51
|
Tao NN, Zhang ZZ, Ren JH, Zhang J, Zhou YJ, Wai Wong VK, Kwan Law BY, Cheng ST, Zhou HZ, Chen WX, Xu HM, Chen J. Overexpression of ubiquitin-conjugating enzyme E2 L3 in hepatocellular carcinoma potentiates apoptosis evasion by inhibiting the GSK3β/p65 pathway. Cancer Lett 2020; 481:1-14. [PMID: 32268166 DOI: 10.1016/j.canlet.2020.03.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 01/07/2023]
Abstract
UBE2L3 is a ubiquitin-conjugating protein belonging to the E2 family that consists of 153 amino acid residues. In this study, we found that UBE2L3 was generally upregulated in clinical HCC samples compared to non-tumour samples and that there was a strong association between high UBE2L3 expression and tumour size, clinical grade and prognosis in HCC patients. UBE2L3 depletion inhibited the proliferation and induced the apoptosis of HCC cells. At the molecular level, we observed that UBE2L3 depletion enhanced the protein stability of GSK3β, thus promoting the expression and activation of GSK3β. Subsequently, activated GSK3β phosphorylated p65 and promoted its nuclear translocation to increase the expression of target genes, including PUMA, Bax, Bim, Bad, and Bid. In vivo, knockout of UBE2L3 in HCC cells inhibited tumour growth in orthotopic liver injection nude mouse models. Moreover, inhibition of p65 or GSK3β significantly restored the effects induced by UBE2L3 knockout in HCC. Together, this study reveals the stimulatory effect of UBE2L3 on HCC cell proliferation, suggesting that UBE2L3 may be an important pro-tumorigenic factor in liver carcinogenesis and a potential therapeutic target of HCC.
Collapse
Affiliation(s)
- Na-Na Tao
- Department of Infectious Disease, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Clinical Laboratory, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Zhen-Zhen Zhang
- Department of Infectious Disease, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Hua Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Juan Zhang
- Department of Clinical Laboratory, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yu-Jiao Zhou
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Sheng-Tao Cheng
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Hong-Zhong Zhou
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wei-Xian Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong-Mei Xu
- Department of Infectious Disease, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Juan Chen
- Department of Infectious Disease, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
52
|
Zhao Y, Zhu X, Han Z, Zhang Y, Dong T, Li Y, Dong J, Wei H, Li X. Comparative analysis of transcriptomes from different coloration of Chinese mitten crab Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2020; 98:515-521. [PMID: 32001357 DOI: 10.1016/j.fsi.2020.01.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 06/10/2023]
Abstract
Chinese mitten crab Eriocheir sinensis is probably the most important freshwater cultured crab in China. A tiny minority of brownish-orange individuals have been discovered in the long period of artificial breeding history of E. sinensiss. Those mutants are usually accompanied with slow growth rate, low molting frequency and poor survival rate, which may be the results of growth defects and immunodeficiency. To better understand the relationship between body color determination and the immune system as well as the related genes expression in E. sinensiss, we performed the whole-body transcriptome analysis in different color of first stage zoea (ZI) larvae using next-generation sequencing (NGS) technology. We randomly assembled 175.40 and 177.52million clean reads from the wild and mutant ZIs, respectively. Finally, we identified 7153 differentially expressed genes (DEGs) (p < 0.05), with 5194 up-regulated and 1959 down-regulated. A total of 13 KEGG pathways related to immune system were detected among 248 pathways. Except the first whole-body RNA sequencing of color-specific transcriptomes for E. sinensis, this study will offer a better understanding of the underlying molecular mechanisms of interaction between color determination and the immune system.
Collapse
Affiliation(s)
- Yingying Zhao
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Xiaochen Zhu
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Zhibin Han
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yazhao Zhang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Tengfei Dong
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yingdong Li
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Jing Dong
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Hua Wei
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Xiaodong Li
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China; Panjin Guanghe Crab Industry Co.Ltd., Panjin, 124000, China.
| |
Collapse
|
53
|
Advancing the Role of Gamma-Tocotrienol as Proteasomes Inhibitor: A Quantitative Proteomic Analysis of MDA-MB-231 Human Breast Cancer Cells. Biomolecules 2019; 10:biom10010019. [PMID: 31877708 PMCID: PMC7022772 DOI: 10.3390/biom10010019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 12/24/2022] Open
Abstract
Tocotrienol, an analogue of vitamin E has been known for its numerous health benefits and anti-cancer effects. Of the four isoforms of tocotrienols, gamma-tocotrienol (γT3) has been frequently reported for their superior anti-tumorigenic activity in both in vitro and in vivo studies, when compared to its counterparts. In this study, the effect of γT3 treatment in the cytoplasmic and nuclear fraction of MDA-MB-231 human breast cancer cells were assessed using the label-free quantitative proteomics analysis. The cytoplasmic proteome results revealed the ability of γT3 to inhibit a group of proteasome proteins such as PSMA, PSMB, PSMD, and PSME. The inhibition of proteasome proteins is known to induce apoptosis in cancer cells. As such, the findings from this study suggest γT3 as a potential proteasome inhibitor that can overcome deficiencies in growth-inhibitory or pro-apoptotic molecules in breast cancer cells. The nuclear proteome results revealed the involvement of important nuclear protein complexes which hardwire the anti-tumorigenesis mechanism in breast cancer following γT3 treatment. In conclusion, this study uncovered the advancing roles of γT3 as potential proteasomes inhibitor that can be used for the treatment of breast cancer.
Collapse
|
54
|
Yerlikaya A, Okur E. An investigation of the mechanisms underlying the proteasome inhibitor bortezomib resistance in PC3 prostate cancer cell line. Cytotechnology 2019; 72:121-130. [PMID: 31863311 DOI: 10.1007/s10616-019-00362-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
The phenomenon of acquired resistance to chemotherapeutic agents is a long-standing conundrum in cancer treatment. To help delineate drug resistance mechanisms and pave the way for the development of novel strategies, we generated a PC3 prostate cancer cell line resistant to proteasome inhibitor bortezomib for the first time. The resistant cells were found to have an IC50 value of 359.6 nM, whereas the IC50 value of parental cells was 82.6 nM after 24 h of treatment with varying doses of bortezomib. The resistant cells were also partly cross-resistant to the novel proteasome inhibitor carfilzomib; however, they were not resistant to widely used chemotherapeutic agent vincristine sulfate, indicating that enhanced cellular drug efflux via the multidrug resistance (MDR) transporters is not the molecular basis of the resistance. Since both bortezomib and carfilzomib target and inhibit the chymotrypsin-related activity residing in the β5 subunit of the proteasome (PSMB5), we next examined its expression and found surprisingly no significant alteration in the expression profile of the mature form. However, a significant increase in the accumulation of the precursor form of PSMB5 in response to 100 nM bortezomib was observed in the parental cells without a significant accumulation in the resistant cells. The results presented here thus suggest that the molecular mechanisms causing resistance to proteasome inhibitors need to be examined in-depth to overcome the resistance to ubiquitin-proteasome pathway inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Azmi Yerlikaya
- Department of Medical Biology, Faculty of Medicine, Kutahya Health Sciences University, Kütahya, Turkey.
| | - Emrah Okur
- Department of Biology, Faculty of Art and Sciences, Dumlupınar University, Kütahya, Turkey
| |
Collapse
|
55
|
Direito I, Fardilha M, Helguero LA. Contribution of the unfolded protein response to breast and prostate tissue homeostasis and its significance to cancer endocrine response. Carcinogenesis 2019; 40:203-215. [PMID: 30596981 DOI: 10.1093/carcin/bgy182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 12/05/2018] [Accepted: 12/14/2018] [Indexed: 12/25/2022] Open
Abstract
Resistant breast and prostate cancers remain a major clinical problem, new therapeutic approaches and better predictors of therapeutic response are clearly needed. Because of the involvement of the unfolded protein response (UPR) in cell proliferation and apoptosis evasion, an increasing number of publications support the hypothesis that impairments in this network trigger and/or exacerbate cancer. Moreover, UPR activation could contribute to the development of drug resistance phenotypes in both breast and prostate cancers. Therefore, targeting this pathway has recently emerged as a promising strategy in anticancer therapy. This review addresses the contribution of UPR to breast and prostate tissues homeostasis and its significance to cancer endocrine response with focus on the current progress on UPR research related to cancer biology, detection, prognosis and treatment.
Collapse
Affiliation(s)
| | - Margarida Fardilha
- Signal Transduction Laboratory, Department of Medical Sciences, Institute for Biomedicine (iBiMED), Universidade de Aveiro, Aveiro, Portugal
| | | |
Collapse
|
56
|
Loss of FBXO9 Enhances Proteasome Activity and Promotes Aggressiveness in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11111717. [PMID: 31684170 PMCID: PMC6895989 DOI: 10.3390/cancers11111717] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
The hematopoietic system is maintained throughout life by stem cells that are capable of differentiating into all hematopoietic lineages. An intimate balance between self-renewal, differentiation, and quiescence is required to maintain hematopoiesis and disruption of this balance can result in malignant transformation. FBXO9, the substrate recognition component from the SCF E3 ubiquitin ligase family, is downregulated in patients with acute myeloid leukemia (AML) compared to healthy bone marrow, and this downregulation is particularly evident in patients with inv(16) AML. To study FBXO9 in malignant hematopoiesis, we generated a conditional knockout mouse model using a novel CRISPR/Cas9 strategy. Deletion of Fbxo9 in the murine hematopoietic system showed no adverse effects on stem and progenitor cell function but in AML lead to markedly accelerated and aggressive leukemia development in mice with inv(16). Not only did Fbxo9 play a role in leukemia initiation but it also functioned to maintain AML activity and promote disease progression. Quantitative mass spectrometry from primary tumors reveals tumors lacking Fbxo9 highly express proteins associated with metastasis and invasion as well as components of the ubiquitin proteasome system. We confirmed that the loss of FBXO9 leads to increased proteasome activity and tumors cells were more sensitive to in vitro proteasome inhibition with bortezomib, suggesting that FBXO9 expression may predict patients’ response to bortezomib.
Collapse
|
57
|
Proteasome subunit α1 overexpression preferentially drives canonical proteasome biogenesis and enhances stress tolerance in yeast. Sci Rep 2019; 9:12418. [PMID: 31455793 PMCID: PMC6712033 DOI: 10.1038/s41598-019-48889-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/13/2019] [Indexed: 02/04/2023] Open
Abstract
The 26S proteasome conducts the majority of regulated protein catabolism in eukaryotes. At the heart of the proteasome is the barrel-shaped 20S core particle (CP), which contains two β-rings sandwiched between two α-rings. Whereas canonical CPs contain α-rings with seven subunits arranged α1-α7, a non-canonical CP in which a second copy of the α4 subunit replaces the α3 subunit occurs in both yeast and humans. The mechanisms that control canonical versus non-canonical CP biogenesis remain poorly understood. Here, we have repurposed a split-protein reporter to identify genes that can enhance canonical proteasome assembly in mutant yeast producing non-canonical α4-α4 CPs. We identified the proteasome subunit α1 as an enhancer of α3 incorporation, and find that elevating α1 protein levels preferentially drives canonical CP assembly under conditions that normally favor α4-α4 CP formation. Further, we demonstrate that α1 is stoichiometrically limiting for α-ring assembly, and that enhancing α1 levels is sufficient to increase proteasome abundance and enhance stress tolerance in yeast. Together, our data indicate that the abundance of α1 exerts multiple impacts on proteasome assembly and composition, and we propose that the limited α1 levels observed in yeast may prime cells for alternative proteasome assembly following environmental stimuli.
Collapse
|
58
|
Kors S, Geijtenbeek K, Reits E, Schipper-Krom S. Regulation of Proteasome Activity by (Post-)transcriptional Mechanisms. Front Mol Biosci 2019; 6:48. [PMID: 31380390 PMCID: PMC6646590 DOI: 10.3389/fmolb.2019.00048] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Intracellular protein synthesis, folding, and degradation are tightly controlled processes to ensure proper protein homeostasis. The proteasome is responsible for the degradation of the majority of intracellular proteins, which are often targeted for degradation via polyubiquitination. However, the degradation rate of proteins is also affected by the capacity of proteasomes to recognize and degrade these substrate proteins. This capacity is regulated by a variety of proteasome modulations including (1) changes in complex composition, (2) post-translational modifications, and (3) altered transcription of proteasomal subunits and activators. Various diseases are linked to proteasome modulation and altered proteasome function. A better understanding of these modulations may offer new perspectives for therapeutic intervention. Here we present an overview of these three proteasome modulating mechanisms to give better insight into the diversity of proteasomes.
Collapse
Affiliation(s)
- Suzan Kors
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Karlijne Geijtenbeek
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Eric Reits
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sabine Schipper-Krom
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
59
|
Mármol I, Quero J, Rodríguez-Yoldi MJ, Cerrada E. Gold as a Possible Alternative to Platinum-Based Chemotherapy for Colon Cancer Treatment. Cancers (Basel) 2019; 11:cancers11060780. [PMID: 31195711 PMCID: PMC6628079 DOI: 10.3390/cancers11060780] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
Due to the increasing incidence and high mortality associated with colorectal cancer (CRC), novel therapeutic strategies are urgently needed. Classic chemotherapy against CRC is based on oxaliplatin and other cisplatin analogues; however, platinum-based therapy lacks selectivity to cancer cells and leads to deleterious side effects. In addition, tumor resistance to oxaliplatin is related to chemotherapy failure. Gold(I) derivatives are a promising alternative to platinum complexes, since instead of interacting with DNA, they target proteins overexpressed on tumor cells, thus leading to less side effects than, but a comparable antitumor effect to, platinum derivatives. Moreover, given the huge potential of gold nanoparticles, the role of gold in CRC chemotherapy is not limited to gold(I) complexes. Gold nanoparticles have been found to be able to overcome multidrug resistance along with reduced side effects due to a more efficient uptake of classic drugs. Moreover, the use of gold nanoparticles has enhanced the effect of traditional therapies such as radiotherapy, photothermal therapy, or photodynamic therapy, and has displayed a potential role in diagnosis as a consequence of their optic properties. Herein, we have reviewed the most recent advances in the use of gold(I) derivatives and gold nanoparticles in CRC therapy.
Collapse
Affiliation(s)
- Inés Mármol
- Department of Pharmacology and Physiology, University of Zaragoza, CIBERobn, IIS Aragón IA2, 50013 Zaragoza, Spain.
| | - Javier Quero
- Department of Pharmacology and Physiology, University of Zaragoza, CIBERobn, IIS Aragón IA2, 50013 Zaragoza, Spain.
| | - María Jesús Rodríguez-Yoldi
- Department of Pharmacology and Physiology, University of Zaragoza, CIBERobn, IIS Aragón IA2, 50013 Zaragoza, Spain.
| | - Elena Cerrada
- Deparment of Inorganic Chemistry, University of Zaragoza, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, University of Zaragoza-CSIC, 50009 Zaragoza, Spain.
| |
Collapse
|
60
|
Motosugi R, Murata S. Dynamic Regulation of Proteasome Expression. Front Mol Biosci 2019; 6:30. [PMID: 31119134 PMCID: PMC6504791 DOI: 10.3389/fmolb.2019.00030] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
The 26S proteasome is a multisubunit complex that catalyzes the degradation of ubiquitinated proteins. The proteasome comprises 33 distinct subunits, all of which are essential for its function and structure. Proteasomes are necessary for various biological processes in cells; therefore, precise regulation of proteasome expression and activity is essential for maintaining cellular health and function. Two decades of research revealed that transcription factors such as Rpn4 and Nrf1 control expression of proteasomes. In this review, we focus on the current understanding and recent findings on the mechanisms underlying the regulation of proteasome expression, as well as the translational regulation of proteasomes.
Collapse
Affiliation(s)
- Ryo Motosugi
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
61
|
Cogo F, Williams R, Burden RE, Scott CJ. Application of nanotechnology to target and exploit tumour associated proteases. Biochimie 2019; 166:112-131. [PMID: 31029743 DOI: 10.1016/j.biochi.2019.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Proteases are hydrolytic enzymes fundamental for a variety of physiological processes, but the loss of their regulation leads to aberrant functions that promote onset and progression of many diseases including cancer. Proteases have been implicated in almost every hallmark of cancer and whilst widely investigated for tumour therapy, clinical adoption of protease inhibitors as drugs remains a challenge due to issues such as off-target toxicity and inability to achieve therapeutic doses at the disease site. Now, nanotechnology-based solutions and strategies are emerging to circumvent these issues. In this review, preclinical advances in approaches to enhance the delivery of protease drugs and the exploitation of tumour-derived protease activities to promote targeting of nanomedicine formulations is examined. Whilst this field is still in its infancy, innovations to date suggest that nanomedicine approaches to protease targeting or inhibition may hold much therapeutic and diagnostic potential.
Collapse
Affiliation(s)
- Francesco Cogo
- Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE, UK
| | - Rich Williams
- Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE, UK
| | - Roberta E Burden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | | |
Collapse
|
62
|
Bánová Vulić R, Zdurienčíková M, Tyčiaková S, Benada O, Dubrovčáková M, Lakota J, Škultéty Ľ. Silencing of carbonic anhydrase I enhances the malignant potential of exosomes secreted by prostatic tumour cells. J Cell Mol Med 2019; 23:3641-3655. [PMID: 30916466 PMCID: PMC6484292 DOI: 10.1111/jcmm.14265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/14/2019] [Accepted: 02/26/2019] [Indexed: 12/17/2022] Open
Abstract
We report results showing that the silencing of carbonic anhydrase I (siCA1) in prostatic (PC3) tumour cells has a significant impact on exosome formation. An increased diameter, concentration and diversity of the produced exosomes were noticed as a consequence of this knock‐down. The protein composition of the exosomes' cargo was also altered. Liquid chromatography and mass spectrometry analyses identified 42 proteins significantly altered in PC3 siCA1 exosomes compared with controls. The affected proteins are mainly involved in metabolic processes, biogenesis, cell component organization and defense/immunity. Interestingly, almost all of them have been described as ‘enhancers' of tumour development through the promotion of cell proliferation, migration and invasion. Thus, our results indicate that the reduced expression of the CA1 protein enhances the malignant potential of PC3 cells.
Collapse
Affiliation(s)
| | | | | | - Oldřich Benada
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | | | - Ján Lakota
- Biomedical Research Center SAS, Bratislava, Slovak Republic.,St. Elizabeth Cancer Institute, Bratislava, Slovak Republic.,Center of Experimental Medicine SAS, Bratislava, Slovak Republic
| | - Ľudovít Škultéty
- Biomedical Research Center SAS, Bratislava, Slovak Republic.,Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| |
Collapse
|
63
|
Chen Y, Chen H, Xie H, Yuan S, Gao C, Yu L, Bi Z. Non‑covalent proteasome inhibitor PI‑1840 induces apoptosis and autophagy in osteosarcoma cells. Oncol Rep 2019; 41:2803-2817. [PMID: 30864717 PMCID: PMC6448088 DOI: 10.3892/or.2019.7040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2019] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma (OS) is the predominant form of primary bone malignancy in children and adolescents. Although the combination of chemotherapy and modified surgical therapy leads to marked improvements in the survival rate, the therapeutic outcomes remain unsatisfactory. Therefore, the identification of novel drugs with higher efficacy and fewer side‑effects is urgently required. Proteasome inhibitors have been approved by the Food and Drug Administration (FDA) for the treatment of certain cancers, although none of them are directed against OS. Non‑covalent proteasome inhibitors, such as PI‑1840, are superior to covalent ones in numerous respects in view of their chemical structure; however, to date, no studies have been published on the effects of non‑covalent proteasome inhibitors on OS cells. In the present study, the antineoplastic effects of PI‑1840 were systematically evaluated in the OS cell lines, MG‑63 and U2‑OS. Cell viability and morphological changes were assessed by Cell Counting Kit‑8 (CCK‑8) and live/dead assays. The cell cycle was analyzed using flow cytometry (FCM) and western blot analysis (assessing the levels of the proteins p21, p27, and the tyrosine kinase, WEE1). The extent of cell apoptosis and autophagy were assessed by FCM, western blot analysis [of the apoptosis‑associated proteins, microtubule‑associated protein 1 light chain 3 α (LC3) and Beclin1], and mRFP‑GFP‑LC3 adenovirus transfection assay. Transwell and wound healing assays, and western blot analysis of the matrix metalloproteinases (MMPs)2 and 9 were performed to preliminarily evaluate the migration and invasion capability of the cells. In the present study, our results revealed that PI‑1840 inhibited the proliferation of OS cells and induced apoptosis, partly due to attenuation of the nuclear factor‑κB (NF‑κB) pathway. In addition, PI‑1840‑induced autophagy was detected, and inhibiting the autophagy of the OS cells led to an increase in the survival rate of the U2‑OS cells rather than of the MG‑63 cells. Furthermore, PI‑1840 attenuated the migration and invasion capabilities of the OS cells. In conclusion, the present study revealed PI‑1840 to be a promising drug for the treatment of OS.
Collapse
Affiliation(s)
- Yuxi Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hongjun Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hui Xie
- Teaching Experiment Center of Biotechnology, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Shaohui Yuan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chuanbo Gao
- Department of Orthopedic Surgery, The Fifth Hospital of Harbin, Heilongjiang 150001, P.R. China
| | - Lei Yu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhenggang Bi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
64
|
Mylavarapu S, Kumar H, Kumari S, Sravanthi LS, Jain M, Basu A, Biswas M, Mylavarapu SVS, Das A, Roy M. Activation of Epithelial-Mesenchymal Transition and Altered β-Catenin Signaling in a Novel Indian Colorectal Carcinoma Cell Line. Front Oncol 2019; 9:54. [PMID: 30828563 PMCID: PMC6385509 DOI: 10.3389/fonc.2019.00054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer is the third major cause of cancer-related mortality worldwide. The upward trend in incidence and mortality rates, poor sensitivity to conventional therapies and a dearth of early diagnostic parameters pose a huge challenge in the management of colorectal cancer in India. Due to the high level of genetic diversity present in the Indian population, unraveling the genetic contributions toward pathogenesis is key for understanding the etiology of colorectal cancer and in reversing this trend. We have established a novel cell line, MBC02, from an Indian colorectal cancer patient and have carried out extensive molecular characterization to unravel the pathological alterations in this cell line. In-depth molecular analysis of MBC02 revealed suppression of E-cadherin expression, concomitant with overexpression of EMT related molecules, which manifested in the form of highly migratory and invasive cells. Loss of membrane-tethered E-cadherin released β-catenin from the adherens junction resulting in its cytoplasmic and nuclear accumulation and consequently, upregulation of c-Myc. MBC02 also showed dramatic transcriptional upregulation of β-catenin. Remarkably, we observed significantly elevated proteasome activity that perhaps co-evolved to compensate for the unnaturally high mRNA level of β-catenin to regulate the increased protein load. In addition, there was substantial misregulation of other clinically relevant signaling pathways that have clinical relevance in the pathogenesis of colorectal cancer. Our findings pave the way toward understanding the molecular differences that could define pathogenesis in cancers originating in the Indian population.
Collapse
Affiliation(s)
- Sanghamitra Mylavarapu
- Invictus Oncology Pvt. Ltd., New Delhi, India.,Department of Biotechnology, Delhi Technological University, New Delhi, India
| | - Harsh Kumar
- Regional Centre for Biotechnology, Faridabad, India.,School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | | | | | - Misti Jain
- Division of Cancer Biology, MITRARxDx India Pvt. Ltd., Bangalore, India
| | - Aninda Basu
- Division of Cancer Biology, MITRARxDx India Pvt. Ltd., Bangalore, India
| | - Manjusha Biswas
- Department of Molecular Pathology, MITRARxDx India Pvt. Ltd., Bangalore, India
| | - Sivaram V S Mylavarapu
- Regional Centre for Biotechnology, Faridabad, India.,School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, New Delhi, India
| | - Monideepa Roy
- Invictus Oncology Pvt. Ltd., New Delhi, India.,India Innovation Research Center, New Delhi, India
| |
Collapse
|
65
|
Monteleone E, Orecchia V, Corrieri P, Schiavone D, Avalle L, Moiso E, Savino A, Molineris I, Provero P, Poli V. SP1 and STAT3 Functionally Synergize to Induce the RhoU Small GTPase and a Subclass of Non-canonical WNT Responsive Genes Correlating with Poor Prognosis in Breast Cancer. Cancers (Basel) 2019; 11:cancers11010101. [PMID: 30654518 PMCID: PMC6356433 DOI: 10.3390/cancers11010101] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is a heterogeneous disease whose clinical management is very challenging. Although specific molecular features characterize breast cancer subtypes with different prognosis, the identification of specific markers predicting disease outcome within the single subtypes still lags behind. Both the non-canonical Wingless-type MMTV Integration site (WNT) and the Signal Transducer and Activator of Transcription (STAT)3 pathways are often constitutively activated in breast tumors, and both can induce the small GTPase Ras Homolog Family Member U RhoU. Here we show that RhoU transcription can be triggered by both canonical and non-canonical WNT ligands via the activation of c-JUN N-terminal kinase (JNK) and the recruitment of the Specificity Protein 1 (SP1) transcription factor to the RhoU promoter, identifying for the first time SP1 as a JNK-dependent mediator of WNT signaling. RhoU down-regulation by silencing or treatment with JNK, SP1 or STAT3 inhibitors leads to impaired migration and invasion in basal-like MDA-MB-231 and BT-549 cells, suggesting that STAT3 and SP1 can cooperate to induce high RhoU expression and enhance breast cancer cells migration. Moreover, in vivo concomitant binding of STAT3 and SP1 defines a subclass of genes belonging to the non-canonical WNT and the Interleukin (IL)-6/STAT3 pathways and contributing to breast cancer aggressiveness, suggesting the relevance of developing novel targeted therapies combining inhibitors of the STAT3 and WNT pathways or of their downstream mediators.
Collapse
Affiliation(s)
- Emanuele Monteleone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Valeria Orecchia
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Paola Corrieri
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Davide Schiavone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Enrico Moiso
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Aurora Savino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Ivan Molineris
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy.
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| |
Collapse
|
66
|
Astakhova TM, Morozov AV, Erokhov PA, Mikhailovskaya MI, Akopov SB, Chupikova NI, Safarov RR, Sharova NP. Combined Effect of Bortezomib and Menadione Sodium Bisulfite on Proteasomes of Tumor Cells: The Dramatic Decrease of Bortezomib Toxicity in a Preclinical Trial. Cancers (Basel) 2018; 10:E351. [PMID: 30257462 PMCID: PMC6209890 DOI: 10.3390/cancers10100351] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/12/2018] [Accepted: 09/22/2018] [Indexed: 11/16/2022] Open
Abstract
Tumor growth is associated with elevated proteasome expression and activity. This makes proteasomes a promising target for antitumor drugs. Current antitumor drugs such as bortezomib that inhibit proteasome activity have significant side effects. The purpose of the present study was to develop effective low-toxic antitumor compositions with combined effects on proteasomes. For compositions, we used bortezomib in amounts four and ten times lower than its clinical dose, and chose menadione sodium bisulfite (MSB) as the second component. MSB is known to promote oxidation of NADH, generate superoxide radicals, and as a result damage proteasome function in cells that ensure the relevance of MSB use for the composition development. The proteasome pool was investigated by the original native gel electrophoresis method, proteasome chymotrypsin-like activity-by Suc-LLVY-AMC-hydrolysis. For the compositions, we detected 10 and 20 μM MSB doses showing stronger proteasome-suppressing and cytotoxic in cellulo effects on malignant cells than on normal ones. MSB indirectly suppressed 26S-proteasome activity in cellulo, but not in vitro. At the same time, MSB together with bortezomib displayed synergetic action on the activity of all proteasome forms in vitro as well as synergetic antitumor effects in cellulo. These findings determine the properties of the developed compositions in vivo: antitumor efficiency, higher (against hepatocellular carcinoma and mammary adenocarcinoma) or comparable to bortezomib (against Lewis lung carcinoma), and drastically reduced toxicity (LD50) relative to bortezomib. Thus, the developed compositions represent a novel generation of bortezomib-based anticancer drugs combining high efficiency, low general toxicity, and a potentially expanded range of target tumors.
Collapse
Affiliation(s)
- Tatiana M Astakhova
- Laboratory of Biochemistry of Ontogenesis Processes, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia.
| | - Alexey V Morozov
- Laboratory of Regulation of Intracellular Proteolysis, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, 119991 Moscow, Russia.
| | - Pavel A Erokhov
- Laboratory of Biochemistry of Ontogenesis Processes, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia.
| | - Maria I Mikhailovskaya
- Laboratory of Biochemistry of Ontogenesis Processes, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia.
| | - Sergey B Akopov
- Laboratory of Human Genes Structure and Functions, Shemyakin⁻Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 16/10 Miklukho-Maklay Street, 117997 Moscow, Russia.
| | - Natalia I Chupikova
- Laboratory of Biochemistry of Ontogenesis Processes, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia.
| | - Ruslan R Safarov
- Laboratory of Biochemistry of Ontogenesis Processes, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia.
| | - Natalia P Sharova
- Laboratory of Biochemistry of Ontogenesis Processes, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia.
| |
Collapse
|
67
|
Solcia E, Necchi V, Sommi P, Ricci V. Proteasome-Rich PaCS as an Oncofetal UPS Structure Handling Cytosolic Polyubiquitinated Proteins. In Vivo Occurrence, in Vitro Induction, and Biological Role. Int J Mol Sci 2018; 19:ijms19092767. [PMID: 30223470 PMCID: PMC6164709 DOI: 10.3390/ijms19092767] [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: 08/29/2018] [Accepted: 09/11/2018] [Indexed: 11/16/2022] Open
Abstract
In this article, we outline and discuss available information on the cellular site and mechanism of proteasome interaction with cytosolic polyubiquitinated proteins and heat-shock molecules. The particulate cytoplasmic structure (PaCS) formed by barrel-like particles, closely reproducing in vivo the high-resolution structure of 26S proteasome as isolated in vitro, has been detected in a variety of fetal and neoplastic cells, from living tissue or cultured cell lines. Specific trophic factors and interleukins were found to induce PaCS during in vitro differentiation of dendritic, natural killer (NK), or megakaryoblastic cells, apparently through activation of the MAPK-ERK pathway. Direct interaction of CagA bacterial oncoprotein with proteasome was shown inside the PaCSs of a Helicobacter pylori-infected gastric epithelium, a finding suggesting a role for PaCS in CagA-mediated gastric carcinogenesis. PaCS dissolution and autophagy were seen after withdrawal of inducing factors. PaCS-filled cell blebs and ectosomes were found in some cells and may represent a potential intercellular discharge and transport system of polyubiquitinated antigenic proteins. PaCS differs substantially from the inclusion bodies, sequestosomes, and aggresomes reported in proteinopathies like Huntington or Parkinson diseases, which usually lack PaCS. The latter seems more linked to conditions of increased cell proliferation/differentiation, implying an increased functional demand to the ubiquitin–proteasome system.
Collapse
Affiliation(s)
- Enrico Solcia
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
- Pathologic Anatomy Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy.
| | - Vittorio Necchi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
- Centro Grandi Strumenti, University of Pavia, 27100 Pavia, Italy.
| | - Patrizia Sommi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
| | - Vittorio Ricci
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
68
|
Quero J, Cabello S, Fuertes T, Mármol I, Laplaza R, Polo V, Gimeno MC, Rodriguez-Yoldi MJ, Cerrada E. Proteasome versus Thioredoxin Reductase Competition as Possible Biological Targets in Antitumor Mixed Thiolate-Dithiocarbamate Gold(III) Complexes. Inorg Chem 2018; 57:10832-10845. [DOI: 10.1021/acs.inorgchem.8b01464] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Javier Quero
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Facultad de Veterinaria, CIBERobn, IIS, Aragón IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Silvia Cabello
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - Teresa Fuertes
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Facultad de Veterinaria, CIBERobn, IIS, Aragón IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Inés Mármol
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Facultad de Veterinaria, CIBERobn, IIS, Aragón IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Ruben Laplaza
- Departamento de Química Física, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Victor Polo
- Departamento de Química Física, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - M. Concepción Gimeno
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| | - M. Jesús Rodriguez-Yoldi
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Facultad de Veterinaria, CIBERobn, IIS, Aragón IA2, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain
| |
Collapse
|
69
|
Abstract
The billions of proteins inside a eukaryotic cell are organized among dozens of sub-cellular compartments, within which they are further organized into protein complexes. The maintenance of both levels of organization is crucial for normal cellular function. Newly made proteins that fail to be segregated to the correct compartment or assembled into the appropriate complex are defined as orphans. In this review, we discuss the challenges faced by a cell of minimizing orphaned proteins, the quality control systems that recognize orphans, and the consequences of excess orphans for protein homeostasis and disease.
Collapse
|
70
|
Dubois C, Lecomte C, Ruys SPD, Kuzmic M, Della-Vedova C, Dubourg N, Galas S, Frelon S. Precoce and opposite response of proteasome activity after acute or chronic exposure of C. elegans to γ-radiation. Sci Rep 2018; 8:11349. [PMID: 30054490 PMCID: PMC6063909 DOI: 10.1038/s41598-018-29033-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
Species are chronically exposed to ionizing radiation, a natural phenomenon which can be enhanced by human activities. The induced toxicity mechanisms still remain unclear and seem depending on the mode of exposure, i.e. acute and chronic. To better understand these phenomena, studies need to be conducted both at the subcellular and individual levels. Proteins, functional molecules in organisms, are the targets of oxidative damage (especially via their carbonylation (PC)) and are likely to be relevant biomarkers. After exposure of Caenorhabditis elegans to either chronic or acute γ rays we showed that hatching success is impacted after acute but not after chronic irradiation. At the molecular level, the carbonylated protein level in relation with dose was slightly different between acute and chronic exposure whereas the proteolytic activity is drastically modified. Indeed, whereas the 20S proteasome activity is inhibited by acute irradiation from 0.5 Gy, it is activated after chronic irradiation from 1 Gy. As expected, the 20S proteasome activity is mainly modified by irradiation whereas the 26S and 30S activity are less changed. This study provides preliminaries clues to understand the role of protein oxidation and proteolytic activity in the radiation-induced molecular mechanisms after chronic versus acute irradiation in C. elegans.
Collapse
Affiliation(s)
- Cécile Dubois
- IRSN/PSE-ENV/SRTE - Laboratoire d'ecotoxicologie des radionucléides - BP3, 13115, St Paul lez Durance Cedex, France
| | - Catherine Lecomte
- IRSN/PSE-ENV/SRTE - Laboratoire d'ecotoxicologie des radionucléides - BP3, 13115, St Paul lez Durance Cedex, France
| | - Sébastien Pyr Dit Ruys
- IRSN/PSE-ENV/SRTE - Laboratoire d'ecotoxicologie des radionucléides - BP3, 13115, St Paul lez Durance Cedex, France
| | - Mira Kuzmic
- IRSN/PSE-ENV/SRTE - Laboratoire d'ecotoxicologie des radionucléides - BP3, 13115, St Paul lez Durance Cedex, France
| | | | - Nicolas Dubourg
- IRSN/PSE-ENV/SRTE - Laboratoire d'ecotoxicologie des radionucléides - BP3, 13115, St Paul lez Durance Cedex, France
| | - Simon Galas
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Sandrine Frelon
- IRSN/PSE-ENV/SRTE - Laboratoire d'ecotoxicologie des radionucléides - BP3, 13115, St Paul lez Durance Cedex, France.
| |
Collapse
|
71
|
Tsvetkov P, Adler J, Myers N, Biran A, Reuven N, Shaul Y. Oncogenic addiction to high 26S proteasome level. Cell Death Dis 2018; 9:773. [PMID: 29991718 PMCID: PMC6039477 DOI: 10.1038/s41419-018-0806-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/15/2022]
Abstract
Proteasomes are large intracellular complexes responsible for the degradation of cellular proteins. The altered protein homeostasis of cancer cells results in increased dependency on proteasome function. The cellular proteasome composition comprises the 20S catalytic complex that is frequently capped with the 19S regulatory particle in forming the 26S proteasome. Proteasome inhibitors target the catalytic barrel (20S) and thus this inhibition does not allow the deconvolution of the distinct roles of 20S versus 26S proteasomes in cancer progression. We examined the degree of dependency of cancer cells specifically to the level of the 26S proteasome complex. Oncogenic transformation of human and mouse immortalized cells with mutant Ras induced a strong posttranscriptional increase of the 26S proteasome subunits, giving rise to high 26S complex levels. Depletion of a single subunit of the 19S RP was sufficient to reduce the 26S proteasome level and lower the cellular 26S/20S ratio. Under this condition the viability of the Ras-transformed MCF10A cells was severely compromised. This observation led us to hypothesize that cancer cell survival is dependent on maximal utilization of its 26S proteasomes. We validated this possibility in a large number of cancer cell lines and found that partial reduction of the 26S proteasome level impairs viability in all cancer cells examined and was not correlated with cell doubling time or reduction efficiency. Interstingly, normal human fibroblasts are refractory to the same type of 26S proteasome reduction. The suppression of 26S proteasomes in cancer cells activated the UPR and caspase-3 and cells stained positive with Annexin V. In addition, suppression of the 26S proteasome resulted in cellular proteasome redistribution, cytoplasm shrinkage, and nuclear deformation, the hallmarks of apoptosis. The observed tumor cell-specific addiction to the 26S proteasome levels sets the stage for future strategies in exploiting this dependency in cancer therapy.
Collapse
Affiliation(s)
- Peter Tsvetkov
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel.,Broad Institute of MIT and Harvard, 415 Main St., Cambridge, MA, 02142, USA
| | - Julia Adler
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Nadav Myers
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Assaf Biran
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Nina Reuven
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
72
|
Okur E, Yerlikaya A. A novel and effective inhibitor combination involving bortezomib and OTSSP167 for breast cancer cells in light of label-free proteomic analysis. Cell Biol Toxicol 2018; 35:33-47. [PMID: 29948483 DOI: 10.1007/s10565-018-9435-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/06/2018] [Indexed: 01/23/2023]
Abstract
PURPOSE The 26S proteasome plays important roles in many intracellular processes and is therefore a critical intracellular cellular target for anticancer treatments. The primary aim of the current study was to identify critical proteins that may play roles in opposing the antisurvival effect of the proteasome inhibitor bortezomib together with the calcium-chelator BAPTA-AM in cancer cells using label-free LC-MS/MS. In addition, based on the results of the proteomic technique, a novel and more effective inhibitor combination involving bortezomib as well as OTSSP167 was developed for breast cancer cells. METHODS AND RESULTS Using label-free LC-MS/MS, it was found that expressions of 1266 proteins were significantly changed between the experimental groups. Among these proteins were cell division cycle 5-like (Cdc5L) and drebrin-like (DBNL). We then hypothesized that inhibition of the activities of these two proteins may lead to more effective anticancer inhibitor combinations in the presence of proteasomal inhibition. In fact, as presented in the current study, Cdc5L phosphorylation inhibitor CVT-313 and DBNL phosphorylation inhibitor OTSSP167 were highly cytotoxic in 4T1 breast cancer cells and their IC50 values were 20.1 and 43 nM, respectively. Under the same experimental conditions, the IC50 value of BAPTA-AM was found 19.9 μM. Using WST 1 cytotoxicity assay, it was determined that 10 nM bortezomib + 10 nM CVT-313 was more effective than the control, the single treatments, or than 5 nM bortezomib + 5 nM CVT-313. Similarly, 10 nM bortezomib + 10 nM OTSSP167 was more cytotoxic than the control, the monotherapies, 5 nM bortezomib + 5 nM OTSSP167, or than 5 nM bortezomib + 10 nM OTSSP167, indicating that bortezomib + OTSSP167 was also more effective than bortezomib + CVT-313 in a dose-dependent manner. Furthermore, the 3D spheroid model proved that bortezomib + OTSSP167 was more effective than the monotherapies as well as bortezomib + CVT-313 and bortezomib + BAPTA-AM combinations. Finally, the effect of bortezomib + OTSSP167 combination was tested on MDA-MB-231 breast cancer cells, and it similarly determined that 20 nM bortezomib +40 nM OTSSP167 combination completely blocked the formation of 3D spheroids. CONCLUSIONS Altogether, the results presented here indicate that bortezomib + OTSSP167 is a novel and effective combination and may be tested further for cancer treatment in vivo and in clinical settings.
Collapse
Affiliation(s)
- Emrah Okur
- Art and Science Faculty, Department of Biology, Kütahya Dumlupınar University, Kütahya, Turkey
| | - Azmi Yerlikaya
- Faculty of Medicine, Department of Medical Biology, Kütahya Health Sciences University, Kütahya, Turkey.
| |
Collapse
|
73
|
Zhao F, Yang B, Wang J, Zhang R, Liu J, Yin F, Xu W, He C. Incidence and risk of cardiac toxicities in patients with relapsed and refractory multiple myeloma treated with carfilzomib. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1525-1531. [PMID: 29881259 PMCID: PMC5985801 DOI: 10.2147/dddt.s159818] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Purpose Carfilzomib has been approved for use in relapsed and refractory multiple myeloma (RRMM). Cardiac toxicities have been reported with the use of carfilzomib. We aimed to determine the overall incidence and risk of cardiac toxicities in RRMM patients treated with carfilzomib using a meta-analysis. Methods We searched several databases for relevant articles. Prospective trials evaluating carfilzomib in RRMM patients with adequate data on cardiac toxicities were included for analysis. Pooled incidence, Peto ORs, and 95% CIs were calculated according to the heterogeneity of selected studies. Results A total of 2,607 RRMM patients from eight prospective trials were included. The pooled incidence of all-grade congestive heart failure (CHF) and ischemic heart disease (IHD) related to carfilzomib in RRMM patients was 5.5% (95% CI: 4.3%–6.9%) and 2.7% (95% CI: 1.1%–6.7%), respectively. In addition, the use of carfilzomib significantly increased all-grade (Peto OR 2.33, 95% CI: 1.56–3.48, p<0.001) and high-grade (Peto OR 3.22, 95% CI: 1.84–5.61, p<0.001) CHF when compared to controls, whereas there was no significantly increased risk of developing all-grade (Peto OR 1.31, 95% CI: 0.79–2.18, p=0.30) and high-grade (Peto OR 1.41, 95% CI: 0.73–2.72, p=0.31) IHD in RRMM patients receiving carfilzomib. Conclusion The use of carfilzomib in RRMM patients significantly increases the risk of developing CHF but not IHD. Clinicians should be cautious about the risk of CHF associated with carfilzomib to maximize the benefits and minimize the toxicities.
Collapse
Affiliation(s)
- Fang Zhao
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Bo Yang
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Juan Wang
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Rui Zhang
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Jing Liu
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Fenglei Yin
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Weixing Xu
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| | - Chunyuan He
- Department of Hematology, Cangzhou Central Hospital, Cangzhou, People's Republic of China
| |
Collapse
|
74
|
Chen Y, Zhang Y, Guo X. Proteasome dysregulation in human cancer: implications for clinical therapies. Cancer Metastasis Rev 2018; 36:703-716. [PMID: 29039081 DOI: 10.1007/s10555-017-9704-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer cells show heightened dependency on the proteasome for their survival, growth, and spread. Proteasome dysregulation is therefore commonly selected in favor of the development of many types of cancer. The vast abnormalities in a cancer cell, on top of the complexity of the proteasome itself, have enabled a plethora of mechanisms gearing the proteasome to the oncogenic process. Here, we use selected examples to highlight some general mechanisms underlying proteasome dysregulation in cancer, including copy number variations, transcriptional control, epigenetic regulation, and post-translational modifications. Research in this field has greatly advanced our understanding of proteasome regulation and will shed new light on proteasome-based combination therapies for cancer treatment.
Collapse
Affiliation(s)
- Yulin Chen
- Life Sciences Institute of Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Yanan Zhang
- Life Sciences Institute of Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Xing Guo
- Life Sciences Institute of Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China.
| |
Collapse
|
75
|
Chen L, Zhu G, Johns EM, Yang X. TRIM11 activates the proteasome and promotes overall protein degradation by regulating USP14. Nat Commun 2018; 9:1223. [PMID: 29581427 PMCID: PMC5964324 DOI: 10.1038/s41467-018-03499-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 02/19/2018] [Indexed: 12/27/2022] Open
Abstract
The proteasome is a complex protease critical for protein quality control and cell regulation, and its dysfunction is associated with cancer and other diseases. However, the mechanisms that control proteasome activity in normal and malignant cells remain unclear. Here we report that TRIM11 enhances degradation of aberrant and normal regulatory proteins, and augments overall rate of proteolysis. Mechanistically, TRIM11 binds to both the proteasome and USP14, a deubiquitinase that prematurely removes ubiquitins from proteasome-bound substrates and also noncatalytically inhibits the proteasome, and precludes their association, thereby increasing proteasome activity. TRIM11 promotes cell survival and is upregulated upon heat shock. Moreover, TRIM11 is required for tumor growth, and increased expression of TRIM11 correlates with poor clinical survival. These findings identify TRIM11 as an important activator of the proteasome, define a pathway that adjusts proteasome activity, and reveal a mechanism by which tumor cells acquire higher degradative power to support oncogenic growth. The proteasome-bound ubiquitinase USP14 plays an important role in determining proteasome activity and substrate specificity. Here the authors show that TRIM11, a member of the mammalian tripartite motif family, regulates USP14 and is an important activator of the proteasome.
Collapse
Affiliation(s)
- Liang Chen
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Guixin Zhu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eleanor M Johns
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
76
|
Shi K, Zhang JZ, Zhao RL, Yang L, Guo D. PSMD7 downregulation induces apoptosis and suppresses tumorigenesis of esophageal squamous cell carcinoma via the mTOR/p70S6K pathway. FEBS Open Bio 2018; 8:533-543. [PMID: 29632807 PMCID: PMC5881544 DOI: 10.1002/2211-5463.12394] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/03/2018] [Accepted: 01/12/2018] [Indexed: 12/23/2022] Open
Abstract
PSMD7, a 19S proteasome subunit, is overexpressed in most carcinoma cells. It forms a dimer with PSMD14 that functions in the removal of attached ubiquitin chain. However, there is little knowledge about the cellular mechanism of PSMD7 and its exact biological function, especially in cancer cells. In this study, we explored the role of PSMD7 in proliferation, cell cycle, apoptosis, and proteasomal proteolysis in the esophageal squamous cell carcinoma (ESCC) cell line EC9706. Our results showed that PSMD7 was highly expressed in ESCC cells. Downregulation of PSMD7 by lentivirus‐mediated shRNA led to decreased proliferation, increased cell apoptosis, and reduced proteasomal function. Notably, lower expression level of mTOR and p70S6K and suppressed activity of mTOR/p70S6K pathway were detected after PSMD7 downregulation. By contrast, increased expression of p‐mTORSer2448 and p‐p70S6KThr421/Ser424 was discovered upon PSMD7 overexpression in Het‐1A cells. Furthermore, PSMD7 downregulation contributed to decelerated tumor growth, inhibition of proteasomal function, induced cell apoptosis and attenuated activity of mTOR/p70S6K pathway in vivo. These findings suggest that PSMD7 and the mTOR/p70S6K pathway may be a promising candidate for developing therapies for ESCC.
Collapse
Affiliation(s)
- Ke Shi
- Department of Biochemistry and Molecular Biology Henan Medical College China
| | - Jin-Zhong Zhang
- Department of Biochemistry and Molecular Biology Henan Medical College China
| | - Rui-Li Zhao
- Editorial Department of Journal of Henan University of Technology Henan University of Technology Zhengzhou China.,College of Biological Engineering Henan University of Technology Zhengzhou China
| | - Liang Yang
- Department of Microbiology and Immunology and Medicine Henan Medical College China
| | - Dan Guo
- Department of Biochemistry and Molecular Biology Henan Medical College China
| |
Collapse
|
77
|
Li Y, Huang J, Sun J, Xiang S, Yang D, Ying X, Lu M, Li H, Ren G. The transcription levels and prognostic values of seven proteasome alpha subunits in human cancers. Oncotarget 2018; 8:4501-4519. [PMID: 27966459 PMCID: PMC5354849 DOI: 10.18632/oncotarget.13885] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 12/01/2016] [Indexed: 11/25/2022] Open
Abstract
Proteasome alpha subunits (PSMAs) have been shown to participate in the malignant progression of human cancers. However, the expression patterns and prognostic values of individual PSMAs remain elusive in most cancers. In the present study, we investigated the mRNA expression levels of seven PSMAs in different kinds of cancers using Oncomine and The Cancer Genome Atlas (TCGA) databases. The prognostic significance of PSMAs was also determined by Kaplan-Meier Plotter and PrognScan databases. Combined with Oncomine and TCGA, the mRNA expression levels of PSMA1-7 were significantly upregulated in breast, lung, gastric, bladder and head and neck cancer compared with normal tissues. Moreover, only PSMA6 and PSMA5 were not overexpressed in colorectal and kidney cancer, respectively. In survival analyses based on Kaplan-Meier Plotter, PSMA1-7 showed significant prognostic values in breast, lung and gastric cancer. Furthermore, potential correlations between PSMAs and survival outcomes were also observed in ovarian cancer, colorectal cancer and melanoma by Kaplan-Meier Plotter and PrognScan. These data indicated that PSMAs might serve as novel biomarkers and potential therapeutic targets for multiple human cancers. However, further studies are needed to explore the detailed biological functions and molecular mechanisms involved in tumor progression.
Collapse
Affiliation(s)
- Yunhai Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiazheng Sun
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shili Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dejuan Yang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuedong Ying
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengqi Lu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
78
|
Okumura T, Ikeda K, Ujihira T, Okamoto K, Horie-Inoue K, Takeda S, Inoue S. Proteasome 26S subunit PSMD1 regulates breast cancer cell growth through p53 protein degradation. J Biochem 2018; 163:19-29. [PMID: 28992264 DOI: 10.1093/jb/mvx053] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/30/2017] [Indexed: 01/14/2023] Open
Abstract
Endocrine therapy using antiestrogens and aromatase inhibitors is usually efficient to treat patients with hormone-sensitive breast cancer. Many patients with endocrine therapy, however, often acquire resistance. In the present study, we performed functional screening using short hairpin RNA library to dissect genes involved in antiestrogen tamoxifen resistance in MCF-7 breast cancer cells. We identified seven candidate genes that are associated with poor prognosis of breast cancer patients based on clinical dataset. The expression levels of six out of seven genes were higher in 4-hydroxytamoxifen (OHT) resistant MCF-7 (OHTR) cells compared with parental MCF-7 cells. Among the six selected genes, siRNA-mediated knockdown of PSMD1 and TSPAN12 markedly reduced the proliferation of OHTR cells. Notably, the knockdown of proteasome 26S subunit PSMD1 exhibited cell cycle arrest and the accumulation of p53 protein through inhibiting p53 protein degradation. In accordance with p53 accumulation, its target genes p21 and SFN were also upregulated by PSMD1 silencing. Taken together, PSMD1 was identified as a potential gene that plays a role in the development of tamoxifen resistance in breast cancer cells. These findings will provide a new insight for the mechanism underlying endocrine therapy resistance and a prognostic and therapeutic molecular target for advanced breast cancer.
Collapse
Affiliation(s)
- Toshiyuki Okumura
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan.,Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Takafumi Ujihira
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan.,Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
79
|
KOIZUMI S, HAMAZAKI J, MURATA S. Transcriptional regulation of the 26S proteasome by Nrf1. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2018; 94:325-336. [PMID: 30305478 PMCID: PMC6275327 DOI: 10.2183/pjab.94.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/10/2018] [Indexed: 05/21/2023]
Abstract
The 26S proteasome is a large protease complex that selectively degrades ubiquitinated proteins. It comprises 33 distinct subunits, each of which differ in function and structure, and which cannot be substituted by the other subunits. Owing to its complicated structure, the biogenesis of the 26S proteasome is elaborately regulated at the transcription, translation, and molecular assembly levels. Recent studies revealed that Nrf1 (NFE2L1) is a transcription factor that upregulates the expression of all the proteasome subunit genes in a concerted manner, especially during proteasome impairment in mammalian cells. In this review, we summarize current knowledge regarding the transcriptional regulation of the proteasome and recent findings concerning the regulation of Nrf1 transcription activity.
Collapse
Affiliation(s)
- Shun KOIZUMI
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun HAMAZAKI
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeo MURATA
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Correspondence should be addressed: S. Murata, Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan (e-mail: )
| |
Collapse
|
80
|
Guo KY, Han L, Li X, Yang AV, Lu J, Guan S, Li H, Yu Y, Zhao Y, Yang J, Zhang H. Novel proteasome inhibitor delanzomib sensitizes cervical cancer cells to doxorubicin-induced apoptosis via stabilizing tumor suppressor proteins in the p53 pathway. Oncotarget 2017; 8:114123-114135. [PMID: 29371974 PMCID: PMC5768391 DOI: 10.18632/oncotarget.23166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023] Open
Abstract
Cervical cancer, the third most commonly occurring cancer, is the second leading cause of cancer related mortality among women. Aberrant ubiquitination and proteasome activity, both human papillomavirus and tumor derived, have been shown to contribute to tumor angiogenesis, proliferation, and invasion in many cancers, including cervical cancer. Thus, small molecule proteasome inhibitors are a potential and strategic treatment option for cervical cancer. In this study, novel proteasome inhibitor delanzomib (CEP-18770) exhibited potent pro-apoptotic and cytotoxic effects on a panel of cervical cancer cell lines by blocking proteasomal activity. Delanzomib also significantly sensitized cervical cancer cells to treatment of doxorubicin (Dox), a traditional chemotherapeutic agent. Furthermore, proteasome inhibition revealed stabilization of p53 and p53 transcriptional targets and induction of p38/JNK phosphorylation. Additionally, delanzomib worked synergistically with Dox to further upregulate p53 and its downstream targets and enhanced Dox-induced p38 phosphorylation. Our study strongly supports the 26S proteasome as a potential therapeutic target in cervical cancer and proteasome inhibition by delanzomib may be a potential treatment strategy for cervical cancer patients.
Collapse
Affiliation(s)
- Kevin Y Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lili Han
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Gynecology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830001, China
| | - Xinyu Li
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrew V Yang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jiaxiong Lu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shan Guan
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui Li
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yang Yu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanling Zhao
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianhua Yang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hong Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| |
Collapse
|
81
|
Soave CL, Guerin T, Liu J, Dou QP. Targeting the ubiquitin-proteasome system for cancer treatment: discovering novel inhibitors from nature and drug repurposing. Cancer Metastasis Rev 2017; 36:717-736. [PMID: 29047025 PMCID: PMC5722705 DOI: 10.1007/s10555-017-9705-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the past 15 years, the proteasome has been validated as an anti-cancer drug target and 20S proteasome inhibitors (such as bortezomib and carfilzomib) have been approved by the FDA for the treatment of multiple myeloma and some other liquid tumors. However, there are shortcomings of clinical proteasome inhibitors, including severe toxicity, drug resistance, and no effect in solid tumors. At the same time, extensive research has been conducted in the areas of natural compounds and old drug repositioning towards the goal of discovering effective, economical, low toxicity proteasome-inhibitory anti-cancer drugs. A variety of dietary polyphenols, medicinal molecules, metallic complexes, and metal-binding compounds have been found to be able to selectively inhibit tumor cellular proteasomes and induce apoptotic cell death in vitro and in vivo, supporting the clinical success of specific 20S proteasome inhibitors bortezomib and carfilzomib. Therefore, the discovery of natural proteasome inhibitors and researching old drugs with proteasome-inhibitory properties may provide an alternative strategy for improving the current status of cancer treatment and even prevention.
Collapse
Affiliation(s)
- Claire L Soave
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, 540.1 HWCRC, 4100 John R Road, Detroit, MI, 48201-2013, USA
| | - Tracey Guerin
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, 540.1 HWCRC, 4100 John R Road, Detroit, MI, 48201-2013, USA
| | - Jinbao Liu
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, 540.1 HWCRC, 4100 John R Road, Detroit, MI, 48201-2013, USA.
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
82
|
Zahedipour F, Dalirfardouei R, Karimi G, Jamialahmadi K. Molecular mechanisms of anticancer effects of Glucosamine. Biomed Pharmacother 2017; 95:1051-1058. [PMID: 28922722 DOI: 10.1016/j.biopha.2017.08.122] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/09/2017] [Accepted: 08/29/2017] [Indexed: 12/19/2022] Open
Abstract
Glucosamine is an amino sugar that is produced naturally in human body. It is an essential carbohydrate component of many cellular glycoproteins, glycolipids, and glycosaminoglycans (GAGs). This popular over-the-counter supplement is also found in the exoskeleton of crustaceans. Glucosamine and its derivatives have a long history in medicine for inflammatory conditions specially to relieve arthritis. This dietary supplement has numerous biological and pharmacological properties, including anti-inflammatory, antioxidant, anti-aging, anti-fibrotic, neuroprotective and cardioprotective activities. Many studies have shown that glucosamine has anti-cancer activity through influence on biological pathways involved in cell death, apoptosis, cell proliferation, and angiogenesis. Accordingly, this comprehensive review summarizes anti-cancer molecular mechanisms of glucosamine in details.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Razieh Dalirfardouei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gholamreza Karimi
- Pharmaceutical Research Center and School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
83
|
Varga G, Mikala G, Kiss KP, Kosóczki É, Szabó E, Meggyesi N, Balassa K, Kövy P, Tegze B, Szombath G, Tordai A, Andrikovics H, Homolya L, Masszi T. Proteasome Subunit Beta Type 1 P11A Polymorphism Is a New Prognostic Marker in Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:734-742. [PMID: 28733196 DOI: 10.1016/j.clml.2017.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 06/14/2017] [Accepted: 06/27/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Proteasome subunit beta type 1 (PSMB1) rs12717 polymorphism, a single nucleotide polymorphism with unknown functional effect, was recently reported to influence response to bortezomib-based therapy in follicular lymphoma. PATIENTS AND METHODS We retrospectively analyzed the prognostic impact of this polymorphism in 211 consecutively diagnosed multiple myeloma cases, and performed in vitro experiments to look into its functional consequences. RESULTS On univariate analysis, patients carrying the variant G allele showed significantly shorter progression-free survival (PFS) with a pattern suggestive of a gene-dose effect (PFS 26.4, 22.3, and 16.4 months in C/C, C/G, and G/G patients, respectively, P = .002). On multivariate analysis, carrying the G/G genotype was a significant independent risk factor for relapse (hazard ratio [HR] 2.29, P < .001) with a similar trend in C/G carriers (HR 1.33, P = .097) when compared with the major allele carrier C/C cohort. Our subsequent in vitro analyses demonstrated significantly reduced protease activity in proteasomes of individuals with G/G genotype compared with that of C/C carriers, despite that PSMB1 expression and proteasome assembly remained unaltered. Bortezomib exhibited a lower inhibitory capacity on the caspase- and trypsin-like activity of proteasomes from G/G individuals. CONCLUSION Our results show that carriership of PSMB1 rs12717 minor allele is predictive for suboptimal response with bortezomib treatment, which could be explained by less active proteasomes that are less sensitive to bortezomib, and myeloma cells consequently relying on other escape mechanisms to cope with the abundance of misfolded proteins.
Collapse
Affiliation(s)
- Gergely Varga
- 3(rd) Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| | - Gábor Mikala
- Department of Haematology and Stem Cell Transplantation, St. István and St. László Hospital, Budapest, Hungary
| | - Katalin Piroska Kiss
- Laboratory of Molecular Diagnostics, Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - Éva Kosóczki
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Edit Szabó
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Nóra Meggyesi
- Laboratory of Molecular Diagnostics, Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - Katalin Balassa
- Laboratory of Molecular Diagnostics, Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - Petra Kövy
- Laboratory of Molecular Diagnostics, Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - Bálint Tegze
- 3(rd) Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Gergely Szombath
- 3(rd) Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Tordai
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Hajnalka Andrikovics
- Laboratory of Molecular Diagnostics, Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tamás Masszi
- 3(rd) Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
84
|
Structure of the Rpn13-Rpn2 complex provides insights for Rpn13 and Uch37 as anticancer targets. Nat Commun 2017; 8:15540. [PMID: 28598414 PMCID: PMC5494190 DOI: 10.1038/ncomms15540] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/07/2017] [Indexed: 12/16/2022] Open
Abstract
Proteasome-ubiquitin receptor hRpn13/Adrm1 binds and activates deubiquitinating enzyme Uch37/UCHL5 and is targeted by bis-benzylidine piperidone RA190, which restricts cancer growth in mice xenografts. Here, we solve the structure of hRpn13 with a segment of hRpn2 that serves as its proteasome docking site; a proline-rich C-terminal hRpn2 extension stretches across a narrow canyon of the ubiquitin-binding hRpn13 Pru domain blocking an RA190-binding surface. Biophysical analyses in combination with cell-based assays indicate that hRpn13 binds preferentially to hRpn2 and proteasomes over RA190. hRpn13 also exists outside of proteasomes where it may be RA190 sensitive. RA190 does not affect hRpn13 interaction with Uch37, but rather directly binds and inactivates Uch37. hRpn13 deletion from HCT116 cells abrogates RA190-induced accumulation of substrates at proteasomes. We propose that RA190 targets hRpn13 and Uch37 through parallel mechanisms and at proteasomes, RA190-inactivated Uch37 cannot disassemble hRpn13-bound ubiquitin chains.
Collapse
|
85
|
Verathamjamras C, Weeraphan C, Chokchaichamnankit D, Watcharatanyatip K, Subhasitanont P, Diskul-Na-Ayudthaya P, Mingkwan K, Luevisadpaibul V, Chutipongtanate S, Champattanachai V, Svasti J, Srisomsap C. Secretomic profiling of cells from hollow fiber bioreactor reveals PSMA3 as a potential cholangiocarcinoma biomarker. Int J Oncol 2017; 51:269-280. [PMID: 28560424 DOI: 10.3892/ijo.2017.4024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/05/2017] [Indexed: 11/06/2022] Open
Abstract
Cholangiocarcinoma (CCA), derived from the bile duct, occurs with a relatively high incidence in Northeast Thailand. Early diagnosis is still hampered by the lack of sufficient biomarkers. In recent years, biomarker discovery using secretomes has provided interesting results, including our studies on CCA secretomes, especially with three-dimensional cell cultures. Thus, cells cultured using the hollow fiber bioreactor (HFB) with 20 kDa molecular weight cut-off (MWCO) yielded higher quality and quantity of secretomes than those from conditioned media of the monolayer culture (MNC) system. In this study, we employed the HFB culture system with 5 kDa MWCO and compared conditioned media from the HFB and MNC systems using two-dimensional gel electrophoresis, followed by identifying proteins of interest by liquid chromatography and mass spectrometry (LC/MS/MS). Two out of 4 spots of NGAL or lipocalin-2 were found to show highest increase in expression of 19.93-fold and 18.79-fold in HFB compared to MNC. Interestingly, all 14 proteasome subunits including proteasome subunit α type-1 to type-7 and β type-1 to type-7 showed 2.92-fold to 12.13-fold increased expression in HFB. The protein-protein interactions of upregulated proteins were predicted, and one of the main interaction clusters involved 20S proteasome subunits. Proteasome activity in the HFB conditioned media was also found to be higher than that in MNC conditioned media. Three types of proteasome subunit were also validated by immunoblotting and showed higher expression in the HFB system compared to MNC system. Proteasome subunit α type-3 (PSMA3) showed the highest level in plasma of cholangiocarcinoma patients compared to normal and hepatocellular carcinoma patients by immunodetection, and is of interest as a potential biomarker for cholangiocarcinoma.
Collapse
Affiliation(s)
- Chris Verathamjamras
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Churat Weeraphan
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | | | | | | | | | - Kanokwan Mingkwan
- Department of Surgery, Sappasitthiprasong Hospital, Ubon Ratchathani 34000, Thailand
| | - Virat Luevisadpaibul
- Division of Information and Technology, Ubonrak Thonburi Hospital, Ubon Ratchathani 34000, Thailand
| | - Somchai Chutipongtanate
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | | | - Jisnuson Svasti
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Chantragan Srisomsap
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| |
Collapse
|
86
|
Park HJ, Kim SH, Moon DO. Growth inhibition of human breast carcinoma cells by overexpression of regulator of G-protein signaling 4. Oncol Lett 2017; 13:4357-4363. [PMID: 28588709 DOI: 10.3892/ol.2017.6009] [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: 04/13/2015] [Accepted: 07/15/2016] [Indexed: 01/05/2023] Open
Abstract
Breast cancer remains the second largest cause of mortality in women with cancer and does not respond well to conventional therapies. Regulator of G-protein signaling 4 (RGS4) is a GTPase-activating protein of the heterotrimeric Gq and Gi proteins. Altered levels of RGS4 are reportedly linked with several human diseases, including cancer. The present study investigated whether overexpression of RGS4 inhibited the growth of human breast cancer cells. Protein expression was investigated by western blot analysis. Cell viability and apoptosis were analyzed by MTT assay and flow cytometric analysis, respectively. Cell cycle analysis was performed using propidium iodide staining in order to examine the anti-proliferative function of increased RGS4 levels. Next, changes in the expression levels of G2/M cell cycle-related proteins were examined. Overexpression of RGS4 led to the upregulation of phosphorylayed (p)-Ser216 cell division cycle (Cdc)25C and p-Tyr15 Cdc2. Importantly, MG132-induced proteasome blockade prevented degradation of RGS4. Suppression of proliferation was associated with G2/M-phase cell cycle arrest. Furthermore, enhanced endogenous RGS4 protein levels significantly inhibited breast cancer cell growth, which was reversed by a pharmacological inhibitor of RGS4. Taken together, these results suggest that overexpression of RGS4 in human breast cancer cells by molecular means may offer a potential therapeutic approach.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Biology Education, Daegu University, Gyeongsan, Gyeongsangbuk-do 38453, Republic of Korea
| | - Seung-Hyun Kim
- Department of Biology Education, Daegu University, Gyeongsan, Gyeongsangbuk-do 38453, Republic of Korea
| | - Dong-Oh Moon
- Department of Biology Education, Daegu University, Gyeongsan, Gyeongsangbuk-do 38453, Republic of Korea
| |
Collapse
|
87
|
Voutsadakis IA. Proteasome expression and activity in cancer and cancer stem cells. Tumour Biol 2017; 39:101042831769224. [DOI: 10.1177/1010428317692248] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Proteasome is a multi-protein organelle that participates in cellular proteostasis by destroying damaged or short-lived proteins in an organized manner guided by the ubiquitination signal. By being in a central place in the cellular protein complement homeostasis, proteasome is involved in virtually all cell processes including decisions on cell survival or death, cell cycle, and differentiation. These processes are important also in cancer, and thus, the proteasome is an important regulator of carcinogenesis. Cancers include a variety of cells which, according to the cancer stem cell theory, descend from a small percentage of cancer stem cells, alternatively termed tumor-initiating cells. These cells constitute the subsets that have the ability to propagate the whole variety of cancer and repopulate tumors after cytostatic therapies. Proteasome plays a role in cellular processes in cancer stem cells, but it has been found to have a decreased function in them compared to the rest of cancer cells. This article will discuss the transcriptional regulation of proteasome sub-unit proteins in cancer and in particular cancer stem cells and the relationship of the proteasome with the pluripotency that is the defining characteristic of stem cells. Therapeutic opportunities that present from the understanding of the proteasome role will also be discussed.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste. Marie, ON, Canada
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
88
|
Vriend J, Marzban H. The ubiquitin-proteasome system and chromosome 17 in cerebellar granule cells and medulloblastoma subgroups. Cell Mol Life Sci 2017; 74:449-467. [PMID: 27592301 PMCID: PMC11107675 DOI: 10.1007/s00018-016-2354-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
Chromosome 17 abnormalities are often observed in medulloblastomas (MBs), particularly those classified in the consensus Groups 3 and 4. Herein we review MB signature genes associated with chromosome 17 and the relationship of these signature genes to the ubiquitin-proteasome system. While clinical investigators have not focused on the ubiquitin-proteasome system in relation to MB, a substantial amount of data on the topic has been hidden in the form of supplemental datasets of gene expression. A supplemental dataset associated with the Thompson classification of MBs shows that a subgroup of MB with 17p deletions is characterized by reduced expression of genes for several core particle subunits of the beta ring of the proteasome (β1, β4, β5, β7). One of these genes (PSMB6, the gene for the β1 subunit) is located on chromosome 17, near the telomeric end of 17p. By comparison, in the WNT group of MBs only one core proteasome subunit, β6, associated with loss of a gene (PSMB1) on chromosome 6, was down-regulated in this dataset. The MB subgroups with the worst prognosis have a significant association with chromosome 17 abnormalities and irregularities of APC/C cyclosome genes. We conclude that the expression of proteasome subunit genes and genes for ubiquitin ligases can contribute to prognostic classification of MBs. The therapeutic value of targeting proteasome subunits and ubiquitin ligases in the various subgroups of MB remains to be determined separately for each classification of MB.
Collapse
Affiliation(s)
- Jerry Vriend
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Rm134, BMSB, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| | - Hassan Marzban
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Rm134, BMSB, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba (CHRIM), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
89
|
Chroma K, Mistrik M, Moudry P, Gursky J, Liptay M, Strauss R, Skrott Z, Vrtel R, Bartkova J, Kramara J, Bartek J. Tumors overexpressing RNF168 show altered DNA repair and responses to genotoxic treatments, genomic instability and resistance to proteotoxic stress. Oncogene 2016; 36:2405-2422. [DOI: 10.1038/onc.2016.392] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/14/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022]
|
90
|
Colonic Lamina Propria Inflammatory Cells from Patients with IBD Induce the Nuclear Factor-E2 Related Factor-2 Thereby Leading to Greater Proteasome Activity and Apoptosis Protection in Human Colonocytes. Inflamm Bowel Dis 2016; 22:2593-2606. [PMID: 27661668 DOI: 10.1097/mib.0000000000000925] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The antioxidant transcription factor Nrf2 confers broad cytoprotection and has a dual role in tumorigenesis. Enhancing proteasome activity is one mechanism by which Nrf2 can promote cancer development, e.g., colorectal cancer. This study investigated whether this potential oncogenic effect of Nrf2 emerges already from the epithelial adaptation to persistent oxidative stress during inflammatory bowel disease (IBD). METHODS Reactive oxygen species (ROS)-producing inflammatory myeloid cells (IMCs) from colon tissue of patients with IBD were cocultured with human NCM460 colonocytes. ARE-luciferase-, c-H2DCF-DA-assays, Western blotting, and quantitative polymerase chain reaction were performed for assessing Nrf2-activity, intracellular ROS-level, and Nrf2-target gene expression. Proteasome activity was quantified by Suc-LLVY-amido-4-methylcumarin-assay, and apoptosis by caspase-3/-7 assay and PARP1-Western blots. Nrf2, proteasome proteins, and IMCs were analyzed in IBD-tissues by immunohistochemistry. RESULTS IMC-coculture caused a temporary increase of ROS in NCM460, followed by Nrf2 activation and elevated expression of ROS-protecting enzymes (NQO1, GCLC). This was accompanied by Nrf2-dependent expression of proteasome proteins (PSMD4, PSMA5) and an enhanced proteasome activity in IMC-cocultured NCM460. Nrf2-siRNA or the ROS-scavenger Tiron blocked these alterations. Depending on Nrf2-induced proteasome activity, IMC-cocultured NCM460 or Colo320 cancer cells were less sensitive to apoptosis (TRAIL-/etoposide induced). Immunostaining of IBD-tissues confirmed Nrf2 activation in the colonic epithelium within inflamed areas, along with greater proteasome protein expression. CONCLUSIONS IMC/NCM460-coculture experiments and immunohistochemistry of colonic tissues from patients with IBD reveal a Nrf2-dependent adaptation of colon epithelial cells to oxidative stress caused by inflammatory cells. This involves increased proteasome activity and apoptosis resistance that protect from tissue damage due to colitis on one hand, but on the other hand, may favor carcinogenesis.
Collapse
|
91
|
Mokarram P, Shakiba-Jam F, Kavousipour S, Sarabi MM, Seghatoleslam A. Promoter Methylation Status of Two Novel Human Genes, UBE2Q1 and UBE2Q2, in Colorectal Cancer: a New Finding in Iranian Patients. Asian Pac J Cancer Prev 2016; 16:8247-52. [PMID: 26745068 DOI: 10.7314/apjcp.2015.16.18.8247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The ubiquitin-proteasome system (UPS) degrades a variety of proteins which attach to specific signals. The ubiquitination pathway facilitates degradation of damaged proteins and regulates growth and stress responses. This pathway is altered in various cancers, including acute lymphoblastic leukemia, head and neck squamous cell carcinoma and breast cancer. Recently it has been reported that expression of newly characterized human genes, UBE2Q1 and UBE2Q2, putative members of ubiquitin-conjugating enzyme family (E2), has been also changed in colorectal cancer. Epigenetics is one of the fastest-growing areas of science and nowadays has become a central issue in biological studies of diseases. According to the lack of information about the role of epigenetic changes on gene expression profiling of UBE2Q1 and UBE2Q2, and the presence of CpG islands in the promoter of these two human genes, we decided to evaluate the promoter methylation status of these genes as a first step. MATERIALS AND METHODS The promoter methylation status of UBE2Q1 and UBE2Q2 was studied by methylation-specific PCR (MSP) in tumor samples of 60 colorectal cancer patients compared to adjacent normal tissues and 20 non-malignant controls. The frequency of the methylation for each gene was analyzed by chi-square method. RESULTS MSP results revealed that UBE2Q2 gene promoter were more unmethylated, while a higher level of methylated allele was observed for UBE2Q1 in tumor tissues compared to the adjacent normal tissues and the non malignant controls. CONCLUSIONS UBE2Q1 and UBE2Q2 genes show different methylation profiles in CRC cases.
Collapse
Affiliation(s)
- Pooneh Mokarram
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran E-mail :
| | | | | | | | | |
Collapse
|
92
|
Identification of noncovalent proteasome inhibitors with high selectivity for chymotrypsin-like activity by a multistep structure-based virtual screening. Eur J Med Chem 2016; 121:578-591. [DOI: 10.1016/j.ejmech.2016.05.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/04/2016] [Accepted: 05/21/2016] [Indexed: 02/03/2023]
|
93
|
Kwon CH, Park HJ, Choi YR, Kim A, Kim HW, Choi JH, Hwang CS, Lee SJ, Choi CI, Jeon TY, Kim DH, Kim GH, Park DY. PSMB8 and PBK as potential gastric cancer subtype-specific biomarkers associated with prognosis. Oncotarget 2016; 7:21454-21468. [PMID: 26894977 PMCID: PMC5008298 DOI: 10.18632/oncotarget.7411] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/05/2016] [Indexed: 11/25/2022] Open
Abstract
Gastric adenocarcinoma is a common form of cancer associated with a poor prognosis. We analyzed microarray profiling data from 48 patients with gastric adenocarcinoma to characterize gastric cancer subtypes and identify biomarkers associated with prognosis. We identified two major subtypes of gastric adenocarcinoma differentially associated with overall survival (P = 0.025). Genes that were differentially expressed were identified using specific criteria (P < 0.001 and >1.5-fold); expression of 294 and 116 genes was enriched in good and poor prognosis subtypes, respectively. Genes related to translational elongation and cell cycle were upregulated in the poor prognosis group. Of these genes, upregulation of proteasome subunit beta type 8 PSMB8 and PDZ binding kinase PBK was confirmed by real-time reverse transcription-PCR analysis. PSMB8 or PBK knockdown had no effect on gastric cancer cell proliferation but suppressed cell migration and invasion, respectively. Furthermore, immunohistochemistry analysis of 385 gastric cancer patients revealed that increased nuclear expression of PSMB8 and PBK was correlated with depth of invasion, lymph node metastasis, and lower survival rates. Taken together, two gastric adenocarcinoma subtypes were predictive of prognosis. PSMB8 and PBK were predictive of gastric cancer prognosis and could be potential gastric cancer subtype-specific biomarkers.
Collapse
Affiliation(s)
- Chae Hwa Kwon
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Hye Ji Park
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Yu Ri Choi
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Ahrong Kim
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Hye Won Kim
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Jin Hwa Choi
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Chung Su Hwang
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - So Jung Lee
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Chang In Choi
- Department of Surgery, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Tae Yong Jeon
- Department of Surgery, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Dae Hwan Kim
- Department of Surgery, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Gwang Ha Kim
- Department of Internal Medicine, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| | - Do Youn Park
- Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, and BioMedical Research Institute, Pusan National University Hospital, Seo-Gu, Busan, Korea
| |
Collapse
|
94
|
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: 1.2] [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
|
95
|
Pundir S, Vu HY, Solomon VR, McClure R, Lee H. VR23: A Quinoline-Sulfonyl Hybrid Proteasome Inhibitor That Selectively Kills Cancer via Cyclin E-Mediated Centrosome Amplification. Cancer Res 2015; 75:4164-75. [PMID: 26238784 DOI: 10.1158/0008-5472.can-14-3370] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 06/30/2015] [Indexed: 11/16/2022]
Abstract
The proteasome is clinically validated as a target for cancer therapeutics. However, proteasome-inhibitory agents that are cancer selective have yet to be developed. In this study, we report the identification of a safe and effective proteasome inhibitor with selective anticancer properties. We screened a chemical library constructed using a hybrid approach that incorporated a 4-piperazinylquinoline scaffold and a sulfonyl phamarcophore. From this library, we identified 7-chloro-4-(4-(2,4-dinitrophenylsulfonyl)piperazin-1-yl)quinoline (VR23) as a small molecule that potently inhibited the activities of trypsin-like proteasomes (IC50 = 1 nmol/L), chymotrypsin-like proteasomes (IC50 = 50-100 nmol/L), and caspase-like proteasomes (IC50 = 3 μmol/L). Data from molecular docking and substrate competition assays established that the primary molecular target of VR23 was β2 of the 20S proteasome catalytic subunit. Notably, VR23 was structurally distinct from other known proteasome inhibitors and selectively killed cancer cells by apoptosis, with little effect on noncancerous cells. Mechanistic investigations showed that cancer cells exposed to VR23 underwent an abnormal centrosome amplification cycle caused by the accumulation of ubiquitinated cyclin E. In combinations with the clinically approved chymotrypsin-like proteasome inhibitor bortezomib, VR23 produced a synergistic effect in killing multiple myeloma cells, including those that were resistant to bortezomib. VR23 was effective in vivo in controlling multiple myelomas and metastatic breast cancer cells, in the latter case also enhancing the antitumor activity of paclitaxel while reducing its side effects. Overall, our results identify VR23 as a structurally novel proteasome inhibitor with desirable properties as an anticancer agent.
Collapse
Affiliation(s)
- Sheetal Pundir
- Advanced Medical Research Institute of Canada, Sudbury, Ontario, Canada. Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Hai-Yen Vu
- Advanced Medical Research Institute of Canada, Sudbury, Ontario, Canada
| | - V Raja Solomon
- Advanced Medical Research Institute of Canada, Sudbury, Ontario, Canada
| | - Rebecca McClure
- Advanced Medical Research Institute of Canada, Sudbury, Ontario, Canada. Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Hoyun Lee
- Advanced Medical Research Institute of Canada, Sudbury, Ontario, Canada. Department of Medicine, University of Ottawa, Faculty of Medicine, Ottawa, Ontario, Canada. Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada. Northern Ontario School of Medicine, Sudbury, Ontario, Canada.
| |
Collapse
|
96
|
Yang S, Lu M, Chen Y, Meng D, Sun R, Yun D, Zhao Z, Lu D, Li Y. Overexpression of eukaryotic elongation factor 1 alpha-2 is associated with poorer prognosis in patients with gastric cancer. J Cancer Res Clin Oncol 2015; 141:1265-75. [PMID: 25601347 DOI: 10.1007/s00432-014-1897-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/15/2014] [Indexed: 01/14/2023]
Abstract
PURPOSE Eukaryotic elongation factor 1 alpha-2 (eEF1A2) is a protein translation factor involved in protein synthesis. It is overexpressed in various cancers, which indicates potential vital functions in tumorigenesis and progression. Our study aims to investigate the expression levels of eEF1A2 in gastric cancer and its roles in clinical practice. METHODS A total of 129 patients with pathologically confirmed gastric cancer and 24 normal controls were recruited for this study. The expression levels of eEF1A2 in gastric cancer and normal tissues were evaluated by tissue microarrays, quantitative real-time PCR, and western blot analysis. Kaplan-Meier analysis and Cox's proportional hazards model were used in survival analysis. RESULTS Compared with corresponding controls, gastric cancer specimens had significantly increased expressions of eEF1A2 at mRNA and protein levels (both P < 0.05). Moreover, multivariate analysis confirmed that overexpression of eEF1A2 was a significant and independent indicator for predicting poor prognosis of gastric cancer. CONCLUSIONS Our results showed for the first time that overexpression of eEF1A2 was correlated with worse outcomes in gastric cancer patients, suggesting its critical roles in the carcinogenesis of gastric cancer.
Collapse
Affiliation(s)
- Song Yang
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Zhang X, Schulz R, Edmunds S, Krüger E, Markert E, Gaedcke J, Cormet-Boyaka E, Ghadimi M, Beissbarth T, Levine A, Moll U, Dobbelstein M. MicroRNA-101 Suppresses Tumor Cell Proliferation by Acting as an Endogenous Proteasome Inhibitor via Targeting the Proteasome Assembly Factor POMP. Mol Cell 2015; 59:243-57. [DOI: 10.1016/j.molcel.2015.05.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/04/2015] [Accepted: 05/26/2015] [Indexed: 12/21/2022]
|
98
|
Abstract
Defects in the maintenance of protein homeostasis, or proteostasis, has emerged as an underlying feature of a variety of human pathologies, including aging-related diseases. Proteostasis is achieved through the coordinated action of cellular systems overseeing amino acid availability, mRNA translation, protein folding, secretion, and degradation. The regulation of these distinct systems must be integrated at various points to attain a proper balance. In a recent study, we found that the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) pathway, well known to enhance the protein synthesis capacity of cells while concordantly inhibiting autophagy, promotes the production of more proteasomes. Activation of mTORC1 genetically, through loss of the tuberous sclerosis complex (TSC) tumor suppressors, or physiologically, through growth factors or feeding, stimulates a transcriptional program involving the sterol-regulatory element binding protein 1 (SREBP1) and nuclear factor erythroid-derived 2-related factor 1 (NRF1; also known as NFE2L1) transcription factors leading to an increase in cellular proteasome content. As discussed here, our findings suggest that this increase in proteasome levels facilitates both the maintenance of proteostasis and the recovery of amino acids in the face of an increased protein load consequent to mTORC1 activation. We also consider the physiological and pathological implications of this unexpected new downstream branch of mTORC1 signaling.
Collapse
Affiliation(s)
- Yinan Zhang
- a Department of Genetics and Complex Diseases; Harvard T.H. Chan School of Public Health ; Boston , MA , USA
| | | |
Collapse
|
99
|
Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2015; 2:e974463. [PMID: 27308423 PMCID: PMC4904962 DOI: 10.4161/23723556.2014.974463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/12/2023]
Abstract
The so-called "ubiquitin-proteasome system" (UPS) is a multicomponent molecular apparatus that catalyzes the covalent attachment of several copies of the small protein ubiquitin to other proteins that are generally (but not always) destined to proteasomal degradation. This enzymatic cascade is crucial for the maintenance of intracellular protein homeostasis (both in physiological conditions and in the course of adaptive stress responses), and regulates a wide array of signaling pathways. In line with this notion, defects in the UPS have been associated with aging as well as with several pathological conditions including cardiac, neurodegenerative, and neoplastic disorders. As transformed cells often experience a constant state of stress (as a result of the hyperactivation of oncogenic signaling pathways and/or adverse microenvironmental conditions), their survival and proliferation are highly dependent on the integrity of the UPS. This rationale has driven an intense wave of preclinical and clinical investigation culminating in 2003 with the approval of the proteasomal inhibitor bortezomib by the US Food and Drug Administration for use in multiple myeloma patients. Another proteasomal inhibitor, carfilzomib, is now licensed by international regulatory agencies for use in multiple myeloma patients, and the approved indications for bortezomib have been extended to mantle cell lymphoma. This said, the clinical activity of bortezomib and carfilzomib is often limited by off-target effects, innate/acquired resistance, and the absence of validated predictive biomarkers. Moreover, the antineoplastic activity of proteasome inhibitors against solid tumors is poor. In this Trial Watch we discuss the contribution of the UPS to oncogenesis and tumor progression and summarize the design and/or results of recent clinical studies evaluating the therapeutic profile of proteasome inhibitors in cancer patients.
Collapse
Affiliation(s)
- Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “Tor Vergata”
| | - Lorenzo Galluzzi
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
100
|
Pacheco MTF, Berra CM, Morais KLP, Sciani JM, Branco VG, Bosch RV, Chudzinski-Tavassi AM. Dynein function and protein clearance changes in tumor cells induced by a Kunitz-type molecule, Amblyomin-X. PLoS One 2014; 9:e111907. [PMID: 25479096 PMCID: PMC4257547 DOI: 10.1371/journal.pone.0111907] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/02/2014] [Indexed: 01/07/2023] Open
Abstract
Amblyomin-X is a Kunitz-type recombinant protein identified from the transcriptome of the salivary glands of the tick Amblyomma cajennense and has anti-coagulant and antitumoral activity. The supposed primary target of this molecule is the proteasome system. Herein, we elucidated intracellular events that are triggered by Amblyomin-X treatment in an attempt to provide new insight into how this serine protease inhibitor, acting on the proteasome, could be comparable with known proteasome inhibitors. The collective results showed aggresome formation after proteasome inhibition that appeared to occur via the non-exclusive ubiquitin pathway. Additionally, Amblyomin-X increased the expression of various chains of the molecular motor dynein in tumor cells, modulated specific ubiquitin linkage signaling and inhibited autophagy activation by modulating mTOR, LC3 and AMBRA1 with probable dynein involvement. Interestingly, one possible role for dynein in the mechanism of action of Amblyomin-X was in the apoptotic response and its crosstalk with autophagy, which involved the factor Bim; however, we observed no changes in the apoptotic response related to dynein in the experiments performed. The characteristics shared among Amblyomin-X and known proteasome inhibitors included NF-κB blockage and nascent polypeptide-dependent aggresome formation. Therefore, our study describes a Kunitz-type protein that acts on the proteasome to trigger distinct intracellular events compared to classic known proteasome inhibitors that are small-cell-permeable molecules. In investigating the experiments and literature on Amblyomin-X and the known proteasome inhibitors, we also found differences in the structures of the molecules, intracellular events, dynein involvement and tumor cell type effects. These findings also reveal a possible new target for Amblyomin-X, i.e., dynein, and may serve as a tool for investigating tumor cell death associated with proteasome inhibition.
Collapse
Affiliation(s)
- Mario T. F. Pacheco
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Carolina M. Berra
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Kátia L. P. Morais
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
- Department of Biochemistry, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana M. Sciani
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Vania G. Branco
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | - Rosemary V. Bosch
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, Brazil
| | | |
Collapse
|