251
|
Cao Y, Qiu GQ, Wu HQ, Wang ZL, Lin Y, Wu W, Xie XB, Gu WY. Decitabine enhances bortezomib treatment in RPMI 8226 multiple myeloma cells. Mol Med Rep 2016; 14:3469-75. [PMID: 27571872 DOI: 10.3892/mmr.2016.5658] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 06/24/2016] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the interactions between decitabine (DAC) and bortezomib (BTZ) in RPMI 8226 multiple myeloma (MM) cells. Cells were exposed to DAC alone and in combination with BTZ for 48 h. A Cell Counting Kit‑8 assay was performed to assess the rate of proliferation inhibition in the cells. Cell apoptosis was investigated by Annexin V-fluorescein isothiocyanate and propidium iodide staining. Flow cytometry was used to detect the different cell cycle stages. Western blotting was performed to analyze the protein expression levels of poly(ADP‑ribose) polymerase 1 (PARP‑1), caspase‑3, ‑9 and DNA (cytosine‑5‑)‑methyltransferase 1 (DNMT1). Reverse transcription‑quantitative polymerase chain reaction was used to assess DNMT1 gene expression. The combination of DAC and BTZ increased the proliferation inhibition, apoptotic rate and G0‑G1 arrest compared with use of a single therapeutic agent. In addition, the combination treatment enhanced PARP‑1 cleavage, caspase‑3 and caspase‑9 activation and downregulated the protein and mRNA expression levels of DNMT1. Therefore, the current study determined that the combination of BTZ and the epigenetic agent DAC may be a novel therapeutic strategy to improve the efficacy of BTZ in patients with MM.
Collapse
Affiliation(s)
- Yang Cao
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Guo-Qiang Qiu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Hao-Qing Wu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Zhi-Lin Wang
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Yan Lin
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Wei Wu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Xiao-Bao Xie
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| | - Wei-Ying Gu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
252
|
Suryadevara CM, Riccione KA, Sampson JH. Immunotherapy Gone Viral: Bortezomib and oHSV Enhance Antitumor NK-Cell Activity. Clin Cancer Res 2016; 22:5164-5166. [PMID: 27521450 DOI: 10.1158/1078-0432.ccr-16-1666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 11/16/2022]
Abstract
Oncolytic viruses, proteasome inhibitors, and natural killer (NK)-cell immunotherapy have all been studied extensively as monotherapies but have never been evaluated in combination. Synergetic treatment of oncolytic virus-infected glioblastomas with a proteasome inhibitor induces necroptotic cell death to enhance NK-cell immunotherapy, prolonging survival against human glioblastoma. Clin Cancer Res; 22(21); 5164-6. ©2016 AACRSee related article by Yoo et al., p. 5265.
Collapse
Affiliation(s)
- Carter M Suryadevara
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Katherine A Riccione
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - John H Sampson
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina. .,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina.,Department of Biomedical Engineering, Duke University, Durham, North Carolina
| |
Collapse
|
253
|
Affiliation(s)
- Dimitrios C. Ziogas
- Department of Clinical Therapeutics, Alexandra General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Alexandra General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, Alexandra General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, Alexandra General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
254
|
Citrin R, Foster JB, Teachey DT. The role of proteasome inhibition in the treatment of malignant and non-malignant hematologic disorders. Expert Rev Hematol 2016; 9:873-89. [DOI: 10.1080/17474086.2016.1216311] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
255
|
Structural characterization, ROS-inductive and proteasome inhibitory properties of ternary and binary copper(II) complexes of N2- and N2O2-ligands. Inorganica Chim Acta 2016. [DOI: 10.1016/j.ica.2016.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
256
|
Lin CC, Lee MH, Lin JH, Lin ML, Chueh FS, Yu CC, Lin JP, Chou YC, Hsu SC, Chung JG. Crude extract of Rheum palmatum L. Induces cell cycle arrest S phase and apoptosis through mitochondrial-dependent pathways in U-2 OS human osteosarcoma cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:957-969. [PMID: 25689151 DOI: 10.1002/tox.22105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 12/19/2014] [Accepted: 12/21/2014] [Indexed: 06/04/2023]
Abstract
Cancer is the second cause of death in children. Osteosarcoma is the most common primary malignancy of solid bone cancer primarily affecting adolescents and young adults. In the Chinese population, the crude extract of Rheum palmatum L. (CERP) has been used for treating different diseases, including SARS, rheumatoid arthritis, coxsackievirus B3, and human colon cancer cell, pancreatic cancer. There are no reports on CERP and human osteosarcoma cells. The present study examined effects of CERP on cytotoxicity including cell cycle distribution and cell death (apoptosis) in U-2 OS human osteosarcoma cells. CERP significantly induced S phase arrest in U-2 OS cells in a dose-dependent. CERP produced DNA damage and DNA condensation. Other effects of CERP were stimulation of ROS and Ca(2+) , mitochondria impairment, and activation of caspase-3, -8, and -9. CERP increased the levels of Bax, Bak, Bad, cyclin B, Fas, PARP, GRP78, GADD153, AIF, Endo G, Calpain-2, p21, and p27, but decreased the levels of Bcl-2, BCL-X, XIAP, Akt, CDC25A, CDK2, Cyclin A, and Cyclin E of U-2 OS cells. It was also observed that CERP promoted the expression of AIF, Endo G, GADD153, and cytochrome c. These results indicate that CERP has anticancer effects in vitro and provide the foundation for in vivo studies of animal models of osteosarcoma. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 957-969, 2016.
Collapse
Affiliation(s)
- Chin-Chung Lin
- Department of Chinese Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Executive Yuan, Taichung, 420, Taiwan
- General Education Center, Central Taiwan University of Science and Technology, Taichung, 406, Taiwan
| | - Ming-Huei Lee
- General Education Center, Central Taiwan University of Science and Technology, Taichung, 406, Taiwan
- Department of Urology, Feng-Yuan Hospital, Ministry of Health and Welfare, Executive Yuan, Taichung, 420, Taiwan
| | - Ju-Hwa Lin
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Meng-Liang Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Fu-Shin Chueh
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, 413, Taiwan
| | - Chien-Chih Yu
- School of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Jing-Pin Lin
- School of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Yu-Cheng Chou
- Division of Neurosurgical Oncology, Neurological Institute, Taichung Veterans General Hospital, Taichung, 407, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, 970, Taiwan
| | - Shu-Chun Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
257
|
Li L, Xu-Monette ZY, Ok CY, Tzankov A, Manyam GC, Sun R, Visco C, Zhang M, Montes-Moreno S, Dybkaer K, Chiu A, Orazi A, Zu Y, Bhagat G, Richards KL, Hsi ED, Choi WWL, van Krieken JH, Huh J, Ponzoni M, Ferreri AJM, Møller MB, Wang J, Parsons BM, Winter JN, Piris MA, Pham LV, Medeiros LJ, Young KH. Prognostic impact of c-Rel nuclear expression and REL amplification and crosstalk between c-Rel and the p53 pathway in diffuse large B-cell lymphoma. Oncotarget 2016; 6:23157-80. [PMID: 26324762 PMCID: PMC4695110 DOI: 10.18632/oncotarget.4319] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 06/16/2015] [Indexed: 02/06/2023] Open
Abstract
Dysregulated NF-κB signaling is critical for lymphomagenesis. The regulation, function, and clinical relevance of c-Rel/NF-κB activation in diffuse large B-cell lymphoma (DLBCL) have not been well studied. In this study we analyzed the prognostic significance and gene-expression signature of c-Rel nuclear expression as surrogate of c-Rel activation in 460 patients with de novo DLBCL. Nuclear c-Rel expression, observed in 137 (26.3%) DLBCL patients frequently associated with extranoal origin, did not show significantly prognostic impact in the overall- or germinal center B-like-DLBCL cohort, likely due to decreased pAKT and Myc levels, up-regulation of FOXP3, FOXO3, MEG3 and other tumor suppressors coincided with c-Rel nuclear expression, as well as the complicated relationships between NF-κB members and their overlapping function. However, c-Rel nuclear expression correlated with significantly poorer survival in p63+ and BCL-2− activated B-cell-like-DLBCL, and in DLBCL patients with TP53 mutations. Multivariate analysis indicated that after adjusting clinical parameters, c-Rel positivity was a significantly adverse prognostic factor in DLBCL patients with wild type TP53. Gene expression profiling suggested dysregulations of cell cycle, metabolism, adhesion, and migration associated with c-Rel activation. In contrast, REL amplification did not correlate with c-Rel nuclear expression and patient survival, likely due to co-amplification of genes that negatively regulate NF-κB activation. These insights into the expression, prognostic impact, regulation and function of c-Rel as well as its crosstalk with the p53 pathway underscore the importance of c-Rel and have significant therapeutic implications.
Collapse
Affiliation(s)
- Ling Li
- Zhengzhou University, The First Affiliated University Hospital, Zhengzhou, China.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zijun Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruifang Sun
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Mingzhi Zhang
- Zhengzhou University, The First Affiliated University Hospital, Zhengzhou, China
| | | | | | - April Chiu
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Attilio Orazi
- Weill Medical College of Cornell University, New York, NY, USA
| | - Youli Zu
- The Methodist Hospital, Houston, TX, USA
| | - Govind Bhagat
- Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Kristy L Richards
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - William W L Choi
- University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong, China
| | | | - Jooryung Huh
- Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | | | | | | | | | | | - Jane N Winter
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Miguel A Piris
- Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Lan V Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas School of Medicine, Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
258
|
Bortezomib inhibits bacterial and fungal β-carbonic anhydrases. Bioorg Med Chem 2016; 24:4406-4409. [PMID: 27469982 DOI: 10.1016/j.bmc.2016.07.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/16/2016] [Indexed: 12/28/2022]
Abstract
Inhibition of the β-carbonic anhydrases (CAs, EC 4.2.1.1) from pathogenic fungi (Cryptococcus neoformans, Candida albicans, Candida glabrata, Malassezia globosa) and bacteria (three isoforms from Mycobacterium tuberculosis, Rv3273, Rv1284 and Rv3588), as well from the insect Drosophila melanogaster (DmeCA) and the plant Flaveria bidentis (FbiCA1) with the boronic acid peptidomimetic proteosome inhibitor bortezomib was investigated. Bortezomib was a micromolar inhibitor of all these enzymes, with KIs ranging between 1.12 and 11.30μM. Based on recent crystallographic data it is hypothesized that the B(OH)2 moiety of the inhibitor is directly coordinated to the zinc ion from the enzyme active site. The class of boronic acids, an under-investigated type of CA inhibitors, may lead to the development of anti-infectives with a novel mechanism of action, based on the pathogenic organisms CA inhibition.
Collapse
|
259
|
Chidley C, Trauger SA, Birsoy K, O'Shea EK. The anticancer natural product ophiobolin A induces cytotoxicity by covalent modification of phosphatidylethanolamine. eLife 2016; 5. [PMID: 27403889 PMCID: PMC4942256 DOI: 10.7554/elife.14601] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/02/2016] [Indexed: 12/18/2022] Open
Abstract
Phenotypic screens allow the identification of small molecules with promising anticancer activity, but the difficulty in characterizing the mechanism of action of these compounds in human cells often undermines their value as drug leads. Here, we used a loss-of-function genetic screen in human haploid KBM7 cells to discover the mechanism of action of the anticancer natural product ophiobolin A (OPA). We found that genetic inactivation of de novo synthesis of phosphatidylethanolamine (PE) mitigates OPA cytotoxicity by reducing cellular PE levels. OPA reacts with the ethanolamine head group of PE in human cells to form pyrrole-containing covalent cytotoxic adducts and these adducts lead to lipid bilayer destabilization. Our characterization of this unusual cytotoxicity mechanism, made possible by unbiased genetic screening in human cells, suggests that the selective antitumor activity displayed by OPA may be due to altered membrane PE levels in cancer cells. DOI:http://dx.doi.org/10.7554/eLife.14601.001 Many of the medications that are available to treat cancer are either collected from natural sources or inspired by molecules existing in nature. While it is often challenging to understand how these natural compounds selectively kill cancer cells, characterizing these mechanisms is essential if researchers are to develop new anticancer drugs and treatments based on these compounds. Ophiobolin A is a compound naturally made by a fungus in order to attack plant cells. It is also able to potently kill cancer cells from humans. In particular, ophiobolin A is a promising candidate for treatment of a type of brain tumor called glioblastomas, which are notoriously difficult to treat with existing medications. Using a newly developed method, Chidley et al. have now tested which components of human cancer cells are important for ophiobolin A to exert its killing effect. The method revealed that ophiobolin A was less able to kill cancer cells if the cells had lower levels of a molecule called phosphatidylethanolamine in their surface membranes. This observation led Chidley et al. to show that ophiobolin A enters the membrane of human cancer cells and combines chemically with phosphatidylethanolamine to form a new composite molecule. Further experiments showed that the formation of this composite molecule disrupted a model membrane, which suggests that ophiobolin A kills cancer cells by breaking their membranes. The next challenge is to understand exactly how the composite molecule kills cancer cells via membrane disruption. It also remains unclear if the anticancer activity of ophiobolin A results from cancer cells having a membrane composition that is different from normal cells, and why this difference arises in the first place. DOI:http://dx.doi.org/10.7554/eLife.14601.002
Collapse
Affiliation(s)
- Christopher Chidley
- Faculty of Arts and Sciences Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States.,Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| | - Sunia A Trauger
- Small Molecule Mass Spectrometry Facility, Faculty of Arts and Sciences Center for Systems Biology, Harvard University, Cambridge, United States
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York, United States
| | - Erin K O'Shea
- Faculty of Arts and Sciences Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States.,Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| |
Collapse
|
260
|
Postiglione I, Barra F, Aloj SM, Palumbo G. Photodynamic therapy with 5-aminolaevulinic acid and DNA damage: unravelling roles of p53 and ABCG2. Cell Prolif 2016; 49:523-38. [PMID: 27389299 DOI: 10.1111/cpr.12274] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/13/2016] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES In spite of high sensitivity of A549 cells (p53(+/+) ) to lethal effects of photodynamic therapy with 5-aminolaevulinic acid (5-ALA/PDT), DNA damage was observed only in H1299 cells (p53(-/-) ), suggesting that p53 may exert a protective effect. Studies on human colon adenocarcinoma cell lines HCT-116, and their cognate knockouts for p53, were not entirely consistent with the assumption above. Exploring alternative explanations for such conflicting behaviour, we observed that expression of the ATP-binding cassette G2 (ABCG2), a regulator of cell component efflux, had important effects on PDT-generated DNA injury in PC3 cells (prostate) which are p53(-/-) and positive for ABCG2. Addition of an ABCG2 inhibitor in ABCG2 positive A549 (p53(+/+) ) and PC3 (p53(-/-) ) cells eliminated resistance to DNA damage. MATERIALS AND METHODS All cell lines investigated were incubated with 5-ALA and irradiated. Effects of PDT were evaluated assessing residual cell viability, cell-cycle profiles, PpIX localization, comet assay and Western blotting. Identical measurements were made in the presence of ABCG2 inhibitor, in cells expressing the transporter. RESULTS Our data show that cell aptitude to defend its DNA from PDT-induced injury was mainly ruled by ABCG2 expression. These findings, while providing helpful information in predicting effectiveness of 5-ALA/PDT, may indicate a way to shift PDT from a palliative to a more effective approach in anti-cancer therapy.
Collapse
Affiliation(s)
- I Postiglione
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - F Barra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - S M Aloj
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - G Palumbo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
261
|
Ng CH, Chan CW, Lai JW, Ooi IH, Chong KV, Maah MJ, Seng HL. Enantiomeric pair of copper(II) polypyridyl-alanine complexes: Effect of chirality on their interaction with biomolecules. J Inorg Biochem 2016; 160:1-11. [DOI: 10.1016/j.jinorgbio.2016.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 03/11/2016] [Accepted: 04/03/2016] [Indexed: 10/22/2022]
|
262
|
Zhang Z, Wang H, Wang Q, Yan M, Wang H, Bi C, Sun S, Fan Y. Anticancer activity and computational modeling of ternary copper (II) complexes with 3-indolecarboxylic acid and 1,10-phenanthroline. Int J Oncol 2016; 49:691-9. [DOI: 10.3892/ijo.2016.3542] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/16/2016] [Indexed: 11/06/2022] Open
|
263
|
Zhu W, Zhan D, Wang L, Ma D, Cheng M, Wang H, Zhao J, Cai Y, Cheng Z. Proteasome inhibitor MG132 potentiates TRAIL-induced apoptosis in gallbladder carcinoma GBC-SD cells via DR5-dependent pathway. Oncol Rep 2016; 36:845-52. [PMID: 27277541 DOI: 10.3892/or.2016.4839] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/08/2016] [Indexed: 11/06/2022] Open
Abstract
TRAIL is a tumor-selective apoptosis-inducing cytokine playing a vital role in the surveillance and elimination of some tumor cells. However, some tumors are resistant to TRAIL treatment. Proteasome inhibitor MG132 exhibits anti-proliferative and pro-apoptotic properties in many tumors. In this study, we demonstrated that proteasome inhibitor MG132 in vitro and in vivo potentiates TRAIL-induced apoptosis in gallbladder carcinoma GBC-SD cells. MG132 was able to inhibit the proliferation of GBC-SD cells and induce apoptosis in a dose-dependent manner. The induction of apoptosis by proteasome inhibitor MG132 was mainly through the extrinsic apoptotic pathways of caspase activation such as caspase-8, caspase-3 and PARP cleavage. In addition, this process was also dependent on the upregulation of death receptor 5 (DR5), which promoted TRAIL-induced apoptosis in GBC-SD cells. Taken together, these findings indicate that MG132 possesses anti-gallbladder cancer potential that correlate with regulation of DR5-dependent pathway, and suggest that MG132 may be a promising agent for sensitizing GBC-SD cells to TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- Weiping Zhu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Dihua Zhan
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Dening Ma
- Department of Liver Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Mingrong Cheng
- Department of General Surgery, Pudong New Area District Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Huipeng Wang
- Department of General Surgery, The Affiliated Shanghai Fifth People's Hospital of Fudan University, Shanghai 200240, P.R. China
| | - Jiaying Zhao
- Department of General Surgery, The Affiliated Shanghai Fifth People's Hospital of Fudan University, Shanghai 200240, P.R. China
| | - Yuankun Cai
- Department of General Surgery, The Affiliated Shanghai Fifth People's Hospital of Fudan University, Shanghai 200240, P.R. China
| | - Zhijian Cheng
- Department of General Surgery, The Affiliated Shanghai Fifth People's Hospital of Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
264
|
Next-generation proteasome inhibitor MLN9708 sensitizes breast cancer cells to doxorubicin-induced apoptosis. Sci Rep 2016; 6:26456. [PMID: 27217076 PMCID: PMC4877646 DOI: 10.1038/srep26456] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 04/29/2016] [Indexed: 12/26/2022] Open
Abstract
Doxorubicin (Dox), one of the most effective chemotherapy drug for cancer treatment, is limited by its severe side effects and chemoresistance. Dox induces DNA damage and leads to significant proteomic changes in the cancer cells, which makes the ubiquitin-proteasome system a potential target to enhance the efficacy of Dox therapy. The unsuccessful clinical trials of proteasome inhibitor PS-341 (bortezomib) in solid tumors led to the invention of MLN9708 (ixazomib), an orally bioavailable next-generation proteasome inhibitor with improved pharmacokinetic and pharmacodynamic features. In this preclinical study, we used eight human breast cancer cell lines, which represent the major molecular subtypes of breast cancer, to validate the cytotoxic effects of MLN9708, alone and in combination with Dox. We found that MLN9708 had cytotoxic effects, induced autophagy and MKP-1 expression, and enhanced Dox-induced apoptosis in these cell lines. MLN9708 also enhanced Dox-induced JNK and p38 phosphorylation and inhibited Dox-induced IκBα degradation. Our in vitro results suggest that MLN9708 has antitumor effects in breast cancer and can sensitize breast cancer cells to Dox treatment. This promising combination may be an effective and feasible therapeutic option for treating breast cancer and warrants clinical validation.
Collapse
|
265
|
Matsuki-Muramoto Y, Nozawa K, Uomori K, Sekigawa I, Takasaki Y. Bortezomib treatment prevents glomerulosclerosis associated with lupus nephritis in a murine model through suppressive effects on the immune and renin-angiotensin systems. Mod Rheumatol 2016; 27:77-86. [PMID: 27166507 DOI: 10.3109/14397595.2016.1170957] [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: 01/01/2023]
Abstract
OBJECTIVE To clarify the mechanisms underlying lupus nephritis (LN) amelioration following bortezomib treatment. METHODS Bortezomib was administered subcutaneously every 3 days to NZB/W F1 mice, and the serum anti-double stranded (ds) deoxyribonucleic acid (DNA) antibody titers and proteinuria levels were measured. The renal samples and the splenocytes were examined histologically or used for real-time quantitative reverse transcription-polymerase chain reaction analysis after 18 weeks of treatment. Serum cytokine and anti-dsDNA antibody levels were measured using flow cytometry and enzyme-linked immunoassays every 3 weeks. Transforming growth factor (TGF)-β, angiotensin II type-1 receptor (AT1R), and type I collagen expression levels in the glomeruli were evaluated using immunohistochemistry. RESULTS Bortezomib reduced the serum anti-dsDNA antibody titers and the proteinuria levels. It prevented inflammatory cell infiltrations into and the deposition of immunoglobulin G within the glomeruli. Bortezomib reduced the interferon-γ, interleukin (IL)-4, and IL-10 levels in the serum and the ribonucleic acid expression levels for these cytokines within the splenocytes. Bortezomib prevented type I collagen synthesis by downregulating TGF-β and AT1R expression in the glomeruli. CONCLUSIONS Bortezomib exerts multiple immunosuppressive effects and thus ameliorates LN. Furthermore, bortezomib can prevent glomerulosclerosis formation in NZB/W F1 mice through suppressive effects on the renin-angiotensin system.
Collapse
Affiliation(s)
- Yuko Matsuki-Muramoto
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| | - Kazuhisa Nozawa
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| | - Kaori Uomori
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| | - Iwao Sekigawa
- b Department of Internal Medicine and Rheumatology , Juntendo University Urayasu Hospital , Chiba , Japan , and.,c Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan
| | - Yoshinari Takasaki
- a Department of Rheumatology , Juntendo University School of Medicine , Tokyo , Japan
| |
Collapse
|
266
|
Song Y, Ray A, Li S, Das DS, Tai YT, Carrasco RD, Chauhan D, Anderson KC. Targeting proteasome ubiquitin receptor Rpn13 in multiple myeloma. Leukemia 2016; 30:1877-86. [PMID: 27118409 DOI: 10.1038/leu.2016.97] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/01/2016] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Abstract
Proteasome inhibitor bortezomib is an effective therapy for relapsed and newly diagnosed multiple myeloma (MM); however, dose-limiting toxicities and the development of resistance can limit its long-term utility. Recent research has focused on targeting ubiquitin receptors upstream of 20S proteasome, with the aim of generating less toxic therapies. Here we show that 19S proteasome-associated ubiquitin receptor Rpn13 is more highly expressed in MM cells than in normal plasma cells. Rpn13-siRNA (small interfering RNA) decreases MM cell viability. A novel agent RA190 targets Rpn13 and inhibits proteasome function, without blocking the proteasome activity or the 19S deubiquitylating activity. CRISPR/Cas9 Rpn13-knockout demonstrates that RA190-induced activity is dependent on Rpn13. RA190 decreases viability in MM cell lines and patient MM cells, inhibits proliferation of MM cells even in the presence of bone marrow stroma and overcomes bortezomib resistance. Anti-MM activity of RA190 is associated with induction of caspase-dependent apoptosis and unfolded protein response-related apoptosis. MM xenograft model studies show that RA190 is well tolerated, inhibits tumor growth and prolongs survival. Combining RA190 with bortezomib, lenalidomide or pomalidomide induces synergistic anti-MM activity. Our preclinical data validates targeting Rpn13 to overcome bortezomib resistance, and provides the framework for clinical evaluation of Rpn13 inhibitors, alone or in combination, to improve patient outcome in MM.
Collapse
Affiliation(s)
- Y Song
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - A Ray
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - S Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - D S Das
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y T Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - R D Carrasco
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D Chauhan
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - K C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
267
|
Proteasome inhibitor-adapted myeloma cells are largely independent from proteasome activity and show complex proteomic changes, in particular in redox and energy metabolism. Leukemia 2016; 30:2198-2207. [PMID: 27118406 PMCID: PMC5097071 DOI: 10.1038/leu.2016.102] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/21/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022]
Abstract
Adaptive resistance of myeloma to proteasome inhibition represents a clinical challenge, whose biology is poorly understood. Proteasome mutations were implicated as underlying mechanism, while an alternative hypothesis based on low activation status of the unfolded protein response was recently suggested (IRE1/XBP1-low model). We generated bortezomib- and carfilzomib-adapted, highly resistant multiple myeloma cell clones (AMO-BTZ, AMO-CFZ), which we analyzed in a combined quantitative and functional proteomic approach. We demonstrate that proteasome inhibitor-adapted myeloma cells tolerate subtotal proteasome inhibition, irrespective of a proteasome mutation, and uniformly show an 'IRE1/XBP1-low' signature. Adaptation of myeloma cells to proteasome inhibitors involved quantitative changes in >600 protein species with similar patterns in AMO-BTZ and AMO-CFZ cells: proteins involved in metabolic regulation, redox homeostasis, and protein folding and destruction were upregulated, while apoptosis and transcription/translation were downregulated. The quantitatively most upregulated protein in AMO-CFZ cells was the multidrug resistance protein (MDR1) protein ABCB1, and carfilzomib resistance could be overcome by MDR1 inhibition. We propose a model where proteasome inhibitor-adapted myeloma cells tolerate subtotal proteasome inhibition owing to metabolic adaptations that favor the generation of reducing equivalents, such as NADPH, which is supported by oxidative glycolysis. Proteasome inhibitor resistance may thus be targeted by manipulating the energy and redox metabolism.
Collapse
|
268
|
Mehdad A, Xavier Reis G, Souza AA, Barbosa JARG, Ventura MM, de Freitas SM. A Bowman-Birk inhibitor induces apoptosis in human breast adenocarcinoma through mitochondrial impairment and oxidative damage following proteasome 20S inhibition. Cell Death Discov 2016; 2:15067. [PMID: 27551492 PMCID: PMC4979482 DOI: 10.1038/cddiscovery.2015.67] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/19/2015] [Accepted: 12/03/2015] [Indexed: 12/29/2022] Open
Abstract
Proteasome inhibitors are emerging as a new class of chemopreventive agents and have gained huge importance as potential pharmacological tools in breast cancer treatment. Improved understanding of the role played by proteases and their specific inhibitors in humans offers novel and challenging opportunities for preventive and therapeutic intervention. In this study, we demonstrated that the Bowman-Birk protease inhibitor from Vigna unguiculata seeds, named black-eyed pea trypsin/chymotrypsin Inhibitor (BTCI), potently suppresses human breast adenocarcinoma cell viability by inhibiting the activity of proteasome 20S. BTCI induced a negative growth effect against a panel of breast cancer cells, with a concomitant cytostatic effect at the G2/M phase of the cell cycle and an increase in apoptosis, as observed by an augmented number of cells at the sub-G1 phase and annexin V-fluorescin isothiocyanate (FITC)/propidium iodide (PI) staining. In contrast, BTCI exhibited no cytotoxic effect on normal mammary epithelial cells. Moreover, the increased levels of intracellular reactive oxygen species (ROS) and changes in the mitochondrial membrane potential in cells treated with BTCI indicated mitochondrial damage as a crucial cellular event responsible for the apoptotic process. The higher activity of caspase in tumoral cells treated with BTCI in comparison with untreated cells suggests that BTCI induces apoptosis in a caspase-dependent manner. BTCI affected NF-kB target gene expression in both non invasive and invasive breast cancer cell lines, with the effect highly pronounced in the invasive cells. An increased expression of interleukin-8 (IL-8) in both cell lines was also observed. Taken together, these results suggest that BTCI promotes apoptosis through ROS-induced mitochondrial damage following proteasome inhibition. These findings highlight the pharmacological potential and benefit of BTCI in breast cancer treatment.
Collapse
Affiliation(s)
- A Mehdad
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - Giselle Xavier Reis
- Faculty of Medicine, Department of Molecular
Pathology, University of Brasilia, Brasilia, Brazil
| | - AA Souza
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - JARG Barbosa
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - MM Ventura
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - SM de Freitas
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| |
Collapse
|
269
|
Boland K, Flanagan L, McCawley N, Pabari R, Kay EW, McNamara DA, Murray F, Byrne AT, Ramtoola Z, Concannon CG, Prehn JHM. Targeting the 19S proteasomal subunit, Rpt4, for the treatment of colon cancer. Eur J Pharmacol 2016; 780:53-64. [PMID: 26997367 DOI: 10.1016/j.ejphar.2016.03.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 11/20/2022]
Abstract
Deregulation of the ubiquitin-proteasome pathway has been frequently observed in a number of malignancies. Using quantitative Western blotting of normal and matched tumour tissue, we here identified a significant increase in the 19S proteasome subunit Rpt4 in response to chemoradiation in locally advanced rectal cancer patients with unfavourable outcome. We therefore explored the potential of Rpt4 reduction as a therapeutic strategy in colorectal cancer (CRC). Utilizing siRNA to down regulate Rpt4 expression, we show that silencing of Rpt4 reduced proteasomal activity and induced endoplasmic reticulum stress. Gene silencing of Rpt4 also inhibited cell proliferation, reduced clonogenic survival and induced apoptosis in HCT-116 colon cancer cells. We next developed a cell penetrating peptide-based nanoparticle delivery system to achieve in vivo gene silencing of Rpt4. Administration of Rpt4 siRNA nanoparticles reduced tumour growth and improved survival in a HCT-116 colon cancer xenograft tumour model in vivo. Collectively, our data suggest that inhibition of Rpt4 represents a novel strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Karen Boland
- Centre for Systems Medicine and Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland; Department of Gastroenterology, Beaumont Hospital, Beaumont, Dublin 9, Ireland
| | - Lorna Flanagan
- Centre for Systems Medicine and Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Niamh McCawley
- Centre for Systems Medicine and Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland; Department of Surgery, Beaumont Hospital, Beaumont, Dublin 9, Ireland
| | - Ritesh Pabari
- School of Pharmacy, Royal College of Surgeons in Ireland, York House, York Street, Dublin 2, Ireland
| | - Elaine W Kay
- Department of Pathology, Beaumont Hospital, Beaumont, Dublin 9, Ireland
| | | | - Frank Murray
- Department of Gastroenterology, Beaumont Hospital, Beaumont, Dublin 9, Ireland
| | - Annette T Byrne
- Centre for Systems Medicine and Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Zebunnissa Ramtoola
- School of Pharmacy, Royal College of Surgeons in Ireland, York House, York Street, Dublin 2, Ireland
| | - Caoimhín G Concannon
- Centre for Systems Medicine and Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Jochen H M Prehn
- Centre for Systems Medicine and Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
270
|
Scaffold protein FHL2 facilitates MDM2-mediated degradation of IER3 to regulate proliferation of cervical cancer cells. Oncogene 2016; 35:5106-18. [PMID: 26973248 PMCID: PMC5399145 DOI: 10.1038/onc.2016.54] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 12/29/2015] [Accepted: 02/08/2016] [Indexed: 12/15/2022]
Abstract
The expression of immediate early response 3 (IER3), a protein with a short half-life, is rapidly induced by various cellular stimuli. We recently reported that IER3 induces the apoptosis of cervical cancer cells and that its expression is downregulated in patients with cervical cancer. However, the molecular mechanism involved in the rapid degradation of IER3 remains unknown. Here, we demonstrate that MDM2 is an E3 ligase that interacts with IER3 and promotes its ubiquitination, followed by proteasomal degradation. Polyubiquitination of the conserved lysine 60 of IER3 is essential for its degradation. In addition, four and a half LIM domains protein 2 (FHL2) binds to both IER3 and MDM2, allowing for efficient MDM2-mediated IER3 degradation by facilitating an association between MDM2 and IER3. Moreover, IER3 induces cell cycle arrest in cervical cancer cells and its activity is further enhanced in cells in which FHL2 or MDM2 was silenced, thereby preventing IER3 degradation. The E6 and E7 oncoproteins of human papilloma virus 18 regulated IER3 expression. FHL2 expression was significantly higher in the squamous epithelium of cervical carcinoma tissues than in non-cancerous cervical tissues, whereas cervical carcinoma expression of IER3 was downregulated in this region. Thus, we determined the molecular mechanism responsible for IER3 degradation, involving a ternary complex of IER3, MDM2 and FHL2, which may contribute to cervical tumor growth. Furthermore, we demonstrated that FHL2 serves as a scaffold for E3 ligase and its substrate during the ubiquitination reaction, a function that has not been previously reported for this protein.
Collapse
|
271
|
Wilson DL, Meininger I, Strater Z, Steiner S, Tomlin F, Wu J, Jamali H, Krappmann D, Götz MG. Synthesis and Evaluation of Macrocyclic Peptide Aldehydes as Potent and Selective Inhibitors of the 20S Proteasome. ACS Med Chem Lett 2016; 7:250-5. [PMID: 26985310 DOI: 10.1021/acsmedchemlett.5b00401] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/13/2016] [Indexed: 12/31/2022] Open
Abstract
This research explores the first design and synthesis of macrocyclic peptide aldehydes as potent inhibitors of the 20S proteasome. Two novel macrocyclic peptide aldehydes based on the ring-size of the macrocyclic natural product TMC-95 were prepared and evaluated as inhibitors of the 20S proteasome. Both compounds inhibited in the low nanomolar range and proved to be selective for the proteasome over other serine and cysteine proteases, particularly when compared to linear analogues with similar amino acid sequences. In HeLa cells, both macrocycles efficiently inhibited activation of nuclear factor-κB (NF-κB) transcription factor by blocking proteasomal degradation of the inhibitor protein IκBα after cytokine stimulation. Due to their covalent mechanism of binding these compounds represent a 1000-fold increase in inhibitory potency over previously reported noncovalently binding TMC-95 analogues. Molecular modeling of the macrocyclic peptides confirms the preference of the large S3 pocket for large, hydrophobic residues and the ability to exploit this to improve selectivity of proteasome inhibitors.
Collapse
Affiliation(s)
- David L. Wilson
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Isabel Meininger
- Research
Unit Cellular Signal Integration, Institute of Molecular Toxicology
and Pharmacology, Helmholtz Zentrum München−German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Zack Strater
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Stephanie Steiner
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Frederick Tomlin
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Julia Wu
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Haya Jamali
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Daniel Krappmann
- Research
Unit Cellular Signal Integration, Institute of Molecular Toxicology
and Pharmacology, Helmholtz Zentrum München−German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Marion G. Götz
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| |
Collapse
|
272
|
Wei D, Tang M, Zhan CG. Fundamental reaction pathway and free energy profile of proteasome inhibition by syringolin A (SylA). Org Biomol Chem 2016; 13:6857-65. [PMID: 26018983 DOI: 10.1039/c5ob00737b] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, molecular dynamics (MD) simulations and first-principles quantum mechanical/molecular mechanical free energy (QM/MM-FE) calculations have been performed to uncover the fundamental reaction pathway of proteasome with a representative inhibitor syringolin A (SylA). The calculated results reveal that the reaction process consists of three steps. The first step is a proton transfer process, activating Thr1-O(γ) directly by Thr1-N(z) to form a zwitterionic intermediate. The next step is a nucleophilic attack on the olefin carbon of SylA by the negatively charged Thr1-O(γ) atom. The last step is a proton transfer from Thr1-N(z) to another olefin carbon of SylA to complete the inhibition reaction process. The calculated free energy profile demonstrates that the second step should be the rate-determining step and has the highest free energy barrier of 24.6 kcal mol(-1), which is reasonably close to the activation free energy (∼22.4-23.0 kcal mol(-1)) derived from the available experimental kinetic data. In addition, our computational results indicate that no water molecule can assist the rate-determining step, since the second step is not involved in a proton transfer process. The obtained mechanistic insights should be valuable for understanding the inhibition process of proteasome by SylA and structurally related inhibitors at a molecular level, and thus provide a solid mechanistic base and valuable clues for future rational design of novel, more potent inhibitors of proteasome.
Collapse
Affiliation(s)
- Donghui Wei
- Department of Chemistry, Zhengzhou University, Daxue Road, Zhengzhou, Henan 450052, China
| | | | | |
Collapse
|
273
|
Rahme GJ, Zhang Z, Young AL, Cheng C, Bivona EJ, Fiering SN, Hitoshi Y, Israel MA. PDGF Engages an E2F-USP1 Signaling Pathway to Support ID2-Mediated Survival of Proneural Glioma Cells. Cancer Res 2016; 76:2964-76. [DOI: 10.1158/0008-5472.can-15-2157] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/11/2016] [Indexed: 11/16/2022]
|
274
|
Upregulation of CCL2 via ATF3/c-Jun interaction mediated the Bortezomib-induced peripheral neuropathy. Brain Behav Immun 2016; 53:96-104. [PMID: 26554515 DOI: 10.1016/j.bbi.2015.11.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/27/2015] [Accepted: 11/07/2015] [Indexed: 11/22/2022] Open
Abstract
Bortezomib (BTZ) is a frequently used chemotherapeutic drug for the treatment of refractory multiple myeloma and hematological neoplasms. The mechanism by which the administration of BTZ leads to painful peripheral neuropathy remains unclear. In present study, we found that application of BTZ at 0.4 mg/kg for consecutive 5 days significantly increased the expression of CCL2 in DRG, and intrathecal administration of neutralizing antibody against CCL2 inhibited the mechanical allodynia induced by BTZ. We also found an increased expression of c-Jun in DRG, and that inhibition of c-Jun signaling prevented the CCL2 upregulation and mechanical allodynia in the rats treated with BTZ. Furthermore, the results with luciferase assay in vitro and ChIP assay in vivo showed that c-Jun might be essential for BTZ-induced CCL2 upregulation via binding directly to the specific position of the ccl2 promoter. In addition, the present results showed that an upregulated expression of ATF3 was co-expressed with c-Jun in the DRG neurons, and the enhanced interaction between c-Jun and ATF3 was observed in DRG in the rats treated with BTZ. Importantly, pretreatment with ATF3 siRNA significantly inhibited the recruitment of c-Jun to the ccl2 promoter in the rats treated with BTZ. Taken together, these findings suggested that upregulation of CCL2 resulting from the enhanced interaction between c-Jun and ATF3 in DRG contributed to BTZ-induced mechanical allodynia.
Collapse
|
275
|
Infante JR, Mendelson DS, Burris HA, Bendell JC, Tolcher AW, Gordon MS, Gillenwater HH, Arastu-Kapur S, Wong HL, Papadopoulos KP. A first-in-human dose-escalation study of the oral proteasome inhibitor oprozomib in patients with advanced solid tumors. Invest New Drugs 2016; 34:216-24. [DOI: 10.1007/s10637-016-0327-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
|
276
|
Bachmann AS, Opoku-Ansah J, Ibarra-Rivera TR, Yco LP, Ambadi S, Roberts CC, Chang CEA, Pirrung MC. Syrbactin Structural Analog TIR-199 Blocks Proteasome Activity and Induces Tumor Cell Death. J Biol Chem 2016; 291:8350-62. [PMID: 26907687 DOI: 10.1074/jbc.m115.710053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Indexed: 01/19/2023] Open
Abstract
Multiple myeloma is an aggressive hematopoietic cancer of plasma cells. The recent emergence of three effective FDA-approved proteasome-inhibiting drugs, bortezomib (Velcade®), carfilzomib (Kyprolis®), and ixazomib (Ninlaro®), confirms that proteasome inhibitors are therapeutically useful against neoplastic disease, in particular refractory multiple myeloma and mantle cell lymphoma. This study describes the synthesis, computational affinity assessment, and preclinical evaluation of TIR-199, a natural product-derived syrbactin structural analog. Molecular modeling and simulation suggested that TIR-199 covalently binds each of the three catalytic subunits (β1, β2, and β5) and revealed key interaction sites. In vitro and cell culture-based proteasome activity measurements confirmed that TIR-199 inhibits the proteasome in a dose-dependent manner and induces tumor cell death in multiple myeloma and neuroblastoma cells as well as other cancer types in the NCI-60 cell panel. It is particularly effective against kidney tumor cell lines, with >250-fold higher anti-tumor activities than observed with the natural product syringolin A. In vivo studies in mice revealed a maximum tolerated dose of TIR-199 at 25 mg/kg. The anti-tumor activity of TIR-199 was confirmed in hollow fiber assays in mice. Adverse drug reaction screens in a kidney panel revealed no off-targets of concern. This is the first study to examine the efficacy of a syrbactin in animals. Taken together, the results suggest that TIR-199 is a potent new proteasome inhibitor with promise for further development into a clinical drug for the treatment of multiple myeloma and other forms of cancer.
Collapse
Affiliation(s)
- André S Bachmann
- From the Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, the Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii, Hilo, Hawaii 96720, the Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, Hawaii 96822,
| | - John Opoku-Ansah
- the Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii, Hilo, Hawaii 96720
| | | | - Lisette P Yco
- From the Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, the Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii, Hilo, Hawaii 96720, the Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, Hawaii 96822
| | - Sudhakar Ambadi
- the Department of Chemistry, University of California, Riverside, California 92521, and
| | - Christopher C Roberts
- the Department of Chemistry, University of California, Riverside, California 92521, and
| | - Chia-En A Chang
- the Department of Chemistry, University of California, Riverside, California 92521, and
| | - Michael C Pirrung
- the Department of Chemistry, University of California, Riverside, California 92521, and the Department of Pharmaceutical Sciences, University of California, Irvine, California 92697
| |
Collapse
|
277
|
Liu Z, Sneve M, Haroldson TA, Smith JP, Drewes LR. Regulation of Monocarboxylic Acid Transporter 1 Trafficking by the Canonical Wnt/β-Catenin Pathway in Rat Brain Endothelial Cells Requires Cross-talk with Notch Signaling. J Biol Chem 2016; 291:8059-69. [PMID: 26872974 DOI: 10.1074/jbc.m115.710277] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 12/20/2022] Open
Abstract
The transport of monocarboxylate fuels such as lactate, pyruvate, and ketone bodies across brain endothelial cells is mediated by monocarboxylic acid transporter 1 (MCT1). Although the canonical Wnt/β-catenin pathway is required for rodent blood-brain barrier development and for the expression of associated nutrient transporters, the role of this pathway in the regulation of brain endothelial MCT1 is unknown. Here we report expression of nine members of the frizzled receptor family by the RBE4 rat brain endothelial cell line. Furthermore, activation of the canonical Wnt/β-catenin pathway in RBE4 cells via nuclear β-catenin signaling with LiCl does not alter brain endothelialMct1mRNA but increases the amount of MCT1 transporter protein. Plasma membrane biotinylation studies and confocal microscopic examination of mCherry-tagged MCT1 indicate that increased transporter results from reduced MCT1 trafficking from the plasma membrane via the endosomal/lysosomal pathway and is facilitated by decreased MCT1 ubiquitination following LiCl treatment. Inhibition of the Notch pathway by the γ-secretase inhibitorN-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycinet-butyl ester negated the up-regulation of MCT1 by LiCl, demonstrating a cross-talk between the canonical Wnt/β-catenin and Notch pathways. Our results are important because they show, for the first time, the regulation of MCT1 in cerebrovascular endothelial cells by the multifunctional canonical Wnt/β-catenin and Notch signaling pathways.
Collapse
Affiliation(s)
- Zejian Liu
- From the Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, Minnesota 55812 and
| | - Mary Sneve
- From the Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, Minnesota 55812 and
| | - Thomas A Haroldson
- From the Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, Minnesota 55812 and
| | - Jeffrey P Smith
- the Department of Biology, Colorado State University, Pueblo, Colorado 81001
| | - Lester R Drewes
- From the Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, Minnesota 55812 and
| |
Collapse
|
278
|
Plasma Levels of Monocyte Chemotactic Protein-1 Are Associated with Clinical Features and Angiogenesis in Patients with Multiple Myeloma. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7870590. [PMID: 26925413 PMCID: PMC4748063 DOI: 10.1155/2016/7870590] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/26/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023]
Abstract
The aim of this pilot study was to determine the plasma levels of monocyte chemotactic protein-1 (MCP-1) and possible associations with angiogenesis and the main clinical features of untreated patients with multiple myeloma (MM). ELISA was used to determine plasma MCP-1 levels in 45 newly diagnosed MM patients and 24 healthy controls. The blood vessels were highlighted by immunohistochemical staining, and computer-assisted image analysis was used for more objective and accurate determination of two parameters of angiogenesis: microvessel density (MVD) and total vascular area (TVA). The plasma levels of MCP-1 were compared to these parameters and the presence of anemia, renal dysfunction, and bone lesions. A significant positive correlation was found between plasma MCP-1 concentrations and TVA (p = 0.02). The MCP-1 levels were significantly higher in MM patients with evident bone lesions (p = 0.01), renal dysfunction (p = 0.02), or anemia (p = 0.04). Therefore, our preliminary results found a positive association between plasma MCP-1 levels, angiogenesis (expressed as TVA), and clinical features in patients with MM. However, additional prospective studies with a respectable number of patients should be performed to authenticate these results and establish MCP-1 as a possible target of active treatment.
Collapse
|
279
|
Milone MR, Pucci B, Bifulco K, Iannelli F, Lombardi R, Ciardiello C, Bruzzese F, Carriero MV, Budillon A. Proteomic analysis of zoledronic-acid resistant prostate cancer cells unveils novel pathways characterizing an invasive phenotype. Oncotarget 2016; 6:5324-41. [PMID: 25481874 PMCID: PMC4467152 DOI: 10.18632/oncotarget.2694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/04/2014] [Indexed: 12/22/2022] Open
Abstract
Proteomic analysis identified differentially expressed proteins between zoledronic acid-resistant and aggressive DU145R80 prostate cancer (PCa) cells and their parental DU145 cells. Ingenuity Pathway Analysis (IPA) showed a strong relationship between the identified proteins within a network associated with cancer and with homogeneous cellular functions prevalently related with regulation of cell organization, movement and consistent with the smaller and reduced cell-cell contact morphology of DU145R80 cells. The identified proteins correlated in publically available human PCa genomic data with increased tumor expression and aggressiveness. DU145R80 exhibit also a clear increase of alpha-v-(αv) integrin, and of urokinase receptor (uPAR), both included within the same network of the identified proteins. Interestingly, the actin-rich structures localized at the cell periphery of DU145R80 cells are rich of Filamin A, one of the identified proteins and uPAR which, in turn, co-localizes with αv-integrin, in podosomes and/or invadopodia. Notably, the invasive feature of DU145R80 may be prevented by blocking anti-αv antibody. Overall, we unveil a signaling network that physically links the interior of the nucleus via the cytoskeleton to the extracellular matrix and that could dictate PCa aggressiveness suggesting novel potential prognostic markers and therapeutic targets for PCa patients.
Collapse
Affiliation(s)
- Maria Rita Milone
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Biagio Pucci
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Katia Bifulco
- Neoplastic Progression Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Federica Iannelli
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Rita Lombardi
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Chiara Ciardiello
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Maria Vincenza Carriero
- Neoplastic Progression Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Alfredo Budillon
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy.,Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| |
Collapse
|
280
|
Śledź P, Baumeister W. Structure-Driven Developments of 26S Proteasome Inhibitors. Annu Rev Pharmacol Toxicol 2016; 56:191-209. [DOI: 10.1146/annurev-pharmtox-010814-124727] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Paweł Śledź
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany;
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany;
| |
Collapse
|
281
|
Kanoh N. Photo-cross-linked small-molecule affinity matrix as a tool for target identification of bioactive small molecules. Nat Prod Rep 2016; 33:709-18. [DOI: 10.1039/c5np00117j] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review describes the status of the photo-cross-linked small-molecule affinity matrix while providing a useful tutorial for academic and industrial chemical biologists who are involved or interested in drug target identification.
Collapse
Affiliation(s)
- Naoki Kanoh
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai 980-8578
- Japan
| |
Collapse
|
282
|
Momose I, Kawada M. The therapeutic potential of microbial proteasome inhibitors. Int Immunopharmacol 2015; 37:23-30. [PMID: 26589840 DOI: 10.1016/j.intimp.2015.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 11/16/2022]
Abstract
The proteasome influences cellular homeostasis through the degradation of regulatory proteins, many of which are also involved in disease pathogenesis. In particular, numerous regulatory proteins associated with tumor growth, such as cyclins, cyclin-dependent kinase inhibitors, tumor suppressors, and NF-κB inhibitors are degraded by the proteasome. Proteasome inhibitors can stabilize these regulatory proteins, resulting in the suppression of tumor development and the regulation of immune responses. Thus, proteasome inhibitors are promising candidate antitumor agents and immune-regulatory agents. Bortezomib is the first-in-class proteasome inhibitor approved for the treatment of multiple myeloma. Despite its high efficiency, however, a large proportion of patients do not attain sufficient clinical response due to toxicity and drug resistance. Therefore, the development of new proteasome inhibitors with improved pharmacological properties is needed. Natural products produced by microorganisms are a promising source of such compounds. This review provides an overview of proteasome inhibitors produced by microorganisms, with special focus on inhibitors isolated from actinomycetes.
Collapse
Affiliation(s)
- Isao Momose
- Institute of Microbial Chemistry (BIKAKEN), Numazu, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan.
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan; Institute of Microbial Chemistry (BIKAKEN), Tokyo, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| |
Collapse
|
283
|
Wang Y, Zheng Z, Zhang J, Wang Y, Kong R, Liu J, Zhang Y, Deng H, Du X, Ke Y. A Novel Retinoblastoma Protein (RB) E3 Ubiquitin Ligase (NRBE3) Promotes RB Degradation and Is Transcriptionally Regulated by E2F1 Transcription Factor. J Biol Chem 2015; 290:28200-28213. [PMID: 26442585 DOI: 10.1074/jbc.m115.655597] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Indexed: 12/25/2022] Open
Abstract
Retinoblastoma protein (RB) plays critical roles in tumor suppression and is degraded through the proteasomal pathway. However, E3 ubiquitin ligases responsible for proteasome-mediated degradation of RB are largely unknown. Here we characterize a novel RB E3 ubiquitin ligase (NRBE3) that binds RB and promotes RB degradation. NRBE3 contains an LXCXE motif and bound RB in vitro. NRBE3 interacted with RB in cells when proteasome activity was inhibited. NRBE3 promoted RB ubiquitination and degradation via the ubiquitin-proteasome pathway. Importantly, purified NRBE3 ubiquitinated recombinant RB in vitro, and a U-box was identified as essential for its E3 activity. Surprisingly, NRBE3 was transcriptionally activated by E2F1/DP1. Consequently, NRBE3 affected the cell cycle by promoting G1/S transition. Moreover, NRBE3 was up-regulated in breast cancer tissues. Taken together, we identified NRBE3 as a novel ubiquitin E3 ligase for RB that might play a role as a potential oncoprotein in human cancers.
Collapse
Affiliation(s)
- Yingshuang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| | - Zongfang Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| | - Jingyi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)
| | - You Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| | - Ruirui Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| | - Jiangying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| | - Ying Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| | - Hongkui Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xiaojuan Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Genetics Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
284
|
Liu R, Lu S, Deng Y, Yang S, He S, Cai J, Qiang F, Chen C, Zhang W, Zhao S, Qian L, Mao G, Wang Y. PSMB4 expression associates with epithelial ovarian cancer growth and poor prognosis. Arch Gynecol Obstet 2015; 293:1297-307. [PMID: 26439929 DOI: 10.1007/s00404-015-3904-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/24/2015] [Indexed: 12/30/2022]
Abstract
PURPOSE In this study, we investigated the expression and role of PSMB4 in human epithelial ovarian cancer(EOC). METHODS Western blot was used to evaluate the expression of PSMB4 in EOC tissues, and immunohistochemical analysis was performed on 115 cases of ovarian cancers. Then, we used Fisher exact test to analyze the correlation between PSMB4 and clinicopathological parameters. Starvation and re-feeding assay was used to assess cell cycle. CCK-8 assay and plate colony formation assay showed the influence of PSMB4 on proliferation of EOC cells. RESULTS The expression of PSMB4 in EOC tissues was higher than normal ovary tissues and was significantly associated with clinical pathologic variables. Kaplan-Meier curve showed that high expression of PSMB4 was related to poor prognosis of EOC patients. Starvation and re-feeding assay suggested that PSMB4 played a critical role in EOC cell proliferation. CCK-8 assay and plate colony formation assay showed that EOC cells treated with PSMB4-siRNA reduced cell proliferation of EOC cells. Additionally, PSMB4 knockdown decreased NF-κB activity. PSMB4 also regulated the expression of NF-κB mediated proteins, including cyclin D1, and cyclin E which involved in cell proliferation. CONCLUSIONS Our findings implied that PSMB4 is involved in the progression of EOC and could serve as potential therapeutical target of EOC. These data suggested that PSMB4 may promote cell proliferation via the NF-κB-target gene in EOC.
Collapse
Affiliation(s)
- Rong Liu
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Shumin Lu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Yan Deng
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Shuyun Yang
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Song He
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jing Cai
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Fulin Qiang
- Department of Gynecologic Oncology, Nantong University Cancer Hospital, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Chen Chen
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Weiwei Zhang
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Shuyang Zhao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Li Qian
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Guoxin Mao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China.
| | - Yingying Wang
- Department of Pathogen Biology, Medical College, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, People's Republic of China.
| |
Collapse
|
285
|
Grigoreva TA, Tribulovich VG, Garabadzhiu AV, Melino G, Barlev NA. The 26S proteasome is a multifaceted target for anti-cancer therapies. Oncotarget 2015; 6:24733-49. [PMID: 26295307 PMCID: PMC4694792 DOI: 10.18632/oncotarget.4619] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 07/10/2015] [Indexed: 12/30/2022] Open
Abstract
Proteasomes play a critical role in the fate of proteins that are involved in major cellular processes, including signal transduction, gene expression, cell cycle, replication, differentiation, immune response, cellular response to stress, etc. In contrast to non-specific degradation by lysosomes, proteasomes are highly selective and destroy only the proteins that are covalently labelled with small proteins, called ubiquitins. Importantly, many diseases, including neurodegenerative diseases and cancers, are intimately connected to the activity of proteasomes making them an important pharmacological target. Currently, the vast majority of inhibitors are aimed at blunting the proteolytic activities of proteasomes. However, recent achievements in solving structures of proteasomes at very high resolution provided opportunities to design new classes of small molecules that target other physiologically-important enzymatic activities of proteasomes, including the de-ubiquitinating one. This review attempts to catalog the information available to date about novel classes of proteasome inhibitors that may have important pharmacological ramifications.
Collapse
Affiliation(s)
- Tatyana A Grigoreva
- St. Petersburg State Technological Institute (Technical University), St. Petersburng, Russia
| | | | | | - Gerry Melino
- St. Petersburg State Technological Institute (Technical University), St. Petersburng, Russia
- University of Rome Tor Vergata, Roma, Italy
| | | |
Collapse
|
286
|
Tundo GR, Sbardella D, Ciaccio C, De Pascali S, Campanella V, Cozza P, Tarantino U, Coletta M, Fanizzi FP, Marini S. Effect of cisplatin on proteasome activity. J Inorg Biochem 2015; 153:253-258. [PMID: 26387966 DOI: 10.1016/j.jinorgbio.2015.08.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/26/2015] [Accepted: 08/28/2015] [Indexed: 02/01/2023]
Abstract
Cisplatin is a widely used chemotherapy drug which exerts cytotoxic activity by affecting both nuclear and cytosolic pathways. Herewith, we report, for the first time, that cisplatin inhibits proteasome activity in vitro. Cisplatin induces a dose dependent inhibition of the three enzymatic activities of proteasome (i.e., the chymotrypsin-like activity, the trypsin-like activity and the caspase-like activity). Moreover, cisplatin administration to neuroblastoma cells brings about a fast loss of proteasome particle activity, which is followed by a de novo synthesis of proteasome. Lastly, we report that the simultaneous administration of lactacystin and cisplatin enhances the cytotoxicity of cisplatin alone. The overall bulk of data opens to an intriguing scenario, concerning the biological effects of cisplatin in the control of cellular life, which goes beyond the well established genotoxic effect.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy; CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy
| | - D Sbardella
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy; CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy
| | - C Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy; CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy
| | - S De Pascali
- CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy; Department of Environmental Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - V Campanella
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy
| | - P Cozza
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy
| | - U Tarantino
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy; Center for Space Biomedicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy; CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy; Center for Space Biomedicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy
| | - F P Fanizzi
- CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy; Department of Environmental Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy; CIRCMSB, Via C. Ulpiani 27, I-70125 Bari, Italy; Center for Space Biomedicine, University of Roma Tor Vergata, Via Montpellier 1, I-00133 Roma, Italy.
| |
Collapse
|
287
|
NF-κB signaling in cancer stem cells: a promising therapeutic target? Cell Oncol (Dordr) 2015; 38:327-39. [PMID: 26318853 DOI: 10.1007/s13402-015-0236-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are regulated by several signaling pathways that ultimately control their maintenance and expansion. NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) forms a protein complex that controls DNA transcription and, as such, plays an important role in proliferation, inflammation, angiogenesis, invasion and metastasis. The NF-κB signaling pathway, which has been found to be constitutively activated in CSCs from a variety of cancers, participates in the maintenance, expansion, proliferation and survival of CSCs. Targeted disruption of this pathway may profoundly impair the adverse phenotype of CSCs and may provide a therapeutic opportunity to remove the CSC fraction. In particular, it may be attractive to use specific NF-κB inhibitors in chronic therapeutic schemes to reduce disease progression. Exceptional low toxicity profiles of these inhibitors are a prerequisite for use in combined treatment regimens and to avoid resistance. CONCLUSION Although still preliminary, recent evidence shows that such targeted strategies may be useful in adjuvant chemo-preventive settings.
Collapse
|
288
|
Postiglione I, Chiaviello A, Barra F, Roscetto E, Soriano AA, Catania MR, Palumbo G, Pierantoni GM. Mitochondrial Malfunctioning, Proteasome Arrest and Apoptosis in Cancer Cells by Focused Intracellular Generation of Oxygen Radicals. Int J Mol Sci 2015; 16:20375-91. [PMID: 26343643 PMCID: PMC4613209 DOI: 10.3390/ijms160920375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/10/2015] [Accepted: 08/21/2015] [Indexed: 12/29/2022] Open
Abstract
Photofrin/photodynamic therapy (PDT) at sub-lethal doses induced a transient stall in proteasome activity in surviving A549 (p53+/+) and H1299 (p53−/−) cells as indicated by the time-dependent decline/recovery of chymotrypsin-like activity. Indeed, within 3 h of incubation, Photofrin invaded the cytoplasm and localized preferentially within the mitochondria. Its light activation determined a decrease in mitochondrial membrane potential and a reversible arrest in proteasomal activity. A similar result is obtained by treating cells with Antimycin and Rotenone, indicating, as a common denominator of this effect, the ATP decrease. Both inhibitors, however, were more toxic to cells as the recovery of proteasomal activity was incomplete. We evaluated whether combining PDT (which is a treatment for killing tumor cells, per se, and inducing proteasome arrest in the surviving ones) with Bortezomib doses capable of sustaining the stall would protract the arrest with sufficient time to induce apoptosis in remaining cells. The evaluation of the mitochondrial membrane depolarization, residual proteasome and mitochondrial enzymatic activities, colony-forming capabilities, and changes in protein expression profiles in A549 and H1299 cells under a combined therapeutic regimen gave results consistent with our hypothesis.
Collapse
Affiliation(s)
- Ilaria Postiglione
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Angela Chiaviello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Federica Barra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Emanuela Roscetto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Amata A Soriano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Maria Rosaria Catania
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Giuseppe Palumbo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
| | - Giovanna Maria Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy.
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Naples 80131, Italy.
| |
Collapse
|
289
|
Larsen JT, Kumar S. Evolving Paradigms in the Management of Multiple Myeloma: Novel Agents and Targeted Therapies. ACTA ACUST UNITED AC 2015; 3:47-68. [PMID: 27182478 PMCID: PMC4837942 DOI: 10.1007/s40487-015-0009-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma cell disorder defined by bone marrow infiltration and osteolytic bone lesions and is the second most common hematologic malignancy after non-Hodgkin lymphoma. The landscape of MM treatment was transformed at the dawn of the twenty-first century by the introduction of novel agents including proteasome inhibitors (bortezomib) and immunomodulatory drugs (thalidomide, lenalidomide), which have prolonged the survival of MM patients. The recently revised International Myeloma Working Group diagnostic criteria for MM added validated biomarkers (clonal bone marrow plasma cell ≥60%, involved:uninvolved serum free light chain ratio ≥100, or >1 focal lesion on magnetic resonance imaging) to identify near inevitable progression to symptomatic MM requiring therapy. In addition, the definition of myeloma-defining CRAB features (hypercalcemia, renal failure, anemia, and bone lesions) has been refined based on advances in imaging and laboratory techniques since the 2003 IMWG consensus. Despite expanded treatment options, MM remains an incurable disease. Drug resistance and clonal evolution remain problematic, and novel therapeutic agents are needed. New approaches to myeloma treatment include anti-CD38 antibodies, next generation proteasome inhibitors, epigenetic modulation with histone deacetylase inhibitors, and targeting the tumor microenvironment. In this article, the diagnosis, staging, and prognostic stratification of newly diagnosed MM will be reviewed. Clinical data pertaining to the emerging targeted agents will be discussed, and a suggested framework for integration of these new therapeutic options will be provided.
Collapse
Affiliation(s)
- Jeremy T Larsen
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN USA
| | - Shaji Kumar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
290
|
HUANG WEIYI, ZHONG WEIQING, XU JUN, SU BENHUA, HUANG GUANGHUI, DU JIAJUN, LIU QI. Lentivirus-mediated gene silencing of NOB1 suppresses non-small cell lung cancer cell proliferation. Oncol Rep 2015; 34:1510-6. [DOI: 10.3892/or.2015.4132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/28/2015] [Indexed: 11/05/2022] Open
|
291
|
Huang Z, Wu Y, Zhou X, Xu J, Zhu W, Shu Y, Liu P. Efficacy of therapy with bortezomib in solid tumors: a review based on 32 clinical trials. Future Oncol 2015; 10:1795-807. [PMID: 25303058 DOI: 10.2217/fon.14.30] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The ubiquitin-proteasome system is a major pathway for protein degradation, so that proteasome is now considered as an important target for drug discovery. Bortezomib, the first US FDA-approved proteasome inhibitor now used as a front-line treatment for multiple myeloma. To better understand the effects of bortezomib in cancer treatment, we carried out a review based on 32 published clinical trials to determine whether bortezomib will benefit patients with solid tumors. Information of complete response, partial response, stable disease and objective response rate was collected to assess clinical outcomes. A lack of therapeutic effects was observed when bortezomib was used as a single agent. Meanwhile, when bortezomib treatment was combined with other agents, bortezomib offered no statistically significant response versus these agents alone. High-quality studies are required to better understand the clinically effects of bortezomib and the development of a new generation of proteasome inhibitors is absolutely necessary.
Collapse
Affiliation(s)
- Zebo Huang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
292
|
Kraus M, Bader J, Geurink PP, Weyburne ES, Mirabella AC, Silzle T, Shabaneh TB, van der Linden WA, de Bruin G, Haile SR, van Rooden E, Appenzeller C, Li N, Kisselev AF, Overkleeft H, Driessen C. The novel β2-selective proteasome inhibitor LU-102 synergizes with bortezomib and carfilzomib to overcome proteasome inhibitor resistance of myeloma cells. Haematologica 2015; 100:1350-60. [PMID: 26069288 DOI: 10.3324/haematol.2014.109421] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/05/2015] [Indexed: 12/31/2022] Open
Abstract
Proteasome inhibitor resistance is a challenge for myeloma therapy. Bortezomib targets the β5 and β1 activity, but not the β2 activity of the proteasome. Bortezomib-resistant myeloma cells down-regulate the activation status of the unfolded protein response, and up-regulate β2 proteasome activity. To improve proteasome inhibition in bortezomib-resistant myeloma and to achieve more efficient UPR activation, we have developed LU-102, a selective inhibitor of the β2 proteasome activity. LU-102 inhibited the β2 activity in intact myeloma cells at low micromolar concentrations without relevant co-inhibition of β1 and β5 proteasome subunits. In proteasome inhibitor-resistant myeloma cells, significantly more potent proteasome inhibition was achieved by bortezomib or carfilzomib in combination with LU-102, compared to bortezomib/carfilzomib alone, resulting in highly synergistic cytotoxic activity of the drug combination via endoplasmatic reticulum stress-induced apoptosis. Combining bortezomib/carfilzomib with LU-102 significantly prolonged proteasome inhibition and increased activation of the unfolded protein response and IRE1-a activity. IRE1-α has recently been shown to control myeloma cell differentiation and bortezomib sensitivity (Leung-Hagesteijn, Cancer Cell 24:3, 289-304). Thus, β2-selective proteasome inhibition by LU-102 in combination with bortezomib or carfilzomib results in synergistic proteasome inhibition, activation of the unfolded protein response, and cytotoxicity, and overcomes bortezomib/carfilzomib resistance in myeloma cells in vitro.
Collapse
Affiliation(s)
- Marianne Kraus
- Experimental Oncology and Hematology, Department of Oncology and Hematology, Kantonsspital St.Gallen, Switzerland
| | - Juergen Bader
- Experimental Oncology and Hematology, Department of Oncology and Hematology, Kantonsspital St.Gallen, Switzerland
| | - Paul P Geurink
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, the Netherlands
| | - Emily S Weyburne
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Anne C Mirabella
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Tobias Silzle
- Experimental Oncology and Hematology, Department of Oncology and Hematology, Kantonsspital St.Gallen, Switzerland
| | - Tamer B Shabaneh
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Wouter A van der Linden
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, the Netherlands
| | - Gerjan de Bruin
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, the Netherlands
| | - Sarah R Haile
- Clinical Trials Unit, Kantonsspital St. Gallen, Switzerland Department of Hematology, Division of Biostatistics, Institute for Social and Preventive Medicine, University of Zurich, Switzerland
| | - Eva van Rooden
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Christina Appenzeller
- Experimental Oncology and Hematology, Department of Oncology and Hematology, Kantonsspital St.Gallen, Switzerland
| | - Nan Li
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, the Netherlands
| | - Alexei F Kisselev
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Herman Overkleeft
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, the Netherlands
| | - Christoph Driessen
- Experimental Oncology and Hematology, Department of Oncology and Hematology, Kantonsspital St.Gallen, Switzerland Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, the Netherlands
| |
Collapse
|
293
|
Abstract
The goal of this study was to determine whether combined targeted therapies, specifically those against the Notch, hedgehog and ubiquitin-proteasome pathways, could overcome ovarian cancer chemoresistance. Chemoresistant ovarian cancer cells were exposed to gamma-secretase inhibitors (GSI-I, Compound E) or the proteasome inhibitor bortezomib, alone and in combination with the hedgehog antagonist, LDE225. Bortezomib, alone and in combination with LDE225, was evaluated for effects on paclitaxel efficacy. Cell viability and cell cycle analysis were assessed by MTT assay and propidium iodide staining, respectively. Proteasome activity and gene expression were determined by luminescence assay and qPCR, respectively. Studies demonstrated that GSI-I, but not Compound E, inhibited proteasome activity, similar to bortezomib. Proteasome inhibition decreased hedgehog target genes (PTCH1, GLI1 and GLI2) and increased LDE225 sensitivity in vitro. Bortezomib, alone and in combination with LDE225, increased paclitaxel sensitivity through apoptosis and G2/M arrest. Expression of the multi-drug resistance gene ABCB1/MDR1 was decreased and acetylation of α-tubulin, a marker of microtubule stabilization, was increased following bortezomib treatment. HDAC6 inhibitor tubastatin-a demonstrated that microtubule effects are associated with hedgehog inhibition and sensitization to paclitaxel and LDE225. These results suggest that proteasome inhibition, through alteration of microtubule dynamics and hedgehog signaling, can reverse taxane-mediated chemoresistance.
Collapse
|
294
|
Tumor vascular targeted liposomal-bortezomib minimizes side effects and increases therapeutic activity in human neuroblastoma. J Control Release 2015; 211:44-52. [PMID: 26031842 DOI: 10.1016/j.jconrel.2015.05.286] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/22/2015] [Accepted: 05/27/2015] [Indexed: 12/19/2022]
Abstract
Neuroblastoma is a childhood cancer with poor long-term prognosis in advanced stages. A major aim in neuroblastoma therapy is to develop targeted drug delivery systems to ameliorate drug therapeutic index and efficacy. In this study, a novel bortezomib (BTZ) liposomal formulation was set-up and characterized. Since BTZ is freely permeable across the lipidic bilayer, an amino-lactose (LM) was synthesized as complexing agent to entrap BTZ inside the internal aqueous compartment of stealth liposomes. High encapsulation efficiency was achieved by a loading method based on the formation of boronic esters between the boronic acid moiety of BTZ and the hydroxyl groups of LM. Next, NGR peptides were linked to the liposome surface as a targeting-ligand for the tumor endothelial cell marker, aminopeptidase N. Liposomes were characterized for size, Z-potential, polydispersity index, drug content, and release. Lyophilization in the presence of cryoprotectants (trehalose, sucrose) was also examined in terms of particle size changes and drug leakage. BTZ was successfully loaded into non-targeted (SL[LM-BTZ]) and targeted (NGR-SL[LM-BTZ]) liposomes with an entrapment efficiency of about 68% and 57%, respectively. These nanoparticles were suitable for intravenous administration, presenting an average diameter of 170nm and narrow polydispersity. Therefore, orthotopic NB-bearing mice were treated with 1.0 or 1.5mg/kg of BTZ, either in free form or encapsulated into liposomes. BTZ loaded liposomes showed a significant reduction of drug systemic adverse effects with respect to free drug, even at the highest dose tested. Moreover, mice treated with 1.5mg/kg of NGR-SL[LM-BTZ] lived statistically longer than untreated mice (P=0.0018) and SL[LM-BTZ]-treated mice (P=0.0256). Our results demonstrate that the novel vascular targeted BTZ formulation is endowed with high therapeutic index and low toxicity, providing a new tool for future applications in neuroblastoma clinical studies.
Collapse
|
295
|
Breast cancer cell line MCF7 escapes from G1/S arrest induced by proteasome inhibition through a GSK-3β dependent mechanism. Sci Rep 2015; 5:10027. [PMID: 25941117 PMCID: PMC4419540 DOI: 10.1038/srep10027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/24/2015] [Indexed: 12/18/2022] Open
Abstract
Targeting the ubiquitin proteasome pathway has emerged as a rational approach in the treatment of human cancers. Autophagy has been described as a cytoprotective mechanism to increase tumor cell survival under stress conditions. Here, we have focused on the role of proteasome inhibition in cell cycle progression and the role of autophagy in the proliferation recovery. The study was performed in the breast cancer cell line MCF7 compared to the normal mammary cell line MCF10A. We found that the proteasome inhibitor MG132 induced G1/S arrest in MCF10A, but G2/M arrest in MCF7 cells. The effect of MG132 on MCF7 was reproduced on MCF10A cells in the presence of the glycogen synthase kinase 3β (GSK-3β) inhibitor VII. Similarly, MCF7 cells overexpressing constitutively active GSK-3β behaved like MCF10A cells. On the other hand, MCF10A cells remained arrested after MG132 removal while MCF7 recovered the proliferative capacity. Importantly, this recovery was abolished in the presence of the autophagy inhibitor 3-methyladenine (3-MA). Thus, our results support the relevance of GSK-3β and autophagy as two targets for controlling cell cycle progression and proliferative capacity in MCF7, highlighting the co-treatment of breast cancer cells with 3-MA to synergize the effect of the proteasome inhibition.
Collapse
|
296
|
Ying H, Qin A, Cheng TS, Pavlos NJ, Rea S, Dai K, Zheng MH. Disulfiram attenuates osteoclast differentiation in vitro: a potential antiresorptive agent. PLoS One 2015; 10:e0125696. [PMID: 25928135 PMCID: PMC4416043 DOI: 10.1371/journal.pone.0125696] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Disulfiram (DSF), a cysteine modifying compound, has long been clinically employed for the treatment of alcohol addiction. Mechanistically, DSF acts as a modulator of MAPK and NF-κB pathways signaling pathways. While these pathways are crucial for osteoclast (OC) differentiation, the potential influence of DSF on OC formation and function has not been directly assessed. Here, we explore the pharmacological effects of DSF on OC differentiation, activity and the modulation of osteoclastogenic signaling cascades. We first analyzed cytotoxicity of DSF on bone marrow monocytes isolated from C57BL/6J mice. Upon the establishment of optimal dosage, we conducted osteoclastogenesis and bone resorption assays in the presence or absence of DSF treatment. Luciferase assays in RAW264.7 cells were used to examine the effects of DSF on major transcription factors activation. Western blot, reverse transcription polymerase chain reaction, intracellular acidification and proton influx assays were employed to further dissect the underlying mechanism. DSF treatment dose-dependently inhibited both mouse and human osteoclastogenesis, especially at early stages of differentiation. This inhibition correlated with a decrease in the expression of key osteoclastic marker genes including CtsK, TRAP, DC-STAMP and Atp6v0d2 as well as a reduction in bone resorption in vitro. Suppression of OC differentiation was found to be due, at least in part, to the blockade of several key receptor activators of nuclear factor kappa-B ligand (RANKL)-signaling pathways including ERK, NF-κB and NFATc1. On the other hand, DSF failed to suppress intracellular acidification and proton influx in mouse and human osteoclasts using acridine orange quenching and microsome-based proton transport assays. Our findings indicate that DSF attenuates OC differentiation via the collective suppression of several key RANKL-mediated signaling cascades, thus making it an attractive agent for the treatment of OC-mediated disorders.
Collapse
Affiliation(s)
- Hua Ying
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
| | - An Qin
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
- Department of Orthopaedics, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tak S. Cheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
| | - Nathan J. Pavlos
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
| | - Sarah Rea
- Harry Perkins Institute of Medical Research, Perth, Australia
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - Kerong Dai
- Department of Orthopaedics, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (MHZ); (KD)
| | - Ming H. Zheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
- * E-mail: (MHZ); (KD)
| |
Collapse
|
297
|
Teicher BA, Tomaszewski JE. Proteasome inhibitors. Biochem Pharmacol 2015; 96:1-9. [PMID: 25935605 DOI: 10.1016/j.bcp.2015.04.008] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/16/2015] [Indexed: 12/20/2022]
Abstract
Proteasome inhibitors have a 20 year history in cancer therapy. The first proteasome inhibitor, bortezomib (Velcade, PS-341), a break-through multiple myeloma treatment, moved rapidly through development from bench in 1994 to first approval in 2003. Bortezomib is a reversible boronic acid inhibitor of the chymotrypsin-like activity of the proteasome. Next generation proteasome inhibitors include carfilzomib and oprozomib which are irreversible epoxyketone proteasome inhibitors; and ixazomib and delanzomib which are reversible boronic acid proteasome inhibitors. Two proteasome inhibitors, bortezomib and carfilzomib are FDA approved drugs and ixazomib and oprozomib are in late stage clinical trials. All of the agents are potent cytotoxics. The disease focus for all the proteasome inhibitors is multiple myeloma. This focus arose from clinical observations made in bortezomib early clinical trials. Later preclinical studies confirmed that multiple myeloma cells were indeed more sensitive to proteasome inhibitors than other tumor cell types. The discovery and development of the proteasome inhibitor class of anticancer agents has progressed through a classic route of serendipity and scientific investigation. These agents are continuing to have a major impact in their treatment of hematologic malignancies and are beginning to be explored as potential treatment agent for non-cancer indications.
Collapse
Affiliation(s)
- Beverly A Teicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, United States.
| | - Joseph E Tomaszewski
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, United States
| |
Collapse
|
298
|
Proteomics in cancer biomarkers discovery: challenges and applications. DISEASE MARKERS 2015; 2015:321370. [PMID: 25999657 PMCID: PMC4427011 DOI: 10.1155/2015/321370] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/15/2015] [Accepted: 02/18/2015] [Indexed: 01/28/2023]
Abstract
With the introduction of recent high-throughput technologies to various fields of science and medicine, it is becoming clear that obtaining large amounts of data is no longer a problem in modern research laboratories. However, coherent study designs, optimal conditions for obtaining high-quality data, and compelling interpretation, in accordance with the evidence-based systems biology, are critical factors in ensuring the emergence of good science out of these recent technologies. This review focuses on the proteomics field and its new perspectives on cancer research. Cornerstone publications that have tremendously helped scientists and clinicians to better understand cancer pathogenesis; to discover novel diagnostic and/or prognostic biomarkers; and to suggest novel therapeutic targets will be presented. The author of this review aims at presenting some of the relevant literature data that helped as a step forward in bridging the gap between bench work results and bedside potentials. Undeniably, this review cannot include all the work that is being produced by expert research groups all over the world.
Collapse
|
299
|
Gill KA, Berrué F, Arens JC, Carr G, Kerr RG. Cystargolides, 20S Proteasome Inhibitors Isolated from Kitasatospora cystarginea. JOURNAL OF NATURAL PRODUCTS 2015; 78:822-826. [PMID: 25769015 DOI: 10.1021/np501060k] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Two novel β-lactone-containing natural products, cystargolides A (1) and B (2), were isolated from the actinomycete Kitasatospora cystarginea. The production of these two natural products was highlighted using a methodology associating liquid chromatography-high-resolution mass spectrometry (LC-HRMS) analysis and the statistical analysis tool principal component analysis (PCA). Their structures were elucidated by interpretation of NMR experiments and tandem mass spectrometry. The absolute configurations of the amino acid residues were determined using Marfey's method, and the relative configurations of the β-lactone substituents were determined on the basis of the vicinal (3)J(HH) coupling value. Due to the presence of the β-lactone, 1 and 2 were evaluated for their ability to inhibit the human 20S proteasome. 1 and 2 both inhibited the 20S proteasome in vitro with IC50 values of 0.35 and 0.93 μM, respectively.
Collapse
Affiliation(s)
- Krista A Gill
- †Department of Chemistry, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Fabrice Berrué
- †Department of Chemistry, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
- ‡Nautilus Biosciences Canada Inc., DRC 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Jennifer C Arens
- §Department of Biomedical Sciences, Atlantic Veterinary College, 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Gavin Carr
- ‡Nautilus Biosciences Canada Inc., DRC 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Russell G Kerr
- †Department of Chemistry, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
- ‡Nautilus Biosciences Canada Inc., DRC 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
- §Department of Biomedical Sciences, Atlantic Veterinary College, 550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| |
Collapse
|
300
|
Tsimokha AS, Kulichkova VA, Karpova EV, Zaykova JJ, Aksenov ND, Vasilishina AA, Kropotov AV, Antonov A, Barlev NA. DNA damage modulates interactions between microRNAs and the 26S proteasome. Oncotarget 2015; 5:3555-67. [PMID: 25004448 PMCID: PMC4116502 DOI: 10.18632/oncotarget.1957] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
26S proteasomes are known as major non-lysosomal cellular machines for coordinated and specific destruction of ubiquitinylated proteins. The proteolytic activities of proteasomes are controlled by various post-translational modifications in response to environmental cues, including DNA damage. Besides proteolysis, proteasomes also associate with RNA hydrolysis and splicing. Here, we extend the functional diversity of proteasomes by showing that they also dynamically associate with microRNAs (miRNAs) both in the nucleus and cytoplasm of cells. Moreover, DNA damage induced by an anti-cancer drug, doxorubicin, alters the repertoire of proteasome-associated miRNAs, enriching the population of miRNAs that target cell cycle checkpoint regulators and DNA repair proteins. Collectively, these data uncover yet another potential mode of action for proteasomes in the cell via their dynamic association with microRNAs.
Collapse
Affiliation(s)
- Anna S Tsimokha
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | | | | | | | | | | | | | | | - Nikolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; Department of Biochemistry, University of Leicester, Leicester, LE1 9HN; Molecular Pharmacology laboratory, Saint-Petersburg Institute of Technology, Saint-Petersburg 190013, Russia
| |
Collapse
|