Network


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

Hotspot


Dive into the research topics where Joel G. Turner is active.

Publication


Featured researches published by Joel G. Turner.


Biochemical Pharmacology | 2012

Nuclear export of proteins and drug resistance in cancer

Joel G. Turner; Jana L. Dawson; Daniel M. Sullivan

The intracellular location of a protein is crucial to its normal functioning in a cell. Cancer cells utilize the normal processes of nuclear-cytoplasmic transport through the nuclear pore complex of a cell to effectively evade anti-neoplastic mechanisms. CRM1-mediated export is increased in various cancers. Proteins that are exported in cancer include tumor-suppressive proteins such as retinoblastoma, APC, p53, BRAC1, FOXO proteins, INI1/hSNF5, galectin-3, Bok, nucleophosmin, RASSF2, Merlin, p21(CIP), p27(KIP1), N-WASP/FAK, estradiol receptor and Tob, drug targets topoisomerase I and IIα and BCR-ABL, and the molecular chaperone protein Hsp90. Here, we review in detail the current processes and known structures involved in the export of a protein through the nuclear pore complex. We also discuss the export receptor molecule CRM1 and its binding to the leucine-rich nuclear export signal of the cargo protein and the formation of a nuclear export trimer with RanGTP. The therapeutic potential of various CRM1 inhibitors will be addressed, including leptomycin B, ratjadone, KOS-2464, and specific small molecule inhibitors of CRM1, N-azolylacrylate analogs, FOXO export inhibitors, valtrate, acetoxychavicol acetate, CBS9106, and SINE inhibitors. We will also discuss examples of how drug resistance may be reversed by targeting the exported proteins topoisomerase IIα, BCR-ABL, and galectin-3. As effective and less toxic CRM1 export inhibitors become available, they may be used as both single agents and in combination with current chemotherapeutic drugs. We believe that the future development of low-toxicity, small-molecule CRM1 inhibitors may provide a new approach to treating cancer.


Oncogene | 1997

Activation of c-Src by receptor tyrosine kinases in human colon cancer cells with high metastatic potential

Weiguang Mao; Rosalyn B. Irby; Domenico Coppola; Ling Fu; Marek Wloch; Joel G. Turner; Hua Yu; Roy Garcia; Richard Jove; Timothy J. Yeatman

Recent data suggest that signal transduction may have a critical role in the development and regulation of the metastatic phenotype. Here, we investigated the role of c-Src activation in the process of human colon cancer metastasis to the liver. Our data, derived from two different sets of human colon cancer cell line metastatic variants, suggest that not only do highly-metastatic cells display constitutively elevated c-Src protein kinase activity when compared to poorly metastatic cells, but also that receptor tyrosine kinases participate in the ligand-activation of c-Src above basal levels. Specifically, the epidermal growth factor receptor (EGFR), p185HER2/Neu and the hepatocyte growth factor receptor (c-Met) appear to be linked to the process because they preferentially activate c-Src in highly-metastatic cells. EGFR was found to associate with c-Src in colon cancer cells and specific inhibitors of the EGFR resulted in a reduction of c-Src activity to basal levels. In addition, c-Src transfectants displayed partially-activated EGFRs, suggesting a feedback role for c-Src in the regulation of the EGFR. p185HER2/Neu was also identified in immunocomplexes of c-Src following ligand activation of the EGFR, but only in highly-metastatic cells. Collectively, these observations suggest a paradigm whereby c-Src interacts with multiple cell-surface growth factors in a catalytic fashion for the development of tumor cells with metastatic potential.


Journal of Immunology | 2001

Anti-CD40 Antibody Induces Antitumor and Antimetastatic Effects: The Role of NK Cells

Joel G. Turner; Alexander L. Rakhmilevich; Lyudmila Burdelya; Zane C. Neal; Michael Imboden; Paul M. Sondel; Hua Yu

We assessed the effect of the stimulatory anti-CD40 Ab on NK cell activation in vivo and the therapeutic potential of activated NK cells in tumor-bearing mice. Single-dose i.p. injection of the anti-CD40 Ab resulted in production of IL-12 and IFN-γ in vivo, followed by a dramatic increase in NK cell cytolytic activity in PBLs. NK cell activation by anti-CD40 Ab was also observed in CD40 ligand knockout mice. Because NK cells express CD40 ligand but not CD40, our results suggest that NK activation is mediated by increased cytokine production upon CD40 ligation of APCs. Treatment of tumor-bearing mice with anti-CD40 Ab resulted in substantial antitumor and antimetastatic effects in three tumor models. Depletion of NK cells with anti-asialo GM1 Ab reduced or abrogated the observed antitumor effects in all the tested models. These results indicate that a stimulatory CD40 Ab indirectly activates NK cells, which can produce significant antitumor and antimetastatic effects.


Current Medicinal Chemistry | 2008

CRM1-mediated nuclear export of proteins and drug resistance in cancer.

Joel G. Turner; Daniel M. Sullivan

Expression levels of intact tumor suppressor proteins and molecular targets of anti-neoplastic agents are critical in defining cancer cell drug sensitivity; however, the intracellular location of a specific protein may be as important. Many tumor suppressor proteins must be present in the cell nucleus to perform their policing activities or for the cell to respond to chemotherapeutic agents. Nuclear proteins needed to prevent cancer initiation or progression or to optimize chemotherapeutic response include the tumor suppressor proteins p53, APC/beta-catenin, and FOXO family genes; negative regulators of cell cycle progression and survival such as p21(CIP1) and p27(KIP1;) and chemotherapeutic targets such as DNA topoisomerases I and IIalpha. Mislocalization of a nuclear protein into the cytoplasm can render it ineffective as a tumor suppressor or as a target for chemotherapy. Blocking nuclear export of any or all of these proteins may restore tumor suppression or apoptosis or, for topoisomerases I and IIalpha, reverse drug resistance to inhibitors of these enzymes. During disease progression or in response to the tumor environment, cancer cells appear to acquire intracellular mechanisms to export anti-cancer nuclear proteins. These mechanisms generally involve modification of nuclear proteins, causing the proteins to reveal leucine-rich nuclear export signal protein sequences. Subsequent export is mediated by CRM1. This review defines the general processes involved in nuclear export mediated by CRM1/RanGTP (exportin/XPO1), examines the functions of individual tumor suppressor nuclear proteins and nuclear targets of chemotherapy, and explores potential mechanisms of cancer cells to induce export of these proteins. Novel drugs that could potentially counteract nuclear export of specific proteins are also discussed.


Molecular Carcinogenesis | 2005

Osteopontin induces multiple changes in gene expression that reflect the six "hallmarks of cancer" in a model of breast cancer progression.

Amy C. Cook; Alan B. Tuck; Susan McCarthy; Joel G. Turner; Rosalyn B. Irby; Gregory C. Bloom; Timothy J. Yeatman; Ann F. Chambers

Tumor progression is a multistep process, which enables cells to evolve from benign to malignant tumors. This progression has been suggested to depend on six essential characteristics identified as the “hallmarks of cancer,” which include: self‐sufficiency in growth signals, insensitivity to growth‐inhibitory signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. Osteopontin (OPN) is an integrin‐binding protein that has been shown to be associated with the progression of several cancer types, and to play an important functional role in various aspects of malignancy, particularly tissue invasion and metastasis. Here we studied genes regulated by OPN in a model of human breast cancer using oligonucleotide microarray technology by comparing the gene‐expression profiles of 21NT mammary carcinoma cells transfected to overexpress OPN versus mock‐transfected control cells. From over 12,000 human genes, we identified 99 known human genes differentially regulated by OPN whose expression changed by at least 1.5‐fold and showed statistically significant differences in mean expression levels between groups. Functional classification of these genes into the hallmarks of cancer categories showed that OPN can affect the expression of genes involved in all six categories in this model. Furthermore, we were able to validate the expression of 18/19 selected candidate genes by quantitative real‐time PCR, further supporting our microarray findings. This study provides the first evidence that OPN can lead to numerous gene expression changes that influence multiple aspects of tumor progression and malignant growth.


Cancer Research | 2004

Clusterin-Mediated Apoptosis Is Regulated by Adenomatous Polyposis Coli and Is p21 Dependent but p53 Independent

Tingan Chen; Joel G. Turner; Susan McCarthy; Maurizio Scaltriti; Saverio Bettuzzi; Timothy J. Yeatman

Clusterin is a widely expressed glycoprotein that has been paradoxically observed to have both pro- and antiapoptotic functions. Recent reports suggest this apparent dichotomy of function may be related to two different isoforms, one secreted and cytoplasmic, the other nuclear. To clarify the functional role of clusterin in regulating apoptosis, we examined its expression in human colon cancer tissues and in human colon cancer cell lines. We additionally explored its expression and activity using models of adenomatous polyposis coli (APC)- and chemotherapy-induced apoptosis. Clusterin RNA and protein levels were decreased in colon cancer tissues largely devoid of wild-type APC when compared with matched normal tissue controls, suggesting a means for invasive cancers to avoid apoptosis. Conversely, induction of apoptosis by expression of wild-type APC or by treatment with chemotherapy led to increased clusterin RNA and protein levels localizing to apoptotic nuclei. We found that transient transfection of clusterin to colon cancer cell lines directly enhanced basal and chemotherapy-induced apoptosis. Clusterin-induced apoptosis was inhibited by antisense clusterin and was found to be highly dependent on p21 but not p53 expression, yet a deficit in p21 can be subverted by clusterin transfection. Collectively, these data support the hypothesis that nuclear clusterin function is proapoptotic when induced by APC or chemotherapy in the context of p21 expression. Absent of p21, clusterin in not induced, and apoptosis is significantly inhibited. These data support a potential therapeutic role for clusterin in enhancing chemotherapy-induced apoptosis and in promoting apoptosis in cells deficient in p21.


Clinical Cancer Research | 2005

Synergistic Interaction between Histone Deacetylase and Topoisomerase II Inhibitors Is Mediated through Topoisomerase IIβ

Douglas C. Marchion; Elona Bicaku; Joel G. Turner; Adil Daud; Daniel M. Sullivan; Pamela N. Munster

Background: DNA topoisomerase II inhibitors and poisons are among the most efficacious drugs for the treatment of cancer. Sensitivity of cancer cells to the cytotoxic effects of topoisomerase II targeting agents is thought to depend on the expression of the topoisomerase IIα isoform, and drug resistance is often associated with loss or mutation of topoisomerase IIα. Histone deacetylase inhibitors (HDACi) are a novel class of compounds that potentiate the antitumor effects of topoisomerase II–targeting agents. Methods: The interaction between HDACi and topoisomerase II–targeting agents in cancer cells was evaluated as a function of topoisomerase IIα and topoisomerase IIβ expression. Topoisomerase II isoforms were selectively depleted using small interfering RNA and antisense. Drug-induced formation of cleavable complexes involving topoisomerase IIα and topoisomerase IIβ was evaluated by trapped-in-agarose DNA immunostaining and band depletion assays in the presence and absence of HDACi. Results: Preexposure to HDACi increased the cytotoxicity of topoisomerase II poisons. This was associated with a down-regulation of topoisomerase IIα expression but had no effects on topoisomerase IIβ. In the setting of HDACi-induced chromatin decondensation and topoisomerase IIα depletion, topoisomerase II poison cytotoxicity was mediated through topoisomerase IIβ cleavable complex formation. The HDACi-induced sensitization was also observed in cells with target-specific resistance to topoisomerase II poisons. Conclusions: The recruitment of topoisomerase IIβ as a target may overcome primary or emergent drug resistance to topoisomerase II–targeting agents and hence may broaden the applicability of this important class of anticancer agents.


Cancer Research | 2009

Human multiple myeloma cells are sensitized to topoisomerase II inhibitors by CRM1 inhibition.

Joel G. Turner; Douglas C. Marchion; Jana L. Dawson; Michael F. Emmons; Lori A. Hazlehurst; Peter Washausen; Daniel M. Sullivan

Topoisomerase IIalpha (topo IIalpha) is exported from the nucleus of human myeloma cells by a CRM1-dependent mechanism at cellular densities similar to those found in patient bone marrow. When topo IIalpha is trafficked to the cytoplasm, it is not in contact with the DNA; thus, topo IIalpha inhibitors are unable to induce DNA-cleavable complexes and cell death. Using a CRM1 inhibitor or a CRM1-specific small interfering RNA (siRNA), we were able to block nuclear export of topo IIalpha as shown by immunofluorescence microscopy. Human myeloma cell lines and patient myeloma cells isolated from bone marrow were treated with a CRM1 inhibitor or CRM1-specific siRNA and exposed to doxorubicin or etoposide at high cell densities. CRM1-treated cell lines or myeloma patient cells were 4-fold more sensitive to topo II poisons as determined by an activated caspase assay. Normal cells were not significantly affected by CRM1-topo II inhibitor combination treatment. Cell death was correlated with increased DNA double-strand breaks as shown by the comet assay. Band depletion assays of CRM1 inhibitor-exposed myeloma cells showed increased topo IIalpha covalently bound to DNA. Topo IIalpha knockdown by a topo IIalpha-specific siRNA abrogated the CRM1-topo II therapy synergistic effect. These results suggest that blocking topo IIalpha nuclear export sensitizes myeloma cells to topo II inhibitors. This method of sensitizing myeloma cells suggests a new therapeutic approach to multiple myeloma.


Molecular Cancer Therapeutics | 2009

HDAC2 regulates chromatin plasticity and enhances DNA vulnerability

Douglas C. Marchion; Elona Bicaku; Joel G. Turner; Morgen Schmitt; Dawn R. Morelli; Pamela N. Munster

Histone deacetylases (HDAC) may have a prominent role in the development of cancer and the response to anticancer therapy. However, the therapeutic relevance and tissue specificity of individual HDAC enzymes remain largely unknown. HDAC inhibitors may function as sensitizing agents to chemotherapies that target DNA through their effects on chromatin structure and plasticity. Here, we report a new role for HDAC2 as a regulator of chromatin compaction status and the mediator of HDAC inhibitor-induced sensitization to chemotherapy. The selective depletion of HDAC2 by small interfering RNA led to reduced expression of heterochromatin maintenance proteins and morphologic changes indicative of chromatin decondensation. Furthermore, depletion of HDAC2 but not HDAC1 or HDAC6 was sufficient to sensitize breast cancer cells to topoisomerase inhibitor-induced apoptosis. The levels of HDAC2 expression appear to correlate with the degree of HDAC inhibitor-induced histone acetylation in a surrogate tissue in patients. These data suggest that HDAC2 may be a relevant pharmacologic and biological target for combination therapy involving drugs that target DNA.[Mol Cancer Ther 2009;8(4):794–801]


Seminars in Cancer Biology | 2014

Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents

Joel G. Turner; Jana L. Dawson; Christopher L. Cubitt; Rachid Baz; Daniel M. Sullivan

Nuclear-cytoplasmic trafficking of proteins is a significant factor in the development of cancer and drug resistance. Subcellular localization of exported proteins linked to cancer development include those involved in cell growth and proliferation, apoptosis, cell cycle regulation, transformation, angiogenesis, cell adhesion, invasion, and metastasis. Here, we examined the basic mechanisms involved in the export of proteins from the nucleus to the cytoplasm. All proteins over 40kDa use the nuclear pore complex to gain entry or exit from the nucleus, with the primary nuclear export molecule involved in these processes being chromosome region maintenance 1 (CRM1, exportin 1 or XPO1). Proteins exported from the nucleus must possess a hydrophobic nuclear export signal (NES) peptide that binds to a hydrophobic groove containing an active-site Cys528 in the CRM1 protein. CRM1 inhibitors function largely by covalent modification of the active site Cys528 and prevent binding to the cargo protein NES. In the absence of a CRM1 inhibitor, CRM1 binds cooperatively to the NES of the cargo protein and RanGTP, forming a trimer that is actively transported out of the nucleus by facilitated diffusion. Nuclear export can be blocked by CRM1 inhibitors, NES peptide inhibitors or by preventing post-translational modification of cargo proteins. Clinical trials using the classic CRM1 inhibitor leptomycin B proved too toxic for patients; however, a new generation of less toxic small molecule inhibitors is being used in clinical trials in patients with both hematological malignancies and solid tumors. Additional trials are being initiated using small-molecule CRM1 inhibitors in combination with chemotherapeutics such as pegylated liposomal doxorubicin. In this review, we present evidence that combining the new CRM1 inhibitors with other classes of therapeutics may prove effective in the treatment of cancer. Potential combinatorial therapies discussed include the use of CRM1 inhibitors and the addition of alkylating agents (melphalan), anthracyclines (doxorubicin and daunomycin), BRAF inhibitors, platinum drugs (cisplatin and oxaliplatin), proteosome inhibitors (bortezomib and carfilzomib), or tyrosine-kinase inhibitors (imatinib). Also, the sequence of treatment may be important for combination therapy. We found that the most effective treatment regimen involved first priming the cancer cells with the CRM1 inhibitor followed by doxorubicin, bortezomib, carfilzomib, or melphalan. This order sensitized both de novo and acquired drug-resistant cancer cell lines.

Collaboration


Dive into the Joel G. Turner's collaboration.

Top Co-Authors

Avatar

Daniel M. Sullivan

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jana L. Dawson

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Hua Yu

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

William S. Dalton

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Kenneth H. Shain

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven Grant

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Yun Dai

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge