Network


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

Hotspot


Dive into the research topics where Kimberly Batten is active.

Publication


Featured researches published by Kimberly Batten.


PLOS ONE | 2011

Multipotent Capacity of Immortalized Human Bronchial Epithelial Cells

Oliver Delgado; Aadil Kaisani; Monica Spinola; Xian Jin Xie; Kimberly Batten; John D. Minna; Woodring E. Wright; Jerry W. Shay

While the adult murine lung utilizes multiple compartmentally restricted progenitor cells during homeostasis and repair, much less is known about the progenitor cells from the human lung. Translating the murine stem cell model to humans is hindered by anatomical differences between species. Here we show that human bronchial epithelial cells (HBECs) display characteristics of multipotent stem cells of the lung. These HBECs express markers indicative of several epithelial types of the adult lung when experimentally tested in cell culture. When cultured in three different three-dimensional (3D) systems, subtle changes in the microenvironment result in unique responses including the ability of HBECs to differentiate into multiple central and peripheral lung cell types. These new findings indicate that the adult human lung contains a multipotent progenitor cell whose differentiation potential is primarily dictated by the microenvironment. The HBEC system is not only important in understanding mechanisms for specific cell lineage differentiation, but also for examining changes that correlate with human lung diseases including lung cancer.


Clinical Cancer Research | 2013

A targeted RNAi screen of the breast cancer genome identifies KIF14 and TLN1 as genes that modulate docetaxel chemosensitivity in triple-negative breast cancer

Stina Mui Singel; Crystal Cornelius; Kimberly Batten; Gail Fasciani; Woodring E. Wright; Lawrence Lum; Jerry W. Shay

Purpose: To identify biomarkers within the breast cancer genome that may predict chemosensitivity in breast cancer. Experimental Design: We conducted an RNA interference (RNAi) screen within the breast cancer genome for genes whose loss-of-function enhanced docetaxel chemosensitivity in an estrogen receptor–negative, progesterone receptor–negative, and Her2-negative (ER−, PR−, and Her2−, respectively) breast cancer cell line, MDA-MB-231. Top candidates were tested for their ability to modulate chemosensitivity in 8 breast cancer cell lines and to show in vivo chemosensitivity in a mouse xenograft model. Results: From ranking chemosensitivity of 328 short hairpin RNA (shRNA) MDA-MB-231 cell lines (targeting 133 genes with known somatic mutations in breast cancer), we focused on the top two genes, kinesin family member 14 (KIF14) and talin 1 (TLN1). KIF14 and TLN1 loss-of-function significantly enhanced chemosensitivity in four triple-negative breast cancer (TNBC) cell lines (MDA-MB-231, HCC38, HCC1937, and Hs478T) but not in three hormone receptor–positive cell lines (MCF7, T47D, and HCC1428) or normal human mammary epithelial cells (HMEC). Decreased expression of KIF14, but not TLN1, also enhanced docetaxel sensitivity in a Her2-amplified breast cancer cell line, SUM190PT. Higher KIF14 and TLN1 expressions are found in TNBCs compared with the other clinical subtypes. Mammary fat pad xenografts of KIF14- and TLN1-deficient MDA-MB-231 cells revealed reduced tumor mass compared with control MDA-MB-231 cells after chemotherapy. KIF14 expression is also prognostic of relapse-free and overall survival in representative breast cancer expression arrays. Conclusion: KIF14 and TLN1 are modulators of response to docetaxel and potential therapeutic targets in TNBC. Clin Cancer Res; 19(8); 2061–70. ©2013 AACR.


Radiation Research | 2010

CDDO-Me Protects against Space Radiation-Induced Transformation of Human Colon Epithelial Cells

Ugur Eskiocak; Sang Bum Kim; Andres I. Roig; Erin Kitten; Kimberly Batten; Crystal Cornelius; Ying S. Zou; Woodring E. Wright; Jerry W. Shay

Abstract Radiation-induced carcinogenesis is a major concern both for astronauts on long-term space missions and for cancer patients being treated with therapeutic radiation. Exposure to radiation induces oxidative stress and chronic inflammation, which are critical initiators and promoters of carcinogenesis. Many studies have demonstrated that non-steroidal anti-inflammatory drugs and antioxidants can reduce the risk of radiation-induced cancer. In this study, we found that a synthetic triterpenoid, CDDO-Me (bardoxolone methyl), was able to protect human colon epithelial cells (HCECs) against radiation-induced transformation. HCECs that were immortalized by ectopic expression of hTERT and cdk4 and exhibit trisomy for chromosome 7 (a non-random chromosome change that occurs in 37% of premalignant colon adenomas) can be transformed experimentally with one combined exposure to 2 Gy of protons at 1 GeV/nucleon followed 24 h later by 50 cGy of 56Fe ions at 1 GeV/nucleon. Transformed cells showed an increase in proliferation rate and in both anchorage-dependent and independent colony formation ability. A spectrum of chromosome aberrations was observed in transformed cells, with 40% showing loss of 17p (e.g. loss of one copy of p53). Pretreatment of cells with pharmacological doses of CDDO-Me, which has been shown to induce antioxidative as well as anti-inflammatory responses, prevented the heavy-ion-induced increase in proliferation rate and anchorage-dependent and independent colony formation efficiencies. Taken together, these results demonstrate that experimentally immortalized human colon epithelial cells with a non-random chromosome 7 trisomy are valuable premalignant cellular reagents that can be used to study radiation-induced colorectal carcinogenesis. The utility of premalignant HCECs to test novel compounds such as CDDO-Me that can be used to protect against radiation-induced neoplastic transformation is also demonstrated.


Breast Cancer Research | 2014

Receptor-interacting protein kinase 2 promotes triple-negative breast cancer cell migration and invasion via activation of nuclear factor-kappaB and c-Jun N-terminal kinase pathways.

Stina Mui Singel; Kimberly Batten; Crystal Cornelius; Gaoxiang Jia; Gail Fasciani; Summer Barron; Woodring E. Wright; Jerry W. Shay

IntroductionMetastasis is the main cause of breast cancer morbidity and mortality. Processes that allow for tumor cell migration and invasion are important therapeutic targets. Here we demonstrate that receptor-interacting protein kinase 2 (RIP2), a kinase known to be involved in inflammatory processes, also has novel roles in cancer cell migration and invasion.MethodsA total of six breast cancer expression databases, including The Cancer Genome Atlas, were assessed for RIP2 expression among various clinical subtypes and its role as a prognostic biomarker. mRNA fluorescence in situ hybridization (FISH) for RIP2 was performed on 17 stage III breast cancers to determine if there was a correlation between RIP2 expression and lymph node involvement. RNA-interference was used to knock-down RIP2 expression in MDA-MB-231, Htb126, SUM149PT, MCF7, T47D, and HCC1428 cells. Cell migration and invasion were measured in vitro by scratch/wound healing and transwell migration assays. A xenograft mouse model was used to assess tumor growth and chemosensitivity to docetaxel in vivo in MDA-MB-231 cells with and without RIP2 small hairpin RNA knockdown. Western blot and immunofluorescence imaging were used to evaluate protein expressions.ResultsInterrogation of expression databases showed that RIP2 expression is significantly over-expressed in triple-negative breast cancers (TNBC: estrogen-receptor (ER) negative, progesterone-receptor (PR) negative, Her2/neu- (Her2) negative), compared to other clinical subtypes. High RIP2 expression correlates with worse progression-free survival using a combined breast cancer expression array dataset consisting of 946 patients. Multivariate analysis shows RIP2 as an independent prognostic biomarker. Knock-down of RIP2 significantly decreases migration in both scratch/wound healing and transwell migration assays in MDA-MB-231, Htb126, SUM149PT, MCF7, and T47D cells and is correlated with decreased Nuclear Factor-kappaB and c-Jun N-terminal kinase (JNK) activation. Finally, RIP2 knock-down leads to increased sensitivity to docetaxel and decreased tumor mass and lung metastases in a xenograft mouse model.ConclusionThese results highlight RIP2 as a pro-metastasis kinase in patients with advanced breast cancer. These results also illustrate a novel role for this kinase in addition to its known role in inflammation, and suggest that targeting RIP2 may improve outcomes in advanced breast cancer patients, in which it is overexpressed.


Clinical Cancer Research | 2014

Radiation-Enhanced Lung Cancer Progression in a Transgenic Mouse Model of Lung Cancer Is Predictive of Outcomes in Human Lung and Breast Cancer

Oliver Delgado; Kimberly Batten; James A. Richardson; Xian Jin Xie; Adi F. Gazdar; Aadil Kaisani; Luc Girard; Carmen Behrens; Milind Suraokar; Gail Fasciani; Woodring E. Wright; Michael D. Story; Ignacio I. Wistuba; John D. Minna; Jerry W. Shay

Purpose: Carcinogenesis is an adaptive process between nascent tumor cells and their microenvironment, including the modification of inflammatory responses from antitumorigenic to protumorigenic. Radiation exposure can stimulate inflammatory responses that inhibit or promote carcinogenesis. The purpose of this study is to determine the impact of radiation exposure on lung cancer progression in vivo and assess the relevance of this knowledge to human carcinogenesis. Experimental Design: K-rasLA1 mice were irradiated with various doses and dose regimens and then monitored until death. Microarray analyses were performed using Illumina BeadChips on whole lung tissue 70 days after irradiation with a fractionated or acute dose of radiation and compared with age-matched unirradiated controls. Unique group classifiers were derived by comparative genomic analysis of three experimental cohorts. Survival analyses were performed using principal component analysis and k-means clustering on three lung adenocarcinoma, three breast adenocarcinoma, and two lung squamous carcinoma annotated microarray datasets. Results: Radiation exposure accelerates lung cancer progression in the K-rasLA1 lung cancer mouse model with dose fractionation being more permissive for cancer progression. A nonrandom inflammatory signature associated with this progression was elicited from whole lung tissue containing only benign lesions and predicts human lung and breast cancer patient survival across multiple datasets. Immunohistochemical analyses suggest that tumor cells drive predictive signature. Conclusions: These results demonstrate that radiation exposure can cooperate with benign lesions in a transgenic model of cancer by affecting inflammatory pathways, and that clinically relevant similarities exist between human lung and breast carcinogenesis. Clin Cancer Res; 20(6); 1610–22. ©2014 AACR.


Journal of Biomolecular Screening | 2012

Development of Methods for Quantitative Comparison of Pooled shRNAs by Mass Sequencing

Hirotoshi Hoshiyama; Jun Tang; Kimberly Batten; Guanghua Xiao; Jean Marie Rouillard; Jerry W. Shay; Yang Xie; Woodring E. Wright

Pooled short-hairpin RNA (shRNA) library screening is a powerful tool for identifying a set of genes in biological pathways that require stable expression to produce a desired phenotype. Massive parallel sequencing of half-hairpins has proven highly variable and has not given satisfactory results concerning the relative abundance of different shRNAs before and after selection. Here, the authors describe a method for quantitative comparison of half-hairpins from pooled shRNAs in the mir30-based pGIPZ vector that is analyzed by massive parallel sequencing. Introducing a multiplexing code and refining the sample preparation scheme resulted in the predicted ability to detect twofold enrichments. These improvements should permit half-hairpin sequencing to analyze either dropout screens or selective pooled shRNA screens of limited stringency to analyze phenotypes not accessible in transient experiments.


Scientific Reports | 2016

Organotypic culture in three dimensions prevents radiation-induced transformation in human lung epithelial cells.

Mariam El-Ashmawy; Melissa Coquelin; Krishna Luitel; Kimberly Batten; Jerry W. Shay

The effects of radiation in two-dimensional (2D) cell culture conditions may not recapitulate tissue responses as modeled in three-dimensional (3D) organotypic culture. In this study, we determined if the frequency of radiation-induced transformation and cancer progression differed in 3D compared to 2D culture. Telomerase immortalized human bronchial epithelial cells (HBECs) with shTP53 and mutant KRas expression were exposed to various types of radiation (gamma, +H, 56Fe) in either 2D or 3D culture. After irradiation, 3D structures were dissociated and passaged as a monolayer followed by measurement of transformation, cell growth and expression analysis. Cells irradiated in 3D produced significantly fewer and smaller colonies in soft agar than their 2D-irradiated counterparts (gamma P = 0.0004; +H P = 0.049; 56Fe P < 0.0001). The cell culture conditions did not affect cell killing, the ability of cells to survive in a colony formation assay, and proliferation rates after radiation—implying there was no selection against cells in or dissociated from 3D conditions. However, DNA damage repair and apoptosis markers were increased in 2D cells compared to 3D cells after radiation. Ideally, expanding the utility of 3D culture will allow for a better understanding of the biological consequences of radiation exposure.


Neoplasia | 2018

Telomerase-Mediated Strategy for Overcoming Non–Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance

Ilgen Mender; Ryan LaRanger; Krishna Luitel; Michael Peyton; Luc Girard; Tsung Po Lai; Kimberly Batten; Crystal Cornelius; Maithili P. Dalvi; Michael Ramirez; Wenting Du; Lani F. Wu; Steven J. Altschuler; Rolf A. Brekken; Elisabeth D. Martinez; John D. Minna; Woodring E. Wright; Jerry W. Shay

Standard and targeted cancer therapies for late-stage cancer patients almost universally fail due to tumor heterogeneity/plasticity and intrinsic or acquired drug resistance. We used the telomerase substrate nucleoside precursor, 6-thio-2′-deoxyguanosine (6-thio-dG), to target telomerase-expressing non–small cell lung cancer cells resistant to EGFR-inhibitors and commonly used chemotherapy combinations. Colony formation assays, human xenografts as well as syngeneic and genetically engineered immune competent mouse models of lung cancer were used to test the effect of 6-thio-dG on targeted therapy– and chemotherapy-resistant lung cancer human cells and mouse models. We observed that erlotinib-, paclitaxel/carboplatin-, and gemcitabine/cisplatin-resistant cells were highly sensitive to 6-thio-dG in cell culture and in mouse models. 6-thio-dG, with a known mechanism of action, is a potential novel therapeutic approach to prolong disease control of therapy-resistant lung cancer patients with minimal toxicities.


Nature Communications | 2018

NOVA1 regulates hTERT splicing and cell growth in non-small cell lung cancer

Andrew T. Ludlow; Mandy S. Wong; Jerome D. Robin; Kimberly Batten; Laura Yuan; Tsung Po Lai; Nicole Dahlson; Lu Zhang; Ilgen Mender; Enzo Tedone; Mohammed E. Sayed; Woodring E. Wright; Jerry W. Shay

Alternative splicing is dysregulated in cancer and the reactivation of telomerase involves the splicing of TERT transcripts to produce full-length (FL) TERT. Knowledge about the splicing factors that enhance or silence FL hTERT is lacking. We identified splicing factors that reduced telomerase activity and shortened telomeres using a siRNA minigene reporter screen and a lung cancer cell bioinformatics approach. A lead candidate, NOVA1, when knocked down resulted in a shift in hTERT splicing to non-catalytic isoforms, reduced telomerase activity, and progressive telomere shortening. NOVA1 knockdown also significantly altered cancer cell growth in vitro and in xenografts. Genome engineering experiments reveal that NOVA1 promotes the inclusion of exons in the reverse transcriptase domain of hTERT resulting in the production of FL hTERT transcripts. Utilizing hTERT splicing as a model splicing event in cancer may provide new insights into potentially targetable dysregulated splicing factors in cancer.Splicing of the telomerase transcript, hTERT, can be altered in cancer cells. Here the authors report NOVA1, as a splicing regulator that directly interacts with hTERT pre-mRNA enhancing the number of full-length transcripts and thus telomerase activity in lung cancer cells.


Molecular Cancer Research | 2016

Abstract B22: Acyl-CoA synthetase long-chain family member 3 dependent lipid homeostasis is required for mutant KRAS driven lung cancer

Mahesh S. Padanad; Georgia Konstantinidou; Chendong Yang; Margherita Melegari; Niranjan Venkateswaran; Kimberly Batten; Kenneth Huffman; Jerry W. Shay; John D. Minna; Ralph J. DeBerardinis; Pier Paolo Scaglioni

Lung cancer is the leading cause of cancer related deaths in the USA and worldwide. Lung tumorigenesis is a multistep process that involves several genetic aberrations. Activating mutations of the proto-oncogene KRAS (mutant KRAS) occur in ~30% of the cases of human non-small cell lung cancer (NSCLC), which is associated with aggressive, therapy-resistant disease. Despite the recent discovery of low affinity inhibitors, mutant KRAS is a challenging therapeutic target and there is a dearth of therapeutic options for these tumors. Mutant KRAS not only promotes tumorigenesis but also the survival of established lung cancer, both in mouse models and in certain human NSCLC lines. Therefore, in the absence of clinically-relevant effective inhibitors of mutant KRAS, there has been an intense clinical interest in the development of inhibitors of its downstream effectors. Importantly, mutant KRAS cancer cells undergo oncogene-directed metabolic reprogramming in order to meet the energetic and biosynthetic challenges of cell survival, growth and proliferation. Activation of certain pathways of fatty acid synthesis has been observed in many cancer types including lung cancer. Till date, fatty acid synthase (FASN) has been the candidate for drug development. Unfortunately, the inhibitors against FASN have poor pharmacokinetics and target related toxicity concerns. There is an urgent need for discovery of additional targets that inhibit lipid metabolism specifically in cancer cells that could be exploited for therapeutic gain. The goal of our study was to identify the cellular networks that mediate the maintenance of mutant KRAS lung cancer which further could be used as high priority therapeutic targets. To this end, we functionally analyzed the transcriptome of transgenic mouse lung tumors manipulated in vivo to undergo mutant Kras extinction, providing isogenic comparisons between mutant KRAS extinguished versus non-extinguished tumors for the discovery of new therapeutic targets. We determined that mutant KRAS controls tumor metabolism by regulating lipid homeostasis. We found that Acyl-CoA synthetase long-chain family member 3 (ACSL3), which converts fatty acids into fatty Acyl-CoA esters, the substrate for lipid synthesis and β-oxidation, is required for the survival of mutant KRAS lung cancer cells. These effects were not due to generalized toxicity, since we did not observe them in immortalized human bronchoalveolar cells and several NSCLC cells expressing wild type KRAS and in Acsl3 null mouse embryonic fibroblasts. We confirmed that ACSL3 is a mutant KRAS responsive gene expressed in the respiratory epithelium, in lung cancer cells and in primary human cancers. With mechanistic experiments we determined that mutant KRAS stimulates, in an ACSL3-dependent manner, the uptake and retention of fatty acids by lung cancer cells as well as their β-oxidation. As predicted by these experiments, ACSL3 is essential for the ability of mutant KRAS human lung cancer cells to form colonies in soft agar or to establish xenografts in immunocompromised mice. In addition, our preliminary results shows reduced tumor size and tumor burden in KrasG12D;Acsl3-/- mice. The detailed characterization of these mice is currently in progress. Our data demonstrate that mutant KRAS reprograms lipid homeostasis in lung cancer, establishing a cancer specific metabolic vulnerability. Thus, ACSL3 could be a viable therapeutic target for NSCLC driven by mutant KRAS. Citation Format: Mahesh S. Padanad, Georgia Konstantinidou, Chendong Yang, Margherita Melegari, Niranjan Venkateswaran, Kimberly Batten, Kenneth E. Huffman, Jerry W. Shay, John D. Minna, Ralph J. DeBerardinis, Pier P. Scaglioni. Acyl-CoA synthetase long-chain family member 3 dependent lipid homeostasis is required for mutant KRAS driven lung cancer. [abstract]. In: Proceedings of the AACR Special Conference: Metabolism and Cancer; Jun 7-10, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(1_Suppl):Abstract nr B22.

Collaboration


Dive into the Kimberly Batten's collaboration.

Top Co-Authors

Avatar

Jerry W. Shay

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Woodring E. Wright

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Crystal Cornelius

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

John D. Minna

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lawrence Lum

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stina Mui Singel

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gail Fasciani

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andres I. Roig

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Georgia Konstantinidou

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Oliver Delgado

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge