1
|
Chen M, Chen X, Shui Y, Ji C, Guo W. Carfilzomib inhibits the progression of hepatocellular cancer by upregulating GADD45α expression. Oncol Lett 2025; 29:208. [PMID: 40070787 PMCID: PMC11894515 DOI: 10.3892/ol.2025.14955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent and lethal cancers affecting humans. Currently, there are limited effective treatments available for HCC. Carfilzomib, a proteasome inhibitor, is known to exert anti-HCC activities; however, its underlying mechanisms of action remain unclear. In the present study, the efficacy of carfilzomib against HCC was evaluated and its underlying mechanisms were explored. Cell Counting Kit-8, 5-ethynyl-2-deoxyuridine staining and colony formation assays were employed to analyze the antiproliferative effect of carfilzomib on MHCC-97H and Huh7 cells. Additionally, flow cytometry was used to assess the effect of carfilzomib on the cell cycle and Transwell assays were used to evaluate the effect of carfilzomib on cell migration and invasion. Western blotting was utilized to examine the protein expression levels associated with cell cycle arrest. Furthermore, short hairpin RNA (shRNA) transfection was used to investigate the role of DNA damage inducible α (GADD45α) on carfilzomib-induced cell cycle arrest. A xenograft tumor model using nude mice was employed to evaluate the anti-HCC activity of carfilzomib in vivo. The findings demonstrated that carfilzomib inhibited proliferation, invasion and migration in both MHCC-97H and Huh7 cells. In addition, carfilzomib caused cell cycle arrest by suppressing the expression of cyclin A2, cyclin E1 and cyclin-dependent kinases 2 and 4. Carfilzomib also upregulated the expression of GADD45α, activated the MAPK pathway and inhibited GADD45α through shRNA abolished carfilzomib-induced cell cycle arrest in HCC cells. In addition, carfilzomib inhibited Huh7 cell growth in vivo. To conclude, the present research revealed that carfilzomib inhibits the progression of HCC cells by upregulating GADD45α expression, suggesting that carfilzomib could be a potential chemotherapeutic agent against HCC.
Collapse
Affiliation(s)
- Mingxun Chen
- Liver Transplantation Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Hepatobiliary and Pancreatic Minimally Invasive Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xu Chen
- Department of Hepatobiliary and Pancreatic Minimally Invasive Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yifang Shui
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chunyong Ji
- Department of Hepatobiliary and Pancreatic Minimally Invasive Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
2
|
Shrivastava S, Ratnacaram CK. Targeting the TGF-β-p21 axis: a critical regulator of bleomycin-induced cell cycle arrest and apoptosis in vitro-implication for progressive cervical cancer therapy. Med Oncol 2025; 42:85. [PMID: 40019671 DOI: 10.1007/s12032-025-02624-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Cervical cancer signifies a global health concern and is a major cause of cancer mortality in women worldwide. Dysregulation of apoptotic pathway and cell cycle play a role in cancer development. Aberrant signalling pathways leads to complex mechanisms leading to the severity. Bleomycin is an anti-tumor glycopeptide molecule which exhibits impressive cytotoxicity in cancer cells. However, its modulating significance on TGF-β induced cell cycle arrest and apoptosis in cervical cancer remains elusive. We confirmed the cytotoxicity, anti proliferative effect of bleomycin in HeLa cells. Meanwhile, bleomycin also led to the suppression of cell migration, invasion. Relative gene expression quantification for cell cycle regulatory, apoptotic, and TGF-β member's genes was done by qRT-PCR. Bleomycin markedly downregulated the expression of cyclin A2, cyclin B1 and cell cycle arrest at G2/M phase in vitro. Dual AO/EB and Propidium iodide staining was done to evaluate early and late apoptosis in cervical cancer cells. Bleomycin prompts early and late apoptosis in cervical cancer cells by chromatin condensation and blebbing. Mechanistically, bleomycin activates TGF-β induced p21 cascade by upregulation of GADD45A and GDF11, stabilizing p53, to induce cell cycle arrest and apoptosis. Bleomycin induces DNA damage triggering TGF-β pathway. This study can broaden our understanding of the signalling mechanisms that could develop effective strategies for cancer therapy. Elucidation of these pathways in cervical cancer may ultimately lead to novel and more effective treatments. Here, we highlight apoptosis-inducing drug as a therapeutic strategy to regulate the process of carcinogenesis, and regulating apoptosis could benefit cancer treatment and prevention.
Collapse
Affiliation(s)
- Somi Shrivastava
- Cancer Research and Therapeutics Division, Yenepoya Research Centre, Yenepoya (Deemed to Be) University, Mangalore, 575018, Karnataka, India
| | - Chandrahas Koumar Ratnacaram
- Cancer Research and Therapeutics Division, Yenepoya Research Centre, Yenepoya (Deemed to Be) University, Mangalore, 575018, Karnataka, India.
- Specialised Research Unit, Yenepoya Medical College, Yenepoya (Deemed to Be) University, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
3
|
de Oliveira Silva N, de Lima LVA, de Oliveira LM, da Silva MF, de Aguiar AP, Semprebon SC, Favaron PO, Lepri SR, Felicidade I, Mantovani MS. Cellular and molecular antiproliferative effects in 2D monolayer and 3D-cultivated HT-29 cells treated with zerumbone. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1561-1573. [PMID: 37672080 DOI: 10.1007/s00210-023-02701-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023]
Abstract
Zerumbone (ZER) is a phytochemical isolated from plants of the Zingiberaceae family. Numerous studies have demonstrated its diverse pharmacological properties, particularly its potent antitumorigenic activity. This study aimed to assess the antiproliferative effects of ZER on HT-29 cells cultivated in both two-dimensional (2D) monolayer and three-dimensional (3D) spheroid culture systems. The evaluation of growth (size), cell death, and cell cycle arrest in 3D spheroid HT-29 cells was correlated with mRNA expression data. Treatment of 2D cells revealed that ZER exhibited cytotoxicity at concentrations above 30 µM, and an IC50 of 83.54 µM (24-h post-ZER treatment) effectively suppressed cell migration. In the 3D model, ZER induced an increase in spheroid volume over a 72-h period attributed to disaggregation and reconfiguration of characteristic zones. Analysis of cell death demonstrated a significant rise in apoptotic cells after 24 h of ZER treatment, along with cell cycle arrest in the G1 phase. Furthermore, ZER treatment resulted in alterations in mRNA expression, affecting key signaling pathways involved in cell death (BCL2 and BBC3), endoplasmic reticulum stress (ERN1), DNA damage (GADD45A), cell cycle regulation (CDKN1A, NFKB1, MYC, and TP53), and autophagy (BECN1 and SQSTM1). These findings suggested that ZER holds promise as a potential candidate for the development of novel anticancer agents that can modulate crucial cell signaling pathways. Additionally, the use of the 3D culture system proved to be a valuable tool in our investigation.
Collapse
Affiliation(s)
- Nayane de Oliveira Silva
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Luan Vitor Alves de Lima
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Liana Martins de Oliveira
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Matheus Felipe da Silva
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Amanda Passuello de Aguiar
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Simone Cristine Semprebon
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Phelipe Oliveira Favaron
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Sandra Regina Lepri
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Ingrid Felicidade
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Mario Sergio Mantovani
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Paraná, Brazil.
| |
Collapse
|
4
|
Rana M, Perotti A, Bisset LM, Smith JD, Lamden E, Khan Z, Ismail MK, Ellis K, Armstrong KA, Hodder SL, Bertoli C, Meneguello L, de Bruin RAM, Morris JR, Romero-Canelon I, Tucker JHR, Hodges NJ. A ferrocene-containing nucleoside analogue targets DNA replication in pancreatic cancer cells. Metallomics 2022; 14:mfac041. [PMID: 35689667 PMCID: PMC9320222 DOI: 10.1093/mtomcs/mfac041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/11/2022] [Indexed: 11/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease that remains refractory to existing treatments including the nucleoside analogue gemcitabine. In the current study we demonstrate that an organometallic nucleoside analogue, the ferronucleoside 1-(S,Rp), is cytotoxic in a panel of PDAC cell lines including gemcitabine-resistant MIAPaCa2, with IC50 values comparable to cisplatin. Biochemical studies show that the mechanism of action is inhibition of DNA replication, S-phase cell cycle arrest and stalling of DNA-replication forks, which were directly observed at single molecule resolution by DNA-fibre fluorography. In agreement with this, transcriptional changes following treatment with 1-(S,Rp) include activation of three of the four genes (HUS1, RAD1, RAD17) of the 9-1-1 check point complex clamp and two of the three genes (MRE11, NBN) that form the MRN complex as well as activation of multiple downstream targets. Furthermore, there was evidence of phosphorylation of checkpoint kinases 1 and 2 as well as RPA1 and gamma H2AX, all of which are considered biochemical markers of replication stress. Studies in p53-deficient cell lines showed activation of CDKN1A (p21) and GADD45A by 1-(S,Rp) was at least partially independent of p53. In conclusion, because of its potency and activity in gemcitabine-resistant cells, 1-(S,Rp) is a promising candidate molecule for development of new treatments for PDAC.
Collapse
Affiliation(s)
- Marium Rana
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- School of Chemistry, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Alessio Perotti
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Lucy M Bisset
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - James D Smith
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Emma Lamden
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Zahra Khan
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Media K Ismail
- Department of pharmacy, college of pharmacy, Knowledge University, 44001 Erbil, Kurdistan Region, Iraq
| | - Katherine Ellis
- Institute of Cancer and Genomic Sciences, and The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Katie A Armstrong
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Samantha L Hodder
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Cosetta Bertoli
- MRC Laboratory or Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Leticia Meneguello
- MRC Laboratory or Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Robertus A M de Bruin
- MRC Laboratory or Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Joanna R Morris
- Institute of Cancer and Genomic Sciences, and The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Isolda Romero-Canelon
- School of Pharmacy, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - James H R Tucker
- School of Chemistry, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nikolas J Hodges
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
5
|
Zhao L, Yan C, Kong S, Jia T, Chu Z, Yang L, Wu J, Geng S, Guo K. Biosafety and differentially expressed genes analysis of melanoma cells treated with cold atmospheric plasma. JOURNAL OF BIOPHOTONICS 2022; 15:e202100403. [PMID: 35261164 DOI: 10.1002/jbio.202100403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
Cold atmospheric plasma (CAP) has attracted increasing attention due to its anti-bacterial and anti-tumor effects. Melanoma is an aggressive malignancy with increasing incidence rate and poor prognosis. Evaluating cell viability, apoptosis rate and reactive species injection efficiency of melanoma cells and human keratinocyte cells (HaCaT) treated with CAP to analyze biological safety of CAP. RNA-sequencing (RNA-seq) of A875 cells before and after treatment was performed to further explore the anti-tumor mechanism of CAP. CAP had a more significant biological effect on melanoma cells than HaCaT cells by inhibiting proliferation and promoting apoptosis. RNA-sequencing analysis showed that besides MAPK and p53 apoptotic signaling pathways, necroptosis and autophagy also played important roles in CAP-induced melanoma cells death. CAP can selectively kill melanoma cells and has good biosafety cytologically. Besides apoptosis, CAP can induce cell death via autophagy and necroptosis.
Collapse
Affiliation(s)
- Lihong Zhao
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cong Yan
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuzhen Kong
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Jia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhaowei Chu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Yang
- Department of Dermatology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jian Wu
- State Key Laboratory of Electrical Insulation and Power Equipment, Xi'an Jiaotong University, Xi'an, China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kun Guo
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
6
|
Patel K, Murray MG, Whelan KA. Roles for GADD45 in Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1360:23-39. [DOI: 10.1007/978-3-030-94804-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
7
|
Soni V, Adhikari M, Simonyan H, Lin L, Sherman JH, Young CN, Keidar M. In Vitro and In Vivo Enhancement of Temozolomide Effect in Human Glioblastoma by Non-Invasive Application of Cold Atmospheric Plasma. Cancers (Basel) 2021; 13:4485. [PMID: 34503293 PMCID: PMC8430547 DOI: 10.3390/cancers13174485] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive forms of adult brain cancers and is highly resistant to treatment, with a median survival of 12-18 months after diagnosis. The poor survival is due to its infiltrative pattern of invasion into the normal brain parenchyma, the diffuse nature of its growth, and its ability to quickly grow, spread, and relapse. Temozolomide is a well-known FDA-approved alkylating chemotherapy agent used for the treatment of high-grade malignant gliomas, and it has been shown to improve overall survival. However, in most cases, the tumor relapses. In recent years, CAP has been used as an emerging technology for cancer therapy. The purpose of this study was to implement a combination therapy of CAP and TMZ to enhance the effect of TMZ and apparently sensitize GBMs. In vitro evaluations in TMZ-sensitive and resistant GBM cell lines established a CAP chemotherapy enhancement and potential sensitization effect across various ranges of CAP jet application. This was further supported with in vivo findings demonstrating that a single CAP jet applied non-invasively through the skull potentially sensitizes GBM to subsequent treatment with TMZ. Gene functional enrichment analysis further demonstrated that co-treatment with CAP and TMZ resulted in a downregulation of cell cycle pathway genes. These observations indicate that CAP can be potentially useful in sensitizing GBM to chemotherapy and for the treatment of glioblastoma as a non-invasive translational therapy.
Collapse
Affiliation(s)
- Vikas Soni
- Department of Mechanical and Aerospace Engineering, MPNL, The George Washington University, Washington, DC 20052, USA; (V.S.); (M.A.); (L.L.); (J.H.S.)
| | - Manish Adhikari
- Department of Mechanical and Aerospace Engineering, MPNL, The George Washington University, Washington, DC 20052, USA; (V.S.); (M.A.); (L.L.); (J.H.S.)
| | - Hayk Simonyan
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA;
| | - Li Lin
- Department of Mechanical and Aerospace Engineering, MPNL, The George Washington University, Washington, DC 20052, USA; (V.S.); (M.A.); (L.L.); (J.H.S.)
| | - Jonathan H. Sherman
- Department of Mechanical and Aerospace Engineering, MPNL, The George Washington University, Washington, DC 20052, USA; (V.S.); (M.A.); (L.L.); (J.H.S.)
| | - Colin N. Young
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA;
| | - Michael Keidar
- Department of Mechanical and Aerospace Engineering, MPNL, The George Washington University, Washington, DC 20052, USA; (V.S.); (M.A.); (L.L.); (J.H.S.)
| |
Collapse
|
8
|
Mendanha D, Vieira de Castro J, Moreira J, Costa BM, Cidade H, Pinto M, Ferreira H, Neves NM. A New Chalcone Derivative with Promising Antiproliferative and Anti-Invasion Activities in Glioblastoma Cells. Molecules 2021; 26:molecules26113383. [PMID: 34205043 PMCID: PMC8199914 DOI: 10.3390/molecules26113383] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma (GBM) is the most common and most deadly primary malignant brain tumor. Current therapies are not effective, the average survival of GBM patients after diagnosis being limited to few months. Therefore, the discovery of new treatments for this highly aggressive brain cancer is urgently needed. Chalcones are synthetic and naturally occurring compounds that have been widely investigated as anticancer agents. In this work, three chalcone derivatives were tested regarding their inhibitory activity and selectivity towards GBM cell lines (human and mouse) and a non-cancerous mouse brain cell line. The chalcone 1 showed the most potent and selective cytotoxic effects in the GBM cell lines, being further investigated regarding its ability to reduce critical hallmark features of GBM and to induce apoptosis and cell cycle arrest. This derivative showed to successfully reduce the invasion and proliferation capacity of tumor cells, both key targets for cancer treatment. Moreover, to overcome potential systemic side effects and its poor water solubility, this compound was encapsulated into liposomes. Therapeutic concentrations were incorporated retaining the potent in vitro growth inhibitory effect of the selected compound. In conclusion, our results demonstrated that this new formulation can be a promising starting point for the discovery of new and more effective drug treatments for GBM.
Collapse
Affiliation(s)
- Daniel Mendanha
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (D.M.); (J.V.d.C.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal;
| | - Joana Vieira de Castro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (D.M.); (J.V.d.C.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal;
| | - Joana Moreira
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (J.M.); (H.C.); (M.P.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Bruno M. Costa
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal;
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Honorina Cidade
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (J.M.); (H.C.); (M.P.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (J.M.); (H.C.); (M.P.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Helena Ferreira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (D.M.); (J.V.d.C.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal;
- Correspondence: (H.F.); (N.M.N.)
| | - Nuno M. Neves
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (D.M.); (J.V.d.C.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal;
- Correspondence: (H.F.); (N.M.N.)
| |
Collapse
|
9
|
Anti-tumour activity of deer growing antlers and its potential applications in the treatment of malignant gliomas. Sci Rep 2021; 11:42. [PMID: 33420194 PMCID: PMC7794318 DOI: 10.1038/s41598-020-79779-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/10/2020] [Indexed: 02/05/2023] Open
Abstract
A recent study showed that antlers have evolved a high rate of growth due to the expression of proto-oncogenes and that they have also evolved to express several tumour suppressor genes to control the risk of cancer. This may explain why deer antler velvet (DAV) extract shows anti-tumour activity. The fast growth of antler innervation through the velvet in close association to blood vessels provides a unique environment to study the fast but non-cancerous proliferation of heterogeneous cell populations. We set out to study the anti-cancer effect of DAV in glioblastoma (GB) cell lines in comparison with temozolomide, a chemotherapeutic drug used to treat high-grade brain tumours. Here we report, for the first time, that DAV extract from the tip, but not from mid-parts of the antler, exhibits an anti-tumour effect in GB cell lines (T98G and A172) while being non-toxic in non-cancerous cell lines (HEK293 and HACAT). In T98G cells, DAV treatment showed reduced proliferation (37.5%) and colony-formation capacity (84%), inhibited migration (39%), induced changes in cell cycle progression, and promoted apoptosis. The anticancer activity of DAV extract as demonstrated by these results may provide a new therapeutic strategy for GB treatment.
Collapse
|
10
|
Atiq A, Parhar I. Anti-neoplastic Potential of Flavonoids and Polysaccharide Phytochemicals in Glioblastoma. Molecules 2020; 25:E4895. [PMID: 33113890 PMCID: PMC7660188 DOI: 10.3390/molecules25214895] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Clinically, gliomas are classified into four grades, with grade IV glioblastoma multiforme being the most malignant and deadly, which accounts for 50% of all gliomas. Characteristically, glioblastoma involves the aggressive proliferation of cells and invasion of normal brain tissue, outcomes as poor patient prognosis. With the current standard therapy of glioblastoma; surgical resection and radiotherapy followed by adjuvant chemotherapy with temozolomide, it remains fatal, because of the development of drug resistance, tumor recurrence, and metastasis. Therefore, the need for the effective therapeutic option for glioblastoma remains elusive. Previous studies have demonstrated the chemopreventive role of naturally occurring pharmacological agents through preventing or reversing the initiation phase of carcinogenesis or arresting the cancer progression phase. In this review, we discuss the role of natural phytochemicals in the amelioration of glioblastoma, with the aim to improve therapeutic outcomes, and minimize the adverse side effects to improve patient's prognosis and enhancing their quality of life.
Collapse
Affiliation(s)
- Ayesha Atiq
- Brain Research Institute Monash Sunway (BRIMS), Jeffery Cheah School of Medicine, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia;
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway (BRIMS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
11
|
Herbener VJ, Burster T, Goreth A, Pruss M, von Bandemer H, Baisch T, Fitzel R, Siegelin MD, Karpel-Massler G, Debatin KM, Westhoff MA, Strobel H. Considering the Experimental use of Temozolomide in Glioblastoma Research. Biomedicines 2020; 8:E151. [PMID: 32512726 PMCID: PMC7344626 DOI: 10.3390/biomedicines8060151] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Temozolomide (TMZ) currently remains the only chemotherapeutic component in the approved treatment scheme for Glioblastoma (GB), the most common primary brain tumour with a dismal patient's survival prognosis of only ~15 months. While frequently described as an alkylating agent that causes DNA damage and thus-ultimately-cell death, a recent debate has been initiated to re-evaluate the therapeutic role of TMZ in GB. Here, we discuss the experimental use of TMZ and highlight how it differs from its clinical role. Four areas could be identified in which the experimental data is particularly limited in its translational potential: 1. transferring clinical dosing and scheduling to an experimental system and vice versa; 2. the different use of (non-inert) solvent in clinic and laboratory; 3. the limitations of established GB cell lines which only poorly mimic GB tumours; and 4. the limitations of animal models lacking an immune response. Discussing these limitations in a broader biomedical context, we offer suggestions as to how to improve transferability of data. Finally, we highlight an underexplored function of TMZ in modulating the immune system, as an example of where the aforementioned limitations impede the progression of our knowledge.
Collapse
Affiliation(s)
- Verena J. Herbener
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Alicia Goreth
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Maximilian Pruss
- Department of Gynecology and Obstetrics, Medical Faculty, University Hospital of the Heinrich-Heine-University Duesseldorf, D-40225 Duesseldorf, Germany;
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Hélène von Bandemer
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Rahel Fitzel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Markus D. Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| |
Collapse
|
12
|
Cai Y, Zhang W, Zhang R, Cui X, Fang J. Combined Use of Three Machine Learning Modeling Methods to Develop a Ten-Gene Signature for the Diagnosis of Ventilator-Associated Pneumonia. Med Sci Monit 2020; 26:e919035. [PMID: 32031163 PMCID: PMC7020762 DOI: 10.12659/msm.919035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND This study aimed to use three modeling methods, logistic regression analysis, random forest analysis, and fully-connected neural network analysis, to develop a diagnostic gene signature for the diagnosis of ventilator-associated pneumonia (VAP). MATERIAL AND METHODS GSE30385 from the Gene Expression Omnibus (GEO) database identified differentially expressed genes (DEGs) associated with patients with VAP. Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment identified the molecular functions of the DEGs. The least absolute shrinkage and selection operator (LASSO) regression analysis algorithm was used to select key genes. Three modeling methods, including logistic regression analysis, random forest analysis, and fully-connected neural network analysis, also known as also known as the feed-forward multi-layer perceptron (MLP), were used to identify the diagnostic gene signature for patients with VAP. RESULTS Sixty-six DEGs were identified for patients who had VAP (VAP+) and who did not have VAP (VAP-). Ten essential or feature genes were identified. Upregulated genes included matrix metallopeptidase 8 (MMP8), arginase 1 (ARG1), haptoglobin (HP), interleukin 18 receptor 1 (IL18R1), and NLR family apoptosis inhibitory protein (NAIP). Down-regulated genes included complement factor D (CFD), pleckstrin homology-like domain family A member 2 (PHLDA2), plasminogen activator, urokinase (PLAU), laminin subunit beta 3 (LAMB3), and dual-specificity phosphatase 2 (DUSP2). Logistic regression, random forest, and MLP analysis showed receiver operating characteristic (ROC) curve area under the curve (AUC) values of 0.85, 0.86, and 0.87, respectively. CONCLUSIONS Logistic regression analysis, random forest analysis, and MLP analysis identified a ten-gene signature for the diagnosis of VAP.
Collapse
Affiliation(s)
- Yunfang Cai
- Department of Anesthesia, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Wen Zhang
- Department of Anesthesia, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Runze Zhang
- Department of Anesthesia, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoying Cui
- Department of Anesthesia, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Jun Fang
- Department of Anesthesia, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
13
|
Honorato JR, Hauser-Davis RA, Saggioro EM, Correia FV, Sales-Junior SF, Soares LOS, Lima LDR, Moura-Neto V, Lopes GPDF, Spohr TCLDS. Role of Sonic hedgehog signaling in cell cycle, oxidative stress, and autophagy of temozolomide resistant glioblastoma. J Cell Physiol 2019; 235:3798-3814. [PMID: 31613002 DOI: 10.1002/jcp.29274] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
The first-line chemotherapy treatment for Glioblastoma (GBM) - the most aggressive and frequent brain tumor - is temozolomide (TMZ). The Sonic hedgehog (SHH) pathway is involved with GBM tumorigenesis and TMZ chemoresistance. The role of SHH pathway inhibition in the potentiation of TMZ's effects using T98G, U251, and GBM11 cell lines is investigated herein. The combination of GANT-61 and TMZ over 72 hr suggested a synergistic effect. All TMZ-resistant cell lines displayed a significant decrease in cell viability, increased DNA fragmentation and loss of membrane integrity. For T98G cells, G2 /M arrest was observed, while U251 cells presented a significant increase in reactive oxygen species production and catalase activity. All the cell lines presented acidic vesicles formation correlated to Beclin-1 overexpression. The combined treatment also enhanced GLI1 expression, indicating the presence of select resistant cells. The selective inhibition of the SHH pathway potentiated the cytotoxic effect of TMZ, thus becoming a promising in vitro strategy for GBM treatment.
Collapse
Affiliation(s)
- Jessica R Honorato
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rachel A Hauser-Davis
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Enrico M Saggioro
- Departamento de Saneamento e Saúde Ambiental, Escola Nacional de Saúde Pública (ENSP), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Fábio V Correia
- Departamento de Ciências Naturais, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Sidney F Sales-Junior
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública (ENSP), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Lorena O S Soares
- Departamento de Ciências Naturais, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Leandro da R Lima
- Departamento de Ciências Naturais, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle P de F Lopes
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Biotecnologia Marinha, Instituto de Estudos do Mar Almirante Paulo Moreira (IEAPM)/Coordenação de Pesquisa, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Tania C L de S Spohr
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Vengoji R, Macha MA, Nimmakayala RK, Rachagani S, Siddiqui JA, Mallya K, Gorantla S, Jain M, Ponnusamy MP, Batra SK, Shonka N. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:266. [PMID: 31215502 PMCID: PMC6582495 DOI: 10.1186/s13046-019-1264-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022]
Abstract
Background Glioblastoma (GBM) is an aggressive brain tumor with universal recurrence and poor prognosis. The recurrence is largely driven by chemoradiation resistant cancer stem cells (CSCs). Epidermal growth factor receptor (EGFR) and its mutant EGFRvIII are amplified in ~ 60% and ~ 30% of GBM patients, respectively; however, therapies targeting EGFR have failed to improve disease outcome. EGFRvIII-mediated cross-activation of tyrosine kinase receptor, cMET, regulates GBM CSC maintenance and promote tumor recurrence. Here, we evaluated the efficacy of pan-EGFR inhibitor afatinib and Temozolomide (TMZ) combination on GBM in vitro and in vivo. Methods We analyzed the effect of afatinib and temozolomide (TMZ) combination on GBM cells U87MG and U251 engineered to express wild type (WT) EGFR, EGFRvIII or EGFRvIII dead kinase, CSCs isolated from U87 and U87EGFRvIII in vitro. The therapeutic utility of the drug combination was investigated on tumor growth and progression using intracranially injected U87EGFRvIII GBM xenografts. Results Afatinib and TMZ combination synergistically inhibited the proliferation, clonogenic survival, motility, invasion and induced senescence of GBM cells compared to monotherapy. Mechanistically, afatinib decreased U87EGFRvIII GBM cell proliferation and motility/invasion by inhibiting EGFRvIII/AKT, EGFRvIII/JAK2/STAT3, and focal adhesion kinase (FAK) signaling pathways respectively. Interestingly, afatinib specifically inhibited EGFRvIII-cMET crosstalk in CSCs, resulting in decreased expression of Nanog and Oct3/4, and in combination with TMZ significantly decreased their self-renewal property in vitro. More interestingly, afatinib and TMZ combination significantly decreased the xenograft growth and progression compared to single drug alone. Conclusion Our study demonstrated significant inhibition of GBM tumorigenicity, CSC maintenance in vitro, and delayed tumor growth and progression in vivo by combination of afatinib and TMZ. Our results warrant evaluation of this drug combination in EGFR and EGFRvIII amplified GBM patients. Electronic supplementary material The online version of this article (10.1186/s13046-019-1264-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
15
|
Shi Q, Sutariya V, Varghese Gupta S, Bhatia D. GADD45α-targeted suicide gene therapy driven by synthetic CArG promoter E9NS sensitizes NSCLC cells to cisplatin, resveratrol, and radiation regardless of p53 status. Onco Targets Ther 2019; 12:3161-3170. [PMID: 31114253 PMCID: PMC6497884 DOI: 10.2147/ott.s192061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
Background: GADD45α is a tumor suppressor protein often upregulated by environmental stresses and DNA-damage agents to cause growth arrest, apoptosis, tumor growth inhibition, and anti-angiogenesis. A novel suicide gene therapy vector pE9NS.G45α was engineered by cloning GADD45α opening reading frame downstream to the synthetic CArG promoter E9NS, which contains nine repeats of CArG element with modified core A/T sequence and functions as a molecular switch to drive the expression of GADD45α. The current study aims to determine the efficacy of this suicide gene therapy vector in combination with cisplatin, resveratrol, and radiation in NSCLC cell lines with various p53 statuses. Methods: Three NSCLC cell lines, H1299 (deleted p53), A549 (wild-type p53), and H23 (mutated p53), were examined in the present investigation to represent NSCLC with different p53 functions. MTT assay was conducted to select suitable doses of cisplatin, resveratrol, and radiation for gene therapy, and dual luciferase assay was performed to validate the activation of promoter E9NS. The efficacy of gene therapy combinations was evaluated by the amount of GADD45α expression, cell survival, and apoptosis. Results: All the combinations successfully activated promoter E9NS to elevate intracellular GADD45α protein levels and subsequently enhanced cell viability reduction and apoptosis induction regardless of p53 status. Conclusion: Our study demonstrates that GADD45α-targeted suicide gene therapy controlled by synthetic promoter E9NS sensitizes NSCLC cells to cisplatin, resveratrol, and radiation and is effective against NSCLC at least in vitro.
Collapse
Affiliation(s)
- Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, People's Republic of China
| | | | | | - Deepak Bhatia
- Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, VA, USA
| |
Collapse
|
16
|
Kaina B, Christmann M. DNA repair in personalized brain cancer therapy with temozolomide and nitrosoureas. DNA Repair (Amst) 2019; 78:128-141. [PMID: 31039537 DOI: 10.1016/j.dnarep.2019.04.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/16/2022]
Abstract
Alkylating agents have been used since the 60ties in brain cancer chemotherapy. Their target is the DNA and, although the DNA of normal and cancer cells is damaged unselectively, they exert tumor-specific killing effects because of downregulation of some DNA repair activities in cancer cells. Agents exhibiting methylating properties (temozolomide, procarbazine, dacarbazine, streptozotocine) induce at least 12 different DNA lesions. These are repaired by damage reversal mechanisms involving the alkyltransferase MGMT and the alkB homologous protein ALKBH2, and through base excision repair (BER). There is a strong correlation between the MGMT expression level and therapeutic response in high-grade malignant glioma, supporting the notion that O6-methylguanine and, for nitrosoureas, O6-chloroethylguanine are the most relevant toxic damages at therapeutically relevant doses. Since MGMT has a significant impact on the outcome of anti-cancer therapy, it is a predictive marker of the effectiveness of methylating anticancer drugs, and clinical trials are underway aimed at assessing the influence of MGMT inhibition on the therapeutic success. Other DNA repair factors involved in methylating drug resistance are mismatch repair, DNA double-strand break (DSB) repair by homologous recombination (HR) and DSB signaling. Base excision repair and ALKBH2 might also contribute to alkylating drug resistance and their downregulation may have an impact on drug sensitivity notably in cells expressing a high amount of MGMT and at high doses of temozolomide, but the importance in a therapeutic setting remains to be shown. MGMT is frequently downregulated in cancer cells (up to 40% in glioblastomas), which is due to CpG promoter methylation. Astrocytoma (grade III) are frequently mutated in isocitrate dehydrogenase (IDH1). These tumors show a surprisingly good therapeutic response. IDH1 mutation has an impact on ALKBH2 activity thus influencing DNA repair. A master switch between survival and death is p53, which often retains transactivation activity (wildtype) in malignant glioma. The role of p53 in regulating survival via DNA repair and the routes of death are discussed and conclusions as to cancer therapeutic options were drawn.
Collapse
Affiliation(s)
- Bernd Kaina
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| | - Markus Christmann
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| |
Collapse
|
17
|
Maurer GD, Heller S, Wanka C, Rieger J, Steinbach JP. Knockdown of the TP53-Induced Glycolysis and Apoptosis Regulator (TIGAR) Sensitizes Glioma Cells to Hypoxia, Irradiation and Temozolomide. Int J Mol Sci 2019; 20:ijms20051061. [PMID: 30823646 PMCID: PMC6429390 DOI: 10.3390/ijms20051061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
The TP53-induced glycolysis and apoptosis regulator (TIGAR) has been shown to decrease glycolysis, to activate the pentose phosphate pathway, and to provide protection against oxidative damage. Hypoxic regions are considered characteristic of glioblastoma and linked with resistance to current treatment strategies. Here, we established that LNT-229 glioma cell lines stably expressed shRNA constructs targeting TIGAR, and exposed them to hypoxia, irradiation and temozolomide. The disruption of TIGAR enhanced levels of reactive oxygen species and cell death under hypoxic conditions, as well as the effectiveness of irradiation and temozolomide. In addition, TIGAR was upregulated by HIF-1α. As a component of a complex network, TIGAR contributes to the metabolic adjustments that arise from either spontaneous or therapy-induced changes in tumor microenvironment.
Collapse
Affiliation(s)
- Gabriele D Maurer
- Dr. Senckenberg Institute of Neurooncology and University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany.
| | - Sonja Heller
- Dr. Senckenberg Institute of Neurooncology and University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany.
| | - Christina Wanka
- Dr. Senckenberg Institute of Neurooncology and University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany.
| | - Johannes Rieger
- Dr. Senckenberg Institute of Neurooncology and University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany.
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, University Hospital Tuebingen, Eberhard Karls University, 72076 Tuebingen, Germany.
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology and University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Ferreira J, Ramos AA, Almeida T, Azqueta A, Rocha E. Drug resistance in glioblastoma and cytotoxicity of seaweed compounds, alone and in combination with anticancer drugs: A mini review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 48:84-93. [PMID: 30195884 DOI: 10.1016/j.phymed.2018.04.062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/19/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Glioblastomas (GBM) are one of the most aggressive tumor of the central nervous system with an average life expectancy of only 1-2 years after diagnosis, even with the use of advanced treatments with surgery, radiation, and chemotherapy. There are several anticancer drugs with alkylating properties that have been used in the therapy of malignant gliomas. Temozolomide (TMZ) is one of them, widely used even in combination with ionizing radiation. However, the main disadvantage of using these types of drugs in the treatment of GBM is the development of cancer drug resistance. Research of bioactive compounds with anticancer activity has been heavily explored. PURPOSE This review focuses on a carotenoid and a phlorotannin present in seaweed, namely fucoxanthin and phloroglucinol, and their anticancer activity against glioblastoma. The combination of natural compounds with conventional drugs is also discussed. CONCLUSION Several natural compounds existing in seaweeds, such as fucoxanthin and phoroglucinol, have shown cytotoxic activity in models in vitro and in vivo, acting through different molecular mechanisms, such as antioxidant, antiproliferative, DNA damage/DNA repair, proapoptotic, antiangiogenic and antimetastic. Within the scope of interactions with conventional drugs, there are evidences that some seaweed compounds could be used to potentiate the action of anticancer drugs. However, their effects and mechanisms of action, alone or in combination with anticancer drugs, namely TMZ, in glioblastoma cell, still few explored and require more attention due to the unquestionable high potential of these marine compounds.
Collapse
Affiliation(s)
- Joana Ferreira
- Team of Histomorphology, Physiopathology and Applied Toxicology, CIIMAR - Interdisciplinary Center for Marine and Environmental Research, U.Porto - University of Porto, Avenida General Norton de Matos s/n, Matosinhos 4450-208, Portugal; Laboratory of Histology and Embryology, Department of Microscopy, ICBAS - Institute of Biomedical Sciences Abel Salazar, U.Porto - University of Porto, Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal; FCUP - Faculty of Sciences, U.Porto - University of Porto (U.Porto), Rua do Campo Alegre, Porto 4169-007, Portugal
| | - Alice Abreu Ramos
- Team of Histomorphology, Physiopathology and Applied Toxicology, CIIMAR - Interdisciplinary Center for Marine and Environmental Research, U.Porto - University of Porto, Avenida General Norton de Matos s/n, Matosinhos 4450-208, Portugal; Laboratory of Histology and Embryology, Department of Microscopy, ICBAS - Institute of Biomedical Sciences Abel Salazar, U.Porto - University of Porto, Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal.
| | - Tânia Almeida
- Team of Histomorphology, Physiopathology and Applied Toxicology, CIIMAR - Interdisciplinary Center for Marine and Environmental Research, U.Porto - University of Porto, Avenida General Norton de Matos s/n, Matosinhos 4450-208, Portugal; Laboratory of Histology and Embryology, Department of Microscopy, ICBAS - Institute of Biomedical Sciences Abel Salazar, U.Porto - University of Porto, Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal; FCUP - Faculty of Sciences, U.Porto - University of Porto (U.Porto), Rua do Campo Alegre, Porto 4169-007, Portugal
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, University of Navarra, C/ Irunlarrea, CP 31008 Pamplona, Navarra, Spain
| | - Eduardo Rocha
- Team of Histomorphology, Physiopathology and Applied Toxicology, CIIMAR - Interdisciplinary Center for Marine and Environmental Research, U.Porto - University of Porto, Avenida General Norton de Matos s/n, Matosinhos 4450-208, Portugal; Laboratory of Histology and Embryology, Department of Microscopy, ICBAS - Institute of Biomedical Sciences Abel Salazar, U.Porto - University of Porto, Rua de Jorge Viterbo Ferreira, n° 228, Porto 4050-313, Portugal
| |
Collapse
|
19
|
Deng S, Li Y, Yi G, Lei B, Guo M, Xiang W, Chen Z, Liu Y, Qi S. Overexpression of COX7A2 is associated with a good prognosis in patients with glioma. J Neurooncol 2017; 136:41-50. [PMID: 29079956 DOI: 10.1007/s11060-017-2637-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/14/2017] [Indexed: 12/15/2022]
Abstract
Cytochrome c oxidase subunit 7A2 (COX7A2) is a nuclear-encoded polypeptide involved in assembly and regulation of cytochrome c oxidase (COX). Changes in the respiratory chain as big complex are known to be associated with cancer, but little research has been performed to discover COX7A2 as a prognostic marker in glioma. In the present study, we investigated COX7A2 expression and its prognostic significance in glioma. Glioma surgical tissue samples were taken from 126 patients who had been followed up from 4 to 51 months. Immunohistochemistry were used to test COX7A2 expression in the 126 tumor samples. Eighty-six of 126 (68.3%) paraffin-embedded glioma biopsies showed high expression of COX7A2. Statistical analysis displayed that there was significant difference of COX7A2 expression level in patients categorized according to WHO classification. Kaplan-Meier survival analysis revealed that patients with higher COX7A2 expression had longer overall survival time and better prognosis. R2: microarray analysis based on Tumor Glioma French 284 database, Tumor Glioblastoma TCGA 540 database, and Tumor Glioma Kawaguchi 50 database testified that high expression of COX7A2 is associated with a good prognosis in patients with glioma. Multivariate analysis showed that COX7A2 high expression was an independent prognostic indicator for survival. Our results suggest that COX7A2 could be served as a valuable prognostic marker of glioma.
Collapse
Affiliation(s)
- Shengze Deng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yaomin Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Bingxi Lei
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Manlan Guo
- Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wei Xiang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ziyang Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yawei Liu
- Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838#, Guangzhou, 510515, Guangdong, China. .,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|