Network


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

Hotspot


Dive into the research topics where Colin D. Crean is active.

Publication


Featured researches published by Colin D. Crean.


Clinical Cancer Research | 2007

OSU-03012, a novel celecoxib derivative, is cytotoxic to myeloma cells and acts through multiple mechanisms

Shuhong Zhang; Attaya Suvannasankha; Colin D. Crean; Valerie L. White; Amy J. Johnson; Ching-Shih Chen; Sherif S. Farag

Purpose: OSU-03012 is a novel celecoxib derivative, without cyclooxygenase-2 inhibitory activity, capable of inducing apoptosis in various cancer cells types, and is being developed as an anticancer drug. We investigated the in vitro activity of OSU-03012 in multiple myeloma (MM) cells. Experimental Design: U266, ARH-77, IM-9, and RPMI-8226, and primary myeloma cells were exposed to OSU-03012 for 6, 24, or 72 h. Cytotoxicity, caspase activation, apoptosis, and effects on intracellular signaling pathways were assessed. Results: OSU-03012 was cytotoxic to MM cells with mean LC50 3.69 ± 0.23 and 6.25 ± 0.86 μmol/L and at 24 h for primary MM cells and cell lines, respectively. As a known PDK-1 inhibitor, OSU-03012 inhibited the PI3K/Akt pathway with downstream effects on BAD, GSK-3β, FoxO1a, p70S6K, and MDM-2. However, transfection of MM cells with constitutively active Akt failed to protect against cell death, indicating activity against other pathways is important. Phospho (p)-signal transducers and activators of transcription 3 and p-MAP/ERK kinase 1/2 were down-regulated, suggesting that OSU-03012 also inhibited the Janus-activated kinase 2/signal transducer and activator of transcription 3 and mitogen-activated protein kinase pathways. Although expression of Bcl-2 proteins was unchanged, OSU-03012 also down-regulated survivin and X-linked inhibitor of apoptosis (XIAP), and also induced G2 cell cycle arrest with associated reductions in cyclins A and B. Finally, although OSU-03012 induced cleavage of caspases 3, 8 and 9, caspase inhibition did not prevent cell death. Conclusions: We conclude that OSU-03012 has potent activity against MM cells and acts via different mechanisms in addition to phosphoinositide-3-kinase/Akt pathway inhibition. These studies provide rationale for the clinical investigation of OSU-03012 in MM.


Clinical Cancer Research | 2008

Antimyeloma Effects of a Sesquiterpene Lactone Parthenolide

Attaya Suvannasankha; Colin D. Crean; Rajasubramaniam Shanmugam; Sherif S. Farag; Rafat Abonour; H. Scott Boswell; Harikrishna Nakshatri

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.


Mammalian Genome | 1998

Expression and chromosomal localization of the Requiem gene

Theodore G. Gabig; Colin D. Crean; Alison Klenk; Haiyan Long; Neal G. Copeland; Debra J. Gilbert; Nancy A. Jenkins; Danielle Quincey; Fabienne Parente; Françoise Lespinasse; Georges F. Carle; Patrick Gaudray; Chang X. Zhang; Alain Calender; Jo Hoeppener; Koen Kas; Rajesh V. Thakker; Filip Farnebo; Bin Tean Teh; Catharina Larsson; Fredrik Piehl; Jacob Lagercrantz; Shideh Khodaei; Emma Carson; Günther Weber

Abstract. Apoptosis in murine myeloid cell lines requires the expression of the Requiem gene, which encodes a putative zinc finger protein. We detected the protein in both cytoplasmic and nuclear subcellular fractions of murine myeloid cells and human K562 leukemia cells, which suggests that the protein might have a function distinct from a transcription factor. This distribution did not alter upon apoptosis induction by IL-3 deprivation. As an approach to investigate its role in development, we determined the spatio-temporal expression pattern in the mouse. Expression was detected in various tissues in earlier gestational age; however, confined to testes, spleen, thymus, and part of the hippocampus in the adult mouse. The expression profile is consistent with a functional role during rapid growth and cell turnover, and in agreement with a regulatory function for hematopoietic cells. The human cDNA clone sequenced showed high homology to its murine counterpart and extended the open reading frame by 20 codons upstream. The gene is located in the proximal region of mouse Chromosome (Chr) 19. In the homologous human region at 11q13, it is located at about 150 kb centromeric from MLK3.


British Journal of Haematology | 2010

Preclinical activity of a novel multiple tyrosine kinase and aurora kinase inhibitor, ENMD-2076, against multiple myeloma.

Xiaojing Wang; Anthony L. Sinn; Karen E. Pollok; George E. Sandusky; Shuhong Zhang; Li Chen; Jing Liang; Colin D. Crean; Attaya Suvannasankha; Rafat Abonour; Carolyn Sidor; Mark R. Bray; Sherif S. Farag

ENMD‐2076 is a novel, orally‐active molecule that has been shown to have significant activity against aurora and multiple receptor tyrosine kinases. We investigated the activity of ENMD‐2076 against multiple myeloma (MM) cells in vitro and in vivo. ENMD‐2076 showed significant cytotoxicity against MM cell lines and primary cells, with minimal cytotoxicity to haematopoietic progenitors. ENMD‐2076 inhibited the phosphoinositide 3‐kinase/AKT pathway and downregulated survivin and X‐linked inhibitor of apoptosis as early as 6 h after treatment. With longer treatment (24–48 h), ENMD‐2076 also inhibited aurora A and B kinases, and induced G2/M cell cycle arrest. In non‐obese diabetic/severe combined immunodeficient mice implanted with H929 human plasmacytoma xenografts, oral treatment with ENMD‐2076 (50, 100, 200 mg/kg per day) resulted in a dose‐dependent inhibition of tumour growth. Immunohistochemical staining of excised tumours showed significant reduction in phospho‐Histone 3 (pH3), Ki‐67, and angiogenesis, and also a significant increase in cleaved caspase‐3 at all dose levels compared to tumours from vehicle‐treated mice. In addition, a significant reduction in p‐FGFR3 was observed on Western blot. ENMD‐2076 shows significant activity against MM cells in vitro and in vivo, and acts on several pathways important for myeloma cell growth and survival. These results provide preclinical rationale for clinical investigation of ENMD‐2076 in MM.


Cell Death & Differentiation | 2005

TNF α resistance in MCF-7 breast cancer cells is associated with altered subcellular localization of p21 CIP1 and p27 KIP1

Zhuo Wang; Hiromitsu Kishimoto; Poornima Bhat-Nakshatri; Colin D. Crean; Harikrishna Nakshatri

TNF α resistance in MCF-7 breast cancer cells is associated with altered subcellular localization of p21 CIP1 and p27 KIP1


Clinical Cancer Research | 2013

Temozolomide-Mediated DNA Methylation in Human Myeloid Precursor Cells: Differential Involvement of Intrinsic and Extrinsic Apoptotic Pathways

Haiyan Wang; Shanbao Cai; Aaron Ernstberger; Barbara J. Bailey; Wang Mz; Cai W; Goebel Ws; Czader Mb; Colin D. Crean; Attaya Suvannasankha; Shokolenkoc I; Glenn L. Wilson; Arthur R. Baluyut; Lindsey D. Mayo; Karen E. Pollok

Purpose: An understanding of how hematopoietic cells respond to therapy that causes myelosuppression will help develop approaches to prevent this potentially life-threatening toxicity. The goal of this study was to determine how human myeloid precursor cells respond to temozolomide (TMZ)-induced DNA damage. Experimental Design: We developed an ex vivo primary human myeloid precursor cells model system to investigate the involvement of cell-death pathways using a known myelosuppressive regimen of O6-benzylguanine (6BG) and TMZ. Results: Exposure to 6BG/TMZ led to increases in p53, p21, γ-H2AX, and mitochondrial DNA damage. Increases in mitochondrial membrane depolarization correlated with increased caspase-9 and -3 activities following 6BG/TMZ treatment. These events correlated with decreases in activated AKT, downregulation of the DNA repair protein O6-methylguanine–DNA methyltransferase (MGMT), and increased cell death. During myeloid precursor cell expansion, FAS/CD95/APO1(FAS) expression increased over time and was present on approximately 100% of the cells following exposure to 6BG/TMZ. Although c-flipshort, an endogenous inhibitor of FAS-mediated signaling, was decreased in 6BG/TMZ–treated versus control, 6BG-, or TMZ alone–treated cells, there were no changes in caspase-8 activity. In addition, there were no changes in the extent of cell death in myeloid precursor cells exposed to 6BG/TMZ in the presence of neutralizing or agonistic anti-FAS antibodies, indicating that FAS-mediated signaling was not operative. Conclusions: In human myeloid precursor cells, 6BG/TMZ–initiated apoptosis occurred by intrinsic, mitochondrial-mediated and not extrinsic, FAS-mediated apoptosis. Human myeloid precursor cells represent a clinically relevant model system for gaining insight into how hematopoietic cells respond to chemotherapeutics and offer an approach for selecting effective chemotherapeutic regimens with limited hematopoietic toxicity. Clin Cancer Res; 19(10); 2699–709. ©2013 AACR.


Surgery | 2015

Targeted nuclear factor-kappaB suppression enhances gemcitabine response in human pancreatic tumor cell line murine xenografts.

Joshua A. Waters; Jesus M. Matos; Michele T. Yip-Schneider; Juan R. Aguilar-Saavedra; Colin D. Crean; Joal D. Beane; Ryan P. Dumas; Attaya Suvannasankha; C. Max Schmidt

BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an almost uniformly fatal malignancy characterized by resistance to chemotherapy. Currently, gemcitabine is the agent used most commonly but demonstrates only a partial response. The transcription factor nuclear factor-kappaB (NF-κB), known to be involved in the inflammatory response, is constitutively activated in PDAC and further activated by gemcitabine. Our aim was to examine the effects of targeted NF-κB suppression on gemcitabine resistance using an in vivo tumor growth model. METHODS To suppress the NF-κB pathway, the mutant IκBα super-repressor protein was stably expressed in PaCa-2 human PDAC cells. Athymic mice were injected subcutaneously with IκBα-super-repressor (SR) or vector-expressing PaCa-2 cells and randomized to receive phosphate-buffered saline (PBS) or 100 mg/kg gemcitabine(gem) for 4 weeks. RESULTS The mean increase in tumor volume was 47 mm(3) (89%) and 196 mm(3) (326%) in gem/SR and gem/vector groups, respectively (P = .03). The PBS-treated groups demonstrated greater tumor growth, ∼340 mm(3) (850%) increase, in both PBS/vector and PBS/SR groups. Intratumoral NF-κB activity was decreased in gem/SR compared with the gem/vector group (P = .04). Decreased Ki-67 positivity was noted in gem/SR (49%) versus gem/vector tumors (73%) (P = .04), with no difference in apoptosis (apoptag, P = .3) or angiogenesis (CD31+, P = .9). CONCLUSION Stable IκBα-SR expression in vivo potentiated the antitumor effects of gemcitabine, resulting in decreased tumor growth in association with decreased cell proliferation. Molecular suppression of the NF-κB pathway decreases successfully gemcitabine resistance in a relatively chemoresistant PDAC line. Thus, NF-κB-targeted agents may complement gemcitabine-based therapies and decrease chemoresistance in patients with PDAC.


Cancer Research | 2016

Cell adhesion molecule CD166 drives malignant progression and osteolytic disease in multiple myeloma

Linlin Xu; Khalid S. Mohammad; Hao Wu; Colin D. Crean; Bradley Poteat; Ying-Hua Cheng; Angelo A. Cardoso; Christophe Machal; Helmut Hanenberg; Rafat Abonour; Melissa A. Kacena; John M. Chirgwin; Attaya Suvannasankha; Edward F. Srour

Multiple myeloma is incurable once osteolytic lesions have seeded at skeletal sites, but factors mediating this deadly pathogenic advance remain poorly understood. Here, we report evidence of a major role for the cell adhesion molecule CD166, which we discovered to be highly expressed in multiple myeloma cell lines and primary bone marrow cells from patients. CD166+ multiple myeloma cells homed more efficiently than CD166- cells to the bone marrow of engrafted immunodeficient NSG mice. CD166 silencing in multiple myeloma cells enabled longer survival, a smaller tumor burden, and less osteolytic lesions, as compared with mice bearing control cells. CD166 deficiency in multiple myeloma cell lines or CD138+ bone marrow cells from multiple myeloma patients compromised their ability to induce bone resorption in an ex vivo organ culture system. Furthermore, CD166 deficiency in multiple myeloma cells also reduced the formation of osteolytic disease in vivo after intratibial engraftment. Mechanistic investigation revealed that CD166 expression in multiple myeloma cells inhibited osteoblastogenesis of bone marrow-derived osteoblast progenitors by suppressing Runx2 gene expression. Conversely, CD166 expression in multiple myeloma cells promoted osteoclastogenesis by activating TRAF6-dependent signaling pathways in osteoclast progenitors. Overall, our results define CD166 as a pivotal director in multiple myeloma cell homing to the bone marrow and multiple myeloma progression, rationalizing its further study as a candidate therapeutic target for multiple myeloma treatment. Cancer Res; 76(23); 6901-10. ©2016 AACR.


Journal of Proteomics & Bioinformatics | 2018

Proteomic Characterization of Plasma Cells from Patients with Multiple Myeloma

Attaya Suvannasankha; Colin D. Crean; Heather M Leyes; Sariya Wongsaengsak; Guihong Qi; Jong Won Kim; Mu Wang

Introduction: Quantitative proteomics approaches have provided insight into biomarkers of cancer and other diseases with high sensitivity, high specificity, and high analytical precision. Multiple Myeloma is an incurable, fatal blood cancers characterized by clonal expansion of plasma cells in the bone marrow. Current multiple myeloma proteomic research mainly focuses on serum biomarkers, not plasma cells, due to technical difficulties including a requirement for tumor cell isolation from bone marrow aspirates, tumor cell paucity and poor in vitro survival after isolation. Materials and methods: A global proteomic analysis was performed using sorted bone marrow plasma cells from normal donors and multiple myeloma patients and a large-scale quantitative mass spectrometry platform. A selected panel of up- and down-regulated proteins were validated by multiple-reaction-monitoring. Results: We identified a panel of 18 up- and down-regulated potential biomarkers of multiple myeloma, which can be further clinically validated for their potential use as disease-specific biomarkers or signature molecules for monitoring disease progression. Conclusion: The study demonstrates a good example of using proteomics as a tool for the development of clinical biomarkers for diagnosis, prognosis, and drug target discovery.


Journal of Hematology & Oncology | 2018

Growth factor independence 1 expression in myeloma cells enhances their growth, survival, and osteoclastogenesis

Daniela N. Petrusca; Denise Toscani; Feng-Ming Wang; Cheolkyu Park; Colin D. Crean; Judith Anderson; Silvia Marino; Khalid S. Mohammad; Dan Zhou; Rebecca Silbermann; Quanhong Sun; Noriyoshi Kurihara; Deborah L. Galson; Nicola Giuliani; G. David Roodman

BackgroundIn spite of major advances in treatment, multiple myeloma (MM) is currently an incurable malignancy due to the emergence of drug-resistant clones. We previously showed that MM cells upregulate the transcriptional repressor, growth factor independence 1 (Gfi1), in bone marrow stromal cells (BMSCs) that induces prolonged inhibition of osteoblast differentiation. However, the role of Gfi1 in MM cells is unknown.MethodsHuman primary CD138+ and BMSC were purified from normal donors and MM patients’ bone marrow aspirates. Gfi1 knockdown and overexpressing cells were generated by lentiviral-mediated shRNA. Proliferation/apoptosis studies were done by flow cytometry, and protein levels were determined by Western blot and/or immunohistochemistry. An experimental MM mouse model was generated to investigate the effects of MM cells overexpressing Gfi1 on tumor burden and osteolysis in vivo.ResultsWe found that Gfi1 expression is increased in patient’s MM cells and MM cell lines and was further increased by co-culture with BMSC, IL-6, and sphingosine-1-phosphate. Modulation of Gfi1 in MM cells had major effects on their survival and growth. Knockdown of Gfi1 induced apoptosis in p53-wt, p53-mutant, and p53-deficient MM cells, while Gfi1 overexpression enhanced MM cell growth and protected MM cells from bortezomib-induced cell death. Gfi1 enhanced cell survival of p53-wt MM cells by binding to p53, thereby blocking binding to the promoters of the pro-apoptotic BAX and NOXA genes. Further, Gfi1-p53 binding could be blocked by HDAC inhibitors. Importantly, inoculation of MM cells overexpressing Gfi1 in mice induced increased bone destruction, increased osteoclast number and size, and enhanced tumor growth.ConclusionsThese results support that Gfi1 plays a key role in MM tumor growth, survival, and bone destruction and contributes to bortezomib resistance, suggesting that Gfi1 may be a novel therapeutic target for MM.

Collaboration


Dive into the Colin D. Crean's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge