1
|
Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B, Teresa M, Bornmann W, Tsao T, Bergamo P, Mak DH, Chen W, McCubrey J, Tafuri A, Andreeff M. Retraction Note: MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia 2024; 38:2072. [PMID: 39025987 PMCID: PMC11420930 DOI: 10.1038/s41375-024-02339-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Affiliation(s)
- M Konopleva
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - M Milella
- Division of Medical Oncology A, Regina Elena National Cancer Institute, Rome, Italy
| | - P Ruvolo
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - J C Watts
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - M R Ricciardi
- Hematology, Department of Biotecnologie Cellulari ed Ematologia, Sapienza, University of Rome, Rome, Italy
| | - B Korchin
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - McQueen Teresa
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - William Bornmann
- Department of Experimental Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - T Tsao
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - P Bergamo
- Division of Medical Oncology A, Regina Elena National Cancer Institute, Rome, Italy
| | - D H Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - W Chen
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - J McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - A Tafuri
- Hematology, Department of Biotecnologie Cellulari ed Ematologia, Sapienza, University of Rome, Rome, Italy
| | - M Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Tagliari de Oliveira S, Binato R, Ellen Broto G, Tomie Takakura E, Navarro Gordan Ferreira Martins L, Abdelhay E, Panis C. Transcriptome of bone marrow-Derived stem cells reveals new inflammatory mediators related to increased survival in patients with multiple myeloma. Cytokine 2024; 179:156613. [PMID: 38643632 DOI: 10.1016/j.cyto.2024.156613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/23/2024]
Abstract
Although multiple myeloma (MM) is a neoplasm that leads affected individuals to death, little is known about why some patients survive much longer than others. In this context, we investigated the transcriptomic profile of bone marrow hematopoietic stem cells obtained from MM patients and compared the clinical outcomes of death and survival six months after bone marrow transplantation. The leukapheresis products of 39 patients with MM eligible for autologous transplantation were collected and analyzed. After extraction, the RNA was analyzed using the GeneChip Human Exon 1.0 Array method. The transcriptome profile was analyzed in silico, and the differentially expressed signaling pathways of interest were validated. The results showed a difference in the expression of inflammation-related genes, immune response processes, and the oxidative stress pathway. The in silico study also pointed out the involvement of the NFκB transcription factor in the possible modulation of these genes. We chose to validate molecules participating in these processes, including the cytokines TNF-α, IFN-γ, and TGF-β1; in addition, we measured the levels of oxidative stress mediators (pro-oxidant profile and the total antioxidant capacity). TNF-α levels were significantly reduced in patients who died and were over 50 years old at diagnosis, as well as in patients with plasmacytoma. Increased TNF-α was detected in patients with very high levels of β2-microglobulin. IFN-γ reduction was observed in patients with a complete response to treatment compared to those with a very good response. Patients with plasmacytoma who died also had an increased pro-oxidant profile. These data show the profile of inflammatory response markers that are altered in patients with MM who die quickly and serve as a basis for the development of future studies of markers to predict better survival in this disease.
Collapse
Affiliation(s)
- Stefania Tagliari de Oliveira
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil; Rede de Assistência a Saúde Metropolitana de Sarandi - Programa de Residência Médica em Clínica Médica - Sarandi - Paraná, Brazil
| | - Renata Binato
- Instituto Nacional de Câncer, Centro de Transplante de Medula Óssea, Rio de Janeiro, Brazil
| | - Geise Ellen Broto
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil
| | - Erika Tomie Takakura
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil
| | | | - Eliana Abdelhay
- Instituto Nacional de Câncer, Centro de Transplante de Medula Óssea, Rio de Janeiro, Brazil
| | - Carolina Panis
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Unioeste - Francisco Beltrão - Paraná, Brazil.
| |
Collapse
|
3
|
Mazzera L, Abeltino M, Lombardi G, Cantoni AM, Jottini S, Corradi A, Ricca M, Rossetti E, Armando F, Peli A, Ferrari A, Martinelli G, Scupoli MT, Visco C, Bonifacio M, Ripamonti A, Gambacorti-Passerini C, Bonati A, Perris R, Lunghi P. MEK1/2 regulate normal BCR and ABL1 tumor-suppressor functions to dictate ATO response in TKI-resistant Ph+ leukemia. Leukemia 2023; 37:1671-1685. [PMID: 37386079 PMCID: PMC10400427 DOI: 10.1038/s41375-023-01940-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 07/01/2023]
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) remains a clinical challenge in Ph-positive variants of chronic myeloid leukemia. We provide mechanistic insights into a previously undisclosed MEK1/2/BCR::ABL1/BCR/ABL1-driven signaling loop that may determine the efficacy of arsenic trioxide (ATO) in TKI-resistant leukemic patients. We find that activated MEK1/2 assemble into a pentameric complex with BCR::ABL1, BCR and ABL1 to induce phosphorylation of BCR and BCR::ABL1 at Tyr360 and Tyr177, and ABL1, at Thr735 and Tyr412 residues thus provoking loss of BCR's tumor-suppression functions, enhanced oncogenic activity of BCR::ABL1, cytoplasmic retention of ABL1 and consequently drug resistance. Coherently, pharmacological blockade of MEK1/2 induces dissociation of the pentameric MEK1/2/BCR::ABL1/BCR/ABL1 complex and causes a concurrent BCRY360/Y177, BCR::ABL1Y360/Y177 and cytoplasmic ABL1Y412/T735 dephosphorylation thereby provoking the rescue of the BCR's anti-oncogenic activities, nuclear accumulation of ABL1 with tumor-suppressive functions and consequently, growth inhibition of the leukemic cells and an ATO sensitization via BCR-MYC and ABL1-p73 signaling axes activation. Additionally, the allosteric activation of nuclear ABL1 was consistently found to enhance the anti-leukemic effects of the MEK1/2 inhibitor Mirdametinib, which when combined with ATO, significantly prolonged the survival of mice bearing BCR::ABL1-T315I-induced leukemia. These findings highlight the therapeutic potential of MEK1/2-inhibitors/ATO combination for the treatment of TKI-resistant leukemia.
Collapse
Affiliation(s)
- Laura Mazzera
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Manuela Abeltino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Guerino Lombardi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
| | | | - Stefano Jottini
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Attilio Corradi
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Micaela Ricca
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
| | - Elena Rossetti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- National Healthcare Service (SSN-Servizio Sanitario Nazionale) ASL Piacenza, Piacenza, Italy
| | - Federico Armando
- Department of Veterinary Science, University of Parma, Parma, Italy
- University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Angelo Peli
- Department for Life Quality Studies Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Anna Ferrari
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, FC, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, FC, Italy
- Institute of Hematology "L. e A. Seragnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carlo Visco
- Department of Engineering for Innovation Medicine, Section of Hematology-University of Verona, Verona, Italy
| | - Massimiliano Bonifacio
- Department of Engineering for Innovation Medicine, Section of Hematology-University of Verona, Verona, Italy
| | - Alessia Ripamonti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Adult Hematology, IRCCS San Gerardo, Monza, Italy
| | - Carlo Gambacorti-Passerini
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Adult Hematology, IRCCS San Gerardo, Monza, Italy
| | - Antonio Bonati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberto Perris
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Centre for Molecular and Translational Oncology-COMT, University of Parma, Parma, Italy
| | - Paolo Lunghi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
- Centre for Molecular and Translational Oncology-COMT, University of Parma, Parma, Italy.
| |
Collapse
|
4
|
Zhang QQ, Zhang SW, Feng YH, Shi JY. Few-Shot Drug Synergy Prediction With a Prior-Guided Hypernetwork Architecture. IEEE TRANSACTIONS ON PATTERN ANALYSIS AND MACHINE INTELLIGENCE 2023; 45:9709-9725. [PMID: 37027608 DOI: 10.1109/tpami.2023.3248041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Predicting drug synergy is critical to tailoring feasible drug combination treatment regimens for cancer patients. However, most of the existing computational methods only focus on data-rich cell lines, and hardly work on data-poor cell lines. To this end, here we proposed a novel few-shot drug synergy prediction method (called HyperSynergy) for data-poor cell lines by designing a prior-guided Hypernetwork architecture, in which the meta-generative network based on the task embedding of each cell line generates cell line dependent parameters for the drug synergy prediction network. In HyperSynergy model, we designed a deep Bayesian variational inference model to infer the prior distribution over the task embedding to quickly update the task embedding with a few labeled drug synergy samples, and presented a three-stage learning strategy to train HyperSynergy for quickly updating the prior distribution by a few labeled drug synergy samples of each data-poor cell line. Moreover, we proved theoretically that HyperSynergy aims to maximize the lower bound of log-likelihood of the marginal distribution over each data-poor cell line. The experimental results show that our HyperSynergy outperforms other state-of-the-art methods not only on data-poor cell lines with a few samples (e.g., 10, 5, 0), but also on data-rich cell lines.
Collapse
|
5
|
Raimondi V, Iannozzi NT, Burroughs-Garcìa J, Toscani D, Storti P, Giuliani N. A personalized molecular approach in multiple myeloma: the possible use of RAF/RAS/MEK/ERK and BCL-2 inhibitors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:463-479. [PMID: 36071980 PMCID: PMC9446161 DOI: 10.37349/etat.2022.00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is a blood cancer that derives from plasma cells (PCs), which will accumulate in the bone marrow (BM). Over time, several drugs have been developed to treat this disease that is still uncurable. The therapies used to treat the disease target immune activity, inhibit proteasome activity, and involve the use of monoclonal antibodies. However, MM is a highly heterogeneous disease, in fact, there are several mutations in signaling pathways that are particularly important for MM cell biology and that are possible therapeutic targets. Indeed, some studies suggest that MM is driven by mutations within the rat sarcoma virus (RAS) signaling cascade, which regulates cell survival and proliferation. The RAS/proto-oncogene, serine/threonine kinase (RAF)/mitogen-activated extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK signaling pathway is deregulated in several cancers, for which drugs have been developed to inhibit these pathways. In addition to the signaling pathways, the disease implements mechanisms to ensure the survival and consequently a high replicative capacity. This strategy consists in the deregulation of apoptosis. In particular, some cases of MM show overexpression of anti-apoptotic proteins belonging to the B cell lymphoma 2 (BCL-2) family that represent a possible druggable target. Venetoclax is an anti-BCL-2 molecule used in hematological malignancies that may be used in selected MM patients based on their molecular profile. We focused on the possible effects in MM of off-label drugs that are currently used for other cancers with the same molecular characteristics. Their use, combined with the current treatments, could be a good strategy against MM.
Collapse
Affiliation(s)
- Vincenzo Raimondi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | | | | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;Hematology, “Azienda Ospedaliero-Universitaria di Parma”, 43126 Parma, Italy
| |
Collapse
|
6
|
Wang H, Wang X, Xu L, Zhang J. TP53 inhibitor PFTα increases the sensitivity of arsenic trioxide in TP53 wild type tumor cells. FEBS Open Bio 2022; 12:616-626. [PMID: 35030298 PMCID: PMC8886521 DOI: 10.1002/2211-5463.13366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/15/2021] [Accepted: 01/11/2022] [Indexed: 11/08/2022] Open
Abstract
Arsenic trioxide (ATO) has been shown to be effective in treating acute promyelocytic leukemia. TP53 mutated/null tumor cells are more sensitive to ATO treatment compared to tumor cells carrying wild type TP53 gene copies. However, it is unclear whether TP53 inhibitors can increase the sensitivity of TP53 wild type tumor cells to ATO. Here, we show that breast, colon and lung cancer cell lines with mutated/null TP53 are more sensitive to ATO-induced cell growth inhibition than cells with wild type TP53. Moreover, inhibition of TP53 by a TP53 inhibitor, PFTα, increased the ATO sensitivity of TP53 wild type tumor cells, coincident with ATO-induced cell growth arrest and cell apoptosis. Furthermore, combined treatment with ATO and PFTα synergistically inhibited tumor growth in mouse xenografts in vivo. Through microarray transcriptional analysis, we found that ATO-regulated genes were associated with TP53 and cell cycle signaling pathways. Co-treatment with PFTα enhanced ATO induced dynamic transcriptional changes. Overall, our results provide evidences in using TP53 chemical inhibitors to enhance the ATO-mediated therapeutic response against TP53 wild type tumor cells.
Collapse
Affiliation(s)
- Haiwei Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Xinrui Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Liangpu Xu
- Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ji Zhang
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
7
|
Willobee BA, Gaidarski AA, Dosch AR, Castellanos JA, Dai X, Mehra S, Messaggio F, Srinivasan S, VanSaun MN, Nagathihalli NS, Merchant NB. Combined Blockade of MEK and CDK4/6 Pathways Induces Senescence to Improve Survival in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2021; 20:1246-1256. [PMID: 34001634 PMCID: PMC8260447 DOI: 10.1158/1535-7163.mct-19-1043] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/07/2020] [Accepted: 05/14/2021] [Indexed: 11/16/2022]
Abstract
Activating KRAS mutations, a defining feature of pancreatic ductal adenocarcinoma (PDAC), promote tumor growth in part through the activation of cyclin-dependent kinases (CDK) that induce cell-cycle progression. p16INK4a (p16), encoded by the gene CDKN2A, is a potent inhibitor of CDK4/6 and serves as a critical checkpoint of cell proliferation. Mutations in and subsequent loss of the p16 gene occur in PDAC at a rate higher than that reported in any other tumor type and results in Rb inactivation and unrestricted cellular growth. Therefore, strategies targeting downstream RAS pathway effectors combined with CDK4/6 inhibition (CDK4/6i) may have the potential to improve outcomes in this disease. Herein, we show that expression of p16 is markedly reduced in PDAC tumors compared with normal pancreatic or pre-neoplastic tissues. Combined MEK inhibition (MEKi) and CDK4/6i results in sustained downregulation of both ERK and Rb phosphorylation and a significant reduction in cell proliferation compared with monotherapy in human PDAC cells. MEKi with CDK4/6i reduces tumor cell proliferation by promoting senescence-mediated growth arrest, independent of apoptosis in vitro We show that combined MEKi and CDK4/6i treatment attenuates tumor growth in xenograft models of PDAC and improves overall survival over 200% compared with treatment with vehicle or individual agents alone in Ptf1acre/+ ;LSL-KRASG12D/+ ;Tgfbr2flox/flox (PKT) mice. Histologic analysis of PKT tumor lysates reveal a significant decrease in markers of cell proliferation and an increase in senescence-associated markers without any significant change in apoptosis. These results demonstrate that combined targeting of both MEK and CDK4/6 represents a novel therapeutic strategy to synergistically reduce tumor growth through induction of cellular senescence in PDAC.
Collapse
Affiliation(s)
- Brent A Willobee
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Alexander A Gaidarski
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Austin R Dosch
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Jason A Castellanos
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Xizi Dai
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Siddharth Mehra
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Fanuel Messaggio
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Supriya Srinivasan
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Michael N VanSaun
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Nagaraj S Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
8
|
Zhang L, Zhou Y, Kong J, Zhang L, Yuan M, Xian S, Wang Y, Cheng Y, Yang X. Effect of arsenic trioxide on cervical cancer and its mechanisms. Exp Ther Med 2020; 20:169. [PMID: 33101463 PMCID: PMC7579781 DOI: 10.3892/etm.2020.9299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is one of the most common types of gynecological tumor, and thus identifying complementary or substitute treatment methods to treat cervical cancer is important. The present study aimed to evaluate the effect of arsenic trioxide (ATO), a traditional Chinese medicine, on cervical cancer cells and its underlying mechanism. MTT, colony formation and Transwell assays were performed to investigate the effects of different concentrations of ATO on cell proliferation and invasion, respectively. Western blotting and reverse transcription-quantitative PCR were applied to measure hypoxia-inducible factor-1α expression (HIF-1α) expression following ATO treatment. Finally, the effects of HIF-1α knockdown on cervical cancer cell proliferation, apoptosis and invasion were evaluated. The results demonstrated that ATO could inhibit cell proliferation and invasion. Moreover, ATO could induce reactive oxygen species production in a time- and dose-dependent manner. ATO could also promote the apoptosis of cervical cancer cells via HIF-1α. Therefore, the present study may provide a theoretical basis for identifying effective molecular targets for the prevention and treatment of cervical cancer.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, P.R. China.,Department of Obstetrics and Gynecology, Wuhan Children's Hospital and Wuhan Maternal and Child Healthcare Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430060, P.R. China
| | - Yanan Zhou
- Technology Chemical Engineering of Huaiyin Institute, Huai'an, Jiangsu 223001, P.R. China
| | - Jing Kong
- Technology Chemical Engineering of Huaiyin Institute, Huai'an, Jiangsu 223001, P.R. China
| | - Li Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shu Xian
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, P.R. China
| |
Collapse
|
9
|
p73: From the p53 shadow to a major pharmacological target in anticancer therapy. Pharmacol Res 2020; 162:105245. [PMID: 33069756 DOI: 10.1016/j.phrs.2020.105245] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
p73, along with p53 and p63, belongs to the p53 family of transcription factors. Besides the p53-like tumor suppressive activities, p73 has unique roles, namely in neuronal development and differentiation. In addition, the TP73 gene is rarely mutated in tumors. This makes p73 a highly appealing therapeutic target, particularly towards cancers with a null or disrupted p53 pathway. Distinct isoforms are transcribed from the TP73 locus either with (TAp73) and without (ΔNp73) the N-terminal transactivation domain. Conversely to TA tumor suppressors, ΔN proteins exhibit oncogenic properties by inhibiting p53 and TA protein functions. As such, p73 isoforms compose a puzzled and challenging regulatory pathway. This state-of-the-art review affords an update overview on p73 structure, biological functions and pharmacological regulation. Importantly, it addresses the relevance of p73 isoforms in carcinogenesis, highlighting their potential as drug targets in anticancer therapy. A critical discussion of major pharmacological approaches to promote p73 tumor suppressive activities, with relevant survival outcomes for cancer patients, is also provided.
Collapse
|
10
|
Mazzera L, Abeltino M, Lombardi G, Cantoni AM, Ria R, Ricca M, Saltarella I, Naponelli V, Rizzi FMA, Perris R, Corradi A, Vacca A, Bonati A, Lunghi P. Functional interplay between NF-κB-inducing kinase and c-Abl kinases limits response to Aurora inhibitors in multiple myeloma. Haematologica 2019; 104:2465-2481. [PMID: 30948493 PMCID: PMC6959191 DOI: 10.3324/haematol.2018.208280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/03/2019] [Indexed: 12/19/2022] Open
Abstract
Considering that Aurora kinase inhibitors are currently under clinical investigation in hematologic cancers, the identification of molecular events that limit the response to such agents is essential for enhancing clinical outcomes. Here, we discover a NF-κB-inducing kinase (NIK)-c-Abl-STAT3 signaling-centered feedback loop that restrains the efficacy of Aurora inhibitors in multiple myeloma. Mechanistically, we demonstrate that Aurora inhibition promotes NIK protein stabilization via downregulation of its negative regulator TRAF2. Accumulated NIK converts c-Abl tyrosine kinase from a nuclear proapoptotic into a cytoplasmic antiapoptotic effector by inducing its phosphorylation at Thr735, Tyr245 and Tyr412 residues, and, by entering into a trimeric complex formation with c-Abl and STAT3, increases both the transcriptional activity of STAT3 and expression of the antiapoptotic STAT3 target genes PIM1 and PIM2. This consequently promotes cell survival and limits the response to Aurora inhibition. The functional disruption of any of the components of the trimer NIK-c-Abl-STAT3 or the PIM survival kinases consistently enhances the responsiveness of myeloma cells to Aurora inhibitors. Importantly, concurrent inhibition of NIK or c-Abl disrupts Aurora inhibitor-induced feedback activation of STAT3 and sensitizes myeloma cells to Aurora inhibitors, implicating a combined inhibition of Aurora and NIK or c-Abl kinases as potential therapies for multiple myeloma. Accordingly, pharmacological inhibition of c-Abl together with Aurora resulted in substantial cell death and tumor regression in vivo The findings reveal an important functional interaction between NIK, Abl and Aurora kinases, and identify the NIK, c-Abl and PIM survival kinases as potential pharmacological targets for improving the efficacy of Aurora inhibitors in myeloma.
Collapse
Affiliation(s)
- Laura Mazzera
- Department of Medicine and Surgery, University of Parma, Parma
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini," Brescia
| | | | - Guerino Lombardi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini," Brescia
| | | | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari "Aldo Moro" Medical School, Bari
| | - Micaela Ricca
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini," Brescia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari "Aldo Moro" Medical School, Bari
| | | | - Federica Maria Angela Rizzi
- Department of Medicine and Surgery, University of Parma, Parma
- Center for Molecular and Translational Oncology, University of Parma, Parma
| | - Roberto Perris
- Center for Molecular and Translational Oncology, University of Parma, Parma
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Attilio Corradi
- Department of Veterinary Science, University of Parma, Parma
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari "Aldo Moro" Medical School, Bari
| | - Antonio Bonati
- Department of Medicine and Surgery, University of Parma, Parma
- Center for Molecular and Translational Oncology, University of Parma, Parma
| | - Paolo Lunghi
- Center for Molecular and Translational Oncology, University of Parma, Parma
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| |
Collapse
|
11
|
Kim JH, Nam GS, Kim SH, Ryu DS, Lee DS. Orostachys japonicus exerts antipancreatic cancer activity through induction of apoptosis and cell cycle arrest in PANC-1 cells. Food Sci Nutr 2019; 7:3549-3559. [PMID: 31763005 PMCID: PMC6848830 DOI: 10.1002/fsn3.1207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/30/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted therapy at the molecular level is important for pancreatic cancer treatment. This study looked over the anticancer activity of Orostachys japonicus in a human pancreatic cancer cell line, PANC-1. An ethyl acetate fraction containing quercetin, kaempferol, and flavonol glycosides from O. japonicus (OJE) exhibited significant anticancer activity against the PANC-1. OJE activated caspase-3, caspase-8, and caspase-9, leading to the induction of both intrinsic and extrinsic apoptosis pathways. It also inhibited cyclin D1, cyclin B1, and cyclin-dependent kinase 4, representing cell cycle arrest at both G1/S and G2/M phases. In addition, OJE phosphorylated MAPKs such as p38, JNK, and ERK, which are important upstream signaling factors in apoptosis and arrest of cell cycle inducing system. In conclusion, OJE effectively exerted antipancreatic cancer activity via induction of apoptosis directed by both intrinsic and extrinsic pathways and arrest of cell cycle regulated at both G1/S and G2/M stages, which is activated by MAPKs, p38, JNK, and ERK.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| | - Gi Suk Nam
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| | - Sung Hyun Kim
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| | - Deok Seon Ryu
- Department of Biomedical Laboratory ScienceSoonchunhyang UniversityAsanKorea
| | - Dong Seok Lee
- Department of Smart Foods and DrugsGraduate School of Inje UniversityGimhaeKorea
| |
Collapse
|
12
|
Li KN, Zhang YY, Yu YN, Wu HL, Wang Z. Met-Controlled Allosteric Module of Neural Generation as A New Therapeutic Target in Rodent Brain Ischemia. Chin J Integr Med 2019; 27:896-904. [PMID: 31418133 DOI: 10.1007/s11655-019-3182-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate a Met-controlled allosteric module (AM) of neural generation as a potential therapeutic target for brain ischemia. METHODS We selected Markov clustering algorithm (MCL) to mine functional modules in the related target networks. According to the topological similarity, one functional module was predicted in the modules of baicalin (BA), jasminoidin (JA), cholic acid (CA), compared with I/R model modules. This functional module included three genes: Inppl1, Met and Dapk3 (IMD). By gene ontology enrichment analysis, biological process related to this functional module was obtained. This functional module participated in generation of neurons. Western blotting was applied to present the compound-dependent regulation of IMD. Co-immunoprecipitation was used to reveal the relationship among the three members. We used IF to determine the number of newborn neurons between compound treatment group and ischemia/reperfusion group. The expressions of vascular endothelial growth factor (VEGF) and matrix metalloproteinase 9 (MMP-9) were supposed to show the changing circumstances for neural generation under cerebral ischemia. RESULTS Significant reduction in infarction volume and pathological changes were shown in the compound treatment groups compared with the I/R model group (P<0.05). Three nodes in one novel module of IMD were found to exert diverse compound-dependent ischemic-specific excitatory regulatory activities. An anti-ischemic excitatory allosteric module (AME) of generation of neurons (AME-GN) was validated successfully in vivo. Newborn neurons increased in BJC treatment group (P<0.05). The expression of VEGF and MMP-9 decreased in the compound treatment groups compared with the I/R model group (P<0.05). CONCLUSIONS AME demonstrates effectiveness of our pioneering approach to the discovery of therapeutic target. The novel approach for AM discovery in an effort to identify therapeutic targets holds the promise of accelerating elucidation of underlying pharmacological mechanisms in cerebral ischemia.
Collapse
Affiliation(s)
- Kang-Ning Li
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Ying-Ying Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ya-Nan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hong-Li Wu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
13
|
Wei XF, Chen QL, Fu Y, Zhang QK. Wnt and BMP signaling pathways co-operatively induce the differentiation of multiple myeloma mesenchymal stem cells into osteoblasts by upregulating EMX2. J Cell Biochem 2018; 120:6515-6527. [PMID: 30450775 DOI: 10.1002/jcb.27942] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022]
Abstract
Osteoblast differentiation, defined as the process whereby a relatively unspecialized cell acquires the specialized features of an osteoblast, is directly linked to multiple myeloma (MM) bone disease. Wnt and bone morphogenetic protein (BMP) are proved to be implicated in the pathological or defective osteoblast differentiation process. This study aims to test the involvement of Wnt, bone morphogenetic proteins (BMP) pathways, and empty spiracles homeobox 2 (EMX2) in osteoblast differentiation and MM development. Initially, differentially expressed genes in bone marrow mesenchymal stem cells (MSCs) from MM patients and healthy donors were identified using microarray-based gene expression profiling. The functional role of Wnt and BMP in MM was determined. Next, we focused on the co-operative effects of Wnt and BMP on calcium deposition, alkaline phosphatase (ALP) activity, the number of mineralized nodules, and osteocalcin (OCN) content in MSCs. The expression patterns of Wnt and BMP pathway-related genes, EMX2 and osteoblast differentiation-related factors were determined to assess their effects on osteoblast differentiation. Furthermore, regulation of Wnt and BMP in ectopic osteogenesis was also investigated in vivo. An integrated genomic screen suggested that Wnt and BMP regularly co-operate to regulate EMX2 and affect MM. EMX2 was downregulated in MSCs. The activated Wnt and BMP resulted in more calcium salt deposits, mineralized nodules, and a noted increased in ALP activity and OCN content by upregulating EMX2, leading to induced differentiation of MSCs into osteoblasts. Collectively, this study demonstrated that Wnt and BMP pathways could co-operatively stimulate differentiation of MSCs into osteoblasts and inhibit MM progression, representing potential targets for MM treatment.
Collapse
Affiliation(s)
- Xiao-Fang Wei
- Department of Hematology, Gansu Provincial Hospital, Lanzhou, China
| | - Qiao-Lin Chen
- Department of Hematology, Gansu Provincial Hospital, Lanzhou, China
| | - Yuan Fu
- Department of Hematology, Gansu Provincial Hospital, Lanzhou, China
| | - Qi-Ke Zhang
- Department of Hematology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
14
|
Abramson HN. Kinase inhibitors as potential agents in the treatment of multiple myeloma. Oncotarget 2018; 7:81926-81968. [PMID: 27655636 PMCID: PMC5348443 DOI: 10.18632/oncotarget.10745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Recent years have witnessed a dramatic increase in the number of therapeutic options available for the treatment of multiple myeloma (MM) - from immunomodulating agents to proteasome inhibitors to histone deacetylase (HDAC) inhibitors and, most recently, monoclonal antibodies. Used in conjunction with autologous hematopoietic stem cell transplantation, these modalities have nearly doubled the disease's five-year survival rate over the last three decades to about 50%. In spite of these advances, MM still is considered incurable as resistance and relapse are common. While small molecule protein kinase inhibitors have made inroads in the therapy of a number of cancers, to date their application to MM has been less than successful. Focusing on MM, this review examines the roles played by a number of kinases in driving the malignant state and the rationale for target development in the design of a number of kinase inhibitors that have demonstrated anti-myeloma activity in both in vitro and in vivo xenograph models, as well as those that have entered clinical trials. Among the targets and their inhibitors examined are receptor and non-receptor tyrosine kinases, cell cycle control kinases, the PI3K/AKT/mTOR pathway kinases, protein kinase C, mitogen-activated protein kinase, glycogen synthase kinase, casein kinase, integrin-linked kinase, sphingosine kinase, and kinases involved in the unfolded protein response.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
15
|
Cholujova D, Bujnakova Z, Dutkova E, Hideshima T, Groen RW, Mitsiades CS, Richardson PG, Dorfman DM, Balaz P, Anderson KC, Jakubikova J. Realgar nanoparticles versus ATO arsenic compounds induce in vitro and in vivo activity against multiple myeloma. Br J Haematol 2017; 179:756-771. [PMID: 29048129 DOI: 10.1111/bjh.14974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/01/2017] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM), a B cell malignancy characterized by clonal proliferation of plasma cells in the bone marrow, remains incurable despite the use of novel and conventional therapies. In this study, we demonstrated MM cell cytotoxicity triggered by realgar (REA; As4 S4 ) nanoparticles (NREA) versus Arsenic trioxide (ATO) against MM cell lines and patient cells. Both NREA and ATO showed in vivo anti-MM activity, resulting in significantly decreased tumour burden. The anti-MM activity of NREA and ATO is associated with apoptosis, evidenced by DNA fragmentation, depletion of mitochondrial membrane potential, cleavage of caspases and anti-apoptotic proteins. NREA induced G2 /M cell cycle arrest and modulation of cyclin B1, p53 (TP53), p21 (CDKN1A), Puma (BBC3) and Wee-1 (WEE1). Moreover, NREA induced modulation of key regulatory molecules in MM pathogenesis including JNK activation, c-Myc (MYC), BRD4, and histones. Importantly, NREA, but not ATO, significantly depleted the proportion and clonogenicity of the MM stem-like side population, even in the context of the bone marrow stromal cells. Finally, our study showed that both NREA and ATO triggered synergistic anti-MM activity when combined with lenalidomide or melphalan. Taken together, the anti-MM activity of NREA was more potent compared to ATO, providing the preclinical framework for clinical trials to improve patient outcome in MM.
Collapse
Affiliation(s)
- Danka Cholujova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovakia
| | | | | | - Teru Hideshima
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Richard W Groen
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Constantine S Mitsiades
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David M Dorfman
- Department of Medicine, Harvard Medical School, Boston, MA, USA.,Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter Balaz
- Institute of Geotechnics SAS, Košice, Slovakia
| | - Kenneth C Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jana Jakubikova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovakia.,Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Shen Y, Sun Y, Zhang L, Liu H. Effects of DTX3L on the cell proliferation, adhesion, and drug resistance of multiple myeloma cells. Tumour Biol 2017; 39:1010428317703941. [PMID: 28653881 DOI: 10.1177/1010428317703941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cell adhesion-mediated drug resistance is an important factor that influences the effects of chemotherapy in multiple myeloma. DTX3L, a ubiquitin ligase, plays a key role in cell-cycle-related process. Here, we found that the expression of DTX3L gradually increased during the proliferation of myeloma cells, which resulted in arrest of the cell cycle in the G1 phase and promoted the adherence of myeloma cells to fibronectin or bone marrow stromal cells. In addition, silencing of DTX3L improved sensitivity to chemotherapy drugs in multiple myeloma cell lines adherent to bone marrow stromal cells and increased the expression of caspase-3 and poly-adenosine diphosphate-ribose polymerase, two markers of apoptosis. Finally, we also found that DTX3L expression was regulated by focal adhesion kinase. Taken together, the results of this study show that DTX3L plays an important role in the proliferation and cell adhesion-mediated drug resistance of multiple myeloma cells and as such may play a key role in the development of multiple myeloma.
Collapse
Affiliation(s)
- Yaodong Shen
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Yuxiang Sun
- 2 Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| | - Linlin Zhang
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong, People's Republic of China.,2 Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| | - Hong Liu
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| |
Collapse
|
17
|
Bone marrow stroma-induced resistance of chronic lymphocytic leukemia cells to arsenic trioxide involves Mcl-1 upregulation and is overcome by inhibiting the PI3Kδ or PKCβ signaling pathways. Oncotarget 2016; 6:44832-48. [PMID: 26540567 PMCID: PMC4792595 DOI: 10.18632/oncotarget.6265] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/22/2015] [Indexed: 11/25/2022] Open
Abstract
CLL remains an incurable disease in spite of the many new compounds being studied. Arsenic trioxide (ATO) induces apoptosis in all CLL cell types and could constitute an efficient therapy. To further explore this, we have studied the influence of stromal cells, key components of the CLL microenvironment, on the response of CLL cells to ATO. Bone marrow stromal cells induced CLL cell resistance to 2 μM ATO and led to activation of Lyn, ERK, PI3K and PKC, as well as NF-κB and STAT3. Mcl-1, Bcl-xL, and Bfl-1 were also upregulated after the co-culture. Inhibition experiments indicated that PI3K and PKC were involved in the resistance to ATO induced by stroma. Moreover, idelalisib and sotrastaurin, specific inhibitors for PI3Kδ and PKCβ, respectively, inhibited Akt phosphorylation, NF-κB/STAT3 activation and Mcl-1 upregulation, and rendered cells sensitive to ATO. Mcl-1 was central to the mechanism of resistance to ATO, since: 1) Mcl-1 levels correlated with the CLL cell response to ATO, and 2) blocking Mcl-1 expression or function with specific siRNAs or inhibitors overcame the protecting effect of stroma. We have therefore identified the mechanism involved in the CLL cell resistance to ATO induced by bone marrow stroma and show that idelalisib or sotrastaurin block this mechanism and restore sensibility to ATO. Combination of ATO with these inhibitors may thus constitute an efficient treatment for CLL.
Collapse
|
18
|
Lionetti M, Barbieri M, Todoerti K, Agnelli L, Marzorati S, Fabris S, Ciceri G, Galletti S, Milesi G, Manzoni M, Mazzoni M, Greco A, Tonon G, Musto P, Baldini L, Neri A. Molecular spectrum of BRAF, NRAS and KRAS gene mutations in plasma cell dyscrasias: implication for MEK-ERK pathway activation. Oncotarget 2016; 6:24205-17. [PMID: 26090869 PMCID: PMC4695180 DOI: 10.18632/oncotarget.4434] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/31/2015] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma (MM) is a clinically and genetically heterogeneous plasma cell (PC) malignancy. Whole-exome sequencing has identified therapeutically targetable mutations such as those in the mitogen-activated protein kinase (MAPK) pathway, which are the most prevalent MM mutations. We used deep sequencing to screen 167 representative patients with PC dyscrasias [132 with MM, 24 with primary PC leukemia (pPCL) and 11 with secondary PC leukemia (sPCL)] for mutations in BRAF, NRAS and KRAS, which were respectively found in 12%, 23.9% and 29.3% of cases. Overall, the MAPK pathway was affected in 57.5% of the patients (63.6% of those with sPCL, 59.8% of those with MM, and 41.7% of those with pPCL). The majority of BRAF variants were comparably expressed at transcript level. Additionally, gene expression profiling indicated the MAPK pathway is activated in mutated patients. Finally, we found that vemurafenib inhibition of BRAF activation in mutated U266 cells affected the expression of genes known to be associated with MM. Our data confirm and extend previous published evidence that MAPK pathway activation is recurrent in myeloma; the finding that it is mediated by BRAF mutations in a significant fraction of patients has potentially immediate clinical implications.
Collapse
Affiliation(s)
- Marta Lionetti
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Marzia Barbieri
- Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Katia Todoerti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Potenza, Italy
| | - Luca Agnelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Simona Marzorati
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Sonia Fabris
- Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Ciceri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Serena Galletti
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giulia Milesi
- Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Martina Manzoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Mara Mazzoni
- Molecular Mechanism Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Angela Greco
- Molecular Mechanism Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Pellegrino Musto
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Potenza, Italy
| | - Luca Baldini
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonino Neri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
19
|
Liu H, Ding L, Shen Y, Zhong F, Wang Q, Xu X. RBQ3 participates in multiple myeloma cell proliferation, adhesion and chemoresistance. Int J Biol Macromol 2016; 91:115-22. [PMID: 27189701 DOI: 10.1016/j.ijbiomac.2016.05.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Cell adhesion mediated drug resistance (CAM-DR) is a major factor that impedes the effect of chemotherapy in multiple myeloma (MM). RBQ3, which is a RB-binding protein, played a crucial role in cell cycle process. Here, we reported that RBQ3 expression was increased gradually during the proliferation process of myeloma cells. Knocking down of RBQ3 resulted in cell cycle arrest in G1 phase and increased myeloma cells adherent to fibronectin or bone marrow stromal cells (BMSCs). Furthermore, silencing of RBQ3 reduced sensitivity to chemotherapeutic drugs in myeloma cell lines adherent to BMSCs and reduced two apoptotic marker proteins cleaved caspase-3 and cleaved PARP expression. Besides, we also found that RBQ3 participated in MAPK/ERK signal transduction pathway. In summary, these results may shed new insights into the role of RBQ3 in the development of multiple myeloma.
Collapse
Affiliation(s)
- Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Linlin Ding
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Yaodong Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Fei Zhong
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Qiru Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong 226361, Jiangsu Province, People's Republic of China.
| |
Collapse
|
20
|
Yu G, Chen X, Chen S, Ye W, Hou K, Liang M. Arsenic trioxide reduces chemo-resistance to 5-fluorouracil and cisplatin in HBx-HepG2 cells via complex mechanisms. Cancer Cell Int 2015; 15:116. [PMID: 26692822 PMCID: PMC4676851 DOI: 10.1186/s12935-015-0269-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023] Open
Abstract
Background Multidrug resistance is one of the major reasons chemotherapy-based treatments failed in hepatitis B virus (HBV) related hepatocellular carcinoma (HCC). Hypoxia is generally associated with tumor chemo-resistance. The aim of the study was to investigate the effect of Arsenic trioxide (As2O3) on the hypoxia-induced chemo-resistance to 5-FU or cisplatin and explored its underlying mechanism in the HBx-HepG2 cells. Methods MTT assay was used to examine the cell viability. Mitochondrial membrane potential (MMP) and cell cycle was examined by flow cytometry. qRT-PCR was employed to observe the mRNA expression level; and western blot assay was used to determine the protein expression level. Results Our results showed that transfection of HBx plasmid established the HBx-HepG2 cells expressing HBx, and the expression of HBx was confirmed by qRT-PCR and western blot. Exposure of HBx-HepG2 cells to hypoxia (5 % O2, 3 % O2, 1 % O2) for 48 h increased the chemo-resistance to 5-fluorouracil (5-FU) (50–1600 µM) and cisplatin (25–800 µM), reduced MMP, and caused the cell cycle arrest at G0/G1 phase in a concentration-dependent manner. Hypoxia also concentration-dependently (5 % O2, 3 % O2, 1 % O2) reduced mRNA expression level of P-glycoprotein (P-gp), multidrug resistance protein (MRP1), lung resistance protein (LRP), and decreased the protein expression level of hypoxia-inducible factor-1α (HIF-1α), P-gp MRP1, and LRP. Following pretreatment with As2O3 at a non-cytotoxic concentration re-sensitized the hypoxia (1 % O2)-induced chemo-resistance to 5-FU and cisplatin in HBx-HepG2 cells. As2O3 pretreatment also prevented MMP reduction and G0/G1 arrest induced by hypoxia. Meanwhile, As2O3 antagonized increase of HIF-1α protein induced by hypoxia, and it also suppresses the increase in expression levels of P-gp, MRP1, and LRP mRNA and proteins. In addition, As2O3 in combination with 5-FU treatment caused up-regulation of DR5, caspase 3, caspase 8, and caspase 9, and down-regulation of BCL-2, but had no effect of DR4. Conclusions Our results may suggest that As2O3 re-sensitizes hypoxia-induced chemo-resistance in HBx-HepG2 via complex pathways, and As2O3 may be a potential agent that given in combination with other anti-drugs for the treatment of HBV related HCC, which is resistant to chemotherapy.
Collapse
Affiliation(s)
- Guifang Yu
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, No. 621, Gangwan Road, Huangpu District, Guangzhou, 510700 China
| | - Xuezhu Chen
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, No. 621, Gangwan Road, Huangpu District, Guangzhou, 510700 China
| | - Shudi Chen
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, No. 621, Gangwan Road, Huangpu District, Guangzhou, 510700 China
| | - Weipeng Ye
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, No. 621, Gangwan Road, Huangpu District, Guangzhou, 510700 China
| | - Kailian Hou
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, No. 621, Gangwan Road, Huangpu District, Guangzhou, 510700 China
| | - Min Liang
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, No. 621, Gangwan Road, Huangpu District, Guangzhou, 510700 China
| |
Collapse
|
21
|
He Y, Wang Y, Liu H, Xu X, He S, Tang J, Huang Y, Miao X, Wu Y, Wang Q, Cheng C. Pyruvate kinase isoform M2 (PKM2) participates in multiple myeloma cell proliferation, adhesion and chemoresistance. Leuk Res 2015; 39:1428-36. [PMID: 26453405 DOI: 10.1016/j.leukres.2015.09.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 09/19/2015] [Accepted: 09/24/2015] [Indexed: 11/26/2022]
Abstract
Cell adhesion mediated drug resistance (CAM-DR) remains the major barrier in human multiple myeloma (MM) therapy. In the present study, we aimed at investigating the role of pyruvate kinase isoform M2 (PKM2) in MM CAM-DR. We determined that PKM2 expression was positively correlated with cell proliferation and knockdown of PKM2 contributed to the increased cell adhesion rate in MM. The enhancement in the adhesion of MM cells to fibronectin or the bone marrow stroma cell line HS-5 cells translated to an increased CAM-DR phenotype. Importantly, we showed that this CAM-DR phenotype was correlated with the phosphorylation of Akt and ERK in MM cells. Taken together, our data shed new light on the molecular mechanism of CAM-DR in MM, and targeting PKM2 may be a novel therapeutic approach for improving the effectiveness of chemotherapy in MM.
Collapse
Affiliation(s)
- Yunhua He
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Yuchan Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong 22600, Jiangsu Province, People's Republic of China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Song He
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jie Tang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Yuejiao Huang
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaobing Miao
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yaxun Wu
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qiru Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Chun Cheng
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
22
|
Li X, Sun WJ. The clinical activity of arsenic trioxide, ascorbic acid, ifosfamide and prednisone combination therapy in patients with relapsed and refractory multiple myeloma. Onco Targets Ther 2015; 8:775-81. [PMID: 25914547 PMCID: PMC4399549 DOI: 10.2147/ott.s81022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
This study aimed to investigate the activity of arsenic trioxide (As2O3) combined with ascorbic acid, ifosfamide, and prednisone chemotherapy in patients with repeatedly relapsed and refractory multiple myeloma (MM). Here, we retrospectively analyzed medical data of 30 MM patients showing progressive disease after receiving at least two previous lines of treatment including an immunomodulatory agent (thalidomide or lenalidomide) and a proteasome inhibitor. There were 19 men and eleven women, aged 54-73 (median 65) years, in this study. The distribution of different isotypes included immunoglobulin G(IgG) (12 patients), IgA (six patients), IgD (three), and light chain (nine patients). All the patients were Durie-Salmon stage III and had relapsed at least three times; the median cycles of prior therapies was 15 (range 10-18). The patients were treated with As2O3, ascorbic acid, and CP (ifosfamide 1 g on day 1, day 3, day 5, and day 7; prednisone 30 mg taken orally for 2 weeks). As2O3 was administered as an intravenous infusion at a dose of 10 mg/d and ascorbic acid at a dose of 2 g/d for 14 days of each 4-week cycle. The results showed that after 2 cycles of therapy, there were five patients that attained partial response, 15 had minimal response, five had no change, and five had progressive disease. The overall response rate was 66.7% (20/30 cases), 50% (10/20 cases), and 40% (2/5 cases), respectively, after 2, 4, and 6 cycles of the therapy. But there were no patients that attained complete remission. The median time of overall survival and progression-free survival were 48 (29-120) and 6 (2-8) months, respectively. The most common treatment-related adverse events included neutropenia, fatigue, anemia, thrombocytopenia, and infection that could be tolerated. The results showed that As2O3 combined with ascorbic acid, ifosfamide, and prednisone chemotherapy may be a choice treatment for repeatedly relapsed and refractory MM patients.
Collapse
Affiliation(s)
- Xin Li
- Department of Hematology and Oncology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wan-Jun Sun
- Department of Hematology, Second Artillery General Hospital, Beijing, People's Republic of China
| |
Collapse
|
23
|
Hong E, Lee E, Kim J, Kwon D, Lim Y. Elevated pressure enhanced TRAIL-induced apoptosis in hepatocellular carcinoma cells via ERK1/2-inactivation. ACTA ACUST UNITED AC 2015; 20:535-48. [DOI: 10.1515/cmble-2015-0030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 06/17/2015] [Indexed: 12/25/2022]
Abstract
AbstractThe high frequency of intrinsic resistance to TNF-related apoptosisinducing ligand (TRAIL) in tumor cell lines has necessitated the development of strategies to sensitize tumors to TRAIL-induced apoptosis. We previously showed that elevated pressure applied as a mechanical stressor enhanced TRAIL-mediated apoptosis in human lung carcinoma cells in vitro and in vivo. This study focused on the effect of elevated pressure on the sensitization of TRAIL-resistant cells and the underlying mechanism. We observed elevated pressure-induced sensitization to TRAIL-mediated apoptosis in Hep3B cells, accompanied by the activation of several caspases and the mitochondrial signaling pathway. Interestingly, the enhanced apoptosis induced by elevated pressure was correlated with suppression of extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) phosphorylation and CREB without any change to other MAPKs. Phosphorylation of Bcl-2-associated death promoter (BAD) also decreased, leading to inhibition of the mitochondrial pathway. To confirm whether the activation of pERK1/2 plays a key role in the TRAIL-sensitizing effect of elevated pressure, Hep3B cells were pre-treated with the ERK1/2-specific inhibitor PD98059 instead of elevated pressure. Co-treatment with PD98059 and TRAIL augmented TRAIL-induced apoptosis and decreased BAD phosphorylation. The inhibition of ERK1/2 activation by elevated pressure and PD98059 also reduced BH3 interacting-domain death agonist (BID), thereby amplifying apoptotic stress at the mitochondrial level. Our results suggest that elevated pressure enhances TRAIL-induced apoptosis of Hep3B cells via specific suppression of ERK1/2 activation among MAPKs.
Collapse
|
24
|
Ravi D, Bhalla S, Gartenhaus RB, Crombie J, Kandela I, Sharma J, Mazar A, Evens AM. The novel organic arsenical darinaparsin induces MAPK-mediated and SHP1-dependent cell death in T-cell lymphoma and Hodgkin lymphoma cells and human xenograft models. Clin Cancer Res 2014; 20:6023-33. [PMID: 25316819 DOI: 10.1158/1078-0432.ccr-14-1532] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Darinaparsin (Zio-101) is a novel organic arsenical compound with encouraging clinical activity in relapsed/refractory T-cell lymphoma (TCL) and Hodgkin lymphoma (HL); however, little is known about its mechanism of action. EXPERIMENTAL DESIGN TCL cell lines (Jurkat, Hut78, and HH) and HL cell lines (L428, L540, and L1236) were examined for in vitro cell death by MTT assay and Annexin V-based flow cytometry. Jurkat and L540-derived xenografts in SCID mice were examined for in vivo tumor inhibition and survival. Biologic effects of darinaparsin on the MAPK pathway were investigated using pharmacologic inhibitors, RNAi and transient transfection for overexpression for SHP1 and MEK. RESULTS Darinaparsin treatment resulted in time- and dose-dependent cytotoxicity and apoptosis in all TCL and HL cell lines. In addition, darinaparsin had more rapid, higher, and sustained intracellular arsenic levels compared with arsenic trioxide via mass spectrometry. In vivo experiments with Jurkat (TCL) and L540 (HL)-derived lymphoma xenografts showed significant inhibition of tumor growth and improved survival in darinaparsin-treated SCID mice. Biologically, darinaparsin caused phosphorylation of ERK (and relevant downstream substrates) primarily by decreasing the inhibitory SHP1 phosphatase and coimmunoprecipitation showed significant ERK/SHP1 interaction. Furthermore, ERK shRNA knockdown or constitutive overexpression of SHP1 resulted in increased apoptosis, whereas cotreatment with pharmacologic MEK inhibitors resulted in synergistic cell death. Conversely, SHP1 blockade (via pharmacologic inhibition or RNAi) and MEK constitutive activation decreased darinaparsin-related cell death. CONCLUSIONS Altogether, these data show that darinaparsin is highly active in HL and TCL and its activity is dependent primarily on MAPK mechanisms.
Collapse
Affiliation(s)
- Dashnamoorthy Ravi
- Molecular Oncology Research Institute and Division of Hematology Oncology, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | - Savita Bhalla
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ronald B Gartenhaus
- Marlene and Stewart Greenebaum Cancer Center, Department of Medicine, University of Maryland, Baltimore, Maryland
| | | | - Irawati Kandela
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Jaya Sharma
- Molecular Oncology Research Institute and Division of Hematology Oncology, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | - Andrew Mazar
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois. Robert H. Lurie Cancer Center, Northwestern University, Chicago, Illinois
| | - Andrew M Evens
- Molecular Oncology Research Institute and Division of Hematology Oncology, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
25
|
Plasmacytomagenesis in Eμ-v-abl transgenic mice is accelerated when apoptosis is restrained. Blood 2014; 124:1099-109. [PMID: 24986687 DOI: 10.1182/blood-2014-04-570770] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mice susceptible to plasma cell tumors provide a useful model for human multiple myeloma. We previously showed that mice expressing an Eµ-v-abl oncogene solely develop plasmacytomas. Here we show that loss of the proapoptotic BH3-only protein Bim or, to a lesser extent, overexpression of antiapoptotic Bcl-2 or Mcl-1, significantly accelerated the development of plasmacytomas and increased their incidence. Disease was preceded by an increased abundance of plasma cells, presumably reflecting their enhanced survival capacity in vivo. Plasmacytomas of each genotype expressed high levels of v-abl and frequently harbored a rearranged c-myc gene, probably as a result of chromosome translocation. As in human multiple myelomas, elevated expression of cyclin D genes was common, and p53 deregulation was rare. Our results for plasmacytomas highlight the significance of antiapoptotic changes in multiple myeloma, which include elevated expression of Mcl-1 and, less frequently, Bcl-2, and suggest that closer attention to defects in Bim expression is warranted.
Collapse
|
26
|
MicroRNA-21 and multiple myeloma: small molecule and big function. Med Oncol 2014; 31:94. [PMID: 24981236 DOI: 10.1007/s12032-014-0094-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 06/21/2014] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is a monoclonal malignant plasma cell disorder with an apparent homogeneity as opposed to leukemia and lymphomas. The recent introduction of thalidomide, lenalidomide and bortezomib has prolonged survival of patients with MM, and drug resistance or relapse of disease is perhaps still the major concern. Deregulation of hundreds of genes and multiple signaling pathways leads to MM pathogenesis and disease progression. While many of these genes and signaling pathways are regulated by microRNAs (miRNAs). miRNAs are small 19-22 nucleotide single-stranded RNAs that either as tumor suppressors or oncogenes play an important role in the progression and pathogenesis of cancer. Among them, microRNA-21 (miR-21) is frequently up-regulated in many cancers. Recent studies have shown that miR-21 displays an important role in the occurrence, development, recurrence and drug resistance of MM. In this review, we aim at summarizing the current knowledge of miR-21 functions in MM, with an emphasis on its laboratory research and clinical research in MM.
Collapse
|
27
|
Amigo-Jiménez I, Bailón E, Ugarte-Berzal E, Aguilera-Montilla N, García-Marco JA, García-Pardo A. Matrix metalloproteinase-9 is involved in chronic lymphocytic leukemia cell response to fludarabine and arsenic trioxide. PLoS One 2014; 9:e99993. [PMID: 24956101 PMCID: PMC4067296 DOI: 10.1371/journal.pone.0099993] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 05/21/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Matrix metalloproteinase-9 (MMP-9) contributes to chronic lymphocytic leukemia (CLL) pathology by regulating cell migration and preventing spontaneous apoptosis. It is not known if MMP-9 is involved in CLL cell response to chemotherapy and we address this in the present study, using arsenic trioxide (ATO) and fludarabine as examples of cytotoxic drugs. METHODS We used primary cells from the peripheral blood of CLL patients and MEC-1 cells stably transfected with an empty vector or a vector containing MMP-9. The effect of ATO and fludarabine was determined by flow cytometry and by the MTT assay. Expression of mRNA was measured by RT-PCR and qPCR. Secreted and cell-bound MMP-9 was analyzed by gelatin zymography and flow cytometry, respectively. Protein expression was analyzed by Western blotting and immunoprecipitation. Statistical analyses were performed using the two-tailed Student's t-test. RESULTS In response to ATO or fludarabine, CLL cells transcriptionally upregulated MMP-9, preceding the onset of apoptosis. Upregulated MMP-9 primarily localized to the membrane of early apoptotic cells and blocking apoptosis with Z-VAD prevented MMP-9 upregulation, thus linking MMP-9 to the apoptotic process. Culturing CLL cells on MMP-9 or stromal cells induced drug resistance, which was overcome by anti-MMP-9 antibodies. Accordingly, MMP-9-MEC-1 transfectants showed higher viability upon drug treatment than Mock-MEC-1 cells, and this effect was blocked by silencing MMP-9 with specific siRNAs. Following drug exposure, expression of anti-apoptotic proteins (Mcl-1, Bcl-xL, Bcl-2) and the Mcl-1/Bim, Mcl-1/Noxa, Bcl-2/Bax ratios were higher in MMP-9-cells than in Mock-cells. Similar results were obtained upon culturing primary CLL cells on MMP-9. CONCLUSIONS Our study describes for the first time that MMP-9 induces drug resistance by modulating proteins of the Bcl-2 family and upregulating the corresponding anti-apoptotic/pro-apoptotic ratios. This is a novel role for MMP-9 contributing to CLL progression. Targeting MMP-9 in combined therapies may thus improve CLL response to treatment.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Apoptosis/drug effects
- Arsenic Trioxide
- Arsenicals/pharmacology
- Arsenicals/therapeutic use
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Down-Regulation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Female
- HEK293 Cells
- Humans
- Hyaluronan Receptors/metabolism
- Integrin alpha4beta1/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/enzymology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Middle Aged
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Oxides/pharmacology
- Oxides/therapeutic use
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-jun/genetics
- Transcription, Genetic/drug effects
- Up-Regulation/drug effects
- Vidarabine/analogs & derivatives
- Vidarabine/pharmacology
- Vidarabine/therapeutic use
Collapse
Affiliation(s)
- Irene Amigo-Jiménez
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Elvira Bailón
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Estefanía Ugarte-Berzal
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Noemí Aguilera-Montilla
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | - Angeles García-Pardo
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
28
|
Ryu DS, Kim SH, Kwon JH, Lee DS. Orostachys japonicus induces apoptosis and cell cycle arrest through the mitochondria-dependent apoptotic pathway in AGS human gastric cancer cells. Int J Oncol 2014; 45:459-69. [PMID: 24789703 DOI: 10.3892/ijo.2014.2404] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/07/2014] [Indexed: 11/06/2022] Open
Abstract
We investigated the anticancer mechanisms of the ethylacetate (EtOAc) fraction from Orostachys japonicus in human gastric cancer (AGS) cells. Flow cytometric analysis revealed that the number of total apoptotic cells following treatment with the EtOAc fraction increased in a dose-dependent manner. In the cell cycle analyses, the EtOAc fraction increased the peak in the sub-G1, indicating apoptosis, and in the G₂/M phases in a dose-dependent manner. In the RT-PCR analysis, the expression of cyclin-dependent kinase 1 (CDK 1) and cyclin B1 decreased in a dose- and time-dependent manner. The results of western blotting revealed that the protein levels of p53, cytochrome c, and cleaved caspase-3, -8 and -9 proteins increased and those of B cell lymphoma-2 (bcl-2) and pro-caspase-3, -8 and -9 proteins decreased in a dose- and time-dependent manner, whereas the levels of bcl-2-associated x protein (bax) remained unchanged. Furthermore, the changes in the levels of pro-caspase-3, -8 and -9 and cleaved caspase-3, -8 and -9 were abolished by the pan-caspase inhibitor Z-VAD-FMK. In addition, phosphorylation of p38 and JNK increased in a time-dependent manner. These results, for the first time, provide an understanding of the potential anticancer activity of the O. japonicus, which functions through the induction of apoptosis and cell cycle arrest.
Collapse
Affiliation(s)
- Deok-Seon Ryu
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Gyungnam 621-749, Republic of Korea
| | - Seon-Hee Kim
- Department of Smart Foods and Drugs, Graduate School of Inje University, Gimhae, Gyungnam 621-749, Republic of Korea
| | - Ji-Hae Kwon
- Department of Smart Foods and Drugs, Graduate School of Inje University, Gimhae, Gyungnam 621-749, Republic of Korea
| | - Dong-Seok Lee
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Gyungnam 621-749, Republic of Korea
| |
Collapse
|
29
|
Wu EJ, Goussetis DJ, Beauchamp E, Kosciuczuk EM, Altman JK, Eklund EA, Platanias LC. Resveratrol enhances the suppressive effects of arsenic trioxide on primitive leukemic progenitors. Cancer Biol Ther 2014; 15:473-8. [PMID: 24496081 DOI: 10.4161/cbt.27824] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Efforts to enhance the antileukemic properties of arsenic trioxide are clinically relevant and may lead to the development of new therapeutic approaches for the management of certain hematological malignancies. We provide evidence that concomitant treatment of acute myeloid leukemia (AML) cells or chronic myeloid leukemia (CML) cells with resveratrol potentiates arsenic trioxide-dependent induction of apoptosis. Importantly, clonogenic assays in methylcellulose demonstrate potent suppressive effects of the combination of these agents on primitive leukemic progenitors derived from patients with AML or CML. Taken together, these findings suggest that combinations of arsenic trioxide with resveratrol may provide an approach for targeting of early leukemic precursors and, possibly, leukemia initiating stem cells.
Collapse
Affiliation(s)
- Edward J Wu
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA
| | - Dennis J Goussetis
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA; Division of Hematology/Oncology; Department of Medicine; Feinberg School of Medicine; Northwestern University; Chicago, IL USA; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL USA
| | - Elspeth Beauchamp
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA; Division of Hematology/Oncology; Department of Medicine; Feinberg School of Medicine; Northwestern University; Chicago, IL USA
| | - Ewa M Kosciuczuk
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA
| | - Jessica K Altman
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA; Division of Hematology/Oncology; Department of Medicine; Feinberg School of Medicine; Northwestern University; Chicago, IL USA; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL USA
| | - Elizabeth A Eklund
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA; Division of Hematology/Oncology; Department of Medicine; Feinberg School of Medicine; Northwestern University; Chicago, IL USA; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center; Northwestern University; Chicago, IL USA; Division of Hematology/Oncology; Department of Medicine; Feinberg School of Medicine; Northwestern University; Chicago, IL USA; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL USA
| |
Collapse
|
30
|
Huang X, Wang Y, Nan X, He S, Xu X, Zhu X, Tang J, Yang X, Yao L, Wang X, Cheng C. The role of the orphan G protein-coupled receptor 37 (GPR37) in multiple myeloma cells. Leuk Res 2013; 38:225-35. [PMID: 24290813 DOI: 10.1016/j.leukres.2013.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 11/04/2013] [Accepted: 11/07/2013] [Indexed: 01/14/2023]
Abstract
The orphan G protein-coupled receptor 37 (GPR37) is homologous to endothelin (ETB-R) and bombesin (GRP-R, NMB-R) receptors. The present study was undertaken to determine the expression and functional significance of GPR37 in human multiple myeloma (MM). We found that GPR37 was lowly expressed in MM cell adhesion model and highly expressed in proliferating cells. In vitro, meddling with the expression of GPR37 affected the CAM-DR by regulating the ability of cell adhesion and the activity of Akt and ERK in MM cells. Further studies indicated the positive role of GPR37 in the proliferation of MM cells.
Collapse
Affiliation(s)
- Xianting Huang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Yuchan Wang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xun Nan
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Song He
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu 226001, People's Republic of China
| | - Xiaohong Xu
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu 226001, People's Republic of China
| | - Xinghua Zhu
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu 226001, People's Republic of China
| | - Jie Tang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xiaojing Yang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Li Yao
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xinxiu Wang
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Chun Cheng
- Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
31
|
Aurora and IKK kinases cooperatively interact to protect multiple myeloma cells from Apo2L/TRAIL. Blood 2013; 122:2641-53. [PMID: 23974204 DOI: 10.1182/blood-2013-02-482356] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Constitutive activation of the canonical and noncanonical nuclear factor-κB (NF-κB) pathways is frequent in multiple myeloma (MM) and can compromise sensitivity to TRAIL. In this study, we demonstrate that Aurora kinases physically and functionally interact with the key regulators of canonical and noncanonical NF-κB pathways IκB kinase β (IKKβ) and IKKα to activate NF-κB in MM, and the pharmacological blockade of Aurora kinase activity induces TRAIL sensitization in MM because it abrogates TRAIL-induced activation of NF-κB. We specifically found that TRAIL induces prosurvival signaling by increasing the phosphorylation state of both Aurora and IKK kinases and their physical interactions, and the blockade of Aurora kinase activity by pan-Aurora kinase inhibitors (pan-AKIs) disrupts TRAIL-induced survival signaling by effectively reducing Aurora-IKK kinase interactions and NF-κB activation. Pan-AKIs consistently blocked TRAIL induction of the antiapoptotic NF-κB target genes A1/Bfl-1 and/or Mcl-1, both important targets for TRAIL sensitization in MM cells. In summary, these results identify a novel interaction between Aurora and IKK kinases and show that these pathways can cooperate to promote TRAIL resistance. Finally, combining pan-AKIs with TRAIL in vivo showed dramatic efficacy in a multidrug-resistant human myeloma xenograft model. These findings suggest that combining Aurora kinase inhibitors with TRAIL may have therapeutic benefit in MM.
Collapse
|
32
|
Soldevilla B, Millán CS, Bonilla F, Domínguez G. The TP73 complex network: ready for clinical translation in cancer? Genes Chromosomes Cancer 2013; 52:989-1006. [PMID: 23913810 DOI: 10.1002/gcc.22095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/28/2013] [Indexed: 01/05/2023] Open
Abstract
TP73 is a member of the TP53 family, whose deregulated expression has been reported in a wide variety of cancers and linked to patients' outcome. The fact that TP73 encodes a complex number of isoforms (TAp73 and ΔTAp73) with opposing functions and the cross-talk with other members of the family (TP53 and TP63) make it difficult to determine its clinical relevance. Here, we review the molecular mechanisms driving TAp73 and ΔTAp73 expression and how these variants inhibit or promote carcinogenesis. We also highlight the intricate interplay between TP53 family members. In addition, we comment on current pharmacological approaches targeting the TP73 pathway and those affecting the TAp73/ΔTAp73 ratio. Finally, we discuss the current data available in the literature that provide evidence on the role of TP73 variants in predicting prognosis. To date, most of the studies that evaluate the status levels of TP73 isoforms have been based on limited-size series. Despite this limitation, these publications highlight the correlation between high levels of the oncogenic forms and failure to respond to chemotherapy and/or shorter survival. Finally, we emphasize the need for studies to evaluate the significance of combining the deregulation of various members of the TP53 family in order to define patient outcome or their responsiveness to specific therapies.
Collapse
Affiliation(s)
- Beatriz Soldevilla
- Servicio de Oncología Médica, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | | | | | | |
Collapse
|
33
|
Bortezomid enhances the efficacy of lidamycin against human multiple myeloma cells. Anticancer Drugs 2013; 24:609-16. [DOI: 10.1097/cad.0b013e3283615006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
34
|
Zhen YZ, Hu G, Zhao YF, Yan F, Li R, Gao JL, Lin YJ. Synergy of Taxol and rhein lysinate associated with the downregulation of ERK activation in lung carcinoma cells. Oncol Lett 2013; 6:525-528. [PMID: 24137360 PMCID: PMC3789082 DOI: 10.3892/ol.2013.1398] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 06/10/2013] [Indexed: 12/13/2022] Open
Abstract
In previous studies we observed that rhein lysinate (RHL), a salt of rhein and lysine that is easily dissolved in water, inhibited the growth of tumor cells in breast cancer, ovarian cancer, hepatocellular carcinoma and cervical cancer. The present study aimed to investigate the effects of RHL on H460 and A549 non-small cell lung cancer (NSCLC) cells using a combination of RHL and Taxol. A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay was used to determine the growth inhibition effect of the drugs in the H460 and A549 cells. Cell apoptosis was analyzed by flow cytometry combined with fluorescein-isothiocyanate-Annexin V/propidium iodide (PI) staining. The expression levels of proteins were detected by western blotting. There was a significant reduction in the proliferation of the NSCLC cell lines treated with a combination of Taxol and RHL. The overall growth inhibition was directly correlated with apoptotic cell death. RHL potentiated Taxol-induced cell killing by reducing extracellular signal-regulated kinase (ERK) activity and increasing the levels of cleaved poly(ADP-ribose) polymerase (PARP) and caspase-3. Notably, the results for the Bcl-2 and NF-κB proteins also showed downregulation in the combined treatment group compared with the single-agent treatment and the untreated control groups. The present results showed that RHL potentiates the growth inhibition induced by Taxol in NSCLC cells and showed that this synergy may be associated with the downregulation of ERK activation.
Collapse
Affiliation(s)
- Yong-Zhan Zhen
- Basic Medical College of Hebei United University, Tangshan, Hebei 063000, P.R. China
| | | | | | | | | | | | | |
Collapse
|
35
|
Modular pharmacology: deciphering the interacting structural organization of the targeted networks. Drug Discov Today 2013; 18:560-6. [DOI: 10.1016/j.drudis.2013.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 12/14/2012] [Accepted: 01/16/2013] [Indexed: 12/24/2022]
|
36
|
Hu J, Huang X, Hong X, Lu Q, Zhu X. Arsenic trioxide inhibits the proliferation of myeloma cell line through notch signaling pathway. Cancer Cell Int 2013; 13:25. [PMID: 23497375 PMCID: PMC3600676 DOI: 10.1186/1475-2867-13-25] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/07/2013] [Indexed: 11/16/2022] Open
Abstract
Arsenic Trioxide (ATO) has shown remarkable efficacy for the treatment of multiple myeloma (MM). However, the mechanism by which ATO exerts its inhibitory effect on the proliferation of myeloma cells remains to be clarified. We study the inhibitory effect of ATO at various concentrations on the proliferation of the myeloma cell line RPMI 8226 and discussed the molecular mechanism of ATO on myeloma cell line. Our results proved that ATO had a significant dose-dependent and time-dependent inhibitory effect on the expressions of the Notch receptor (Notch1) and Notch ligand (Jag2). Data from the real-time PCR assay showed that the mRNA expression levels of the Jag2 gene and its downstream gene Hes1 were both significantly down-regulated after the myeloma cells were treated with ATO while the expression of the tumor suppressor gene PTEN was up-regulated. These results elucidated the molecular mechanism underlying the ATO mediated inhibition of myeloma cell proliferation. This is the first report on the anti-myeloma activity in myeloma cells through inhibition of the Notch signaling pathway.
Collapse
Affiliation(s)
- Jiasheng Hu
- Department of Haematology, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361004, China.
| | | | | | | | | |
Collapse
|
37
|
Galvin JP, Altman JK, Szilard A, Goussetis DJ, Vakana E, Sassano A, Platanias LC. Regulation of the kinase RSK1 by arsenic trioxide and generation of antileukemic responses. Cancer Biol Ther 2013; 14:411-6. [PMID: 23377826 DOI: 10.4161/cbt.23760] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Arsenic Trioxide (As₂O₃) is one of the most effective agents in the treatment of acute promyelocytic leukemia (APL), but has no significant efficacy in other forms of AML. The mechanisms of relative resistance of non-APL cells are not well understood, but emerging evidence suggests that activation of negative feedback regulatory loops and pathways contributes to such resistance. We provide evidence that a signaling cascade involving the kinase RSK1 is engaged in a negative feedback manner during arsenic-treatment of cells and exhibits regulatory effects on growth and survival of AML cells in response to treatment with As₂O₃. Our data demonstrate that pharmacological inhibition or molecular disruption of expression of RSK1 enhances As₂O₃-dependent apoptosis and/or growth inhibition of AML cells. Importantly, combination of a pharmacological inhibitor of RSK and As₂O₃ results in enhanced suppression of primary AML leukemic progenitors. Altogether, our findings suggest an important regulatory role for RSK1 in the generation of the effects of As₂O₃ in AML cells. They also raise the potential of RSK1 targeting in combination with As₂O₃ as a novel approach to promote antileukemic responses.
Collapse
Affiliation(s)
- John P Galvin
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Alterations in glutathione levels and apoptotic regulators are associated with acquisition of arsenic trioxide resistance in multiple myeloma. PLoS One 2012; 7:e52662. [PMID: 23285138 PMCID: PMC3528737 DOI: 10.1371/journal.pone.0052662] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 11/19/2012] [Indexed: 11/19/2022] Open
Abstract
Arsenic trioxide (ATO) has been tested in relapsed/refractory multiple myeloma with limited success. In order to better understand drug mechanism and resistance pathways in myeloma we generated an ATO-resistant cell line, 8226/S-ATOR05, with an IC50 that is 2–3-fold higher than control cell lines and significantly higher than clinically achievable concentrations. Interestingly we found two parallel pathways governing resistance to ATO in 8226/S-ATOR05, and the relevance of these pathways appears to be linked to the concentration of ATO used. We found changes in the expression of Bcl-2 family proteins Bfl-1 and Noxa as well as an increase in cellular glutathione (GSH) levels. At low, clinically achievable concentrations, resistance was primarily associated with an increase in expression of the anti-apoptotic protein Bfl-1 and a decrease in expression of the pro-apoptotic protein Noxa. However, as the concentration of ATO increased, elevated levels of intracellular GSH in 8226/S-ATOR05 became the primary mechanism of ATO resistance. Removal of arsenic selection resulted in a loss of the resistance phenotype, with cells becoming sensitive to high concentrations of ATO within 7 days following drug removal, indicating changes associated with high level resistance (elevated GSH) are dependent upon the presence of arsenic. Conversely, not until 50 days without arsenic did cells once again become sensitive to clinically relevant doses of ATO, coinciding with a decrease in the expression of Bfl-1. In addition we found cross-resistance to melphalan and doxorubicin in 8226/S-ATOR05, suggesting ATO-resistance pathways may also be involved in resistance to other chemotherapeutic agents used in the treatment of multiple myeloma.
Collapse
|
39
|
Therapeutic potential of MEK inhibition in acute myelogenous leukemia: rationale for "vertical" and "lateral" combination strategies. J Mol Med (Berl) 2012; 90:1133-44. [PMID: 22399013 DOI: 10.1007/s00109-012-0886-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 02/08/2012] [Accepted: 02/14/2012] [Indexed: 02/03/2023]
Abstract
In hematological malignancies, constitutive activation of the RAF/MEK/ERK pathway is frequently observed, conveys a poor prognosis, and constitutes a promising target for therapeutic intervention. Here, we investigated the molecular and functional effects of pharmacological MEK inhibition in cell line models of acute myeloid leukemia (AML) and freshly isolated primary AML samples. The small-molecule, ATP-non-competitive, MEK inhibitor PD0325901 markedly inhibited ERK phosphorylation and growth of several AML cell lines and approximately 70 % of primary AML samples. Growth inhibition was due to G(1)-phase arrest and induction of apoptosis. Transformation by constitutively active upstream pathway elements (HRAS, RAF-1, and MEK) rendered FDC-P1 cells exquisitely prone to PD0325901-induced apoptosis. Gene and protein expression profiling revealed a selective effect of PD0325901 on ERK phosphorylation and compensatory upregulation of the RAF/MEK and AKT/p70( S6K ) kinase modules, potentially mediating resistance to drug-induced growth inhibition. Consequently, in appropriate cellular contexts, both "vertical" (i.e., inhibition of RAF and MEK along the MAPK pathway) and "lateral" (i.e., simultaneous inhibition of the MEK/ERK and mTOR pathways) combination strategies may result in synergistic anti-leukemic effects. Overall, MEK inhibition exerts potent growth inhibitory and proapoptotic activity in preclinical models of AML, particularly in combination with other pathway inhibitors. Deeper understanding of the molecular mechanisms of action of MEK inhibitors will likely translate into more effective targeted strategies for the treatment of AML.
Collapse
|
40
|
Xu G, Shen XJ, Pu J, Chu SP, Wang XD, Wu XH, Sun CJ, Zhang X, Zhu BL, Ju SQ. BLyS expression and JNK activation may form a feedback loop to promote survival and proliferation of multiple myeloma cells. Cytokine 2012; 60:505-13. [PMID: 22850273 DOI: 10.1016/j.cyto.2012.06.317] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/31/2012] [Accepted: 06/28/2012] [Indexed: 12/25/2022]
Abstract
B-Lymphocyte stimulator (BLyS), a member of tumor necrosis factor superfamily, is a potent co-activator of B cells in vitro, and in vivo induces B cell proliferation and immunoglobulin secretion. Multiple myeloma (MM) is an incurable malignancy of terminally differentiated B cells (plasma cells). Previous studies have well ascertained that BLyS plays an important contributory role in the pathogenesis and propagation of multiple myeloma by virtue of its ability to promote B cell survival, expansion, and differentiation. However, the intracellular signaling of BLyS in human MM cells remains undefined. This study was designed to see whether there was interaction between MAPK signaling pathway and BLyS expression. It was found that the active protein p-JNK was expressed in KM3, U266 and PBMCs of MM patients, and that the expression of BLyS could be changed by JNK pathway activator and inhibitor. In addition, recombinant BLyS activated JNK pathway, while BLyS siRNA treatment inhibited the activation of JNK pathway. The level of BLyS expression and the activation of JNK pathway were positively correlated. These findings suggest that JNK activation and BLyS expression in MM cells may form a positive feedback loop that promotes the survival and proliferation of MM cells, and these may shed some light on the pathogenesis and treatment of MM.
Collapse
Affiliation(s)
- G Xu
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, JS, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee C, Zhang Q, Zi X, Dash A, Soares MB, Rahmatpanah F, Jia Z, McClelland M, Mercola D. TGF-β mediated DNA methylation in prostate cancer. Transl Androl Urol 2012; 1:78-88. [PMID: 25133096 PMCID: PMC4131550 DOI: 10.3978/j.issn.2223-4683.2012.05.06] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 05/04/2012] [Indexed: 12/15/2022] Open
Abstract
Almost all tumors harbor a defective negative feedback loop of signaling by transforming growth factor-β (TGF-β). Epigenetic mechanisms of gene regulation, including DNA methylation, are fundamental to normal cellular function and also play a major role in carcinogenesis. Recent evidence demonstrated that TGF-β signaling mediates cancer development and progression. Many key events in TGF-β signaling in cancer included auto-induction of TGF-β1 and increased expression of DNA methyltransferases (DNMTs), suggesting that DNA methylation plays a significant role in cancer development and progression. In this review, we performed an extensive survey of the literature linking TGF-β signaling to DNA methylation in prostate cancer. It appeared that almost all DNA methylated genes detected in prostate cancer are directly or indirectly related to TGF-β signaling. This knowledge has provided a basis for our future directions of prostate cancer research and strategies for prevention and therapy for prostate cancer.
Collapse
|
42
|
Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B, McQueen T, Bornmann W, Tsao T, Bergamo P, Mak DH, Chen W, McCubrey J, Tafuri A, Andreeff M. MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia 2012; 26:778-87. [PMID: 22064351 PMCID: PMC3604791 DOI: 10.1038/leu.2011.287] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 08/25/2011] [Accepted: 09/06/2011] [Indexed: 12/29/2022]
Abstract
Recently, strategies for acute myeloid leukemia (AML) therapy have been developed that target anti-apoptotic BCL2 family members using BH3-mimetic drugs such as ABT-737. Though effective against BCL2 and BCL-X(L), ABT-737 poorly inhibits MCL-1. Here we report that, unexpectedly, ABT-737 induces activation of the extracellular receptor activated kinase and induction of MCL-1 in AML cells. MEK inhibitors such as PD0325901 and CI-1040 have been used successfully to suppress MCL-1. We report that PD0325901 blocked ABT-737-induced MCL-1 expression, and when combined with ABT-737 resulted in potent synergistic killing of AML-derived cell lines, primary AML blast and CD34+38-123+ progenitor/stem cells. Finally, we tested the combination of ABT-737 and CI-1040 in a murine xenograft model using MOLM-13 human leukemia cells.Whereas control mice and CI-1040-treated mice exhibited progressive leukemia growth, ABT-737, and to a significantly greater extent, ABT-737+CI-1040 exerted major anti-leukemia activity. Collectively, results demonstrated unexpected anti-apoptotic interaction between the BCL2 family-targeted BH3-mimetic ABT-737 and mitogen-activated protein kinase signaling in AML cells: the BH3 mimetic is not only restrained in its activity by MCL-1, but also induces its expression. However, concomitant inhibition by BH3 mimetics and MEK inhibitors could abrogate this effect and may be developed into a novel and effective therapeutic strategy for patients with AML.
Collapse
Affiliation(s)
- M Konopleva
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bisso A, Collavin L, Del Sal G. p73 as a pharmaceutical target for cancer therapy. Curr Pharm Des 2011; 17:578-90. [PMID: 21391908 PMCID: PMC3267157 DOI: 10.2174/138161211795222667] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 03/03/2011] [Indexed: 02/07/2023]
Abstract
About half of all human tumors contain an inactivating mutation of p53, while in the remaining tumors, the p53 pathway is frequently abrogated by alterations of other components of its signaling pathway. In humans, the p53 tumor suppressor is part of a small gene family that includes two other members, p73 and p63, structurally and functionally related to p53. Accumulating evidences indicate that all p53-family proteins function as molecular hubs of a highly interconnected signaling network that coordinates cell proliferation, differentiation and death in response to physiological inputs and oncogenic stress. Therefore, not only the p53-pathway but the entire “p53-family pathway” is a primary target for cancer drug development. In particular, the p53-related protein p73 has a crucial role in determining cellular responses to chemotherapy, and can vicariate p53 functions in triggering cell death after DNA damage in multiple experimental models. The biology and regulation of p73 is complex, since the TP73 gene incorporates both tumor-suppressive and proto-oncogenic functions. However, the p73 gene is rarely mutated in tumors, so appropriate pharmacological manipulation of the p73 pathway is a very promising approach for cancer therapy. Here we provide an overview of the principal mechanism of p73 regulation, and describe several examples of pharmacological tools that can induce p73 accumulation and function by acting on upstream p73 modulators or displacing inhibitory p73 interactors. A better understanding of how the p73 pathway works is mandatory to discover additional players intervening in this pathway and has important implications for the improvement of cancer treatment with the development of new molecules or with the reposition of currently available drugs.
Collapse
Affiliation(s)
- Andrea Bisso
- Laboratorio Nazionale CIB, AREA Science Park, Padriciano 99, Trieste, TS 34149, Italy
| | | | | |
Collapse
|
44
|
Li Y, Kong D, Bao B, Ahmad A, Sarkar FH. Induction of cancer cell death by isoflavone: the role of multiple signaling pathways. Nutrients 2011; 3:877-96. [PMID: 22200028 PMCID: PMC3244210 DOI: 10.3390/nu3100877] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/29/2011] [Accepted: 10/09/2011] [Indexed: 12/31/2022] Open
Abstract
Soy isoflavones have been documented as dietary nutrients broadly classified as "natural agents" which plays important roles in reducing the incidence of hormone-related cancers in Asian countries, and have shown inhibitory effects on cancer development and progression in vitro and in vivo, suggesting the cancer preventive or therapeutic activity of soy isoflavones against cancers. Emerging experimental evidence shows that isoflavones could induce cancer cell death by regulating multiple cellular signaling pathways including Akt, NF-κB, MAPK, Wnt, androgen receptor (AR), p53 and Notch signaling, all of which have been found to be deregulated in cancer cells. Therefore, homeostatic regulation of these important cellular signaling pathways by isoflavones could be useful for the activation of cell death signaling, which could result in the induction of apoptosis of both pre-cancerous and/or cancerous cells without affecting normal cells. In this article, we have attempted to summarize the current state-of-our-knowledge regarding the induction of cancer cell death pathways by isoflavones, which is believed to be mediated through the regulation of multiple cellular signaling pathways. The knowledge gained from this article will provide a comprehensive view on the molecular mechanism(s) by which soy isoflavones may exert their effects on the prevention of tumor progression and/or treatment of human malignancies, which would also aid in stimulating further in-depth mechanistic research and foster the initiation of novel clinical trials.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, 740 Hudson Webber Cancer Research Center, 4100 John R, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
45
|
Moffatt CE, Whitmore SE, Griffen AL, Leys EJ, Lamont RJ. Filifactor alocis interactions with gingival epithelial cells. Mol Oral Microbiol 2011; 26:365-73. [PMID: 22053964 DOI: 10.1111/j.2041-1014.2011.00624.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An association between the gram-positive anaerobe Filifactor alocis and periodontal disease has recently emerged; however, possible pathogenic mechanisms have not been investigated. In this study we examined the responses of primary cultures of gingival epithelial cells (GECs) to infection with F. alocis. Secretion of the pro-inflammatory cytokines interleukin-1β, interleukin-6 and tumor necrosis factor-α from GECs was stimulated by F. alocis infection. F. alocis also induced apoptosis in GECs through pathways that involved caspase-3 but not caspase-9. Apoptosis was coincident with inhibition of mitogen-activated protein kinase kinase (MEK) activation. These results show that F. alocis has characteristics in common with established periodontal pathogens and has the potential to contribute to periodontal tissue destruction.
Collapse
Affiliation(s)
- C E Moffatt
- Center for Oral Health and Systemic Disease, School of Dentistry, University of Louisville, Louisville, KY 40292, USA
| | | | | | | | | |
Collapse
|
46
|
Chen SJ, Zhou GB, Zhang XW, Mao JH, de Thé H, Chen Z. From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia. Blood 2011; 117:6425-37. [PMID: 21422471 PMCID: PMC3123014 DOI: 10.1182/blood-2010-11-283598] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 03/11/2011] [Indexed: 12/29/2022] Open
Abstract
Arsenic had been used in treating malignancies from the 18th to mid-20th century. In the past 3 decades, arsenic was revived and shown to be able to induce complete remission and to achieve, when combined with all-trans retinoic acid and chemotherapy, a 5-year overall survival of 90% in patients with acute promyelocytic leukemia driven by the t(15;17) translocation-generated promyelocytic leukemia-retinoic acid receptor α (PML-RARα) fusion. Molecularly, arsenic binds thiol residues and induces the formation of reactive oxygen species, thus affecting numerous signaling pathways. Interestingly, arsenic directly binds the C3HC4 zinc finger motif in the RBCC domain of PML and PML-RARα, induces their homodimerization and multimerization, and enhances their interaction with the SUMO E2 conjugase Ubc9, facilitating subsequent sumoylation/ubiquitination and proteasomal degradation. Arsenic-caused intermolecular disulfide formation in PML also contributes to PML-multimerization. All-trans retinoic acid, which targets PML-RARα for degradation through its RARα moiety, synergizes with arsenic in eliminating leukemia-initiating cells. Arsenic perturbs a number of proteins involved in other hematologic malignancies, including chronic myeloid leukemia and adult T-cell leukemia/lymphoma, whereby it may bring new therapeutic benefits. The successful revival of arsenic in acute promyelocytic leukemia, together with modern mechanistic studies, has thus allowed a new paradigm to emerge in translational medicine.
Collapse
Affiliation(s)
- Sai-Juan Chen
- Shanghai Institute of Hematology and State Key Laboratory for Medical Genomics, Rui Jin Hospital/Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Road II, Shanghai, China
| | | | | | | | | | | |
Collapse
|
47
|
Assessment of the involvement of oxidative stress and Mitogen-Activated Protein Kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines. Med Oncol 2011; 29:1161-72. [DOI: 10.1007/s12032-011-9920-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022]
|
48
|
Interruption of the MEK/ERK signaling cascade promotes dihydroartemisinin-induced apoptosis in vitro and in vivo. Apoptosis 2011; 16:511-23. [DOI: 10.1007/s10495-011-0580-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
49
|
Abeltino M, Bonomini S, Bolzoni M, Storti P, Colla S, Todoerti K, Agnelli L, Neri A, Rizzoli V, Giuliani N. The proapoptotic effect of zoledronic acid is independent of either the bone microenvironment or the intrinsic resistance to bortezomib of myeloma cells and is enhanced by the combination with arsenic trioxide. Exp Hematol 2011; 39:55-65. [DOI: 10.1016/j.exphem.2010.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 09/16/2010] [Accepted: 10/18/2010] [Indexed: 11/25/2022]
|
50
|
Yingchun L, Xiujuan Q, Jinglei Q, Ye Z, Jing L, Yuee T, Xuejun H, Kezuo H, Yunpeng L. E3 ubiquitin ligase Cbl-b potentiates the apoptotic action of arsenic trioxide by inhibiting the PI3K/Akt pathway. Braz J Med Biol Res 2010; 44:105-11. [PMID: 21180886 DOI: 10.1590/s0100-879x2010007500142] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 12/06/2010] [Indexed: 11/22/2022] Open
Abstract
Arsenic trioxide (ATO) is a strong inducer of apoptosis in malignant hematological cells. Inducible phosphatidyl inositol 3 kinase (PI3K)-Akt activation promotes resistance to ATO. In the present study, we evaluated whether E3 ubiquitin ligase Cbl-b, a negative regulator of PI3K activation, is involved in the action of ATO. The effect of ATO on cell viability was measured by the Trypan blue exclusion assay or by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis was determined by flow cytometry and protein expression was assayed by Western blotting. ATO decreased the viability of HL60 cells and induced cellular apoptosis, which was accompanied by transient activation of Akt. The PI3K/Akt inhibitor, LY294002, significantly increased ATO-induced apoptosis (P < 0.05). In addition, ATO up-regulated the expression of Cbl-b proteins. Furthermore, ATO inhibited cell viability with an IC50 of 18.54 μM at 24 h in rat basophilic leukemia-2H3 cells. ATO induced cellular apoptosis with transient activation of Akt and Cbl-b was also up-regulated. Rat basophilic leukemia-2H3 cells transfected with a dominant negative (DN) Cbl-b mutation showed overexpression of Cbl-b (DN) and enhanced Akt activation. Compared with cells transfected with vector, ATO-induced apoptosis was decreased and G2/M phase cells were increased at the same concentration (P < 0.05). The PI3K/Akt inhibitor, LY294002, re-sensitized Cbl-b (DN) overexpressing cells to ATO and reversed G2/M arrest (P < 0.05). Taken together, these results suggest that Cbl-b potentiates the apoptotic action of ATO by inhibition of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Li Yingchun
- Department of Medical Oncology, China Medical University
| | | | | | | | | | | | | | | | | |
Collapse
|