1
|
Świderska-Kołacz G, Madej M, Zmorzynski S, Styk W, Surowiec I, Witek B, Wojciechowska A, Czerwik-Marcinkowska J, Nowakowska A. Effects of bortezomib on intracellular antioxidant and apoptosis in HepG2cells. PeerJ 2025; 13:e19235. [PMID: 40313384 PMCID: PMC12045286 DOI: 10.7717/peerj.19235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/10/2025] [Indexed: 05/03/2025] Open
Abstract
Bortezomib, as a proteasome inhibitor, is used in clinical trials related to solid cancers. However, its use is not always associated with a good response to treatment. Taking into account the above, we decided to analyze the effect of the time-dependency (24 vs. 48 h) and the dose-dependency of bortezomib (2, 4, 8 and 16 nM) on apoptosis and activities of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GR), glutathione peroxidase (GPx) and glutathione transferase (GST), as well as concentrations of reduced glutathione (GSH) and malondialdehyde (MDA) in hepatoblastoma cell line (HepG2) cells. We have shown that increasing concentrations of bortezomib caused (I) a gradual decrease in the levels of GSH; (II) changes in MDA concentrations and antioxidant enzymes activities; (III) increase in apoptosis levels in HepG2 cells. We did not find significant association between antioxidant parameters and number of apoptotic cells. Our study showed that the analyzed parameters (such as: CAT, SOD, GR, GPx, GST, GSH, MDA) changed after bortezomib treatment. It is important to search for new anti-cancer therapies based on next-generation proteasome inhibitors. It is possible that the use of proteins associated with oxidative stress will help enhance the action of these inhibitors and will provide a better treatment effect.
Collapse
Affiliation(s)
| | - Magdalena Madej
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | | | - Wojciech Styk
- Academic Laboratory of Psychological Tests, Medical University of Lublin, Lublin, Poland
| | - Iwona Surowiec
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Bożena Witek
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Anna Wojciechowska
- Department of Geobotany and Landscape Planning, Nicolaus Copernicus University of Torun, Toruń, Poland
| | | | - Anna Nowakowska
- Department of Animal Physiology and Neurobiology, Nicolaus Copernicus University of Torun, Toruń, Poland
| |
Collapse
|
2
|
Cebeci E, Yüksel B, Şahin F. Anti-cancer effects of boron derivatives on non-small cell lung cancer. J Trace Elem Med Biol 2025; 88:127627. [PMID: 40073680 DOI: 10.1016/j.jtemb.2025.127627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Lung cancer is the most frequently diagnosed cancer globally, with 2.5 million new cases representing 12.4 % of the total number of new cases. Small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) are two main subtypes of lung cancer that have different growth and dissemination capacities. Treatment options for lung cancer include surgery, radiation therapy, chemotherapy, and targeted therapy, including Bortezomib, an FDA-approved boron-derived agent. To bring novel boron-containing compounds to the armamentarium of lung cancer therapy, this study aimed to unravel the anti-cancer potential of Boric Acid (BA) and Sodium pentaborate pentahydrate (NaB) on Non-Small Cell Lung Cancer cell line A549. According to the results, the survival rates of A549 cells decreased after BA and NaB treatments by 5.5-fold and 5.2-fold, respectively. The number of colony-forming units was reduced by 3-fold and 2.4-fold after BA and NaB treatments, respectively. Gene expressions and protein levels of some pro-apoptotic and tumor suppressor genes were upregulated and some anti-apoptotic genes were downregulated. Cell cycle arrest at the G2/M phase was detected after treatment with both boron. Proliferation markers Ki-67 and PCNA, and checkpoint regulators CDK1 and CDK2 gene expression levels were downregulated after BA and NaB treatments in the A549 cells. Our findings point to a promising selective anti-cancer activity of BA and NaB in the A549 cells without exerting any adverse effects on normal cells. Further in vitro and in vivo studies are required to get detailed information about the anti-cancer mechanism of BA, and NaB on lung cancer.
Collapse
Affiliation(s)
- Emre Cebeci
- Yeditepe University, Department of Genetics and Bioengineering, Faculty of Engineering, Istanbul, Turkey
| | - Büşra Yüksel
- Yeditepe University, Department of Genetics and Bioengineering, Faculty of Engineering, Istanbul, Turkey
| | - Fikrettin Şahin
- Yeditepe University, Department of Genetics and Bioengineering, Faculty of Engineering, Istanbul, Turkey.
| |
Collapse
|
3
|
Zhou SS, Ye YP, Chen Y, Zeng DT, Zheng GC, He RQ, Chi BT, Wang L, Lin Q, Su QY, Dang YW, Chen G, Wei JL. Overexpression pattern, function, and clinical value of proteasome 26S subunit non-ATPase 6 in hepatocellular carcinoma. World J Clin Oncol 2025; 16:99839. [PMID: 39995557 PMCID: PMC11686554 DOI: 10.5306/wjco.v16.i2.99839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND In recent years, many studies have shown that proteasome 26S subunit non-ATPase 6 (PSMD6) plays an important role in the occurrence and development of malignant tumours. Unfortunately, there are no reports on the evaluation of the potential role of PSMD6 in hepatocellular carcinoma (HCC). AIM To comprehensively evaluate the overexpression pattern and clinical significance of PSMD6 in HCC tissues. METHODS This study integrated PSMD6 mRNA expression profiles from 4672 HCC and 3667 non-HCC tissues, along with immunohistochemical scores from 383 HCC and adjacent tissues, to assess PSMD6 overexpression in HCC. Clustered regularly interspaced short palindromic repeats knockout technology evaluated PSMD6's essential role in HCC cell growth. Functional enrichment analysis explored the molecular mechanism of PSMD6 abnormalities in HCC. Drug sensitivity analysis and molecular docking analysed the effect of abnormal expression of PSMD6 on the drug sensitivity of HCC cells. RESULTS The results of 41 external and two internal datasets showed that PSMD6 mRNA (SMD = 0.26, 95%CI: 0.09-0.42, P < 0.05) and protein (SMD = 2.85, 95%CI: 1.19-4.50, P < 0.05) were significantly overexpressed in HCC tissues. The integrated analysis results showed that PSMD6 had a significant overexpression pattern in HCC tissues (SMD = 0.40, 95%CI: 0.15-0.66, P < 0.05). PSMD6 knockout inhibited HCC cell growth (chronos scores < -1). Functional enrichment implicated ribosome biogenesis and RNA splicing. Significant enrichment of signalling pathways such as RNA degradation, ribosomes, and chemical carcinogenesis-reactive oxygen species. Drug sensitivity analysis and a molecular docking model showed that high expression of PSMD6 was associated with the tolerance of HCC cells to drugs such as ML323, sepantronium bromide, and GDC0810. Overexpressed PSMD6 effectively distinguished HCC tissues (AUC = 0.75, 95%CI: 0.71-0.79). CONCLUSION This study was the first to discover that PSMD6 was overexpressed in HCC tissues. PSMD6 is essential for the growth of HCC cells and may be involved in ribosome biogenesis and RNA splicing.
Collapse
Affiliation(s)
- Sheng-Sheng Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yu-Ping Ye
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yi Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Da-Tong Zeng
- Department of Pathology, Redcross Hospital of Yulin City, Yulin 537000, Guangxi Zhuang Autonomous Region, China
| | - Guang-Cai Zheng
- Department of Surgery, Redcross Hospital of Yulin City, Yulin 537000, Guangxi Zhuang Autonomous Region, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Bang-Teng Chi
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Qian Lin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Qin-Yan Su
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jia-Liang Wei
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
4
|
Bian Z, Benjamin MM, Bialousow L, Tian Y, Hobbs GA, Karan D, Choo YM, Hamann MT, Wang X. Targeting sine oculis homeoprotein 1 (SIX1): A review of oncogenic roles and potential natural product therapeutics. Heliyon 2024; 10:e33204. [PMID: 39022099 PMCID: PMC11252760 DOI: 10.1016/j.heliyon.2024.e33204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sine oculis homeoprotein 1 (SIX1), a prominent representative of the homeodomain transcription factors within the SIX family, has attracted significant interest owing to its role in tumorigenesis, cancer progression, and prognostic assessments. Initially recognized for its pivotal role in embryonic development, SIX1 has emerged as a resurgent factor across a diverse set of mammalian cancers. Over the past two decades, numerous investigations have emphasized SIX1's dual significance as a developmental regulator and central player in oncogenic processes. A mounting body of evidence links SIX1 to the initiation of diverse cancers, encompassing enhanced cellular metabolism and advancement. This review provides an overview of the multifaceted roles of SIX1 in both normal development and oncogenic processes, emphasizing its importance as a possible therapeutic target and prognostic marker. Additionally, this review discusses the natural product agents that inhibit various pro-oncogenic mechanisms associated with SIX1.
Collapse
Affiliation(s)
- Zhiwei Bian
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Menny M. Benjamin
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Lucas Bialousow
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Yintai Tian
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - G. Aaron Hobbs
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Dev Karan
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yeun-Mun Choo
- Chemistry Department, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mark T. Hamann
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| |
Collapse
|
5
|
Maddaloni M, Farra R, Dapas B, Felluga F, Benedetti F, Berti F, Drioli S, Vidali M, Cemazar M, Kamensek U, Brancolini C, Murano E, Maremonti F, Grassi M, Biasin A, Rizzolio F, Cavarzerani E, Scaggiante B, Bulla R, Balduit A, Ricci G, Zito G, Romano F, Bonin S, Azzalini E, Baj G, Tierno D, Grassi G. In Vitro and In Vivo Evaluation of the Effects of Drug 2c and Derivatives on Ovarian Cancer Cells. Pharmaceutics 2024; 16:664. [PMID: 38794326 PMCID: PMC11125437 DOI: 10.3390/pharmaceutics16050664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND The identification of novel therapeutic strategies for ovarian cancer (OC), the most lethal gynecological neoplasm, is of utmost urgency. Here, we have tested the effectiveness of the compound 2c (4-hydroxy-2,6-bis(4-nitrobenzylidene)cyclohexanone 2). 2c interferes with the cysteine-dependent deubiquitinating enzyme (DUB) UCHL5, thus affecting the ubiquitin-proteasome-dependent degradation of proteins. METHODS 2c phenotypic/molecular effects were studied in two OC 2D/3D culture models and in a mouse xenograft model. Furthermore, we propose an in silico model of 2c interaction with DUB-UCHL5. Finally, we have tested the effect of 2c conjugated to several linkers to generate 2c/derivatives usable for improved drug delivery. RESULTS 2c effectively impairs the OC cell line and primary tumor cell viability in both 2D and 3D conditions. The effectiveness is confirmed in a xenograft mouse model of OC. We show that 2c impairs proteasome activity and triggers apoptosis, most likely by interacting with DUB-UCHL5. We also propose a mechanism for the interaction with DUB-UCHL5 via an in silico evaluation of the enzyme-inhibitor complex. 2c also reduces cell growth by down-regulating the level of the transcription factor E2F1. Eventually, 2c activity is often retained after the conjugation with linkers. CONCLUSION Our data strongly support the potential therapeutic value of 2c/derivatives in OC.
Collapse
Affiliation(s)
- Marianna Maddaloni
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
| | - Rossella Farra
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
| | - Fulvia Felluga
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (F.F.); (F.B.); (F.B.); (S.D.); (M.V.)
| | - Fabio Benedetti
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (F.F.); (F.B.); (F.B.); (S.D.); (M.V.)
| | - Federico Berti
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (F.F.); (F.B.); (F.B.); (S.D.); (M.V.)
| | - Sara Drioli
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (F.F.); (F.B.); (F.B.); (S.D.); (M.V.)
| | - Mattia Vidali
- Department of Chemical and Pharmaceutical Sciences (DSCF), University of Trieste, 34127 Trieste, Italy; (F.F.); (F.B.); (F.B.); (S.D.); (M.V.)
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (M.C.); (U.K.)
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (M.C.); (U.K.)
| | - Claudio Brancolini
- Laboratory of Epigenomics, Department of Medicine, University of Udine, Piazzale Kolbe 4, 33100 Udine, Italy;
| | | | - Francesca Maremonti
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, 34127 Trieste, Italy; (M.G.); (A.B.)
| | - Alice Biasin
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, 34127 Trieste, Italy; (M.G.); (A.B.)
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30172 Venice, Italy;
| | - Enrico Cavarzerani
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30172 Venice, Italy;
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.B.); (G.B.)
| | - Andrea Balduit
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (A.B.); (G.R.); (G.Z.); (F.R.)
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (A.B.); (G.R.); (G.Z.); (F.R.)
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy; (S.B.); (E.A.)
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (A.B.); (G.R.); (G.Z.); (F.R.)
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (A.B.); (G.R.); (G.Z.); (F.R.)
| | - Serena Bonin
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy; (S.B.); (E.A.)
| | - Eros Azzalini
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy; (S.B.); (E.A.)
| | - Gabriele Baj
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.B.); (G.B.)
| | - Domenico Tierno
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, 34149 Trieste, Italy; (M.M.); (R.F.); (B.D.); (F.M.); (B.S.)
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy; (S.B.); (E.A.)
| |
Collapse
|
6
|
Tonon F, Farra R, Zennaro C, Pozzato G, Truong N, Parisi S, Rizzolio F, Grassi M, Scaggiante B, Zanconati F, Bonazza D, Grassi G, Dapas B. Xenograft Zebrafish Models for the Development of Novel Anti-Hepatocellular Carcinoma Molecules. Pharmaceuticals (Basel) 2021; 14:803. [PMID: 34451900 PMCID: PMC8400454 DOI: 10.3390/ph14080803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common type of tumor and the second leading cause of tumor-related death worldwide. Liver cirrhosis is the most important predisposing factor for HCC. Available therapeutic approaches are not very effective, especially for advanced HCC, which is the most common form of the disease at diagnosis. New therapeutic strategies are therefore urgently needed. The use of animal models represents a relevant tool for preclinical screening of new molecules/strategies against HCC. However, several issues, including animal husbandry, limit the use of current models (rodent/pig). One animal model that has attracted the attention of the scientific community in the last 15 years is the zebrafish. This freshwater fish has several attractive features, such as short reproductive time, limited space and cost requirements for husbandry, body transparency and the fact that embryos do not show immune response to transplanted cells. To date, two different types of zebrafish models for HCC have been developed: the transgenic zebrafish and the zebrafish xenograft models. Since transgenic zebrafish models for HCC have been described elsewhere, in this review, we focus on the description of zebrafish xenograft models that have been used in the last five years to test new molecules/strategies against HCC.
Collapse
Affiliation(s)
- Federica Tonon
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Rossella Farra
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Cristina Zennaro
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Gabriele Pozzato
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Nhung Truong
- Stem Cell Research and Application Laboratory, VNUHCM, University of Science, Ho Chi Minh City 72711, Vietnam;
| | - Salvatore Parisi
- Pathology Unit, CRO Aviano, National Cancer Institute, IRCCS, I 33081 Aviano, Italy; (S.P.); (F.R.)
- Doctoral School in Molecular Biomedicine, University of Trieste, I 34127 Trieste, Italy
| | - Flavio Rizzolio
- Pathology Unit, CRO Aviano, National Cancer Institute, IRCCS, I 33081 Aviano, Italy; (S.P.); (F.R.)
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, I 30170 Mestre, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I 34127 Trieste, Italy;
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I 34149 Trieste, Italy; (B.S.); (B.D.)
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Deborah Bonazza
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
| | - Gabriele Grassi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I 34149 Trieste, Italy; (F.T.); (R.F.); (C.Z.); (G.P.); (F.Z.); (D.B.)
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I 34149 Trieste, Italy; (B.S.); (B.D.)
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I 34149 Trieste, Italy; (B.S.); (B.D.)
| |
Collapse
|
7
|
CDCA2 protects against oxidative stress by promoting BRCA1-NRF2 signaling in hepatocellular carcinoma. Oncogene 2021; 40:4368-4383. [PMID: 34103686 DOI: 10.1038/s41388-021-01855-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023]
Abstract
Hepatocellular carcinoma (HCC) patients mostly suffer from poor survival outcomes. It is necessary to identify effective therapeutic targets to improve prognosis for HCC patients. Here, we report a new factor, CDCA2, in promoting HCC development. CDCA2 amplification is an independent risk factor for the recurrence and survival of HCC patients, which is positively correlated with elevated level of alpha-fetoprotein (AFP), high histological grade, large tumor size, advanced TNM stage, and poor prognosis for HCC patients. In HCC cells, CDCA2 promotes cell growth and inhibits apoptosis. Mechanistically, CDCA2's transcription is activated through the binding of E2F2/E2F8 with its promoter. CDCA2 depletion contributes to the suppression of cell proliferation and induction of apoptosis due to reactive oxygen species (ROS)-mediated stress, which can be reversed by antioxidants N-acetyl cysteine (NAC) and glutathione (GSH). Interestingly, we found that CDCA2 triggers the BRCA1-NRF2 cascade, which elevates antioxidant response and attenuates ROS levels. In response to oxidative stress, CDCA2 promotes BRCA1's chromatin relocalization to NRF2, activating NRF2-driven downstream signaling (HO-1, TXNRD1, and NQO1), which then protects HCC cells against oxidative damage. In conclusion, our results reveal that CDCA2 is a prognostic biomarker for HCC patients, and present the E2F2/E2F8-CDCA2-BRCA1-NRF2-ROS signaling axis that have implications for HCC therapeutics.
Collapse
|
8
|
Li TH, Qin C, Zhao BB, Cao HT, Yang XY, Wang YY, Li ZR, Zhou XT, Wang WB. Identification METTL18 as a Potential Prognosis Biomarker and Associated With Immune Infiltrates in Hepatocellular Carcinoma. Front Oncol 2021; 11:665192. [PMID: 34123827 PMCID: PMC8187872 DOI: 10.3389/fonc.2021.665192] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Methyltransferase-like 18 (METTL18), a METTL family member, is abundant in hepatocellular carcinoma (HCC). Studies have indicated the METTL family could regulate the progress of diverse malignancies while the role of METTL18 in HCC remains unclear. Data of HCC patients were acquired from the cancer genome atlas (TCGA) and gene expression omnibus (GEO). The expression level of METTL18 in HCC patients was compared with normal liver tissues by Wilcoxon test. Then, the logistic analysis was used to estimate the correlation between METTL18 and clinicopathological factors. Besides, Gene Ontology (GO), Gene Set Enrichment Analysis (GSEA), and single-sample Gene Set Enrichment Analysis (ssGSEA) were used to explore relevant functions and quantify the degree of immune infiltration for METTL18. Univariate and Multivariate Cox analyses and Kaplan-Meier analysis were used to estimate the association between METTL18 and prognosis. Besides, by cox multivariate analysis, a nomogram was conducted to forecast the influence of METTL18 on survival rates. METTL18-high was associated with Histologic grade, T stage, Pathologic stage, BMI, Adjacent hepatic tissue inflammation, AFP, Vascular invasion, and TP53 status (P < 0.05). HCC patients with METTL18-high had a poor Overall-Survival [OS; hazard ratio (HR): 1.87, P < 0.001), Disease-Specific Survival (DSS, HR: 1.76, P = 0.015), and Progression-Free Interval (PFI, HR: 1.51, P = 0.006). Multivariate analysis demonstrated that METTL18 was an independent factor for OS (HR: 2.093, P < 0.001), DSS (HR: 2.404, P = 0.015), and PFI (HR: 1.133, P = 0.006). Based on multivariate analysis, the calibration plots and C-indexes of nomograms showed an efficacious predictive effect for HCC patients. GSEA demonstrated that METTL18-high could activate G2M checkpoint, E2F targets, KRAS signaling pathway, and Mitotic Spindle. There was a positive association between the METTL18 and abundance of innate immunocytes (T helper 2 cells) and a negative relation to the abundance of adaptive immunocytes (Dendritic cells, Cytotoxic cells etc.). Finally, we uncovered knockdown of METTL18 significantly suppressed the proliferation, invasion, and migration of HCC cells in vitro. This research indicates that METTL18 could be a novel biomarker to evaluate HCC patients' prognosis and an important regulator of immune responses in HCC.
Collapse
Affiliation(s)
- Tian-Hao Li
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Cheng Qin
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Bang-Bo Zhao
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hong-Tao Cao
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiao-Ying Yang
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuan-Yang Wang
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ze-Ru Li
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xing-Tong Zhou
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei-Bin Wang
- Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Li P, Pan X, Zheng Z, Sun Y, Han Y, Dong J. LINC00271 inhibits epithelial-mesenchymal transition of papillary thyroid cancer cells by downregulating trefoil factor 3 expression. ACTA ACUST UNITED AC 2020. [DOI: 10.31491/apt.2020.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Wang H, Wang L, Tang L, Luo J, Ji H, Zhang W, Zhou J, Li Q, Miao L. Long noncoding RNA SNHG6 promotes proliferation and angiogenesis of cholangiocarcinoma cells through sponging miR-101-3p and activation of E2F8. J Cancer 2020; 11:3002-3012. [PMID: 32226515 PMCID: PMC7086249 DOI: 10.7150/jca.40592] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/02/2020] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CCA) development is an extremely complex process with alterations occurring in numerous genes. SNHG6, a validated lncRNA, has been reported to regulate the expression of multiple tumor-related genes in hepatocellular carcinoma, colorectal cancer and breast cancer. Here, we elucidated the function and possible molecular mechanisms of SNHG6 in human CCA cells. Our results proved that the expression SNHG6 was upregulated in CCA tissues and cell lines. Ectopic expression of SNHG6 promoted cell proliferation, cell cycle progression, migration, and angiogenesis in CCA cells, whereas knockdown of SNHG6 repressed these cellular processes. Further mechanistic studies revealed that SNHG6 could compete with the transcription factor E2F8 to bind with miR-101-3p, thus affecting E2F8 expression. Taken together, these results provided a comprehensive analysis of the role of SNHG6 in CCA cells and offered important clues to understand the key roles of competing endogenous RNA (ceRNA) mechanisms in human cholangiocarcinoma.
Collapse
Affiliation(s)
- Huishan Wang
- Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
| | - Li Wang
- Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
| | - Lingyu Tang
- Taizhou hospital of traditional Chinese medicine, 86 Jichuandong Road, Hailing District, Taizhou 225300, Jiangsu Province, China
| | - Jing Luo
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Ji
- Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
| | - Wen Zhang
- Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
| | - Jian Zhou
- Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
| | - Quanpeng Li
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China
| | - Lin Miao
- Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China
| |
Collapse
|
11
|
Lü Y, Zhang J, Li L, Li S, Yang Z. Carcinogenesis effects of E2F transcription factor 8 (E2F8) in hepatocellular carcinoma outcomes: an integrated bioinformatic report. Biosci Rep 2020; 40:BSR20193212. [PMID: 31990034 PMCID: PMC7012657 DOI: 10.1042/bsr20193212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 02/05/2023] Open
Abstract
This report aimed to investigate the carcinogenesis effects of E2F transcription factor 8 (E2F8) in hepatocellular carcinoma (HCC). E2F8 expression level was compared in Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA) and Oncomine. Survival analysis of E2F8 for HCC were conducted in Kaplan-Meier plotter. Correlations of E2F8 and clinico-pathological features were performed in TCGA. Enrichment of interacted and similar genes with E2F8 was evaluated in Gene Set Enrichment Analysis (GSEA) and Metascape. We found that E2F8 was significantly up-regulated in tumor tissues compared with nontumor tissues (all P < 0.01). Moreover, E2F8 was significantly overexpressed in peripheral blood mononuclear cell (PBMC) in HCC patients than that in healthy individuals (P < 0.001). Meta-analysis in Oncomine database confirmed that E2F8 was significantly higher in HCC tumors (P = 4.28E-08). Additionally, E2F8 elevation significantly correlated with overall survival (OS), recurrence-free survival (RFS), disease-specific survival (DSS) and progression-free survival (PFS) in HCC patients (all P < 0.01). E2F8 level was significantly higher in HCC patients with advanced neoplasm histologic grade, American Joint Committee on Cancer (AJCC) stage and α-fetoprotein (AFP) elevation (all P < 0.05). Cox regression model demonstrated that high E2F8 was an independent risk factor for OS and DFS in HCC patients (HR = 2.16, P = 0.003 and HR = 1.64, P = 0.002, respectively). Enrichment analysis revealed that genes interacted/similar with E2F8 were mainly enriched in cell cycle pathways/biological process. Conclusively, up-regulated in tumors, E2F8 might accelerate tumor progression and result in unfavorable outcomes in HCC patients.
Collapse
Affiliation(s)
- Ying Lü
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Jing Zhang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lei Li
- Department of Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Shun Li
- Department of Laboratory Animal, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zongguo Yang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
12
|
Jusino S, Saavedra HI. Role of E2Fs and mitotic regulators controlled by E2Fs in the epithelial to mesenchymal transition. Exp Biol Med (Maywood) 2019; 244:1419-1429. [PMID: 31575294 DOI: 10.1177/1535370219881360] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a complex cellular process in which epithelial cells acquire mesenchymal properties. EMT occurs in three biological settings: development, wound healing and fibrosis, and tumor progression. Despite occurring in three independent biological settings, EMT signaling shares some molecular mechanisms that allow epithelial cells to de-differentiate and acquire mesenchymal characteristics that confer cells invasive and migratory capacity to distant sites. Here we summarize the molecular mechanism that delineates EMT and we will focus on the role of E2 promoter binding factors (E2Fs) in EMT during tumor progression. Since the E2Fs are presently undruggable due to their control in numerous pivotal cellular functions and due to the lack of selectivity against individual E2Fs, we will also discuss the role of three mitotic regulators and/or mitotic kinases controlled by the E2Fs (NEK2, Mps1/TTK, and SGO1) in EMT that can be useful as drug targets. Impact statement The study of the epithelial to mesenchymal transition (EMT) is an active area of research since it is one of the early intermediates to invasion and metastasis—a state of the cancer cells that ultimately kills many cancer patients. We will present in this review that besides their canonical roles as regulators of proliferation, unregulated expression of the E2F transcription factors may contribute to cancer initiation and progression to metastasis by signaling centrosome amplification, chromosome instability, and EMT. Since our discovery that the E2F activators control centrosome amplification and mitosis in cancer cells, we have identified centrosome and mitotic regulators that may represent actionable targets against EMT and metastasis in cancer cells. This is impactful to all of the cancer patients in which the Cdk/Rb/E2F pathway is deregulated, which has been estimated to be most cancer patients with solid tumors.
Collapse
Affiliation(s)
- Shirley Jusino
- Basic Sciences Department, Division of Pharmacology and Toxicology, Ponce Research Institute, Ponce Health Sciences University, Ponce PR 00732, USA
| | - Harold I Saavedra
- Basic Sciences Department, Division of Pharmacology and Toxicology, Ponce Research Institute, Ponce Health Sciences University, Ponce PR 00732, USA
| |
Collapse
|
13
|
Preclinical efficacy of a novel dual PI3K/mTOR inhibitor, CMG002, alone and in combination with sorafenib in hepatocellular carcinoma. Cancer Chemother Pharmacol 2019; 84:809-817. [PMID: 31385002 DOI: 10.1007/s00280-019-03918-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022]
Abstract
PURPOSE Sorafenib has been the only first systemic drug that improves survival of patients with advanced hepatocellular carcinoma (HCC). However, because the response rate of sorafenib is relatively low, novel therapeutic strategies are needed to improve survival in patients with HCC. This study investigated the effect of CMG002 alone and in combination with sorafenib on HCC in vitro and vivo. METHODS The effect of a newly developed dual PI3K/mTOR inhibitor, CMG002, on the proliferation of Huh-7 and HepG2 HCC cells was investigated using the MTT assay. Western blotting was performed to assess phosphorylation of the key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. HepG2 cells were inoculated into mice, which were treated with vehicle, sorafenib, CMG002, and their combinations. Tumor cell proliferation and tumor angiogenesis were evaluated by immunohistochemical analysis of Ki-67 and CD31, respectively. Tumor cell apoptosis was detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Levels of key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways were evaluated by western blot analysis. RESULTS The combination of sorafenib and CMG002 additively inhibited Huh-7 and HepG2 cell proliferation compared to single-agent treatment. Sorafenib and CMG002 as single agents differentially inhibited or activated key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. The combination of sorafenib and CMG002 inhibited all key enzymes in the two pathways. Treatment with CMG002 for 4 weeks alone and in combination with sorafenib strongly inhibited tumor growth. CMG002 inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Furthermore, these effects were enhanced when CMG002 was combined with sorafenib. CONCLUSIONS The combination of CMG002 and sorafenib significantly inhibited HCC cell proliferation and tumorigenesis by inhibiting the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. These findings suggest that CMG002 to be a potential novel candidate treatment for HCC.
Collapse
|
14
|
Huang YL, Ning G, Chen LB, Lian YF, Gu YR, Wang JL, Chen DM, Wei H, Huang YH. Promising diagnostic and prognostic value of E2Fs in human hepatocellular carcinoma. Cancer Manag Res 2019; 11:1725-1740. [PMID: 30863181 PMCID: PMC6388971 DOI: 10.2147/cmar.s182001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A growing body of evidence suggests that E2Fs, by regulating gene expression related to cell cycle progression and other cellular processes, play a pivotal role in human cancer. However, the distinct roles of each E2F in the development and treatment of hepatocellular carcinoma (HCC) remain unknown. In the present study, the mRNA expression and prognostic value of different E2Fs in HCC are analyzed. MATERIALS AND METHODS Transcriptional and survival data related to E2F expression in patients with HCC were obtained through ONCOMINE and UALCAN databases. Survival analysis plots were drawn with Kaplan-Meier Plotter. The sequence alteration data for E2Fs were obtained from The Cancer Genome Atlas and c-BioPortal. Gene functional enrichment analyses were performed in Database for Annotation, Visualization and Integrated Discovery. RESULTS The mRNA expression levels of E2F1-E2F8 were all significantly upregulated in HCC patients, and high expression of each E2F was obviously related to poor prognosis. Similarly, the expression of E2Fs showed prognostic prediction value in HCC patients with different cancer stages and pathological grades. Moreover, the mutation rate of E2Fs was relatively high in HCC patients, and the DNA sequence alterations primarily occurred in E2F5, E2F3, and E2F6, which were associated with worse overall survival and disease-free survival in HCC patients. Network analysis confirmed that the expression levels of cell cycle-related genes were mostly affected by E2F mutations. CONCLUSION High expression of individual E2Fs was associated with poor prognosis in all liver cancer patients. E2Fs may be exploited as good prognostic targets for comprehensive management of HCC patients, but this notion should be further evaluated in clinical studies.
Collapse
Affiliation(s)
- Yan-Lin Huang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Gang Ning
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Lu-Biao Chen
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Yi-Fan Lian
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Yu-Rong Gu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Jia-Liang Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Dong-Mei Chen
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Huan Wei
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| | - Yue-Hua Huang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,
| |
Collapse
|
15
|
Huang IT, Dhungel B, Shrestha R, Bridle KR, Crawford DHG, Jayachandran A, Steel JC. Spotlight on Bortezomib: potential in the treatment of hepatocellular carcinoma. Expert Opin Investig Drugs 2018; 28:7-18. [PMID: 30474444 DOI: 10.1080/13543784.2019.1551359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION This study reviews the evidence for the use of Bortezomib (BZB), a first-in-class proteasome inhibitor in advanced Hepatocellular carcinoma (HCC). This review aims to delineate the role of BZB within the management of non-surgical and metastatic HCC, either as an alternative or as an adjunct to the current treatment paradigm. AREAS COVERED In addition to BZB pharmacology and mechanism of action, safety and tolerance profiles of the drug obtained from clinical trials are explored. The utility of BZB as a therapeutic agent either alone or in combination with other therapies against HCC, including its application in both preclinical and clinical settings has been reviewed. In particular, we highlight the importance of preclinical evaluation of BZB as a combinatorial agent in synergism with other therapies for the use in the management of HCC. EXPERT OPINION There has been much interest surrounding the use of BZB, a first-in-class proteasome inhibitor for HCC therapy. The discernment of outcomes of BZB clinical trials for HCC need to take into consideration the disease-specific factors that can affect survival outcomes including patient selection and aetiological differences. Further preclinical testing of BZB in combination with other therapeutic modalities can be important for eliciting enhanced anti-HCC effects.
Collapse
Affiliation(s)
- I-Tao Huang
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Bijay Dhungel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Ritu Shrestha
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Kim R Bridle
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Darrell H G Crawford
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Aparna Jayachandran
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Jason C Steel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,c School of Health, Medical and Applied Sciences , CQ University , Rockhampton , Australia
| |
Collapse
|
16
|
Wang J, Tian Y, Chen H, Li H, Zheng S. Key signaling pathways, genes and transcription factors associated with hepatocellular carcinoma. Mol Med Rep 2018; 17:8153-8160. [PMID: 29658607 PMCID: PMC5983994 DOI: 10.3892/mmr.2018.8871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/03/2018] [Indexed: 01/30/2023] Open
Abstract
The purpose of the present study was to investigate the underlying molecular mechanism of hepatocellular carcinoma (HCC) using bioinformatics approaches. The microarray dataset GSE64041 was downloaded from the Gene Expression Omnibus database, which included 60 tumor liver samples and 60 matched control samples. Differentially expressed genes (DEGs) between HCC and control groups were identified. Then functional enrichment analyses, protein‑protein interaction (PPI) network, sub‑network and integrated transcription factor (TF)‑microRNA (miRNA)‑target network analyses were performed for these DEGs. A total of 378 DEGs were obtained, including 101 upregulated and 277 downregulated DEGs. In addition, functional enrichment analysis for DEGs in the sub‑network revealed 'cell division' and 'cell cycle' as key Gene Ontology (GO) terms and pathways. Topoisomerase (DNA) IIα (TOP2A) and integrin subunit α2 (ITGA2) were hub nodes in the PPI network. TOP2A, cyclin dependent kinase 1 (CDK1) and polo like kinase 1 (PLK1) were revealed to be hub nodes in the sub‑network. Finally, 4 TFs including forkhead box M1 (FOXM1), E2F transcription factor 4 (E2F4), SIN3 transcription regulator family member A (SIN3A) and transcription factor 7 like 1 (TCF7L1) were obtained through integrated network analysis. TOP2A, ITGA2, PLK1 and CDK1 may be key genes involved in HCC development. 'Cell division' and 'cell cycle' were indicated to act as key GO terms and Kyoto Encyclopedia of Genes and Genomes pathways in HCC. In addition, FOXM1, TCF7L1, E2F4 and SIN3A were revealed to be key TFs associated with HCC.
Collapse
Affiliation(s)
- Jingcheng Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Yang Tian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Hui Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Hui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
17
|
Roeten MSF, Cloos J, Jansen G. Positioning of proteasome inhibitors in therapy of solid malignancies. Cancer Chemother Pharmacol 2018; 81:227-243. [PMID: 29184971 PMCID: PMC5778165 DOI: 10.1007/s00280-017-3489-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/19/2017] [Indexed: 12/13/2022]
Abstract
Targeting of the protein degradation pathway, in particular, the ubiquitin-proteasome system, has emerged as an attractive novel cancer chemotherapeutic modality. Although proteasome inhibitors have been most successfully applied in the treatment of hematological malignancies, they also received continuing interest for the treatment of solid tumors. In this review, we summarize the current positioning of proteasome inhibitors in the treatment of common solid malignancies (e.g., lung, colon, pancreas, breast, and head and neck cancer), addressing topics of their mechanism(s) of action, predictive factors and molecular mechanisms of resistance.
Collapse
Affiliation(s)
- Margot S F Roeten
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands.
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Location VUmc, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Feliciano A, Garcia-Mayea Y, Jubierre L, Mir C, Hummel M, Castellvi J, Hernández-Losa J, Paciucci R, Sansano I, Sun Y, Ramón Y Cajal S, Kondon H, Soriano A, Segura M, Lyakhovich A, LLeonart ME. miR-99a reveals two novel oncogenic proteins E2F2 and EMR2 and represses stemness in lung cancer. Cell Death Dis 2017; 8:e3141. [PMID: 29072692 PMCID: PMC5680913 DOI: 10.1038/cddis.2017.544] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/22/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022]
Abstract
Lung cancer is one of the most aggressive tumours with very low life expectancy. Altered microRNA expression is found in human tumours because it is involved in tumour growth, progression and metastasis. In this study, we analysed microRNA expression in 47 lung cancer biopsies. Among the most downregulated microRNAs we focussed on the miR-99a characterisation. In vitro experiments showed that miR-99a expression decreases the proliferation of H1650, H1975 and H1299 lung cancer cells causing cell cycle arrest and apoptosis. We identified two novel proteins, E2F2 (E2F transcription factor 2) and EMR2 (EGF-like module-containing, mucin-like, hormone receptor-like 2), downregulated by miR-99a by its direct binding to their 3′-UTR. Moreover, miR-99a expression prevented cancer cell epithelial-to-mesenchymal transition (EMT) and repressed the tumourigenic potential of the cancer stem cell (CSC) population in both these cell lines and mice tumours originated from H1975 cells. The expression of E2F2 and EMR2 at protein level was studied in 119 lung cancer biopsies. E2F2 and EMR2 are preferentially expressed in adenocarcinomas subtypes versus other tumour types (squamous and others). Interestingly, the expression of E2F2 correlates with the presence of vimentin and both E2F2 and EMR2 correlate with the presence of β-catenin. Moreover, miR-99a expression correlates inversely with E2F2 and directly with β-catenin expression in lung cancer biopsies. In conclusion, miR-99a reveals two novel targets E2F2 and EMR2 that play a key role in lung tumourigenesis. By inhibiting E2F2 and EMR2, miR-99a represses in vivo the transition of epithelial cells through an EMT process concomitantly with the inhibition of stemness features and consequently decreasing the CSC population.
Collapse
Affiliation(s)
- Andrea Feliciano
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Luz Jubierre
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Manuela Hummel
- Centre for Genomic Regulation, Core Facilities - Microarray Unit, C/ Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Josep Castellvi
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Hernández-Losa
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Rosanna Paciucci
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Irene Sansano
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Yilin Sun
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Santiago Ramón Y Cajal
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Hiroshi Kondon
- Geriatric Unit, Graduate School of Medicine, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Aroa Soriano
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Miguel Segura
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Alex Lyakhovich
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Institut de Recerca Hospital Vall d'Hebron (VHIR), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
19
|
Cavallaro G, Farra R, Craparo EF, Sardo C, Porsio B, Giammona G, Perrone F, Grassi M, Pozzato G, Grassi G, Dapas B. Galactosylated polyaspartamide copolymers for siRNA targeted delivery to hepatocellular carcinoma cells. Int J Pharm 2017; 525:397-406. [PMID: 28119125 DOI: 10.1016/j.ijpharm.2017.01.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 02/07/2023]
Abstract
The limited efficacy of available treatments for hepatocellular carcinoma (HCC) requires the development of novel therapeutic approaches. We synthesized a novel cationic polymer based on α,β-poly-(N-2-hydroxyethyl)-d,L-aspartamide (PHEA) for drug delivery to HCC cells. The copolymer was synthesized by subsequent derivatization of PHEA with diethylene triamine (DETA) and with a polyethylene glycol (PEG) derivative bearing galactose (GAL) molecules, obtaining the cationic derivative PHEA-DETA-PEG-GAL. PHEA-DETA-PEG-GAL has suitable chemical-physical characteristics for a potential systemic use and can effectively deliver a siRNA (siE2F1) targeted against the transcription factor E2F1, a gene product involved in HCC. The presence of GAL residues in the polyplexes allows the targeting of HCC cells that express the asialo-glycoprotein receptor (ASGP-R). In these cells, but not in ASGP-R non-expressing cells, PHEA-DETA-PEG-GAL/siE2F1 polyplexes induce the reduction of the mRNA and protein levels of E2F1 and of E2F1-regulated genes, all involved in the promotion of the G1/S phase transition. This results in a decrease of cell proliferation with a G1/G0 phase cells accumulation. Notably, removal of GAL residue almost completely abrogates the targeting capacity of the developed polyplexes. In conclusion, the generated polyplexes demonstrate the potential to effectively contributing to the development of novel anti-HCC therapeutic approaches via a siRNA-targeted delivery.
Collapse
Affiliation(s)
- Gennara Cavallaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Rossella Farra
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Emanuela Fabiola Craparo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Carla Sardo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Porsio
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Gaetano Giammona
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | | | - Mario Grassi
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Gabriele Pozzato
- Department of 'Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | | | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Italy
| |
Collapse
|
20
|
Farra R, Scaggiante B, Guerra C, Pozzato G, Grassi M, Zanconati F, Perrone F, Ferrari C, Trotta F, Grassi G, Dapas B. Dissecting the role of the elongation factor 1A isoforms in hepatocellular carcinoma cells by liposome-mediated delivery of siRNAs. Int J Pharm 2017; 525:367-376. [PMID: 28229942 DOI: 10.1016/j.ijpharm.2017.02.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/20/2017] [Accepted: 02/10/2017] [Indexed: 02/08/2023]
Abstract
Eukaryotic elongation factor 1A (eEF1A), a protein involved in protein synthesis, has two major isoforms, eEF1A1 and eEF1A2. Despite the evidences of their involvement in hepatocellular carcinoma (HCC), the quantitative contribution of each of the two isoforms to the disease is unknown. We depleted the two isoforms by means of siRNAs and studied the effects in three different HCC cell lines. Particular care was dedicated to select siRNAs able to target each of the two isoform without affecting the other one. This is not a trivial aspect due to the high sequence homology between eEF1A1 and eEF1A2. The selected siRNAs can specifically deplete either eEF1A1 or eEF1A2. This, in turn, results in an impairment of cell vitality, growth and arrest in the G1/G0 phase of the cell cycle. Notably, these effects are quantitatively superior following eEF1A1 than eEF1A2 depletion. Moreover, functional tests revealed that the G1/G0 block induced by eEF1A1 depletion depends on the down-regulation of the transcription factor E2F1, a known player in HCC. In conclusion, our data indicate that the independent targeting of the two eEF1A isoforms is effective in reducing HCC cell growth and that eEF1A1 depletion may result in a more evident effect.
Collapse
Affiliation(s)
- Rossella Farra
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | | | - Chiara Guerra
- Department of Life Sciences, University of Trieste, Italy
| | - Gabriele Pozzato
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Mario Grassi
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | | | - Cinzia Ferrari
- Department of Clinic-Surgical Sciences, Experimental Surgery Laboratory, University of Pavia, Italy
| | - Francesco Trotta
- Department of Clinic-Surgical Sciences, Experimental Surgery Laboratory, University of Pavia, Italy; U.O. di Chirurgia Generale e Toracica, Ospedale Maggiore, Lodi, Italy
| | | | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Italy
| |
Collapse
|
21
|
Xie Y, Si J, Wang Y, Li H, Di C, Yan J, Ye Y, Zhang Y, Zhang H. E2F is involved in radioresistance of carbon ion induced apoptosis via Bax/caspase 3 signal pathway in human hepatoma cell. J Cell Physiol 2017; 233:1312-1320. [DOI: 10.1002/jcp.26005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/11/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Yi Xie
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
| | - Jing Si
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
| | - Yu‐Pei Wang
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
- Graduate School of University of Chinese Academy of SciencesBeijingChina
| | - Hong‐Yan Li
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
- Graduate School of University of Chinese Academy of SciencesBeijingChina
| | - Cui‐Xia Di
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
| | - Jun‐Fang Yan
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
- Graduate School of University of Chinese Academy of SciencesBeijingChina
| | | | | | - Hong Zhang
- Institute of Modern PhysicsChinese Academy of SciencesLanzhouChina
- CAS Key Laboratory of Heavy Ion Radiation Biology and MedicineInstitute of Modern PhysicsLanzhouGansuChina
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhouChina
- Gansu Wuwei Tumor HospitalWuweiChina
| |
Collapse
|
22
|
Lv Y, Xiao J, Liu J, Xing F. E2F8 is a Potential Therapeutic Target for Hepatocellular Carcinoma. J Cancer 2017; 8:1205-1213. [PMID: 28607595 PMCID: PMC5463435 DOI: 10.7150/jca.18255] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/11/2017] [Indexed: 12/15/2022] Open
Abstract
E2F transcriptional factors are widely expressed in a number of tissues and organs, possessing many regulatory functions related to cellular proliferation, differentiation, DNA repair, cell-cycle and cell apoptosis. E2F8 is a recently identified member of the E2F family with a duplicated DNA-binding domain feature discriminated from E2F1-6, controlling gene expression in a dimerization partner-independent manner. It is indispensable for angiogenesis, lymphangiogenesis and embryonic development. Although E2F8 and E2F7 perform complementary and overlapping functions in many cell metabolisms, E2F8, but not E2F7, overexpresses remarkably in hepatocellular carcinoma (HCC) to facilitate the HCC occurrence and development via activating a E2F1/ Cyclin D1 signaling pathway to regulate the G1- to S-phase transition of cell cycle progression or transcriptionally suppressing CDK1 to induce hepatocyte polyploidization. It also involves closely a variety of cellular physiological functions and pathological processes, which may bring a new breakthrough for the treatment of certain diseases, especially the HCC. Here, we summarize the latest progress of E2F8 on its relevant functions and mechanisms as well as potential application.
Collapse
Affiliation(s)
- Yi Lv
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China.,Key Laboratory of Functional Protein Research of Guangdong, Higher Education Institutes, Jinan University, Guangzhou, China
| | - Jia Xiao
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Jing Liu
- Department of Stomatology, Jinan University, Guangzhou, China
| | - Feiyue Xing
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China.,Key Laboratory of Functional Protein Research of Guangdong, Higher Education Institutes, Jinan University, Guangzhou, China
| |
Collapse
|
23
|
Tonon F, Zennaro C, Dapas B, Carraro M, Mariotti M, Grassi G. Rapid and cost-effective xenograft hepatocellular carcinoma model in Zebrafish for drug testing. Int J Pharm 2016; 515:583-591. [PMID: 27989824 DOI: 10.1016/j.ijpharm.2016.10.070] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023]
Abstract
We developed a novel, rapid and cost-effective Zebrafish xenograft model of hepatocellular carcinoma (HCC) for drug screening in the disease. Following injection into the yolk sack of Zebrafish larvae of the human HCC cell line JHH6 stained by a vital dye, tumor mass growth was followed by fluorescence microscopy and by human Ki67 quantification. Tumor induced neo-angiogenesis was evaluated by alkaline phosphatase staining of the vessels, by using the Tg(fli1:EGFP)y1 strain of Zebrafish and by the quantification of the zebrafish vascular endothelial growth factor and of its receptor. We show that it is feasible to micro-inject JHH6 in Zebrafish larvae, that injected cells can grow for different days and that this induces a marked neo-angiogenesis. Finally, we show that our model allows testing the effects of anti-HCC drugs such as Bortezomib. Compared to more complex HCC mouse models, our model is far less expensive, faster to set up and does not need immunosuppressant treatment. Finally, the model makes use of JHH6, an aggressive form of HCC cell line never tested before in Zebrafish. In conclusion, the possibility to test anti HCC/neo-angiogenesis drugs makes our JHH6 model useful to select therapeutic molecules for a highly vascularized tumor such as HCC.
Collapse
Affiliation(s)
- Federica Tonon
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Cristina Zennaro
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Italy
| | - Michele Carraro
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Massimo Mariotti
- IRCCS Galeazzi Orthopedic Institute Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | | |
Collapse
|
24
|
Scaggiante B, Farra R, Dapas B, Baj G, Pozzato G, Grassi M, Zanconati F, Grassi G. Aptamer targeting of the elongation factor 1A impairs hepatocarcinoma cells viability and potentiates bortezomib and idarubicin effects. Int J Pharm 2016; 506:268-279. [PMID: 27094354 DOI: 10.1016/j.ijpharm.2016.04.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/23/2016] [Accepted: 04/13/2016] [Indexed: 02/05/2023]
Abstract
The high morbidity and mortality of hepatocellular carcinoma (HCC) is mostly due to the limited efficacy of the available therapeutic approaches. Here we explore the anti-HCC potential of an aptamer targeting the elongation factor 1A (eEF1A), a protein implicated in the promotion of HCC. As delivery methods, we have compared the effectiveness of cationic liposome and cholesterol-mediated approaches. A75 nucleotide long aptamer containing GT repetition (GT75) was tested in three HCC cell lines, HepG2, HuH7 and JHH6. When delivered by liposomes, GT75 was able to effectively reducing HCC cells viability in a dose and time dependent fashion. Particular sensitive were JHH6 where increased apoptosis with no effects on cell cycle were observed. GT75 effect was likely due to the interference with eEF1A activity as neither the mRNA nor the protein levels were significantly affected. Notably, cholesterol-mediated delivery of GT75 abrogated its efficacy due to cellular mis-localization as proven by fluorescence and confocal microscopic analysis. Finally, liposome-mediated delivery of GT75 improved the therapeutic index of the anticancer drugs bortezomib and idarubicin. In conclusion, liposome but not cholesterol-mediated delivery of GT75 resulted in an effective delivery of GT75, causing the impairment of the vitality of a panel of HCC derived cells.
Collapse
Affiliation(s)
| | - Rosella Farra
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Italy
| | - Gabriele Baj
- Department of Life Sciences, University of Trieste, Italy
| | - Gabriele Pozzato
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | - Mario Grassi
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | | |
Collapse
|
25
|
Huntington JT, Tang X, Kent LN, Schmidt CR, Leone G. The Spectrum of E2F in Liver Disease--Mediated Regulation in Biology and Cancer. J Cell Physiol 2016; 231:1438-49. [PMID: 26566968 DOI: 10.1002/jcp.25242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022]
Abstract
Uncoordinated cell growth is one of the fundamental concepts in carcinogenesis and occurs secondary to dysregulation of the cell cycle. The E2Fs are a large family of transcription factors and are key regulators of the cell cycle. The activation of E2Fs is intimately regulated by retinoblastoma 1 (RB1). The RB pathway has been implicated in almost every human malignancy. Recently there have been exciting developments in the E2F field using animal models to better understand the role of E2Fs in vivo. Genetic mouse models have proven essential in implicating E2Fs in hepatocellular carcinoma (HCC) and liver disease. In this review, the general structure and function of E2Fs as well as the role for E2Fs in the development of HCC and liver disease is evaluated. Specifically, what is known about E2Fs in human disease is explored in depth, and future directions are discussed.
Collapse
Affiliation(s)
- Justin T Huntington
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Xing Tang
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, Columbus, Ohio.,Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Lindsey N Kent
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, Columbus, Ohio.,Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Carl R Schmidt
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Gustavo Leone
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, Columbus, Ohio.,Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
26
|
Sardo C, Farra R, Licciardi M, Dapas B, Scialabba C, Giammona G, Grassi M, Grassi G, Cavallaro G. Development of a simple, biocompatible and cost-effective Inulin-Diethylenetriamine based siRNA delivery system. Eur J Pharm Sci 2015; 75:60-71. [PMID: 25845631 DOI: 10.1016/j.ejps.2015.03.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/28/2015] [Accepted: 03/24/2015] [Indexed: 02/07/2023]
Abstract
Small interfering RNAs (siRNAs) have the potential to be of therapeutic value for many human diseases. So far, however, a serious obstacle to their therapeutic use is represented by the absence of appropriate delivery systems able to protect them from degradation and to allow an efficient cellular uptake. In this work we developed a siRNA delivery system based on inulin (Inu), an abundant and natural polysaccharide. Inu was functionalized via the conjugation with diethylenetriamine (DETA) residues to form the complex Inu-DETA. We studied the size, surface charge and the shape of the Inu-DETA/siRNA complexes; additionally, the cytotoxicity, the silencing efficacy and the cell uptake-mechanisms were studied in the human bronchial epithelial cells (16HBE) and in the hepatocellular carcinoma derived cells (JHH6). The results presented here indicate that Inu-DETA copolymers can effectively bind siRNAs, are highly cytocompatible and, in JHH6, can effectively deliver functional siRNAs. Optimal delivery is observed using a weight ratio Inu-DETA/siRNA of 4 that corresponds to polyplexes with an average size of 600nm and a slightly negative surface charge. Moreover, the uptake and trafficking mechanisms, mainly based on micropinocytosis and clatrin mediated endocytosis, allow the homogeneous diffusion of siRNA within the cytoplasm of JHH6. Notably, in 16 HBE where the trafficking mechanism (caveolae mediated endocytosis) does not allow an even distribution of siRNA within the cell cytoplasm, no significant siRNA activity is observed. In conclusion, we developed a novel inulin-based siRNA delivery system able to efficiently release siRNA in JHH6 with negligible cytotoxicity thus opening the way for further testing in more complex in vivo models.
Collapse
Affiliation(s)
- C Sardo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - R Farra
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, I-34127 Trieste, Italy
| | - M Licciardi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - B Dapas
- Department of Life Sciences, University Hospital of Cattinara, Strada di Fiume 447, 34100 Trieste, Italy
| | - C Scialabba
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - G Giammona
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - M Grassi
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, I-34127 Trieste, Italy
| | - G Grassi
- Department of Life Sciences, University Hospital of Cattinara, Strada di Fiume 447, 34100 Trieste, Italy.
| | - G Cavallaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
27
|
Chen YJ, Wu H, Shen XZ. The ubiquitin-proteasome system and its potential application in hepatocellular carcinoma therapy. Cancer Lett 2015; 379:245-52. [PMID: 26193663 DOI: 10.1016/j.canlet.2015.06.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 02/07/2023]
Abstract
The ubiquitin-proteasome system (UPS) is a complicated tightly controlled system in charge of degrading 80-90% of proteins, and is central to regulating cellular function and keeping protein homeostasis. Therefore, the components of UPS attract considerable attention as potential targets for hepatocellular carcinoma (HCC) therapy. The clinical success of bortezomib in multiple myeloma and mantle cell lymphoma patients has set the precedent for therapeutically targeting this pathway. This review will provide an overview of the UPS in HCC and the current status of therapeutic strategies.
Collapse
Affiliation(s)
- Yan-Jie Chen
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai 200032, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Hao Wu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai 200032, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Xi-Zhong Shen
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai 200032, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai 200032, China.
| |
Collapse
|
28
|
Farra R, Dapas B, Baiz D, Tonon F, Chiaretti S, Del Sal G, Rustighi A, Elvassore N, Pozzato G, Grassi M, Grassi G. Impairment of the Pin1/E2F1 axis in the anti-proliferative effect of bortezomib in hepatocellular carcinoma cells. Biochimie 2015; 112:85-95. [PMID: 25742740 DOI: 10.1016/j.biochi.2015.02.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/20/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND The modest efficacy of available therapies for Hepatocellular carcinoma (HCC) indicates the need to develop novel therapeutic approaches. For the proteasome inhibitor Bortezomib (BZB), potentially attractive for HCC treatment, the mechanism of action is largely unknown. The BZB effect on E2Fs and the E2Fs control on the peptidylproline cis-trans isomerase (Pin1), prompted us to explore the BZB effect on the Pin1-E2F1 axis. METHODS The tumorigenic cell line HuH7 together with the non-tumorigenic cells IHH and the human pluripotent stem cell derived hepatocytes (hPSC-H), were used as cellular models of HCC and normal liver cells, respectively. RESULTS BZB reduces HuH7 growth as shown by cell counting, cell vitality test and cell cycle analysis; this is paralleled by the decrease of Pin1, E2F1, cyclin A2 and of the hyper-phosphorylated pRB. Pin1-E2F1 axis impairment justifies the anti-proliferative effect since Pin-E2F1 depletion decreases HuH7 growth while the over-expression rescues BZB-induced inhibition of proliferation. Moreover, Pin1-E2F1 promote HuH7 growth via the up-regulation of cyclin D1, cyclin E, cyclin A2, E2F2 and in part E2F3. Finally, in the control cells IHH and hPSC-H, BZB effect on cell vitality is not irrelevant, a fact correlated to the cellular proliferation rate. Thus, BZB effect on healthy liver tissue may not be entirely negligible hence caution should be exercised in its use in liver regeneration processes. CONCLUSION For the first time we prove the functional involvement of the Pin1-E2F1 axis in the anti-proliferative effect of BZB indicating Pin1-E2F as an attractive target to control HCC cell growth.
Collapse
Affiliation(s)
- Rossella Farra
- Department of Industrial Engineering and Information Technology, University of Trieste, V. Valerio 10, 34100 Trieste, Italy
| | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Via Giorgieri, 10, 34127 Trieste, Italy
| | - Daniele Baiz
- International Centre for Genetic Engineering and Biotechnology, Via E. Ramarini 32, 00016 Monterotondo Scalo, Rome, Italy
| | - Federica Tonon
- Department of Industrial Engineering and Information Technology, University of Trieste, V. Valerio 10, 34100 Trieste, Italy
| | - Sara Chiaretti
- Department of Life Sciences, University of Trieste, Via Giorgieri, 10, 34127 Trieste, Italy
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, Via Giorgieri, 10, 34127 Trieste, Italy; Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy
| | - Alessandra Rustighi
- Department of Life Sciences, University of Trieste, Via Giorgieri, 10, 34127 Trieste, Italy; Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, Italy; Venetian Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy
| | - Gabriele Pozzato
- Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume 447, 34134, Trieste, Italy
| | - Mario Grassi
- Department of Industrial Engineering and Information Technology, University of Trieste, V. Valerio 10, 34100 Trieste, Italy
| | - Gabriele Grassi
- Department of Life Sciences, University of Trieste, Via Giorgieri, 10, 34127 Trieste, Italy; Department of Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume 447, 34134, Trieste, Italy.
| |
Collapse
|
29
|
Chen YS, Sun ZW, Meng CC, Tang JZ, Li XY, Wang JF, Liu L, Zhang LJ. Molecular pathogenesis and molecular targeted agents for primary liver cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:914-924. [DOI: 10.11569/wcjd.v23.i6.914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The treatment of primary liver cancer (PLC) is still challenging in China nowadays, and it fundamentally depends on the systematic knowledge of hepatocarcinogenesis. In recent years, the gradual clarification of the molecular pathogenesis of PLC has brought new opportunities and challenges to its treatment. Molecular targeted agents, including tyrosine kinase inhibitors and monoclonal antibodies, have appeared and developed rapidly since the mechanisms were elucidated. These agents have gradually became a preferred choice of treatment of PLC and represents the future trend. In this paper, we will review the molecular pathogenesis of PLC and the targeted agents.
Collapse
|
30
|
Halib N, Mohd Amin MCI, Ahmad I, Abrami M, Fiorentino S, Farra R, Grassi G, Musiani F, Lapasin R, Grassi M. Topological characterization of a bacterial cellulose-acrylic acid polymeric matrix. Eur J Pharm Sci 2014; 62:326-333. [PMID: 24932712 DOI: 10.1016/j.ejps.2014.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/29/2014] [Accepted: 06/04/2014] [Indexed: 02/07/2023]
Abstract
This paper focuses on the micro- and nano-topological organization of a hydrogel, constituted by a mixture of bacterial cellulose and acrylic acid, and intended for biomedical applications. The presence of acrylic acid promotes the formation of two interpenetrated continuous phases: the primary "pores phase" (PP) containing only water and the secondary "polymeric network phase" (PNP) constituted by the polymeric network swollen by the water. Low field Nuclear Magnetic Resonance (LF NMR), rheology, Scanning Electron Microscopy (SEM) and release tests were used to determine the characteristics of the two phases. In particular, we found that this system is a strong hydrogel constituted by 81% (v/v) of PP phase the remaining part being occupied by the PNP phase. Pores diameters span in the range 10-100 μm, the majority of them (85%) falling in the range 30-90 μm. The high PP phase tortuosity indicates that big pores are not directly connected to each other, but their connection is realized by a series of interconnected small pores that rend the drug path tortuous. The PNP is characterized by a polymer volume fraction around 0.73 while mesh size is around 3 nm. The theoretical interpretation of the experimental data coming from the techniques panel adopted, yielded to the micro- and nano-organization of our hydrogel.
Collapse
Affiliation(s)
- N Halib
- Faculty of Dentistry, Universiti Sains Islam Malaysia (USIM), Aras 15, Menara B, Persiaran MPAJ, Jalan Pandan Utama, Pandan Indah, 55100 Kuala Lumpur, Malaysia.
| | - M C I Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia.
| | - I Ahmad
- School of Chemical Science and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia.
| | - M Abrami
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6, 34127 Trieste, Italy.
| | - S Fiorentino
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6, 34127 Trieste, Italy.
| | - R Farra
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6, 34127 Trieste, Italy.
| | - G Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy.
| | - F Musiani
- Scuola Internazionale Superiore di Studi Avanzati (SISSA/ISAS), Via Bonomea 265, I-34136 Trieste, Italy.
| | - R Lapasin
- School of Chemical Science and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia.
| | - M Grassi
- School of Chemical Science and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia.
| |
Collapse
|
31
|
Dapas B, Dall'Acqua S, Bulla R, Agostinis C, Perissutti B, Invernizzi S, Grassi G, Voinovich D. Immunomodulation mediated by a herbal syrup containing a standardized Echinacea root extract: a pilot study in healthy human subjects on cytokine gene expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:1406-1410. [PMID: 24877712 DOI: 10.1016/j.phymed.2014.04.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/03/2014] [Accepted: 04/20/2014] [Indexed: 06/03/2023]
Abstract
In this study, the immunomodulatory effect of a triply standardized Echinacea angustifolia root extract (Polinacea(®)) was evaluated in 10 healthy subjects. Ten ml of syrup containing one hundred mg of extract (corresponding to 4.7 mg of Echinacoside and 8.0mg of a high molecular weight-20,000 Da- polysaccharide) were administered as a herbal syrup once a day for one month. The immunomodulatory effect was evaluated before and after herbal syrup administration evaluating the expression levels of the cytokines IL-2, IL-8, IL-6 and TNF-α. Cytokine expression was studied in lympho-monocytes and in plasma samples measuring the mRNA and protein levels, respectively. The results were analysed by ANOVA and non-parametric Friedman rank sum tests; when possible it was adopted a pair-wise comparisons at different post-treatment times, using the paired t-tests with Holm correction. The correlation between the variations of cytokine plasma levels and the respective mRNA was carried out using a linear regression model. In lympho-monocytes our data indicate the up-regulation of the mRNA levels of IL-2 and IL-8 and the down regulation of the mRNA levels of the pro-inflammatory cytokines TNF-α and IL6. The differential regulation was maximal after 14 days of treatment. IL-2 up-regulation and IL-6 down-regulation were also confirmed at the protein level in plasma. Finally, the up-regulation of the mRNA of IL-2/IL-8 and the down-regulation of IL-6 positively correlated with the protein levels detected in the plasma. In conclusion, this pilot study suggests a relevant role for the standardized Echinacea angustifolia root extract in the control of cytokine expression. This first demonstration of the immuno-modulating activity of Echinacea angustifolia root extract in the healthy subject, supports at least in part the common use of such products as health promoting supplement.
Collapse
Affiliation(s)
- B Dapas
- Department of Life Sciences, Cattinara Hospital, University of Trieste, Strada di Fiume 447, Trieste, Italy
| | - S Dall'Acqua
- Department of Pharmaceutical Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - R Bulla
- Department of Life Sciences, University of Trieste, Via Valerio 28, 34127 Trieste, Italy
| | - C Agostinis
- Department of Life Sciences, University of Trieste, Via Valerio 28, 34127 Trieste, Italy; Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell'Istria 65/1, 34100 Trieste, Italy
| | - B Perissutti
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, P.le Europa 1, 34127 Trieste, Italy
| | - S Invernizzi
- Department of Life Sciences, University of Trieste, Via Valerio 28, 34127 Trieste, Italy
| | - G Grassi
- Department of Life Sciences, Cattinara Hospital, University of Trieste, Strada di Fiume 447, Trieste, Italy
| | - D Voinovich
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, P.le Europa 1, 34127 Trieste, Italy.
| |
Collapse
|
32
|
Dapas B, Grassi M, Grassi G. Can TIE-2 expressing monocytes represent a novel marker for hepatocellular carcinoma? Hepatobiliary Surg Nutr 2014; 3:175-178. [PMID: 25202693 PMCID: PMC4141291 DOI: 10.3978/j.issn.2304-3881.2014.07.07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/01/2014] [Indexed: 12/29/2022]
Abstract
Hepatocellular carcinoma (HCC), the predominant form of primary liver cancer, is a global health problem representing the sixth most common cancer and the third cause of cancer related death worldwide. The number of deaths per year in HCC is comparable to the incidence number, underlying the aggressive behavior of HCC and the modest efficacy of available curative treatments. Effective HCC treatment is problematic also due to the lack of early and specific diagnostic markers. In this regard, particular interest has been put on the tyrosine kinase with Ig and endothelial growth factor (EGF) homology domains 2 (TIE2), a receptor of angiopoietins, predominantly present on endothelial cells but also observed on monocytes [TIE-2-expressing monocytes (TEMs)]. Recently, a work by Matsubara et al. showed that the amount of circulating TEMs is higher in hepatitis virus C (HCV)/HCC patients compared to HCV patients or healthy subjects. Additionally the authors showed that TEMs have a diagnostic potential for HCC. Whereas the molecular mechanisms responsible for this observation remain elusive and further studies are necessary to confirm this finding, the work of Matsubara et al. may contribute to the identification of a novel HCC prognostic and diagnostic marker.
Collapse
|
33
|
Cavallaro G, Licciardi M, Amato G, Sardo C, Giammona G, Farra R, Dapas B, Grassi M, Grassi G. Synthesis and characterization of polyaspartamide copolymers obtained by ATRP for nucleic acid delivery. Int J Pharm 2014; 466:246-257. [PMID: 24631053 DOI: 10.1016/j.ijpharm.2014.03.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/07/2014] [Accepted: 03/09/2014] [Indexed: 01/23/2023]
Abstract
Nucleic acid molecules such as small interfering RNAs (siRNAs) and plasmidic DNAs (pDNAs) have been shown to have the potential to be of therapeutic value in different human diseases. Their practical use is however compromised by the lack of appropriate release systems. Delivered as naked molecules, siRNAs/pDNAs are rapidly degraded by extracellular nucleases thus considerably reducing the amount of molecule which can reach the target cells. Additionally, the anionic charge of the phosphate groups present on the siRNAs/pDNAs backbone, disfavors the interaction with the negatively charged surface of the cell membrane. In this paper we describe the generation of a novel polymer able to deliver both siRNAs and pDNAs. The combined release of these molecules is used in many different experimental settings such as the evaluation of the silencing efficiency of a given siRNA targeted against a given RNA, encoded by the pDNA. The possibility to use the same delivery system is very convenient from the technical point of view and it allows minimizing possible artifacts introduced by the use of different delivery agents for siRNAs and pDNA. The copolymer described here is based on α,β-poly(N-2-hydroxyethyl)-d,l-aspartamide (PHEA) bearing positively chargeable side oligochains, with diethylamino ethyl methacrylate (DEAEMA) as monomer. Monomer polymerization has been obtained by atom transfer radical polymerization (ATRP), a technique which allows the precise polymerization of the monomer. In addition to the chemical-physical characterization of the polymer, we provide evidences of the polymer ability to delivery both siRNAs and pDNA to cultured cells. Whereas additional investigations are necessary to study the delivery mechanisms of this polyplex, the polymer generated represents a novel and convenient device for the delivery of both siRNAs and pDNA.
Collapse
Affiliation(s)
- G Cavallaro
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), University of Palermo, via Archirafi 32, Palermo 90123, Italy.
| | - M Licciardi
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - G Amato
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - C Sardo
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - G Giammona
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - R Farra
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, Trieste I-34127, Italy
| | - B Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy
| | - M Grassi
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, Trieste I-34127, Italy
| | - G Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy
| |
Collapse
|