of the micellar structure to form SCKs. In vitro, physically loaded micellar and SCK nanotherapeutics demonstrated IC50 values against osteosarcoma cell lines, known to metastasize to the lungs (CCH-OS-O and SJSA), similar to the pharmaceutical Taxol formulation. Evaluation of these materials in vivo has provided an understanding of the effects of nanoparticle structure-function relationships on intratracheal delivery and related biodistribution and pharmacokinetics. Overall, we have demonstrated the potential of these novel nanotherapeutics toward future sustained release treatments via administration directly to the sites of lung metastases of osteosarcoma.


Cancer Research | 2009

Cetuximab Attenuates Metastasis and u-PAR Expression in Non-Small Cell Lung Cancer: u-PAR and E-Cadherin are Novel Biomarkers of Cetuximab Sensitivity

Dessislava A. Nikolova; Irfan A. Asangani; Laura D. Nelson; Dennis P.M. Hughes; Doris R. Siwak; Gordon B. Mills; Andrea Harms; Erika Buchholz; Lothar Pilz; Christian Manegold; Heike Allgayer

Cetuximab, which blocks ligand binding to epidermal growth factor receptor (EGFR), is currently being studied as a novel treatment for non-small cell lung cancer (NSCLC). However, its mechanisms of action toward metastasis, and markers of drug sensitivity, have not been fully elucidated. This study was conducted to (a) determine the effect of Cetuximab on invasion and NSCLC-metastasis; (b) investigate urokinase-type plasminogen activator receptor (u-PAR), a major molecule promoting invasion and metastasis, as a target molecule; (c) delineate molecular mediators of Cetuximab-induced metastasis inhibition; and (d) identify biomarkers of drug sensitivity in NSCLC. Cetuximab treatment resulted in reduced growth and Matrigel invasion of H1395 and A549 NSCLC cell lines, in parallel with reduced u-PAR mRNA and protein. u-PAR down-regulation was brought about by suppressing the binding of JunD and c-Jun to u-PAR promoter motif -190/-171 in vivo, and an inhibition of MAP/ERK kinase signaling. Furthermore, Cetuximab inhibited NSCLC proliferation and metastasis to distant organs in vivo as indicated by the chicken embryo metastasis assay. Low E-cadherin and high u-PAR, but not EGFR, was associated with resistance to Cetuximab in seven NSCLC cell lines. Furthermore, siRNA knockdown of u-PAR led to a resensitization to Cetuximab. Moreover, low E-cadherin and high u-PAR was found in 63% of resected tumor tissues of NSCLC patients progressing under Cetuximab therapy. This is the first study to show u-PAR as a target and marker of sensitivity to Cetuximab, and to delineate novel mechanisms leading to metastasis suppression of NSCLC by Cetuximab.


Molecular Cancer | 2012

Triplex DNA-binding proteins are associated with clinical outcomes revealed by proteomic measurements in patients with colorectal cancer.

Laura D. Nelson; Christian Bender; Heiko Mannsperger; Daniel Buergy; Patryk Kambakamba; Giridhar Mudduluru; Ulrike Korf; Dennis P.M. Hughes; Michael W. Van Dyke; Heike Allgayer

BackgroundTri- and tetra-nucleotide repeats in mammalian genomes can induce formation of alternative non-B DNA structures such as triplexes and guanine (G)-quadruplexes. These structures can induce mutagenesis, chromosomal translocations and genomic instability. We wanted to determine if proteins that bind triplex DNA structures are quantitatively or qualitatively different between colorectal tumor and adjacent normal tissue and if this binding activity correlates with patient clinical characteristics.MethodsExtracts from 63 human colorectal tumor and adjacent normal tissues were examined by gel shifts (EMSA) for triplex DNA-binding proteins, which were correlated with clinicopathological tumor characteristics using the Mann-Whitney U, Spearman’s rho, Kaplan-Meier and Mantel-Cox log-rank tests. Biotinylated triplex DNA and streptavidin agarose affinity binding were used to purify triplex-binding proteins in RKO cells. Western blotting and reverse-phase protein array were used to measure protein expression in tissue extracts.ResultsIncreased triplex DNA-binding activity in tumor extracts correlated significantly with lymphatic disease, metastasis, and reduced overall survival. We identified three multifunctional splicing factors with biotinylated triplex DNA affinity: U2AF65 in cytoplasmic extracts, and PSF and p54nrb in nuclear extracts. Super-shift EMSA with anti-U2AF65 antibodies produced a shifted band of the major EMSA H3 complex, identifying U2AF65 as the protein present in the major EMSA band. U2AF65 expression correlated significantly with EMSA H3 values in all extracts and was higher in extracts from Stage III/IV vs. Stage I/II colon tumors (p = 0.024). EMSA H3 values and U2AF65 expression also correlated significantly with GSK3 beta, beta-catenin, and NF- B p65 expression, whereas p54nrb and PSF expression correlated with c-Myc, cyclin D1, and CDK4. EMSA values and expression of all three splicing factors correlated with ErbB1, mTOR, PTEN, and Stat5. Western blots confirmed that full-length and truncated beta-catenin expression correlated with U2AF65 expression in tumor extracts.ConclusionsIncreased triplex DNA-binding activity in vitro correlates with lymph node disease, metastasis, and reduced overall survival in colorectal cancer, and increased U2AF65 expression is associated with total and truncated beta-catenin expression in high-stage colorectal tumors.


PLOS ONE | 2013

Small Molecule ErbB Inhibitors Decrease Proliferative Signaling and Promote Apoptosis in Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia

Mary E. Irwin; Laura D. Nelson; Janice M. Santiago-O’Farrill; Phillip Knouse; Claudia P. Miller; Shana L. Palla; Doris R. Siwak; Gordon B. Mills; Zeev Estrov; Shulin Li; Steven M. Kornblau; Dennis P.M. Hughes; Joya Chandra

The presence of the Philadelphia chromosome in patients with acute lymphoblastic leukemia (Ph+ALL) is a negative prognostic indicator. Tyrosine kinase inhibitors (TKI) that target BCR/ABL, such as imatinib, have improved treatment of Ph+ALL and are generally incorporated into induction regimens. This approach has improved clinical responses, but molecular remissions are seen in less than 50% of patients leaving few treatment options in the event of relapse. Thus, identification of additional targets for therapeutic intervention has potential to improve outcomes for Ph+ALL. The human epidermal growth factor receptor 2 (ErbB2) is expressed in ∼30% of B-ALLs, and numerous small molecule inhibitors are available to prevent its activation. We analyzed a cohort of 129 ALL patient samples using reverse phase protein array (RPPA) with ErbB2 and phospho-ErbB2 antibodies and found that activity of ErbB2 was elevated in 56% of Ph+ALL as compared to just 4.8% of Ph−ALL. In two human Ph+ALL cell lines, inhibition of ErbB kinase activity with canertinib resulted in a dose-dependent decrease in the phosphorylation of an ErbB kinase signaling target p70S6-kinase T389 (by 60% in Z119 and 39% in Z181 cells at 3 µM). Downstream, phosphorylation of S6-kinase was also diminished in both cell lines in a dose-dependent manner (by 91% in both cell lines at 3 µM). Canertinib treatment increased expression of the pro-apoptotic protein Bim by as much as 144% in Z119 cells and 49% in Z181 cells, and further produced caspase-3 activation and consequent apoptotic cell death. Both canertinib and the FDA-approved ErbB1/2-directed TKI lapatinib abrogated proliferation and increased sensitivity to BCR/ABL-directed TKIs at clinically relevant doses. Our results suggest that ErbB signaling is an additional molecular target in Ph+ALL and encourage the development of clinical strategies combining ErbB and BCR/ABL kinase inhibitors for this subset of ALL patients.


Oncogene | 2014

Identification of RBCK1 as a novel regulator of FKBPL: implications for tumor growth and response to tamoxifen

Christopher Donley; Keeva McClelland; Hayley D. McKeen; Laura D. Nelson; Anita Yakkundi; Puthen V. Jithesh; James F. Burrows; Lana McClements; Andrea Valentine; Kevin Prise; Helen O. McCarthy; Tracy Robson

FKBPL has been implicated in processes associated with cancer, including regulation of tumor growth and angiogenesis with high levels of FKBPL prognosticating for improved patient survival. Understanding how FKBPL levels are controlled within the cell is therefore critical. We have identified a novel role for RBCK1 as an FKBPL-interacting protein, which regulates FKBPL stability at the post-translational level via ubiquitination. Both RBCK1 and FKBPL are upregulated by 17-β-estradiol and interact within heat shock protein 90 chaperone complexes, together with estrogen receptor-α (ERα). Furthermore, FKBPL and RBCK1 associate with ERα at the promoter of the estrogen responsive gene, pS2, and regulate pS2 levels. MCF-7 clones stably overexpressing RBCK1 were shown to have reduced proliferation and increased levels of FKBPL and p21. Furthermore, these clones were resistant to tamoxifen therapy, suggesting that RBCK1 could be a predictive marker of response to endocrine therapy. RBCK1 knockdown using targeted small interfering RNA resulted in increased proliferation and increased sensitivity to tamoxifen treatment. Moreover, in support of our in vitro data, analysis of mRNA microarray data sets demonstrated that high levels of FKBPL and RBCK1 correlated with increased patient survival, whereas high RBCK1 predicted for a poor response to tamoxifen. Our findings support a role for RBCK1 in the regulation of FKBPL with important implications for estrogen receptor signaling, cell proliferation and response to endocrine therapy.


Molecular Cancer Research | 2013

Unraveling the role of FOXQ1 in colorectal cancer metastasis.

Mohammed Abba; Nitin Patil; Kabeer Rasheed; Laura D. Nelson; Giridhar Mudduluru; Jorg Hendrick Leupold; Heike Allgayer

Malignant cell transformation, invasion, and metastasis are dependent on the coordinated rewiring of gene expression. A major component in the scaffold of these reprogramming events is one in which epithelial cells lose intercellular connections and polarity to adopt a more motile mesenchymal phenotype, which is largely supported by a robust transcriptional machinery consisting mostly of developmental transcription factors. This study demonstrates that the winged helix transcription factor, FOXQ1, contributes to this rewiring process, in part by directly modulating the transcription of TWIST1, itself a key mediator of metastasis that transcriptionally regulates the expression of important molecules involved in epithelial-to-mesenchymal transition. Forced expression and RNA-mediated silencing of FOXQ1 led to enhanced and suppressed mRNA and protein levels of TWIST1, respectively. Mechanistically, FOXQ1 enhanced the reporter activity of TWIST1 and directly interacted with its promoter. Furthermore, enhanced expression of FOXQ1 resulted in increased migration and invasion in colorectal cancer cell lines, whereas knockdown studies showed the opposite effect. Moreover, using the in vivo chicken chorioallantoic membrane metastasis assay model, FOXQ1 significantly enhanced distant metastasis with minimal effects on tumor growth. Implications: These findings reveal FOXQ1 as a modulator of TWIST1-mediated metastatic phenotypes and support its potential as a biomarker of metastasis. Mol Cancer Res; 11(9); 1017–28. ©2013 AACR.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

FKBPL Is a Critical Antiangiogenic Regulator of Developmental and Pathological Angiogenesis

Anita Yakkundi; Rachel Bennett; Ivette Hernández-Negrete; Jean-Marie Delalande; Mary L. Hanna; Oksana Lyubomska; Kenneth Arthur; Amy Short; Hayley D. McKeen; Laura D. Nelson; Cian M. McCrudden; Ross McNally; Lana McClements; Helen O. McCarthy; Alan J. Burns; Roy Bicknell; Adrien Kissenpfennig; Tracy Robson

Objective— The antitumor effects of FK506-binding protein like (FKBPL) and its extracellular role in angiogenesis are well characterized; however, its role in physiological/developmental angiogenesis and the effect of FKBPL ablation has not been evaluated. This is important as effects of some angiogenic proteins are dosage dependent. Here we evaluate the regulation of FKBPL secretion under angiogenic stimuli, as well as the effect of FKBPL ablation in angiogenesis using mouse and zebrafish models. Approach and Results— FKBPL is secreted maximally by human microvascular endothelial cells and fibroblasts, and this was specifically downregulated by proangiogenic hypoxic signals, but not by the angiogenic cytokines, VEGF or IL8. FKBPL’s critical role in angiogenesis was supported by our inability to generate an Fkbpl knockout mouse, with embryonic lethality occurring before E8.5. However, whilst Fkbpl heterozygotic embryos showed some vasculature irregularities, the mice developed normally. In murine angiogenesis models, including the ex vivo aortic ring assay, in vivo sponge assay, and tumor growth assay, Fkbpl +/− mice exhibited increased sprouting, enhanced vessel recruitment, and faster tumor growth, respectively, supporting the antiangiogenic function of FKBPL. In zebrafish, knockdown of zFkbpl using morpholinos disrupted the vasculature, and the phenotype was rescued with hFKBPL. Interestingly, this vessel disruption was ineffective when zcd44 was knocked-down, supporting the dependency of zFkbpl on zCd44 in zebrafish. Conclusions— FKBPL is an important regulator of angiogenesis, having an essential role in murine and zebrafish blood vessel development. Mouse models of angiogenesis demonstrated a proangiogenic phenotype in Fkbpl heterozygotes.


Cancer Research | 2014

Abstract 3970: EGF has stimulatory and survival effects in osteosarcoma in vitro and pan-ERBB kinase inhibition causes cell growth inhibition in vitro and reduces lung metastases in vivo

Laura D. Nelson; Tiffany N. Lynch; Yanwen Yang; Wei-Lien Wang; Diane Liu; Dafydd G. Thomas; Dennis P.M. Hughes

Osteosarcoma is the most common primary bone tumor in children and adolescents, with recurring disease or lung metastases being the primary cause of death in patients. Expression of EGFR, Her-2, and Her-4 (ERBB1, 2 and 4) have been reported in human primary osteosarcoma, suggesting that they may be contributing to osteosarcoma pathogenesis, but the correlation of expression levels with patient outcomes has been controversial. We have shown that a pan-ERBB small molecule kinase inhibitor causes growth inhibition and apoptosis of osteosarcoma cell in vitro, suggesting a potential therapeutic benefit of using this drug in vivo. In the present study, we show that protein expression levels of EGFR, Her-2 and Her-4 are higher in osteosarcoma lung metastases than in primary tumors by TMA, and that these levels are significantly higher for Her-2 and Her-4. We created eight new osteosarcoma cell lines from primary patient tissues and found that all eight, in addition to six commonly used osteosarcoma cell lines, express detectable levels of EGFR and Her-2, and several express detectable Her-4. EGF ligand stimulation in vitro produces robust phosphorylation of EGFR, Her-2 and Her-4 in most cell lines and moderate activation in others. EGF stimulation also produces phosphorylation of Akt, MAPK and p70S6K in most cell lines and of STAT5 in six cell lines, suggesting that EGF has growth and survival stimulation effects. When adding a pan-ERBB kinase inhibitor, Pf-299804, to cells pre-stimulated with EGF, phosphorylation of EGFR, Her-2, Her-4, Akt, etc. are inhibited in a dose-dependent manner. Reverse phase protein array also shows an increase in pro-apoptotic proteins Bim and cleaved PARP with Pf-299804 treatment, confirmed by Western blots. Mouse xenograft experiments, where cells injected in the tibia spontaneously form lung metastases, showed a significant decrease in three categories of Her-4-positive lung metastases in drug-treated mice based on size: 1-4 cell oligomets (p=0.0055); 5 cell-200 micron micromets (p=0.0276); and greater than 200 micron macromets (p=0.0055). These data suggest that EGF produces mitogenic and survival stimulation in osteosarcoma cells and that pan ERBB-family kinase inhibition causes growth inhibition and apoptosis in vitro and reduction of lung metastases in a mouse model in vivo. This provides evidence that pan-ERBB kinase inhibition has therapeutic potential for osteosarcoma lung metastases. Citation Format: Laura D. Nelson, Tiffany N. Lynch, Yanwen Yang, Wei-Lien Wang, Diane Liu, Dafydd G. Thomas, Dennis P. M. Hughes. EGF has stimulatory and survival effects in osteosarcoma in vitro and pan-ERBB kinase inhibition causes cell growth inhibition in vitro and reduces lung metastases in vivo. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3970. doi:10.1158/1538-7445.AM2014-3970


Cancer Research | 2010

Abstract 4566: Triplex DNA-binding proteins in resected normal and tumor tissues from colorectal cancer patients: Biological and clinical relevance

Laura D. Nelson; Heiko Mannsperger; Christian Bender; Daniel Buergy; Patryk Kambakamba; Giridhar Mudduluru; Dennis P.M. Hughes; Michael W. Van Dyke; Heike Allgayer

This study is to determine if triplex DNA-binding proteins in human cells play an important role in processes that lead to cancer such as genetic instability or growth deregulation. Cytoplasmic and nuclear extracts from 60 colorectal cancer patients’ tumor (T) and normal (N) tissues were isolated and examined by gel shifts (EMSA) for triplex DNA-binding proteins. Total tumor extracts showed significantly higher triplex DNA-binding activities of the major H3 complex than normal tissue extracts (Wilcoxon test). Neither T-staging nor M-staging at diagnosis was associated with any of the factors, but N-Stage was significantly associated with the ratio of tumor/normal (T/N) for the total of cytoplasmic and nuclear extracts. This means all patients without lymph node affectation had decreased binding ratios (T/N) in both cytoplasmic and nuclear extracts. Kaplan-Meier survival analysis using a cut-off of 1.5 (rounded-up median) for the nuclear binding activity ratio (T/N) showed significantly poorer survival for those patients whose nuclear binding was greater than 1.5. This suggests that although the major triplex DNA-binding protein is present in normal tissue extracts, it is more abundant in tumor extracts, and that a specific abundance of the major triplex-binding protein (H3) in the tumor nucleus is associated with poorer patient survival. Biotinylated triplex DNA-protein complexes were isolated from the RKO colorectal cancer cell line with streptavidin-agarose and analyzed by nano-HPLC ESI-MS-MS. We identified 100 and 60 kDa proteins from nuclear extracts as PSF (polypyrimidine tract binding-associated splicing factor) and p54nrb (NonO), known to function as RNA polymerase II-associated splicing factors, bind as heterodimers, and implicated in the regulation of expression of the myc family of oncoproteins and COX2. We also identified a 75 kDa protein from cytoplasmic extracts as U2AF65 (U2 small nuclear RNA auxiliary factor 2 isoform b), also a splicing factor that can directly interact with p54 nrb and shuttle continuously between the nucleus and cytoplasm. To examine differences between tumor and normal tissues in multiple signaling proteins and proteins known to be relevant in colorectal cancer, reverse phase protein array (RPPA), Pearson correlation and cluster analysis of tumor nuclear lysates ranked from lowest to highest H3 EMSA values shows a highly significant correlation between decreased β-catenin expression in tissues that have the lowest H3 EMSA values and increased β-catenin expression in tissues that have the highest H3 EMSA values. We found this pattern in all tissue lysate types: normal-cytoplasm, normal-nucleus, tumor-cytoplasm, and tumor-nucleus. We believe these splicing factors are overexpressed in tumor tissues and could cause deregulated growth of cells by influencing other proteins associated with tumorigenesis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4566.

Collaboration


Dive into the Laura D. Nelson's collaboration.

Top Co-Authors

Avatar

Dennis P.M. Hughes

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tracy Robson

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anita Yakkundi

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Fiona Furlong

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Lana McClements

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Doris R. Siwak

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hayley D. McKeen

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Helen O. McCarthy

Queen's University Belfast

View shared research outputs
Researchain Logo
Decentralizing Knowledge