1
|
Ma L, Huo J, Cao S, Yue Y, Li X, Tian S, Liu L. Knockdown of EBP1 promotes doxorubicin-induced apoptosis in renal clear cell carcinoma cells through activation of the p38/HIF-1α pathway. Oncol Lett 2025; 29:172. [PMID: 39968014 PMCID: PMC11834144 DOI: 10.3892/ol.2025.14918] [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: 10/17/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Kidney clear cell carcinoma (KIRC) is a prevalent urological cancer. Despite substantial improvements in KIRC care, patients with intermediate and advanced stages of the disease lack access to appropriate medications. Doxorubicin is widely used as a chemotherapy drug for the treatment of multiple types of cancer. However, its use is associated with harmful side effects and drug resistance. ErbB3-binding protein (EBP1) is highly expressed in KIRC, and the knockdown of EBP1 reduces the phosphorylation of p38 mitogen-activated protein kinase (p38MAPK) and the expression of HIF-1α. Therefore, the present study aimed to evaluate the effectiveness of combined doxorubicin administration and EBP1 knockdown in KIRC cell lines. The KIRC cell lines 786-O and 769-P were used for the experiments, and short hairpin RNA technology was employed to specifically knock down the expression of the EBP1 gene. After treatment, cells were analyzed by western blotting to detect changes in p38MAPK phosphorylation levels and HIF-1α expression. The results showed that EBP1 knockdown significantly enhanced the antitumor effect of doxorubicin on KIRC cells through the p38MAPK/HIF-1α pathway. In conclusion, the knockdown of EBP1 in combination with doxorubicin may be a potential strategy for the treatment of KIRC.
Collapse
Affiliation(s)
- Lina Ma
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Jiaqi Huo
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Shuxia Cao
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Yuyang Yue
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Xiangdan Li
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Shengri Tian
- Department of Urology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Lan Liu
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, Jilin 133000, P.R. China
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
2
|
HELVACIOĞLU S, HAMİTOĞLU M, YILDIRIM E, KORKUT ŞVURAL, YABA A, AYDIN A. Protective Effects of Rosmarinic Acid and Epigallocatechin Gallate Against Doxorubicin-Induced Cytotoxicity and Genotoxicity in CHO-K1 Cells. Turk J Pharm Sci 2025; 21:536-543. [PMID: 39801063 PMCID: PMC11730000 DOI: 10.4274/tjps.galenos.2024.80552] [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: 12/28/2023] [Accepted: 02/20/2024] [Indexed: 01/16/2025]
Abstract
Objectives The chemotherapeutic drug doxorubicin (DOX) affects not only cancer cells but also healthy cells in an undesirable manner. The purpose of this study was to investigate the protective roles of rosmarinic acid (RA) and Epigallocatechin gallate (EGCG) alone and in combination against DOX-induced oxidative stress, cytotoxicity, and genotoxicity in healthy cells. In addition, this study evaluated the expression of the mammalian target of rapamycin (mTOR) protein in the Chinese hamster ovary cell line (CHO-K1). Materials and Methods Cell viability was analyzed using the WST-1 cytotoxicity assay. mTOR expression in the CHO-K1 cell line was determined by western blotting. DNA damage was analyzed using a comet assay. Reactive oxygen species (ROS) levels were determined microscopically using the dihydroethidium staining method. Results RA demonstrated superior protective effects against DOX-induced cytotoxicity compared to EGCG. Epigallocatechin gallate and RA did not exert genotoxic effects, but DOX increased genotoxicity in CHO-K1. Neither RA nor EGCG exhibited genotoxic effects; however, DOX significantly increased genotoxicity in CHO-K1 cells. Both RA and EGCG markedly reduced DOX-induced genotoxicity, as confirmed by the comet assay. In the DOX-treated group, the expression of mTOR protein was notably suppressed. EGCG further reduced mTOR protein levels when administered alone or in combination with DOX, whereas RA did not exhibit a similar effect. RA decreased intracellular generation of ROS in CHO-K1 cells. However, at high concentrations, Epigallocatechin gallate did not protect against oxidative stress and cell damage due to its prooxidant properties. Conclusion Epigallocatechin gallate and RA are promising plant-derived active components. Another important point is the evaluation of the safety of herbal products. It should be considered that herbal products may increase the toxicity of chemotherapeutic agents.
Collapse
Affiliation(s)
- Sinem HELVACIOĞLU
- İstinye University Faculty of Pharmacy, Department of Pharmaceutical Toxicology, İstanbul, Türkiye
| | - Muhammed HAMİTOĞLU
- Yeditepe University Faculty of Pharmacy, Department of Pharmaceutical Toxicology, İstanbul, Türkiye
| | - Ecem YILDIRIM
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye
| | - Şenay VURAL KORKUT
- Yıldız Technical University Faculty of Arts and Science, Department of Molecular Biology and Genetics, İstanbul, Türkiye
| | - Aylin YABA
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye
| | - Ahmet AYDIN
- Yeditepe University Faculty of Pharmacy, Department of Pharmaceutical Toxicology, İstanbul, Türkiye
| |
Collapse
|
3
|
Joung H, Liu H. 2‑D08 mediates notable anticancer effects through multiple cellular pathways in uterine leiomyosarcoma cells. Oncol Rep 2024; 52:97. [PMID: 38874019 PMCID: PMC11200159 DOI: 10.3892/or.2024.8756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
2',3',4'‑trihydroxyflavone (2‑D08), a SUMO E2 inhibitor, has several biological functions, including anticancer activity, but its effects on uterine leiomyosarcoma (Ut‑LMS) are unknown. The anticancer activity of 2‑D08 was explored in an in vitro model using SK‑LMS‑1 and SK‑UT‑1B cells (human Ut‑LMS cells). Treatment with 2‑D08 inhibited cell viability in a dose‑ and time‑dependent manner and significantly inhibited the colony‑forming ability of Ut‑LMS cells. In SK‑UT‑1B cells treated with 2‑D08, flow cytometric analysis revealed a slight increase in apoptotic rates, while cell cycle progression remained unaffected. Western blotting revealed elevated levels of RIP1, indicating induction of necrosis, but LC3B levels remained unchanged, suggesting no effect on autophagy. A lactate dehydrogenase (LDH) assay confirmed increased LDH release, further supporting the induction of apoptosis and necrosis by 2‑D08 in SK‑UT‑1B cells. 2‑D08‑induced production of reactive oxygen species and apoptosis progression were observed in SK‑LMS‑1 cells. Using Ki67 staining and bromodeoxyuridine assays, it was found that 2‑D08 suppressed proliferation in SK‑LMS‑1 cells, while treatment for 48 h led to cell‑cycle arrest. 2‑D08 upregulated p21 protein expression in SK‑LMS‑1 cells and promoted apoptosis through caspase‑3. Evaluation of α‑SM‑actin, calponin 1 and TAGLN expression indicated that 2‑D08 did not directly initiate smooth muscle phenotypic switching in SK‑LMS‑1 cells. Transcriptome analysis on 2‑D08‑treated SK‑LMS‑1 cells identified significant differences in gene expression and suggested that 2‑D08 modulates cell‑cycle‑ and apoptosis‑related pathways. The analysis identified several differentially expressed genes and significant enrichment for biological processes related to DNA replication and molecular functions associated with the apoptotic process. It was concluded that 2‑D08 exerts antitumor effects in Ut‑LMS cells by modulating multiple signaling pathways and that 2‑D08 may be a promising candidate for the treatment of human Ut‑LMS. The present study expanded and developed knowledge regarding Ut‑LMS management and indicated that 2‑D08 represents a notable finding in the exploration of fresh treatment options for such cancerous tumors.
Collapse
Affiliation(s)
- Hosouk Joung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Hwasun, Jeonnam 58128, Republic of Korea
| | - Hyunju Liu
- Department of Obstetrics and Gynecology, Chosun University College of Medicine, Gwangju 61452, Republic of Korea
- Department of Obstetrics and Gynecology, Chosun University Hospital, Gwangju 61453, Republic of Korea
| |
Collapse
|
4
|
Jalali F, Fakhari F, Sepehr A, Zafari J, Sarajar BO, Sarihi P, Jafarzadeh E. Synergistic anticancer effects of doxorubicin and metformin combination therapy: A systematic review. Transl Oncol 2024; 45:101946. [PMID: 38636389 PMCID: PMC11040171 DOI: 10.1016/j.tranon.2024.101946] [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: 12/20/2023] [Revised: 03/05/2024] [Accepted: 03/24/2024] [Indexed: 04/20/2024] Open
Abstract
INTRODUCTION Doxorubicin (DOX) a chemotherapy drug often leads to the development of resistance, in cancer cells after prolonged treatment. Recent studies have suggested that using metformin plus doxorubicin could result in synergic effects. This study focuses on exploring the co-treat treatment of doxorubicin and metformin for various cancers. METHOD Following the PRISMA guidelines we conducted a literature search using different databases such as Embase, Scopus, Web of Sciences, PubMed, Science Direct and Google Scholar until July 2023. We selected search terms based on the objectives of this study. After screening a total of 30 articles were included. RESULTS The combination of doxorubicin and metformin demonstrated robust anticancer effects, surpassing the outcomes of monotherapy drug treatment. In vitro experiments consistently demonstrated inhibition of cancer cell growth and increased rates of cell death. Animal studies confirmed substantial reductions in tumor growth and improved survival rates, emphasizing the synergistic impact of the combined therapy. The research' discoveries collectively emphasize the capability of the co-treat doxorubicin-metformin as a compelling approach in cancer treatment, highlighting its potential to address medicate resistance and upgrade generally helpful results. CONCLUSION The findings of this study show that the combined treatment regimen including doxorubicin and metformin has significant promise in fighting cancer. The observed synergistic effects suggest that this combination therapy could be valuable, in a setting. This study highlights the need for clinical research to validate and enhance the application of the doxorubicin metformin regimen.
Collapse
Affiliation(s)
- Fereshtehsadat Jalali
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fakhari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Afrah Sepehr
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Behnam Omidi Sarajar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouria Sarihi
- Research Institute of Bioscience and Biotechnology, University of Tabriz, Tabriz, Iran.
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Abedin Y, Fife A, Samuels CA, Wright R, Murphy T, Zhang X, Alpert E, Cheung E, Zhao Q, Einstein MH, Douglas NC. Combined Treatment of Uterine Leiomyosarcoma with Gamma Secretase Inhibitor MK-0752 and Chemotherapeutic Agents Decreases Cellular Invasion and Increases Apoptosis. Cancers (Basel) 2024; 16:2184. [PMID: 38927890 PMCID: PMC11201464 DOI: 10.3390/cancers16122184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Due to limited effective therapeutics for uterine leiomyosarcoma (uLMS), the impact of the gamma secretase inhibitor (GSI) MK-0752 with common chemotherapeutics was explored in uLMS. MTT assays were performed on two human uLMS cell lines, SK-UT-1B and SK-LMS-1, using MK-0752, docetaxel, doxorubicin, and gemcitabine, individually and in combination, to determine cell viability after treatment. Synergistic combinations were used in transwell invasion assays, cell cycle flow cytometry, proliferation assays, and RNA sequencing. In SK-UT-1B, MK-0752 was synergistic with doxorubicin and gemcitabine plus docetaxel. In SK-LMS-1, MK-0752 was synergistic with all individual agents and with the combination of gemcitabine plus docetaxel. MK-0752, gemcitabine, and docetaxel decreased invasion in SK-UT-1B 2.1-fold* and in SK-LMS-1 1.7-fold*. In SK-LMS-1, invasion decreased 1.2-fold* after treatment with MK-0752 and docetaxel and 2.2-fold* after treatment with MK-0752 and doxorubicin. Cell cycle analysis demonstrated increases in the apoptotic sub-G1 population with MK-0752 alone in SK-UT-1B (1.4-fold*) and SK-LMS-1 (2.7-fold**), along with increases with all combinations in both cell lines. The combination treatments had limited effects on proliferation, while MK-0752 alone decreased proliferation in SK-LMS-1 (0.63-fold**). Both MK-0752 alone and in combination altered gene expression and KEGG pathways. In conclusion, the combinations of MK-0752 with either doxorubicin, docetaxel, or gemcitabine plus docetaxel are potential novel therapeutic approaches for uLMS. (* p < 0.05, ** p < 0.01).
Collapse
Affiliation(s)
- Yasmin Abedin
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Alexander Fife
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Cherie-Ann Samuels
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Rasheena Wright
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Trystn Murphy
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Xusheng Zhang
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Emily Alpert
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Emma Cheung
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Mark H. Einstein
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Nataki C. Douglas
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
6
|
Manohar S, Neurohr GE. Too big not to fail: emerging evidence for size-induced senescence. FEBS J 2024; 291:2291-2305. [PMID: 37986656 DOI: 10.1111/febs.16983] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Cellular senescence refers to a permanent and stable state of cell cycle exit. This process plays an important role in many cellular functions, including tumor suppression. It was first noted that senescence is associated with increased cell size in the early 1960s; however, how this contributes to permanent cell cycle exit was poorly understood until recently. In this review, we discuss new findings that identify increased cell size as not only a consequence but also a cause of permanent cell cycle exit. We highlight recent insights into how increased cell size alters normal cellular physiology and creates homeostatic imbalances that contribute to senescence induction. Finally, we focus on the potential clinical implications of these findings in the context of cell cycle arrest-causing cancer therapeutics and speculate on how tumor cell size changes may impact outcomes in patients treated with these drugs.
Collapse
Affiliation(s)
- Sandhya Manohar
- Department of Biology, Institute for Biochemistry, ETH Zürich, Switzerland
| | - Gabriel E Neurohr
- Department of Biology, Institute for Biochemistry, ETH Zürich, Switzerland
| |
Collapse
|
7
|
Wani AK, Singh R, Akhtar N, Prakash A, Nepovimova E, Oleksak P, Chrienova Z, Alomar S, Chopra C, Kuca K. Targeted Inhibition of the PI3K/Akt/mTOR Signaling Axis: Potential for Sarcoma Therapy. Mini Rev Med Chem 2024; 24:1496-1520. [PMID: 38265369 DOI: 10.2174/0113895575270904231129062137] [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: 07/06/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 01/25/2024]
Abstract
Sarcoma is a heterogeneous group of malignancies often resistant to conventional chemotherapy and radiation therapy. The phosphatidylinositol-3-kinase/ protein kinase B /mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway has emerged as a critical cancer target due to its central role in regulating key cellular processes such as cell growth, proliferation, survival, and metabolism. Dysregulation of this pathway has been implicated in the development and progression of bone sarcomas (BS) and soft tissue sarcomas (STS). PI3K/Akt/mTOR inhibitors have shown promising preclinical and clinical activity in various cancers. These agents can inhibit the activation of PI3K, Akt, and mTOR, thereby reducing the downstream signaling events that promote tumor growth and survival. In addition, PI3K/Akt/mTOR inhibitors have been shown to enhance the efficacy of other anticancer therapies, such as chemotherapy and radiation therapy. The different types of PI3K/Akt/mTOR inhibitors vary in their specificity, potency, and side effect profiles and may be effective depending on the specific sarcoma type and stage. The molecular targeting of PI3K/Akt/mToR pathway using drugs, phytochemicals, nanomaterials (NMs), and microbe-derived molecules as Pan-PI3K inhibitors, selective PI3K inhibitors, and dual PI3K/mTOR inhibitors have been delineated. While there are still challenges to be addressed, the preclinical and clinical evidence suggests that these inhibitors may significantly improve patient outcomes. Further research is needed to understand the potential of these inhibitors as sarcoma therapeutics and to continue developing more selective and effective agents to meet the clinical needs of sarcoma patients.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Patrik Oleksak
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Zofia Chrienova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Suliman Alomar
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Králové, Czechia
| |
Collapse
|
8
|
Shah H, Hill TA, Lim J, Fairlie DP. Protease-activated receptor 2 attenuates doxorubicin-induced apoptosis in colon cancer cells. J Cell Commun Signal 2023:10.1007/s12079-023-00791-6. [PMID: 37991681 DOI: 10.1007/s12079-023-00791-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023] Open
Abstract
Drug resistance represents a major problem in cancer treatment. Doxorubicin (adriamycin) is an injectable DNA intercalating drug that halts cancer cell growth by inhibiting topoisomerase 2, but its long-term effectiveness is compromised by onset of resistance. This study demonstrates that expression of the PAR2 gene in human colon adenocarcinoma tissue samples was the highest among 32 different cancer types (n = 10,989), and higher in colon adenocarcinoma tissues (n = 331) than normal colon tissues (n = 308), revealing an association between PAR2 expression and human colon cancer. HT29 cells are a human colorectal adenocarcinoma cell line that is sensitive to the chemotherapeutic drug doxorubicin and also expresses PAR2. We find that PAR2 activation in HT29 cells, either by an endogenous protease agonist (trypsin) or an exogenous peptide agonist (2f-LIGRL-NH2), significantly reduces doxorubicin-induced cell death, reactive oxygen species production, caspase 3/7 activity and cleavage of caspase-8 and caspase-3. Moreover, PAR2-mediated MEK1/2-ERK1/2 pathway induced by 2f-LIGRL-NH2 leads to upregulated anti-apoptotic MCL-1 and Bcl-xL proteins that promote cellular survival. These findings suggest that activation of PAR2 compromises efficacy of doxorubicin in colon cancer. Further support for this conclusion came from experiments with human colon cancer HT29 cells, either with the PAR2 gene deleted or in the presence of a pharmacological antagonist of PAR2, which showed full restoration of all doxorubicin-mediated effects. Together, these findings reveal a strong link between PAR2 activation and signalling in human colon cancer cells and increased survival against doxorubicin-induced cell death. They support PAR2 antagonism as a possible new strategy for enhancing doxorubicin therapy.
Collapse
Affiliation(s)
- Himani Shah
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Timothy A Hill
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Junxian Lim
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia.
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia.
| | - David P Fairlie
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia.
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
9
|
de Nigris F, Meo C, Palinski W. Combination of Genomic Landsscape and 3D Culture Functional Assays Bridges Sarcoma Phenotype to Target and Immunotherapy. Cells 2023; 12:2204. [PMID: 37681936 PMCID: PMC10486752 DOI: 10.3390/cells12172204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
Genomic-based precision medicine has not only improved tumour therapy but has also shown its weaknesses. Genomic profiling and mutation analysis have identified alterations that play a major role in sarcoma pathogenesis and evolution. However, they have not been sufficient in predicting tumour vulnerability and advancing treatment. The relative rarity of sarcomas and the genetic heterogeneity between subtypes also stand in the way of gaining statistically significant results from clinical trials. Personalized three-dimensional tumour models that reflect the specific histologic subtype are emerging as functional assays to test anticancer drugs, complementing genomic screening. Here, we provide an overview of current target therapy for sarcomas and discuss functional assays based on 3D models that, by recapitulating the molecular pathways and tumour microenvironment, may predict patient response to treatments. This approach opens new avenues to improve precision medicine when genomic and pathway alterations are not sufficient to guide the choice of the most promising treatment. Furthermore, we discuss the aspects of the 3D culture assays that need to be improved, such as the standardisation of growth conditions and the definition of in vitro responses that can be used as a cut-off for clinical implementation.
Collapse
Affiliation(s)
- Filomena de Nigris
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Concetta Meo
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Wulf Palinski
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA;
| |
Collapse
|
10
|
Fazzari M, Lunghi G, Henriques A, Callizot N, Ciampa MG, Mauri L, Prioni S, Carsana EV, Loberto N, Aureli M, Mari L, Sonnino S, Chiricozzi E, Di Biase E. GM1 Oligosaccharide Efficacy in Parkinson's Disease: Protection against MPTP. Biomedicines 2023; 11:biomedicines11051305. [PMID: 37238977 DOI: 10.3390/biomedicines11051305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Past evidence has shown that the exogenous administration of GM1 ganglioside slowed neuronal death in preclinical models of Parkinson's disease, a neurodegenerative disorder characterized by the progressive loss of dopamine-producing neurons: however, the physical and chemical properties of GM1 (i.e., amphiphilicity) limited its clinical application, as the crossing of the blood-brain barrier is denied. Recently, we demonstrated that the GM1 oligosaccharide head group (GM1-OS) is the GM1 bioactive portion that, interacting with the TrkA-NGF complex at the membrane surface, promotes the activation of a multivariate network of intracellular events regulating neuronal differentiation, protection, and reparation. Here, we evaluated the GM1-OS neuroprotective potential against the Parkinson's disease-linked neurotoxin MPTP, which destroys dopaminergic neurons by affecting mitochondrial bioenergetics and causing ROS overproduction. In dopaminergic and glutamatergic primary cultures, GM1-OS administration significantly increased neuronal survival, preserved neurite network, and reduced mitochondrial ROS production enhancing the mTOR/Akt/GSK3β pathway. These data highlight the neuroprotective efficacy of GM1-OS in parkinsonian models through the implementation of mitochondrial function and reduction in oxidative stress.
Collapse
Affiliation(s)
- Maria Fazzari
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | | | - Noëlle Callizot
- Neuro-Sys, 410 Chemin Départemental 60, 13120 Gardanne, France
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Nicoletta Loberto
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Luigi Mari
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| | - Erika Di Biase
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, MI, Italy
| |
Collapse
|
11
|
Nagao Y, Yokoi A, Yoshida K, Sugiyama M, Watanabe E, Nakamura K, Kitagawa M, Asano-Inami E, Koya Y, Yoshihara M, Tamauchi S, Shimizu Y, Ikeda Y, Yoshikawa N, Kato T, Yamamoto Y, Kajiyama H. Novel therapeutic strategies targeting UCP2 in uterine leiomyosarcoma. Pharmacol Res 2023; 189:106693. [PMID: 36773710 DOI: 10.1016/j.phrs.2023.106693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Uterine leiomyosarcoma (ULMS) is a malignant stromal tumor arising from the myometrium with a poor prognosis and very limited response to current chemotherapy. This study aimed to identify novel targets for ULMS through a three-step screening process using a chemical library consisting of 1271 Food and Drug Administration-approved drugs. First, we evaluated their inhibitory effects on ULMS cells and identified four candidates: proscillaridin A, lanatoside C, floxuridine, and digoxin. Then, we subcutaneously or orthotopically transplanted SK-UT-1 cells into mice to establish mouse models. In vivo analyses showed that proscillaridin A and lanatoside C exerted a superior antitumor effect. The results of mRNA sequencing showed that uncoupling protein 2 (UCP2) was suppressed in the sirtuin signaling pathway, increasing reactive oxygen species (ROS) and inducing cell death. Moreover, the downregulation of UCP2 induced ROS and suppressed ULMS cell growth. Furthermore, analyses using clinical samples showed that UCP2 expression was significantly upregulated in ULMS tissues than in myoma tissues both at the RNA and protein levels. These findings suggested that UCP2 is a potential therapeutic target and can contribute to the development of novel therapeutic strategies in patients with ULMS.
Collapse
Affiliation(s)
- Yukari Nagao
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan; Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya-shi, Aichi 464-8603, Japan; Japan Science and Technology Agency (JST), FOREST, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan.
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan; Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya-shi, Aichi 464-8603, Japan
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Eri Watanabe
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Kae Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan; Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya-shi, Aichi, 464-8603, Japan
| | - Masami Kitagawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Eri Asano-Inami
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Yusuke Shimizu
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Tomoyasu Kato
- Department of Gynecologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yusuke Yamamoto
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| |
Collapse
|
12
|
Metabolomics Analysis Reveals Novel Targets of Chemosensitizing Polyphenols and Omega-3 Polyunsaturated Fatty Acids in Triple Negative Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24054406. [PMID: 36901842 PMCID: PMC10002396 DOI: 10.3390/ijms24054406] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer with typically poorer outcomes due to its aggressive clinical behavior and lack of targeted treatment options. Currently, treatment is limited to the administration of high-dose chemotherapeutics, which results in significant toxicities and drug resistance. As such, there is a need to de-escalate chemotherapeutic doses in TNBC while also retaining/improving treatment efficacy. Dietary polyphenols and omega-3 polyunsaturated fatty acids (PUFAs) have been demonstrated to have unique properties in experimental models of TNBC, improving the efficacy of doxorubicin and reversing multi-drug resistance. However, the pleiotropic nature of these compounds has caused their mechanisms to remain elusive, preventing the development of more potent mimetics to take advantage of their properties. Using untargeted metabolomics, we identify a diverse set of metabolites/metabolic pathways that are targeted by these compounds following treatment in MDA-MB-231 cells. Furthermore, we demonstrate that these chemosensitizers do not all target the same metabolic processes, but rather organize into distinct clusters based on similarities among metabolic targets. Common themes in metabolic targets included amino acid metabolism (particularly one-carbon and glutamine metabolism) and alterations in fatty acid oxidation. Moreover, doxorubicin treatment alone generally targeted different metabolites/pathways than chemosensitizers. This information provides novel insights into chemosensitization mechanisms in TNBC.
Collapse
|
13
|
Liu Q, Luo L, Gao X, Zhang D, Feng X, Yang P, Li H, Mao S. Co-Delivery of Daunorubicin and Homoharringtonine in Folic Acid Modified-Liposomes for Enhancing Therapeutic Effect on Acute Myeloid Leukemia. J Pharm Sci 2023; 112:123-131. [PMID: 35469834 DOI: 10.1016/j.xphs.2022.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022]
Abstract
Acute myeloid leukemia (AML) remains a threatening disease due to severe complications, drug resistance, and high recurrence rates. Many drug combinations have demonstrated enhanced therapeutic effects in clinical practice. However, it requires complicated dosing regimens and is accompanied by increased toxicity. This study explored the combined effect of two therapeutic agents, daunorubicin (DNR) and homoharringtonine (HHT) in cell viability, apoptosis, and cell cycle in vitro and verified their synergistic effect. We encapsulated the two drugs into liposomes to construct a folic acid-modified co-delivery system (FA-DH-LP) to achieve an effective and safe therapeutic strategy. The FA-DH-LP was prepared by film hydration method. The resultant FA-DH-LP was homogeneously spherical and showed good blood compatibility with high encapsulation efficiency for DNR and HHT. The FA-DH-LP exhibited higher cellular uptake in HL60 and K562 cells and enhanced cytotoxicity than DNR/HHT co-delivery liposomes without folic acid modification (DH-LP) in vitro. In the HL60 subcutaneous xenotransplantation model, FA-DH-LP showed improved tumor targeting ability, anti-leukemia activity and safety profile superior to free combinational drugs and DH-LP after 18-day treatment. The results demonstrated that FA-DH-LP might present a promising delivery strategy to improve the efficacy of the two combinational chemotherapeutics while reducing toxicity.
Collapse
Affiliation(s)
- Qi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lijun Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaofeng Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Di Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xinqian Feng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Peng Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hui Li
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China.
| | - Shengjun Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Tiwari S, Kharbanda S, Singh H. Quatramer™ Mediated Codelivery of PI3-Kδ/HDAC6 Dual Inhibitor Augments the Anti-Cancer Efficacy of Epirubicin in Breast Cancer. Eur J Pharm Biopharm 2022; 179:184-193. [PMID: 36087881 DOI: 10.1016/j.ejpb.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/01/2022] [Accepted: 09/04/2022] [Indexed: 01/04/2023]
Abstract
The disruption and overexpression of phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway in cancer results in tumor growth, metastasis, and survival. Treatment with common anthracyclines has confirmed cancer cells' dependence on PI3K pathway through overexpression of AKT. Moreover, combining HDAC inhibitor with anthracycline has shown the targeting of breast cancer stem cells. Therefore, it has been hypothesized that the co-delivery of PI3-Kδ/HDAC6 dual inhibitor with Epirubicin using polymeric nanoparticle could increase the anti-cancer treatment efficacy with reduced toxicity. Pluronic modified polylactic acid block copolymer (quatramer) was used for encapsulation of PI3-Kδ/HDAC6 and Epirubicin. The co-encapsulated nanoparticles, PI3-Kδ/HDAC6-Epi-NPs have shown size of 99±3 nm, PDI of 0.18±0.07 with a sustained and slow-release profile in non-physiological buffer (PBS, pH 7.4). The in-vitro cell proliferation inhibition studies done on 2D and 3D culture of breast cancer cell lines have confirmed the synergistic effect of PI3-Kδ/HDAC6-Epi-NPs with lower IC50 values compared to PI3-Kδ/HDAC6-NPs and Epi-NPs. Additionally, intravenous twice a week treatment for three weeks with PI3-Kδ/HDAC6-Epi-NPs resulted in complete tumor eradication in the syngeneic breast tumor mice model. In comparison, the PI3-Kδ/HDAC6-NPs and Epi-NPs result in tumor growth inhibition of 15.86% and 81.59%, respectively. These studies predicted that clinical use of PI3-Kδ/HDAC6-Epi-NPs will be effective in breast cancer treatments.
Collapse
Affiliation(s)
- Sachchidanand Tiwari
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Surender Kharbanda
- Dana Farber Cancer Institute, Harvard Medical School, Boston-02115, MA, USA.
| | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India; Department of Biomedical Engineering, All India Institute of Medical Sciences Delhi, New Delhi 110023, India.
| |
Collapse
|
15
|
eIF3a regulation of mTOR signaling and translational control via HuR in cellular response to DNA damage. Oncogene 2022; 41:2431-2443. [PMID: 35279705 PMCID: PMC9035104 DOI: 10.1038/s41388-022-02262-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 01/29/2023]
Abstract
eIF3a (eukaryotic translation initiation factor 3a), a subunit of the eIF3 complex, has been suggested to play a regulatory role in protein synthesis and in cellular response to DNA-damaging treatments. S6K1 is an effector and a mediator of mTOR complex 1 (mTORC1) in regulating protein synthesis and integrating diverse signals into control of cell growth and response to stress. Here, we show that eIF3a regulates S6K1 activity by inhibiting mTORC1 kinase via regulating Raptor synthesis. The regulation of Raptor synthesis is via eIF3a interaction with HuR (human antigen R) and binding of the eIF3a-HuR complex to the 5'-UTR of Raptor mRNA. Furthermore, mTORC1 may mediate eIF3a function in cellular response to cisplatin by regulating synthesis of NER proteins and NER activity. Taken together, we conclude that the mTOR signaling pathway may also be regulated by translational control and mediate eIF3a regulation of cancer cell response to cisplatin by regulating NER protein synthesis.
Collapse
|
16
|
Holliday H, Yang J, Dodson E, Nikolic I, Kamili A, Wheatley M, Deng N, Alexandrou S, Davis TP, Kavallaris M, Caldon CE, McCarroll J, De Preter K, Mestdagh P, Marshall GM, Simpson KJ, Fletcher J, Swarbrick A. miR-99b-5p, miR-380-3p, and miR-485-3p are novel chemosensitizing miRNAs in high-risk neuroblastoma. Mol Ther 2022; 30:1119-1134. [PMID: 34998954 PMCID: PMC8899605 DOI: 10.1016/j.ymthe.2022.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/17/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma is a deadly childhood cancer arising in the developing sympathetic nervous system. High-risk patients are currently treated with intensive chemotherapy, which is curative in only 50% of children and leaves some surviving patients with life-long side effects. microRNAs (miRNAs) are critical regulators of neural crest development and are deregulated during neuroblastoma tumorigenesis, making miRNA-based drugs an attractive therapeutic avenue. A functional screen of >1,200 miRNA mimics was conducted in neuroblastoma cell lines to discover miRNAs that sensitized cells to low doses (30% inhibitory concentration [IC30]) of doxorubicin and vincristine chemotherapy used in the treatment of the disease. Three miRNAs, miR-99b-5p, miR-380-3p, and miR-485-3p, had potent chemosensitizing activity with doxorubicin in multiple models of high-risk neuroblastoma. These miRNAs underwent genomic loss in a subset of neuroblastoma patients, and low expression predicted poor survival outcome. In vitro functional assays revealed each of these miRNAs enhanced the anti-proliferative and pro-apoptotic effects of doxorubicin. We used RNA sequencing (RNA-seq) to show that miR-99b-5p represses neuroblastoma dependency genes LIN28B and PHOX2B both in vitro and in patient-derived xenograft (PDX) tumors. Luciferase reporter assays demonstrate that PHOX2B is a direct target of miR-99b-5p. We anticipate that restoring the function of the tumor-suppressive miRNAs discovered here may be a valuable therapeutic strategy for the treatment of neuroblastoma patients.
Collapse
Affiliation(s)
- Holly Holliday
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jessica Yang
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Eoin Dodson
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Iva Nikolic
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Alvin Kamili
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Madeleine Wheatley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia
| | - Niantao Deng
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Sarah Alexandrou
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Australian Institute for Bioengineering, The University of Queensland, Brisbane, QLD 2072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia; Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - C Elizabeth Caldon
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia; Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Katleen De Preter
- Cancer Research Institute Ghent, Ghent University, Ghent B-9000, Belgium
| | - Pieter Mestdagh
- Cancer Research Institute Ghent, Ghent University, Ghent B-9000, Belgium
| | - Glenn M Marshall
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW 2031, Australia
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Jamie Fletcher
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2031, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Alexander Swarbrick
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia.
| |
Collapse
|
17
|
Carrabotta M, Laginestra MA, Durante G, Mancarella C, Landuzzi L, Parra A, Ruzzi F, Toracchio L, De Feo A, Giusti V, Pasello M, Righi A, Lollini PL, Palmerini E, Donati DM, Manara MC, Scotlandi K. Integrated Molecular Characterization of Patient-Derived Models Reveals Therapeutic Strategies for Treating CIC-DUX4 Sarcoma. Cancer Res 2022; 82:708-720. [PMID: 34903601 PMCID: PMC9359717 DOI: 10.1158/0008-5472.can-21-1222] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/18/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
Capicua-double homeobox 4 (CIC-DUX4)-rearranged sarcomas (CDS) are extremely rare, highly aggressive primary sarcomas that represent a major therapeutic challenge. Patients are treated according to Ewing sarcoma protocols, but CDS-specific therapies are strongly needed. In this study, RNA sequencing was performed on patient samples to identify a selective signature that differentiates CDS from Ewing sarcoma and other fusion-driven sarcomas. This signature was used to validate the representativeness of newly generated CDS experimental models-patient-derived xenografts (PDX) and PDX-derived cell lines-and to identify specific therapeutic vulnerabilities. Annotation analysis of differentially expressed genes and molecular gene validation highlighted an HMGA2/IGF2BP/IGF2/IGF1R/AKT/mTOR axis that characterizes CDS and renders the tumors particularly sensitive to combined treatments with trabectedin and PI3K/mTOR inhibitors. Trabectedin inhibited IGF2BP/IGF2/IGF1R activity, but dual inhibition of the PI3K and mTOR pathways was required to completely dampen downstream signaling mediators. Proof-of-principle efficacy for the combination of the dual AKT/mTOR inhibitor NVP-BEZ235 (dactolisib) with trabectedin was obtained in vitro and in vivo using CDS PDX-derived cell lines, demonstrating a strong inhibition of local tumor growth and multiorgan metastasis. Overall, the development of representative experimental models (PDXs and PDX-derived cell lines) has helped to identify the unique sensitivity of the CDS to AKT/mTOR inhibitors and trabectedin, revealing a mechanism-based therapeutic strategy to fight this lethal cancer. SIGNIFICANCE This study identifies altered HMGA2/IGF2BP/IGF2 signaling in CIC-DUX4 sarcomas and provides proof of principle for combination therapy with trabectedin and AKT/mTOR dual inhibitors to specifically combat the disease.
Collapse
Affiliation(s)
- Marianna Carrabotta
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Giorgio Durante
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Caterina Mancarella
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Lorena Landuzzi
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandro Parra
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Lisa Toracchio
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandra De Feo
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Veronica Giusti
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Michela Pasello
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alberto Righi
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Sarcoma and Novel Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Davide Maria Donati
- Third Orthopaedic Clinic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | | | - Katia Scotlandi
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
18
|
Lee EY, Kim M, Choi BK, Kim DH, Choi I, You HJ. TJP1 Contributes to Tumor Progression through Supporting Cell-Cell Aggregation and Communicating with Tumor Microenvironment in Leiomyosarcoma. Mol Cells 2021; 44:784-794. [PMID: 34764231 PMCID: PMC8627839 DOI: 10.14348/molcells.2021.0130] [Citation(s) in RCA: 7] [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: 05/18/2021] [Revised: 09/01/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022] Open
Abstract
Leiomyosarcoma (LMS) is a mesenchymal malignancy with a complex karyotype. Despite accumulated evidence, the factors contributing to the development of LMS are unclear. Here, we investigated the role of tight-junction protein 1 (TJP1), a membrane-associated intercellular barrier protein during the development of LMS and the tumor microenvironment. We orthotopically transplanted SK-LMS-1 cells and their derivatives in terms of TJP1 expression by intramuscular injection, such as SK-LMS-1 Sh-Control cells and SK-LMS-1 Sh-TJP1. We observed robust tumor growth in mice transplanted with LMS cell lines expressing TJP1 while no tumor mass was found in mice transplanted with SK-LMS-1 Sh-TJP1 cells with silenced TJP1 expression. Tissues from mice were stained and further analyzed to clarify the effects of TJP1 expression on tumor development and the tumor microenvironment. To identify the TJP1-dependent factors important in the development of LMS, genes with altered expression were selected in SK-LMS-1 cells such as cyclinD1, CSF1 and so on. The top 10% of highly expressed genes in LMS tissues were obtained from public databases. Further analysis revealed two clusters related to cell proliferation and the tumor microenvironment. Furthermore, integrated analyses of the gene expression networks revealed correlations among TJP1, CSF1 and CTLA4 at the mRNA level, suggesting a possible role for TJP1 in the immune environment. Taken together, these results imply that TJP1 contributes to the development of sarcoma by proliferation through modulating cell-cell aggregation and communication through cytokines in the tumor microenvironment and might be a beneficial therapeutic target.
Collapse
Affiliation(s)
- Eun-Young Lee
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| | - Minjeong Kim
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Beom K. Choi
- Biomedicine Production Branch, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Dae Hong Kim
- Division of Convergence Technology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| | - Hye Jin You
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea
- Department of Cancer Biomedical Science, National Cancer Center-Graduate School of Cancer Science and Policy (NCC-GCSP), National Cancer Center, Goyang 10408, Korea
| |
Collapse
|
19
|
Anderson ND, Babichev Y, Fuligni F, Comitani F, Layeghifard M, Venier RE, Dentro SC, Maheshwari A, Guram S, Wunker C, Thompson JD, Yuki KE, Hou H, Zatzman M, Light N, Bernardini MQ, Wunder JS, Andrulis IL, Ferguson P, Razak ARA, Swallow CJ, Dowling JJ, Al-Awar RS, Marcellus R, Rouzbahman M, Gerstung M, Durocher D, Alexandrov LB, Dickson BC, Gladdy RA, Shlien A. Lineage-defined leiomyosarcoma subtypes emerge years before diagnosis and determine patient survival. Nat Commun 2021; 12:4496. [PMID: 34301934 PMCID: PMC8302638 DOI: 10.1038/s41467-021-24677-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Leiomyosarcomas (LMS) are genetically heterogeneous tumors differentiating along smooth muscle lines. Currently, LMS treatment is not informed by molecular subtyping and is associated with highly variable survival. While disease site continues to dictate clinical management, the contribution of genetic factors to LMS subtype, origins, and timing are unknown. Here we analyze 70 genomes and 130 transcriptomes of LMS, including multiple tumor regions and paired metastases. Molecular profiling highlight the very early origins of LMS. We uncover three specific subtypes of LMS that likely develop from distinct lineages of smooth muscle cells. Of these, dedifferentiated LMS with high immune infiltration and tumors primarily of gynecological origin harbor genomic dystrophin deletions and/or loss of dystrophin expression, acquire the highest burden of genomic mutation, and are associated with worse survival. Homologous recombination defects lead to genome-wide mutational signatures, and a corresponding sensitivity to PARP trappers and other DNA damage response inhibitors, suggesting a promising therapeutic strategy for LMS. Finally, by phylogenetic reconstruction, we present evidence that clones seeding lethal metastases arise decades prior to LMS diagnosis.
Collapse
Affiliation(s)
- Nathaniel D Anderson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Yael Babichev
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Fabio Fuligni
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, ON, Ontario, Canada
| | - Federico Comitani
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mehdi Layeghifard
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rosemarie E Venier
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Stefan C Dentro
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Anant Maheshwari
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sheena Guram
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Claire Wunker
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - J Drew Thompson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kyoko E Yuki
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Huayun Hou
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Matthew Zatzman
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Nicholas Light
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Marcus Q Bernardini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Jay S Wunder
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- University Musculoskeletal Oncology Unit, Mount Sinai Hospital, Toronto, ON, Canada
| | - Irene L Andrulis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Peter Ferguson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- University Musculoskeletal Oncology Unit, Mount Sinai Hospital, Toronto, ON, Canada
| | | | - Carol J Swallow
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Division of General Surgery, Mount Sinai Hospital, Toronto, ON, Canada
| | - James J Dowling
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Rima S Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Richard Marcellus
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Marjan Rouzbahman
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Brendan C Dickson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Rebecca A Gladdy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Surgery, University of Toronto, Toronto, ON, Canada.
- Division of General Surgery, Mount Sinai Hospital, Toronto, ON, Canada.
| | - Adam Shlien
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, ON, Ontario, Canada.
| |
Collapse
|
20
|
Miallot R, Galland F, Millet V, Blay JY, Naquet P. Metabolic landscapes in sarcomas. J Hematol Oncol 2021; 14:114. [PMID: 34294128 PMCID: PMC8296645 DOI: 10.1186/s13045-021-01125-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolic rewiring offers novel therapeutic opportunities in cancer. Until recently, there was scant information regarding soft tissue sarcomas, due to their heterogeneous tissue origin, histological definition and underlying genetic history. Novel large-scale genomic and metabolomics approaches are now helping stratify their physiopathology. In this review, we show how various genetic alterations skew activation pathways and orient metabolic rewiring in sarcomas. We provide an update on the contribution of newly described mechanisms of metabolic regulation. We underscore mechanisms that are relevant to sarcomagenesis or shared with other cancers. We then discuss how diverse metabolic landscapes condition the tumor microenvironment, anti-sarcoma immune responses and prognosis. Finally, we review current attempts to control sarcoma growth using metabolite-targeting drugs.
Collapse
Affiliation(s)
- Richard Miallot
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Aix Marseille Univ, Marseille, France.
| | - Franck Galland
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Aix Marseille Univ, Marseille, France
| | - Virginie Millet
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Aix Marseille Univ, Marseille, France
| | - Jean-Yves Blay
- Centre Léon Bérard, Lyon 1, Lyon Recherche Innovation contre le Cancer, Université Claude Bernard, Lyon, France
| | - Philippe Naquet
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Aix Marseille Univ, Marseille, France.
| |
Collapse
|
21
|
Coronel-Hernández J, Salgado-García R, Cantú-De León D, Jacobo-Herrera N, Millan-Catalan O, Delgado-Waldo I, Campos-Parra AD, Rodríguez-Morales M, Delgado-Buenrostro NL, Pérez-Plasencia C. Combination of Metformin, Sodium Oxamate and Doxorubicin Induces Apoptosis and Autophagy in Colorectal Cancer Cells via Downregulation HIF-1α. Front Oncol 2021; 11:594200. [PMID: 34123772 PMCID: PMC8187873 DOI: 10.3389/fonc.2021.594200] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/30/2021] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death worldwide in both sexes. Current therapies include surgery, chemotherapy, and targeted therapy; however, prolonged exposure to chemical agents induces toxicity in patients and drug resistance. So, we implemented a therapeutic strategy based on the combination of doxorubicin, metformin, and sodium oxamate called triple therapy (Tt). We found that Tt significantly reduced proliferation by inhibiting the mTOR/AKT pathway and promoted apoptosis and autophagy in CRC derived cells compared with doxorubicin. Several autophagy genes were assessed by western blot; ULK1, ATG4, and LC3 II were overexpressed by Tt. Interestingly, ULK1 was the only one autophagy-related protein gradually overexpressed during Tt administration. Thus, we assumed that there was a post-transcriptional mechanism mediating by microRNAs that regulate UKL1 expression during autophagy activation. Through bioinformatics approaches, we ascertained that ULK1 could be targeted by mir-26a, which is overexpressed in advanced stages of CRC. In vitro experiments revealed that overexpression of mir-26a decreased significantly ULK1, mRNA, and protein expression. Contrariwise, the Tt recovered ULK1 expression by mir-26a decrease. Due to triple therapy repressed mir-26a expression, we hypothesized this drug combination could be involved in mir-26a transcription regulation. Consequently, we analyzed the mir-26a promoter sequence and found two HIF-1α transcription factor recognition sites. We developed two different HIF-1α stabilization models. Both showed mir-26a overexpression and ULK1 reduction in hypoxic conditions. Immunoprecipitation experiments were performed and HIF-1α enrichment was observed in mir-26a promoter. Surprisingly, Tt diminished HIF-1α detection and restored ULK1 mRNA expression. These results reveal an important regulation mechanism controlled by the signaling that activates HIF-1α and that in turn regulates mir-26a transcription.
Collapse
Affiliation(s)
- Jossimar Coronel-Hernández
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-Iztacala, UNAM, Tlalnepantla, Mexico,Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
| | | | - David Cantú-De León
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
| | | | | | | | | | | | | | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-Iztacala, UNAM, Tlalnepantla, Mexico,Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico,*Correspondence: Carlos Pérez-Plasencia,
| |
Collapse
|
22
|
Kasper B, Achee A, Schuster K, Wilson R, van Oortmerssen G, Gladdy RA, Hemming ML, Huang P, Ingham M, Jones RL, Pollack SM, Reinke D, Sanfilippo R, Schuetze SM, Somaiah N, Van Tine BA, Wilky B, Okuno S, Trent J. Unmet Medical Needs and Future Perspectives for Leiomyosarcoma Patients-A Position Paper from the National LeioMyoSarcoma Foundation (NLMSF) and Sarcoma Patients EuroNet (SPAEN). Cancers (Basel) 2021; 13:886. [PMID: 33672607 PMCID: PMC7924026 DOI: 10.3390/cancers13040886] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
As leiomyosarcoma patients are challenged by the development of metastatic disease, effective systemic therapies are the cornerstone of outcome. However, the overall activity of the currently available conventional systemic treatments and the prognosis of patients with advanced or metastatic disease are still poor, making the treatment of this patient group challenging. Therefore, in a joint effort together with patient networks and organizations, namely Sarcoma Patients EuroNet (SPAEN), the international network of sarcoma patients organizations, and the National LeioMyoSarcoma Foundation (NLMSF) in the United States, we aim to summarize state-of-the-art treatments for leiomyosarcoma patients in order to identify knowledge gaps and current unmet needs, thereby guiding the community to design innovative clinical trials and basic research and close these research gaps. This position paper arose from a leiomyosarcoma research meeting in October 2020 hosted by the NLMSF and SPAEN.
Collapse
Affiliation(s)
- Bernd Kasper
- Mannheim University Medical Center, University of Heidelberg, 68167 Mannheim, Germany
| | - Annie Achee
- National LeioMyoSarcoma Foundation (NLMSF), Denver, CO 80222, USA;
| | - Kathrin Schuster
- Sarcoma Patients EuroNet, SPAEN, 61200 Wölfersheim, Germany; (K.S.); (R.W.); (G.v.O.)
| | - Roger Wilson
- Sarcoma Patients EuroNet, SPAEN, 61200 Wölfersheim, Germany; (K.S.); (R.W.); (G.v.O.)
| | | | - Rebecca A. Gladdy
- Department of Surgery, Mount Sinai Hospital, Toronto, ON M5G 1XS, Canada;
| | | | - Paul Huang
- Institute of Cancer Research, London SM2 5NG, UK; (P.H.); (R.L.J.)
| | - Matthew Ingham
- Department of Medicine, Columbia University School of Medicine, New York, NY 10032, USA;
| | - Robin L. Jones
- Institute of Cancer Research, London SM2 5NG, UK; (P.H.); (R.L.J.)
- Royal Marsden Hospital, London SW3 6JJ, UK
| | - Seth M. Pollack
- Northwestern Medicine, Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Denise Reinke
- Sarcoma Alliance for Research through Collaboration (SARC), Ann Arbor, MI 48105, USA;
| | | | - Scott M. Schuetze
- Michigan Medicine Sarcoma Clinic, Rogel Cancer Center, Ann Arbor, MI 48109, USA;
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Care Center, Houston, TX 77030, USA;
| | - Brian A. Van Tine
- Barnes and Jewish Hospital, Washington University in St. Louis, St. Louis, MO 63110, USA;
| | - Breelyn Wilky
- Department of Sarcoma Medical Oncology, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
| | - Scott Okuno
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Jonathan Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA;
| |
Collapse
|
23
|
Anthracycline-induced cardiomyopathy: cellular and molecular mechanisms. Clin Sci (Lond) 2021; 134:1859-1885. [PMID: 32677679 DOI: 10.1042/cs20190653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023]
Abstract
Despite the known risk of cardiotoxicity, anthracyclines are widely prescribed chemotherapeutic agents. They are broadly characterized as being a robust effector of cellular apoptosis in rapidly proliferating cells through its actions in the nucleus and formation of reactive oxygen species (ROS). And, despite the early use of dexrazoxane, no effective treatment strategy has emerged to prevent the development of cardiomyopathy, despite decades of study, suggesting that much more insight into the underlying mechanism of the development of cardiomyopathy is needed. In this review, we detail the specific intracellular activities of anthracyclines, from the cell membrane to the sarcoplasmic reticulum, and highlight potential therapeutic windows that represent the forefront of research into the underlying causes of anthracycline-induced cardiomyopathy.
Collapse
|
24
|
Boichuk S, Bikinieva F, Nurgatina I, Dunaev P, Valeeva E, Aukhadieva A, Sabirov A, Galembikova A. Inhibition of AKT-Signaling Sensitizes Soft Tissue Sarcomas (STS) and Gastrointestinal Stromal Tumors (GIST) to Doxorubicin via Targeting of Homology-Mediated DNA Repair. Int J Mol Sci 2020; 21:E8842. [PMID: 33266502 PMCID: PMC7700672 DOI: 10.3390/ijms21228842] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/23/2022] Open
Abstract
Activation of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR pathway is well documented for a broad spectrum of human malignancies supporting their growth and progression. Accumulating evidence has also implicated AKT as a potent modulator of anti-cancer therapies via regulation of DNA damage response and repair (DDR) induced by certain chemotherapeutic agents and ionizing radiation (IR). In the present study, we examined the role of AKT signaling in regulating of Rad51 turnover and cytotoxic effects of topoisomerase II inhibitor, doxorubicin (Dox) in soft tissue sarcomas (STS) and gastrointestinal stromal tumors (GIST) in vitro. Blocking of AKT signaling (MK-2206) enhanced cytotoxic and pro-apoptotic effects of Dox in vast majority of STS and GIST cell lines. The phosphorylated form of Akt co-immunoprecipitates with Rad51 after Dox-induced DNA damage, whereas Akt inhibition interrupts this interaction and decreases Rad51 protein level by enhancing protein instability via proteasome-dependent degradation. Inhibition of Akt signaling in Dox-treated cells was associated with the increased number of γ-H2AX-positive cells, decrease of Rad51 foci formation and its colocalization with γ-H2AX foci, thereby revealing unsuccessful DDR events. This was also in consistency with an increase of tail moment (TM) and olive tail moment (OTM) in Dox-treated GIST and STS cells cultured in presence of Akt inhibitor after Dox washout. Altogether, our data illustrates that inhibition of AKT signaling is STS and GIST might potentiate the cytotoxic effect of topoisomerase II inhibitors via attenuating the homology-mediated DNA repair.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (F.B.); (I.N.); (P.D.); (A.A.); (A.G.)
- Central Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia;
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (F.B.); (I.N.); (P.D.); (A.A.); (A.G.)
| | - Ilmira Nurgatina
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (F.B.); (I.N.); (P.D.); (A.A.); (A.G.)
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (F.B.); (I.N.); (P.D.); (A.A.); (A.G.)
| | - Elena Valeeva
- Central Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia;
| | - Aida Aukhadieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (F.B.); (I.N.); (P.D.); (A.A.); (A.G.)
| | - Alexey Sabirov
- Department of Pathology, Tatarstan Cancer Center, 420029 Kazan, Russia;
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (F.B.); (I.N.); (P.D.); (A.A.); (A.G.)
| |
Collapse
|
25
|
Zhu H, Shi Y, Jiao X, Yang G, Wang R, Yuan Y. Synergistic antitumor effect of dual PI3K and mTOR inhibitor NVP-BEZ235 in combination with cisplatin on drug-resistant non-small cell lung cancer cell. Oncol Lett 2020; 20:326. [PMID: 33123242 PMCID: PMC7584016 DOI: 10.3892/ol.2020.12189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Cisplatin resistance is an obstacle for the effective treatment of non-small cell lung cancer (NSCLC). The combined use of two or more chemotherapeutic agents displays advantages for the clinical treatment of drug-resistant lung cancer. The present study aimed to assess the synergy of the dual PI3K/Akt/mTOR signaling pathway inhibitor NVP-BEZ235 and cisplatin, a chemotherapeutic agent, on proliferation, apoptosis, cell cycle arrest and protein expression in cisplatin-resistant NSCLC A549/diamminedichloroplatinum resistance (DDP) cells. Cell proliferation was determined by performing Cell Counting Kit 8 and colony formation assays. Combination index (CI) was used to assess the combinatorial effects of NVP-BEZ235 and cisplatin. Cellular apoptosis and cell cycle arrest were detected via flow cytometry. Western blotting was performed to evaluate protein expression levels relative to β-actin. Cisplatin and NVP-BEZ235 displayed the strongest synergy (CI50=0.23) at the mass ratio of 10:1. The half inhibitory concentrations of cisplatin and NVP-BEZ235 at 10:1 were 1.53 and 0.15 µg/ml, respectively. Compared with the control group, the combination of cisplatin and NVP-BEZ235 induced cell apoptosis and inhibited colony formation. Furthermore, compared with the control group, phosphorylation of Akt and p70S6 Kinase was significantly inhibited and cell cycle was arrested at G0G1 phase in the combination treatment group. The expression levels of drug efflux proteins, such as multidrug resistance-associated protein 1 and ATP-binding cassette sub-family G member 2, were significantly decreased when A549/DDP cells were treated with a combination of cisplatin and NVP-BEZ235 compared with the control group. Collectively, the present study indicated that the combined treatment of cisplatin and NVP-BEZ235 displayed synergistic antitumor effects on drug-resistant A549/DDP cells, by which the antiproliferative effects may occur via inhibition of the PI3K/Akt/mTOR signaling pathway and downregulation of drug efflux.
Collapse
Affiliation(s)
- Hao Zhu
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yuhuan Shi
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xiuxiu Jiao
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Gang Yang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Rong Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
26
|
Shapira T, Rankine-Wilson L, Chao JD, Pichler V, Rens C, Pfeifer T, Av-Gay Y. High-Content Screening of Eukaryotic Kinase Inhibitors Identify CHK2 Inhibitor Activity Against Mycobacterium tuberculosis. Front Microbiol 2020; 11:553962. [PMID: 33042061 PMCID: PMC7530171 DOI: 10.3389/fmicb.2020.553962] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
A screen of a eukaryotic kinase inhibitor library in an established intracellular infection model identified a set of drug candidates enabling intracellular killing of Mycobacterium tuberculosis (M.tb). Screen validity was confirmed internally by a Z′ = 0.5 and externally by detecting previously reported host-targeting anti-M.tb compounds. Inhibitors of the CHK kinase family, specifically checkpoint kinase 2 (CHK2), showed the highest inhibition and lowest toxicity of all kinase families. The screen identified and validated DDUG, a CHK2 inhibitor, as a novel bactericidal anti-M.tb compound. CHK2 inhibition by RNAi phenocopied the intracellular inhibitory effect of DDUG. DDUG was active intracellularly against M.tb, but not other mycobacteria. DDUG also had extracellular activity against 4 of 12 bacteria tested, including M.tb. Combined, these observations suggest DDUG acts in tandem against both host and pathogen. Importantly, DDUG’s validation highlights the screening and analysis methodology developed for this screen, which identified novel host-directed anti-M.tb compounds.
Collapse
Affiliation(s)
- Tirosh Shapira
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Leah Rankine-Wilson
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Joseph D Chao
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Virginia Pichler
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Celine Rens
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Tom Pfeifer
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Yossef Av-Gay
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Huang Y, Sun L, Zhu S, Xu L, Liu S, Yuan C, Guo Y, Wang X. Neuroprotection Against Parkinson's Disease Through the Activation of Akt/GSK3β Signaling Pathway by Tovophyllin A. Front Neurosci 2020; 14:723. [PMID: 32742256 PMCID: PMC7364155 DOI: 10.3389/fnins.2020.00723] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most prevalent and life-threatening neurodegenerative disease and mainly characterized by lack of sufficient dopaminergic neurons in the substantia nigra pars compacta (SNc). Although current treatments help to alleviate clinical symptoms, effective therapies preventing neuronal loss remain scarce. Tovophyllin A (TA), one of the xanthones extracted from Garcinia mangostana L. (GM), has recently been reported to play a beneficial role in the therapy of neurodegenerative diseases. In our research, we explored whether TA has protective effects on dopaminergic neurons in PD models. We found that TA significantly reduced apoptotic cell death in primary cortical neurons treated with 1-methyl-4-phenyl pyridinium (MPP+) or paraquat (PQ) in the in vitro PD model. In an in vivo acute PD model induced by 1-methyl4-phenyl-1,2,3,5-tetrahydropyridine (MPTP) treatment, TA also attenuated the resulting behavioral dysfunctions and dopaminergic neuron loss. In the collected brain tissues, TA increased the phosphorylation of Akt and GSK-3β, which may be related to TA-mediated dopaminergic neuronal protective effects. In summary, our results illustrated that TA is a powerful cytoprotective agent for dopaminergic neurons in the MPTP-induced PD model, suggesting TA as a possible therapeutic candidate for PD.
Collapse
Affiliation(s)
- Yanjun Huang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lirong Sun
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liu Xu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuhu Liu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chunhua Yuan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanwu Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuemin Wang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Si W, Lyu J, Liu Z, Wang C, Huang J, Jiang L, Ma T. Cucurbitacin E inhibits cellular proliferation and enhances the chemo-response in gastric cancer by suppressing AKt activation. J Cancer 2019; 10:5843-5851. [PMID: 31737120 PMCID: PMC6843881 DOI: 10.7150/jca.31303] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 08/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background: The incidence and mortality rate of gastric cancer has markedly declined over the past few decades, due to the progress and advances in the development of diagnostic and treatment regimens. However, there is still a large portion of patients who are first diagnosed during the advanced stage of gastric cancer when chemotherapy is needed. Unfortunately, resistance to chemotherapeutic agents is the most frequent occurrence during treatment, which indicates a need for the discovery of novel therapeutic anticancer drugs. Methods: The tumor-suppression effect of eight different cucurbitacins was evaluated in gastric cancer cell lines, and the Cucurbitacin E (CuE) showing the greatest effect was used in further studies to explore the mechanism and potential synergistic effect of Dox both in vitro and in vivo. Results: Compared with other cucurbitacins, CuE showed the greatest antiproliferative activity against the gastric cancer cell lines. Further investigations revealed that CuE suppressed the growth of gastric cancer cell lines through the induction of G2/M arrest and subsequent apoptosis by impairing AKt activation and reducing its expression in gastric cancer cells. Furthermore, our results indicate that CuE can significantly enhance the cytotoxicity of doxorubicin (Dox) both in vitro and in vivo. Conclusion: In summary, we present the first evidence of the efficacy of CuE for the inhibition of gastric cancer growth and the synergistic antitumorigenic effect of CuE and Dox, both in vitro and in vivo.
Collapse
Affiliation(s)
- Wenzhang Si
- Department of General Surgery, Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), Shaoxing, Zhejiang Province, China
| | - Jia Lyu
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Zhengchuang Liu
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province 310014, China
| | - Chunyang Wang
- Genetron Health (Hangzhou) Medical Laboratory Co. Ltd, Hangzhou 310000, China
| | - Jingjing Huang
- Genetron Health (Hangzhou) Medical Laboratory Co. Ltd, Hangzhou 310000, China
| | - Liping Jiang
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province 310014, China
| | - Tonghui Ma
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province 310014, China
| |
Collapse
|
29
|
Huang CT, Hsieh CH, Chung YH, Oyang YJ, Huang HC, Juan HF. Perturbational Gene-Expression Signatures for Combinatorial Drug Discovery. iScience 2019; 15:291-306. [PMID: 31102995 PMCID: PMC6525321 DOI: 10.1016/j.isci.2019.04.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/02/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease that relies on both oncogenic mutations and non-mutated genes for survival, and therefore coined as oncogene and non-oncogene addictions. The need for more effective combination therapies to overcome drug resistance in oncology has been increasingly recognized, but the identification of potentially synergistic drugs at scale remains challenging. Here we propose a gene-expression-based approach, which uses the recurrent perturbation-transcript regulatory relationships inferred from a large compendium of chemical and genetic perturbation experiments across multiple cell lines, to engender a testable hypothesis for combination therapies. These transcript-level recurrences were distinct from known compound-protein target counterparts, were reproducible in external datasets, and correlated with small-molecule sensitivity. We applied these recurrent relationships to predict synergistic drug pairs for cancer and experimentally confirmed two unexpected drug combinations in vitro. Our results corroborate a gene-expression-based strategy for combinatorial drug screening as a way to target non-mutated genes in complex diseases. Compound signatures targeting non-oncogene addiction for combinatorial drug discovery These signatures are reproducible and linked to cancer hallmarks and drug sensitivity Two synergistic drug combinations are experimentally confirmed in vitro
Collapse
Affiliation(s)
- Chen-Tsung Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
| | - Chiao-Hui Hsieh
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Yun-Hsien Chung
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Yen-Jen Oyang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Hsueh-Fen Juan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan; Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan; Department of Life Science, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
30
|
Jiang X, Du Y, Meng X, Zhang H, Zhao D, Zhao L, Chen J, Xiao S, Jiang H. Low-Dose Radiation Enhanced Inhibition of Breast Tumor Xenograft and Reduced Myocardial Injury Induced by Doxorubicin. Dose Response 2018; 16:1559325818813061. [PMID: 30622447 PMCID: PMC6302275 DOI: 10.1177/1559325818813061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/14/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022] Open
Abstract
We reported that low-dose radiation (LDR) alleviated cardiotoxicity of doxorubicin (DOX) via inhibiting myocardial cell apoptosis and oxidative stress in vivo. Here, we tested whether LDR could enhance chemotherapeutic effect of DOX and alleviate myocardial injury induced by DOX by observing cell proliferation, apoptosis, and metastasis of heterotopic tumor in vivo. Mice implanted with 4T1 breast carcinoma cells were given 7.5 mg/kg DOX or 0.9% NaCl solution 72 hours after LDR (0 or 75 mGy). The histology of tumor tissue was observed by hematoxylin and eosin staining, the apoptosis was determined by terminal deoxynucleotidyl transferase–mediated dUTP nick-end labeling, and the expression of Ki67, Bcl-2, Bax, cleaved caspase3, matrix metalloproteinase 2 (MMP2), MMP9, and CD34 was detected by Western blot. Expression of Ki67 and CD34 was also detected by immunohistochemistry. Results showed that cell proliferation of the breast tumor and protein expression of the metastasis-related molecules were significantly reduced and the apoptosis of tumor cells was significantly increased in the LDR + DOX-treated tumor-bearing mice. Pretreatment with LDR significantly prevented DOX-induced cardiotoxicity likely through preventing DOX-induced mitochondrial Bcl2/Bax dyshomeostasis-induced caspase-3 cleavage-dependent apoptosis. These results suggested that LDR not only enhances DOX antitumor effect but also reduces DOX cardiotoxicity, which may potentially overcome the limitation for DOX clinical usage.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Health Examination Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanwei Du
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xinxin Meng
- Department of Health Examination Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongmei Zhang
- Cardiac Intensive Care Unit, First People's Hospital of Shangqiu, Henan, China
| | - Di Zhao
- Department of Health Examination Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Junyu Chen
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shengxiang Xiao
- Emergency Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongyu Jiang
- Department of Health Examination Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
31
|
Renu K, K B S, Parthiban S, S S, George A, P B TP, Suman S, V G A, Arunachalam S. Elevated lipolysis in adipose tissue by doxorubicin via PPARα activation associated with hepatic steatosis and insulin resistance. Eur J Pharmacol 2018; 843:162-176. [PMID: 30452912 DOI: 10.1016/j.ejphar.2018.11.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022]
Abstract
Adipose dysfunction is tightly associated with hepatic insulin resistance and steatosis condition. Doxorubicin would disturb the lipid metabolism both in adipose and liver. Here we projected that doxorubicin would impede lipogenesis and elevated lipolysis in adipose tissue would elevate the circulatory lipid profile and leads to insulin resistance. Further exacerbated lipid profile in circulation would impair the lipid metabolism in hepatic tissue which leads to fatty liver condition and consequently related disease during doxorubicin treatment. Doxorubicin impairs the lipogenesis through PPARγ and augments lipolysis and fatty acid oxidation through ATGL and PPARα in adipose tissue. Increased fatty acid level by adipose tissue in circulation would translocate into the liver and dysregulates AHR, PXR, PPARγ, ATGL and Apo B,which further develop insulin resistance and hepatic steatosis condition. The findings add to the mechanistic role of association between adipose tissue dysfunction and hepatic dysfunction.
Collapse
Affiliation(s)
- Kaviyarasi Renu
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu 632014, India
| | - Sruthy K B
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu 632014, India
| | - Sujitha Parthiban
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu 632014, India
| | - Sugunapriyadharshini S
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu 632014, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - Tirupathi Pichiah P B
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center (LCCC), Georgetown University Medical Center (GUMC), E504, NRB, 3970 Reservoir Rd. NW, Washington, D.C., USA
| | - Abilash V G
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu 632014, India.
| | - Sankarganesh Arunachalam
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India.
| |
Collapse
|
32
|
Lai ZY, Yeo HY, Chen YT, Chang KM, Chen TC, Chuang YJ, Chang SJ. PI3K inhibitor enhances the cytotoxic response to etoposide and cisplatin in a newly established neuroendocrine cervical carcinoma cell line. Oncotarget 2018; 8:45323-45334. [PMID: 28484083 PMCID: PMC5542189 DOI: 10.18632/oncotarget.17335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/12/2017] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Neuroendocrine cervical carcinoma (NECC) is a rare and aggressive subtype of cervical cancer. To date, no NECC cell-based model is available, which hinders the development of new therapeutic strategies for NECC. In this study, we derived a new NECC cell line from an ex vivo biopsy and used it to explore novel drug combination approach for NECC. RESULTS The stable HM-1 cell line displayed high expression levels of the neuroendocrine marker, synaptophysin. HM-1 cell transplantation could induce tumor growth in nude mice. As expected, the combination of etoposide and cisplatin synergistically inhibited HM-1 cell proliferation. Strikingly, when etoposide and cisplatin were combined with PI3K inhibitor BEZ235, the growth of HM-1 cells was significantly reduced. Taken together, the data implied the combination of etoposide and cisplatin with BEZ235 not only inhibited HM-1 cell proliferation but also increased cell apoptosis. MATERIALS AND METHODS A NECC tissue sample from a 75-year-old female patient was processed to derive a primary cell line annotated as HM-1. The features of HM-1 were analyzed to establish its characteristic profile. Next, HM-1 was treated with PI3K inhibitors, BKM120 and/or BEZ235, in combination with two well-known genotoxic drugs, etoposide and/or cisplatin, to evaluate which combination could serve as a more effective treatment approach. Their inhibiting effects on HM-1 were evaluated by cell viability, apoptosis, and target kinase expression. CONCLUSIONS The newly established NECC cell line HM-1 could serve as a cell-based model for NECC research. The synergistic drug combination of PI3K inhibitor with genotoxic drugs might become a potential new treatment strategy against NECC.
Collapse
Affiliation(s)
- Zih-Yin Lai
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan (R.O.C.)
| | - Hsin-Yueh Yeo
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan (R.O.C.)
| | - Ya-Tse Chen
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan (R.O.C.)
| | - Kuo-Ming Chang
- Department of Pathology, Hsinchu MacKay Memorial Hospital, Hsinchu, 30071, Taiwan (R.O.C.)
| | - Tze-Chien Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, 10449, Taiwan (R.O.C.)
| | - Yung-Jen Chuang
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan (R.O.C.)
| | - Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu, 30071, Taiwan (R.O.C.)
| |
Collapse
|
33
|
Kawaguchi K, Igarashi K, Kiyuna T, Miyake K, Miyake M, Murakami T, Chmielowski B, Nelson SD, Russell TA, Dry SM, Li Y, Singh AS, Unno M, Eilber FC, Hoffman RM. Individualized doxorubicin sensitivity testing of undifferentiated soft tissue sarcoma (USTS) in a patient-derived orthotopic xenograft (PDOX) model demonstrates large differences between patients. Cell Cycle 2018; 17:627-633. [PMID: 29384032 DOI: 10.1080/15384101.2017.1421876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Doxorubicin (DOX) is often first-line treatment of undifferentiated/unclassified soft tissue sarcoma (USTS). However, the DOX response rate for USTS patients is low. Individualized precision-medicine technology that could identify DOX responders as well as non-responders would be of high value to cancer patients. In the present study, we established 5 patient-derived orthotopic xenograft (PDOX) nude mouse models from 5 USTS patients and evaluated the efficacy of DOX in each PDOX model. USTS's were grown orthotopically in the right thigh of nude mice to establish the PDOX models. Two weeks after implantation, the mouse models were randomized into two groups of 8 mice each: untreated control; and DOX (3 mg/kg, i.p., once a week for 2 weeks). DOX showed significant growth inhibition in only 2 USTS PDOX models out of 5 (p = 0.0054, p = 0.0055, respectively) on day 14 after initiation. DOX was ineffective in the other 3 PDOX models. However, even in the DOX-sensitive cases, DOX could not regress the PDOX tumors responding to treatment. The present study has important implications since this is the first in vivo study to compare the DOX sensitivity for USTS on multiple patient tumors. We showed that only two of five USTS were responsive to DOX, despite DOX being first line chemotherapy for USTS. The 3 resistant cases should not be treated with DOX clinically, in order to spare the patients' unnecessary toxicity. This PDOX model is useful for precise individualized drug sensitivity testing, especially for rare heterogeneous recalcitrant sarcomas such as USTS.
Collapse
Affiliation(s)
- Kei Kawaguchi
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA.,c Department of Surgery, Graduate School of Medicine , Tohoku University , Sendai , Japan
| | - Kentaro Igarashi
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Tasuku Kiyuna
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Kentaro Miyake
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Masuyo Miyake
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Takashi Murakami
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Bartosz Chmielowski
- d Division of Hematology-Oncology , University of California , Los Angeles , CA , USA
| | - Scott D Nelson
- e Dep artmen t of Pathology , University of California , Los Angeles , CA , USA
| | - Tara A Russell
- f Division of Surgical Oncology , University of California , Los Angeles , CA , USA
| | - Sarah M Dry
- e Dep artmen t of Pathology , University of California , Los Angeles , CA , USA
| | - Yunfeng Li
- e Dep artmen t of Pathology , University of California , Los Angeles , CA , USA
| | - Arun S Singh
- d Division of Hematology-Oncology , University of California , Los Angeles , CA , USA
| | - Michiaki Unno
- c Department of Surgery, Graduate School of Medicine , Tohoku University , Sendai , Japan
| | - Fritz C Eilber
- f Division of Surgical Oncology , University of California , Los Angeles , CA , USA
| | - Robert M Hoffman
- a AntiCancer, Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| |
Collapse
|
34
|
Fourneaux B, Chaire V, Lucchesi C, Karanian M, Pineau R, Laroche-Clary A, Italiano A. Dual inhibition of the PI3K/AKT/mTOR pathway suppresses the growth of leiomyosarcomas but leads to ERK activation through mTORC2: biological and clinical implications. Oncotarget 2018; 8:7878-7890. [PMID: 28002802 PMCID: PMC5352367 DOI: 10.18632/oncotarget.13987] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
The PI3K/AKT/mTOR pathway plays a crucial role in the development of leiomyosarcomas (LMSs). In this study, we tested the efficacy of dual PI3K/mTOR (BEZ235), PI3K (BKM120) and mTOR (everolimus) inhibitors in three human LMS cell lines. In vitro and in vivo studies using LMS cell lines showed that BEZ235 has a significantly higher anti-tumor effect than either BKM120 or everolimus, resulting in a greater reduction in tumor growth and more pronounced inhibitory effects on mitotic activity and PI3K/AKT/mTOR signaling. Strikingly, BEZ235 but neither BKM120 nor everolimus markedly enhanced the ERK pathway. This effect was reproduced by the combination of BKM120 and everolimus, suggesting the involvement of mTORC2 via a PI3K-independent mechanism. Silencing of RICTOR in LMS cells confirmed the role of mTORC2 in the regulation of ERK activity. Combined treatment with BEZ235 and GSK1120212, a potent MEK inhibitor, resulted in synergistic growth inhibition and apoptosis induction in vitro and in vivo. These findings document for the first time that dual PI3K/mTOR inhibition in leiomyosarcomas suppress a negative feedback loop mediated by mTORC2, leading to enhanced ERK pathway activity. Thus, combining a dual PI3K/mTOR inhibitor with MEK inhibitors may be a relevant approach to increase anti-tumor activity and prevent drug resistance in patients with LMS.
Collapse
Affiliation(s)
- Benjamin Fourneaux
- Université de Bordeaux, Bordeaux, France.,Institut National de la Santé et de la Recherche Medicale (INSERM), Institut Bergonié, Bordeaux, France
| | - Vanessa Chaire
- Université de Bordeaux, Bordeaux, France.,Institut National de la Santé et de la Recherche Medicale (INSERM), Institut Bergonié, Bordeaux, France
| | - Carlo Lucchesi
- Institut National de la Santé et de la Recherche Medicale (INSERM), Institut Bergonié, Bordeaux, France
| | - Marie Karanian
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | | | - Audrey Laroche-Clary
- Université de Bordeaux, Bordeaux, France.,Institut National de la Santé et de la Recherche Medicale (INSERM), Institut Bergonié, Bordeaux, France
| | - Antoine Italiano
- Institut National de la Santé et de la Recherche Medicale (INSERM), Institut Bergonié, Bordeaux, France.,Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| |
Collapse
|
35
|
Abstract
RATIONALE Leiomyosarcoma (LMS) is a common type of soft tissue sarcoma. Primary colonic LMS in general is a very rare entity, accounting for 1% to 2% of gastrointestinal malignancies. PATIENT CONCERNS We report a case of 55-year-old female who presented with a sudden onset of sharp right lower quadrant abdominal pain. Electronic colonoscopy showed a normal lumen. However, an abdominal computed tomography scan revealed a mass of soft tissue attenuation inseparable from the ascending colon which appeared as a gastrointestinal stromal tumor (GIST). DIAGNOSES It is important to diagnose LMS definitively by immunohistochemical profiling of smooth muscle actin, desmin, and CD34. INTERVENTIONS She underwent laparotomy and right hemicolectomy, and histology confirmed a colonic LMS. The patient received no oncological treatment after surgery. OUTCOMES No recurrence or metastasis was observed at 5 months postoperatively. It is crucial to identify colonic LMS precisely based on immunohistochemistry, and thereby distinguish it from GIST. LESSONS Further investigation on LMS cases so far is required to establish standard treatment strategies.
Collapse
|
36
|
Gao P, Seebacher NA, Hornicek F, Guo Z, Duan Z. Advances in sarcoma gene mutations and therapeutic targets. Cancer Treat Rev 2017; 62:98-109. [PMID: 29190505 DOI: 10.1016/j.ctrv.2017.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022]
Abstract
Sarcomas are rare and complex malignancies that have been associated with a poor prognostic outcome. Over the last few decades, traditional treatment with surgery and/or chemotherapy has not significantly improved outcomes for most types of sarcomas. In recent years, there have been significant advances in the understanding of specific gene mutations that are important in driving the pathogenesis and progression of sarcomas. Identification of these new gene mutations, using next-generation sequencing and advanced molecular techniques, has revealed a range of potential therapeutic targets. This, in turn, may lead to the development of novel agents targeted to different sarcoma subtypes. In this review, we highlight the advances made in identifying sarcoma gene mutations, including those of p53, RB, PI3K and IDH genes, as well as novel therapeutic strategies aimed at utilizing these mutant genes. In addition, we discuss a number of preclinical studies and ongoing early clinical trials in sarcoma targeting therapies, as well as gene editing technology, which may provide a better choice for sarcoma patient management.
Collapse
Affiliation(s)
- Peng Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Nicole A Seebacher
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Francis Hornicek
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Zheng Guo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhenfeng Duan
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA.
| |
Collapse
|
37
|
PI3K/mTOR dual inhibitor BEZ235 and histone deacetylase inhibitor Trichostatin A synergistically exert anti-tumor activity in breast cancer. Oncotarget 2017; 8:11937-11949. [PMID: 28060760 PMCID: PMC5355316 DOI: 10.18632/oncotarget.14442] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/27/2016] [Indexed: 01/01/2023] Open
Abstract
Molecule-targeted therapy has achieved great progress in cancer therapy. Effective drug combinations are one way to enhance the therapeutic efficacy and combat resistance. Here, we determined the effect of the PI3K/mTOR dual inhibitor BEZ235 and the histone deacetylase inhibitor Trichostatin A (TSA) on human breast cancer. We demonstrated that the combination of BEZ235 and TSA results in significant synergistic growth inhibition of multiple breast cancer cell lines. Mechanistic studies revealed that the combined therapy induced apoptosis in a caspase-dependent manner, which might be related to the further depression of the PI3K/Akt/mTOR signalling pathway. Additionally, co-treatment with BEZ235 and TSA enhanced autophagic cell death by up-regulating the expression of LC3B-II and Beclin-1. The vivo tumour modelling studies revealed that BEZ235 combined with TSA blocked tumour growth without noticeable side effects. These data suggest that the combination of BEZ235 and TSA may be a new selective strategy, which may have significant clinical application in the treatment of breast cancer patients.
Collapse
|
38
|
Abstract
INTRODUCTION Uterine sarcomas are rare cancers, of which the most common entities are leiomyosarcoma and endometrial stromal sarcoma. These two tumors may have overlapping clinical presentation, morphology and immunohistochemical profile, but are increasingly recognized to be two molecularly distinct entities. Endometrial stromal sarcomas are further currently divided into a low-grade and high-grade group based on molecular characteristics. Area covered: This review discusses recent data which shed light on the molecular profile of these two cancers and may aid in understanding their evolution and progression, in the aim of improving their diagnosis and management. Search was through PubMed, with focus on studies published in the last 5 years. Expert commentary: The literature presented and discussed documents rapidly expanding knowledge of the genetic characteristics of leiomyosarcoma and endometrial stromal sarcoma, with an array of molecules and pathways implicated in the biology of these cancers. Several of these molecules are potential therapeutic targets. Assessment of their predictive and prognostic role awaits larger studies.
Collapse
Affiliation(s)
- Ben Davidson
- a Department of Pathology , Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway.,b Institute of Clinical Medicine , University of Oslo , Oslo , Norway
| | - Francesca Micci
- c Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Norwegian Radium Hospital , Oslo University Hospital , Oslo , Norway.,d Centre for Cancer Biomedicine , University of Oslo , Oslo , Norway
| |
Collapse
|
39
|
Cuppens T, Annibali D, Coosemans A, Trovik J, ter Haar N, Colas E, Garcia-Jimenez A, Van de Vijver K, Kruitwagen RP, Brinkhuis M, Zikan M, Dundr P, Huvila J, Carpén O, Haybaeck J, Moinfar F, Salvesen HB, Stukan M, Mestdagh C, Zweemer RP, Massuger LF, Mallmann MR, Wardelmann E, Mints M, Verbist G, Thomas D, Gommé E, Hermans E, Moerman P, Bosse T, Amant F. Potential Targets' Analysis Reveals Dual PI3K/mTOR Pathway Inhibition as a Promising Therapeutic Strategy for Uterine Leiomyosarcomas—an ENITEC Group Initiative. Clin Cancer Res 2017; 23:1274-1285. [DOI: 10.1158/1078-0432.ccr-16-2149] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/03/2016] [Accepted: 11/08/2016] [Indexed: 11/16/2022]
|
40
|
Enhanced Anticancer Activity of PF-04691502, a Dual PI3K/mTOR Inhibitor, in Combination With VEGF siRNA Against Non-small-cell Lung Cancer. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e384. [PMID: 27845769 PMCID: PMC5155322 DOI: 10.1038/mtna.2016.90] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
Abstract
Lung cancer is the leading cause of cancer deaths in both men and women in the United States accounting for about 27% of all cancer deceases. In our effort to develop newer therapy for lung cancer, we evaluated the combinatory antitumor effect of siRNA targeting VEGF and the PI3K/mTOR dual inhibitor PF-04691502. We analyzed the anticancer effect of siRNA VEGF and PF-04691502 combination on proliferation, colony formation and migration of A549 and H460 lung cancer cells. Additionally, we assessed the combination treatment antiangiogenic effect on human umbilical vein endothelial cells. Here, we show for the first time that the antiangiogenic siRNA VEGF potentiates the PF-04691502 anticancer activity against non–small-cell lung cancer. We observed a significant (P < 0.05) decrease in cell viability, colony formation, and migration for the combination comparing with the single drug treatment. We also showed a significant (P < 0.05) enhanced effect of the combination treatment inhibiting angiogenesis progression and tube formation organization compared to the single drug treatment groups. Our findings demonstrated an enhanced synergistic anticancer effect of siRNA VEGF and PF-04691502 combination therapy by targeting two main pathways involved in lung cancer cell survival and angiogenesis which will be useful for future preclinical studies and potentially for lung cancer patient management.
Collapse
|
41
|
Ren ZX, Zhao YF, Cao T, Zhen XC. Dihydromyricetin protects neurons in an MPTP-induced model of Parkinson's disease by suppressing glycogen synthase kinase-3 beta activity. Acta Pharmacol Sin 2016; 37:1315-1324. [PMID: 27374489 DOI: 10.1038/aps.2016.42] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/21/2016] [Indexed: 12/20/2022]
Abstract
AIM It is general believed that mitochondrial dysfunction and oxidative stress play critical roles in the pathology of Parkinson's disease (PD). Dihydromyricetin (DHM), a natural flavonoid extracted from Ampelopsis grossedentata, has recently been found to elicit potent anti-oxidative effects. In the present study, we explored the role of DHM in protecting dopaminergic neurons. METHODS Male C57BL/6 mice were intraperitoneally injected with 1-methyl4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) for 7 d to induce PD. Additionally, mice were treated with either 5 or 10 mg/kg DHM for a total of 13 d (3 d before the start of MPTP, during MPTP administration (7 d) and 3 d after the end of MPTP). For the saline or DHM alone treatment groups, mice were injected with saline or DHM for 13 d. On d 14, behavioral tests (locomotor activity, the rotarod test and the pole test) were administered. After the behavioral tests, the mice were sacrificed, and brain tissue was collected for immunofluorescence staining and Western blotting. In addition, MES23.5 cells were treated with MPP+ and DHM, and evaluated using cell viability assays, reactive oxygen species (ROS) measurements, apoptosis analysis and Western blotting. RESULTS DHM significantly attenuated MPTP-induced mouse behavioral impairments and dopaminergic neuron loss. In the MES23.5 cells, DHM attenuated MPP+-induced cell injury and ROS production in a dose-dependent manner. In addition, DHM increased glycogen synthase kinase-3 beta phosphorylation in a dose- and time-dependent manner, which may be associated with DHM-induced dopaminergic neuronal protection. CONCLUSION The present study demonstrated that DHM is a potent neuroprotective agent for DA neurons by modulating the Akt/GSK-3β pathway, which suggests that DHM may be a promising therapeutic candidate for PD.
Collapse
|