1
|
Gao C, Fan X, Liu Y, Han Y, Liu S, Li H, Zhang Q, Wang Y, Xue F. Comprehensive Analysis Reveals the Potential Roles of CDKN3 in Pancancer and Verification in Endometrial Cancer. Int J Gen Med 2023; 16:5817-5839. [PMID: 38106976 PMCID: PMC10723185 DOI: 10.2147/ijgm.s438479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
Background Cyclin-dependent kinase inhibitor 3 (CDKN3) has been studied in many cancers. However, the comprehensive and systematic pancancer analysis of CDKN3 genes is still lacking. Methods Data were downloaded from online databases. R was used for analysis of the differential expression and gene alteration of CDKN3 and of the associations between CDKN3 expression and survival, signaling pathways, and drug sensitivity. Clinical samples and in vitro experiments were selected for verification. Results CDKN3 expression was higher in most types of cancers, and this phenotype was significantly correlated with poor survival. CDKN3 showed gene alterations and copy number alterations in many cancers and associated with some immune-related pathways and factors. Drug sensitivity analysis elucidated that CDKN3 could be a useful marker for therapy selection. Clinical samples elucidated CDKN3 expressed high in endometrial cancer tissue. In vitro studies showed that CDKN3 induced pro-tumor effect in immune environment and facilitated endometrial cancer cell proliferation and G1/S phase transition. Conclusion CDKN3 has been shown to be highly expressed in most types of cancers and promoted cancer cell progression. CDKN3 may serve as a novel marker in clinical diagnosis, treatment, and prognosis prediction in future.
Collapse
Affiliation(s)
- Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Xiangqin Fan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Department of Obstetrics and Gynecology, Zaozhuang Municipal Hospital, Shandong, People’s Republic of China
| | - Yanyan Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Yanyan Han
- Department of Pathology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Shiqi Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Qiaoling Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
2
|
Cuciniello R, Di Meo F, Sulli M, Demurtas OC, Tanori M, Mancuso M, Villano C, Aversano R, Carputo D, Baldi A, Diretto G, Filosa S, Crispi S. Aglianico Grape Seed Semi-Polar Extract Exerts Anticancer Effects by Modulating MDM2 Expression and Metabolic Pathways. Cells 2023; 12:cells12020210. [PMID: 36672146 PMCID: PMC9856309 DOI: 10.3390/cells12020210] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Grapevine (Vitis vinifera L.) seeds are rich in polyphenols including proanthocyanidins, molecules with a variety of biological effects including anticancer action. We have previously reported that the grape seed semi-polar extract of Aglianico cultivar (AGS) was able to induce apoptosis and decrease cancer properties in different mesothelioma cell lines. Concomitantly, this extract resulted in enriched oligomeric proanthocyanidins which might be involved in determining the anticancer activity. Through transcriptomic and metabolomic analyses, we investigated in detail the anticancer pathway induced by AGS. Transcriptomics analysis and functional annotation allowed the identification of the relevant causative genes involved in the apoptotic induction following AGS treatment. Subsequent biological validation strengthened the hypothesis that MDM2 could be the molecular target of AGS and that it could act in both a p53-dependent and independent manner. Finally, AGS significantly inhibited tumor progression in a xenograft mouse model of mesothelioma, confirming also in vivo that MDM2 could act as molecular player responsible for the AGS antitumor effect. Our findings indicated that AGS, exerting a pro-apoptotic effect by hindering MDM2 pathway, could represent a novel source of anticancer molecules.
Collapse
Affiliation(s)
- Rossana Cuciniello
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino 111, 80131 Naples, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Francesco Di Meo
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino 111, 80131 Naples, Italy
- Department of Medicine, Indiana University School of Medicine, 975 W Walnut Street, Indianapolis, IN 46202, USA
| | - Maria Sulli
- Division of Biotechnology and Agroindustry, Biotechnology Laboratory, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Olivia Costantina Demurtas
- Division of Biotechnology and Agroindustry, Biotechnology Laboratory, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Mirella Tanori
- Division of Health Protection Technologies, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Clizia Villano
- Department of Agricultural Sciences, University of Naples Federico II, Via Università 100, 80055 Portici, Italy
| | - Riccardo Aversano
- Department of Agricultural Sciences, University of Naples Federico II, Via Università 100, 80055 Portici, Italy
| | - Domenico Carputo
- Department of Agricultural Sciences, University of Naples Federico II, Via Università 100, 80055 Portici, Italy
| | - Alfonso Baldi
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino 111, 80131 Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “L. Vanvitelli”, 81055 Caserta, Italy
| | - Gianfranco Diretto
- Division of Biotechnology and Agroindustry, Biotechnology Laboratory, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
- Correspondence: (G.D.); (S.C.)
| | - Stefania Filosa
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino 111, 80131 Naples, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Stefania Crispi
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino 111, 80131 Naples, Italy
- Correspondence: (G.D.); (S.C.)
| |
Collapse
|
3
|
Boccellino M, Ambrosio P, Ballini A, De Vito D, Scacco S, Cantore S, Feola A, Di Donato M, Quagliuolo L, Sciarra A, Galasso G, Crocetto F, Imbimbo C, Boffo S, Di Zazzo E, Di Domenico M. The Role of Curcumin in Prostate Cancer Cells and Derived Spheroids. Cancers (Basel) 2022; 14:cancers14143348. [PMID: 35884410 PMCID: PMC9320241 DOI: 10.3390/cancers14143348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
A major challenge in the clinical management of prostate cancer (PC) is to inhibit tumor growth and prevent metastatic spreading. In recent years, considerable efforts have been made to discover new compounds useful for PC therapy, and promising advances in this field were reached. Drugs currently used in PC therapy frequently induce resistance and PC progresses toward metastatic castration-resistant forms (mCRPC), making it virtually incurable. Curcumin, a commercially available nutritional supplement, represents an attractive therapeutic agent for mCRPC patients. In the present study, we compared the effects of chemotherapeutic drugs such as docetaxel, paclitaxel, and cisplatin, to curcumin, on two PC cell lines displaying a different metastatic potential: DU145 (moderate metastatic potential) and PC-3 (high metastatic potential). Our results revealed a dose-dependent reduction of DU145 and PC-3 cell viability upon treatment with curcumin similar to chemotherapeutic agents (paclitaxel, cisplatin, and docetaxel). Furthermore, we explored the EGFR-mediated signaling effects on ERK activation in DU145 and PC-3 cells. Our results showed that DU145 and PC-3 cells overexpress EGFR, and the treatment with chemotherapeutic agents or curcumin reduced EGFR expression levels and ERK activation. Finally, chemotherapeutic agents and curcumin reduced the size of DU145 and PC-3 spheroids and have the potential to induce apoptosis and also in Matrigel. In conclusion, despite different studies being carried out to identify the potential synergistic curcumin combinations with chemopreventive/therapeutic efficacy for inhibiting PC growth, the results show the ability of curcumin used alone, or in combinatorial approaches, to impair the size and the viability of PC-derived spheroids.
Collapse
Affiliation(s)
- Mariarosaria Boccellino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Pasqualina Ambrosio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
- Correspondence: (A.B.); (S.C.)
| | - Danila De Vito
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (D.D.V.); (S.S.)
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (D.D.V.); (S.S.)
| | - Stefania Cantore
- Independent Researcher, 70129 Bari, Italy
- Correspondence: (A.B.); (S.C.)
| | - Antonia Feola
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Marzia Di Donato
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Lucio Quagliuolo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Antonella Sciarra
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy;
| | - Giovanni Galasso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Felice Crocetto
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (C.I.)
| | - Ciro Imbimbo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (C.I.)
| | - Silvia Boffo
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122-6078, USA;
| | - Erika Di Zazzo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122-6078, USA;
| |
Collapse
|
4
|
Di Meo F, Esposito R, Cuciniello R, Favale G, Arenga M, Ruocco N, Nuzzo G, Fontana A, Filosa S, Crispi S, Costantini M. Organic extract of Geodia cydonium induces cell cycle block in human mesothelioma cells. Oncol Lett 2022; 24:286. [PMID: 35814825 PMCID: PMC9260718 DOI: 10.3892/ol.2022.13406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Francesco Di Meo
- Department of Biology, Agriculture and Food Science, Institute of Biosciences and BioResources‑UOS Naples, National Research Council, I‑80131 Naples, Italy
| | - Roberta Esposito
- Department of Ecosustainable Marine Biotechnology, Zoological Station Anton Dohrn, I‑80121 Naples, Italy
| | - Rossana Cuciniello
- Department of Biology, Agriculture and Food Science, Institute of Biosciences and BioResources‑UOS Naples, National Research Council, I‑80131 Naples, Italy
| | - Gregorio Favale
- Department of Biology, Agriculture and Food Science, Institute of Biosciences and BioResources‑UOS Naples, National Research Council, I‑80131 Naples, Italy
| | - Mario Arenga
- Department of Biology, Agriculture and Food Science, Institute of Biosciences and BioResources‑UOS Naples, National Research Council, I‑80131 Naples, Italy
| | - Nadia Ruocco
- Department of Ecosustainable Marine Biotechnology, Zoological Station Anton Dohrn, I‑80121 Naples, Italy
| | - Genoveffa Nuzzo
- Department of Chemical Sciences and Materials Technologies, Institute of Biomolecular Chemistry, National Research Council, I‑80078 Naples, Italy
| | - Angelo Fontana
- Department of Chemical Sciences and Materials Technologies, Institute of Biomolecular Chemistry, National Research Council, I‑80078 Naples, Italy
| | - Stefania Filosa
- Department of Biology, Agriculture and Food Science, Institute of Biosciences and BioResources‑UOS Naples, National Research Council, I‑80131 Naples, Italy
| | - Stefania Crispi
- Department of Biology, Agriculture and Food Science, Institute of Biosciences and BioResources‑UOS Naples, National Research Council, I‑80131 Naples, Italy
| | - Maria Costantini
- Department of Ecosustainable Marine Biotechnology, Zoological Station Anton Dohrn, I‑80121 Naples, Italy
| |
Collapse
|
5
|
ACTL6A suppresses p21 Cip1 tumor suppressor expression to maintain an aggressive mesothelioma cancer cell phenotype. Oncogenesis 2021; 10:70. [PMID: 34689163 PMCID: PMC8542039 DOI: 10.1038/s41389-021-00362-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Mesothelioma is a poor prognosis cancer of the mesothelial lining that develops in response to exposure to various agents including asbestos. Actin-Like Protein 6A (ACTL6A, BAF53a) is a SWI/SNF regulatory complex protein that is elevated in cancer cells and has been implicated as a driver of cancer cell survival and tumor formation. In the present study, we show that ACTL6A drives mesothelioma cancer cell proliferation, spheroid formation, invasion, and migration, and that these activities are markedly attenuated by ACTL6A knockdown. ACTL6A expression reduces the levels of the p21Cip1 cyclin-dependent kinase inhibitor and tumor suppressor protein. DNA binding studies show that ACTL6A interacts with Sp1 and p53 binding DNA response elements in the p21Cip1 gene promoter and that this is associated with reduced p21Cip1 promoter activity and p21Cip1 mRNA and protein levels. Moreover, ACTL6A suppression of p21Cip1 expression is required for maintenance of the aggressive mesothelioma cancer cell phenotype suggesting that p21Cip1 is a mediator of ACTL6A action. p53, a known inducer of p21Cip1 expression, is involved ACTL6A in regulation of p21Cip1 in some but not all mesothelioma cells. In addition, ACTL6A knockout markedly reduces tumor formation and this is associated with elevated tumor levels of p21Cip1. These findings suggest that ACTL6A suppresses p21Cip1 promoter activity to reduce p21Cip1 protein as a mechanism to maintain the aggressive mesothelioma cell phenotype.
Collapse
|
6
|
H9c2 Cardiomyocytes under Hypoxic Stress: Biological Effects Mediated by Sentinel Downstream Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6874146. [PMID: 34630851 PMCID: PMC8497098 DOI: 10.1155/2021/6874146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 04/13/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022]
Abstract
The association between diabetes and cardiovascular diseases is well known. Related diabetes macro- and microangiopathies frequently induce hypoxia and consequently energy failure to satisfy the jeopardized myocardium basal needs. Additionally, it is widely accepted that diabetes impairs endothelial nitric oxide synthase (eNOS) activity, resulting in diminished nitric oxide (NO) bioavailability and consequent endothelial cell dysfunction. In this study, we analyzed the embryonic heart-derived H9c2 cell response to hypoxic stress after administration of a high glucose concentration to reproduce a condition often observed in diabetes. We observed that 24 h hypoxia exposure of H9c2 cells reduced cell viability compared to cells grown in normoxic conditions. Cytotoxicity and early apoptosis were increased after exposure to high glucose administration. In addition, hypoxia induced a RhoA upregulation and a Bcl-2 downregulation and lowered the ERK activation observed in normoxia at both glucose concentrations. Furthermore, a significant cell proliferation rate increases after the 1400 W iNOS inhibitor administration was observed. Again, hypoxia increased the expression level of myogenin, a marker of skeletal muscle cell differentiation. The cardiomyocyte gene expression profiles and morphology changes observed in response to pathological stimuli, as hypoxia, could lead to improper ventricular remodeling responsible for heart failure. Therefore, understanding cell signaling events that regulate cardiac response to hypoxia could be useful for the discovery of novel therapeutic approaches able to prevent heart diseases.
Collapse
|
7
|
Nuvoli B, Antoniani B, Libener R, Maconi A, Sacconi A, Carosi M, Galati R. Identification of novel COX-2 / CYP19A1 axis involved in the mesothelioma pathogenesis opens new therapeutic opportunities. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:257. [PMID: 34404424 PMCID: PMC8369782 DOI: 10.1186/s13046-021-02050-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/23/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Based on previous studies highlighting that the induction of cyclooxygenase-2 (COX-2) and high prostaglandin E2 (PGE2) levels contribute to the pathogenesis of malignant pleural mesothelioma (MPM), and that aromatase (CYP19A1), an enzyme that plays a key role in estrogen biosynthesis, along with estradiol (E2) were expressed in MPM, this study aimed to investigate the possible interplay between COX-2 and CYP19A1 in the pathogenesis of mesothelioma, as well as the underlying mechanism. METHODS The interaction between COX-2 and CYP19A1 was first investigated on different MPM lines upon PGE2, and COX-2 inhibitor (rofecoxib) treatment by western blot, RT-PCR. The key regulatory pathways involved in the COX-2 and CYP19A1 axis were further studied in MPM cells, after rofecoxib and exemestane (CYP19A1 inhibitor) treatment in monotherapy and in combination, by cell cycle distribution, western blot, and combination index analysis. To explore the role of COX-2/CYP19A1 axis in 3D preclinical models of MPM cells, we analyzed the effect of combination of COX-2 and CYP19A1 inhibitors in mesosphere formation. Immunohistochemical analysis of MPM mesosphere and specimens was utilized to evaluate the involvement of COX-2 on the CYP19A1 activity and the relationship between E2 and COX-2. RESULTS PGE2 or rofecoxib treatment caused in MPM cells an increased or decreased, respectively, CYP19A1 expression at mRNA and protein levels. The effect of rofecoxib and exemestane combination in MPM cell proliferation was synergistic. Activation of caspase-3 and cleavage of PARP confirmed an apoptotic death for MPM cell lines. Increased expression levels of p53, p21, and p27, downregulation of cyclin D1 and inhibition of Akt activation (pAKT) were also found. The antagonistic effect of rofecoxib and exemestane combination found only in one cell line, was reverted by pretreatment with MK2206, a pAKT inhibitor, indicating pAKT as an actionable mediator in the COX-2-CYP19A1 axis. Reduction of size and sphere-forming efficiency in MPM spheres after treatment with both inhibitor and a decrease in COX-2 and E2 staining was found. Moreover, immunohistochemical analysis of 46 MPM samples showed a significant positive correlation between COX-2 and E2. CONCLUSIONS Collectively, the results highlighted a novel COX-2/CYP19A1 axis in the pathogenesis of MPM that can be pharmacologically targeted, consequently opening up new therapeutic options.
Collapse
Affiliation(s)
- Barbara Nuvoli
- grid.417520.50000 0004 1760 5276Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Barbara Antoniani
- grid.417520.50000 0004 1760 5276Anatomy Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Roberta Libener
- Department of Integrated Activities Research and Innovation, SS Antonio and Biagio General Hospital, Alessandria, Italy
| | - Antonio Maconi
- Department of Integrated Activities Research and Innovation, SS Antonio and Biagio General Hospital, Alessandria, Italy
| | - Andrea Sacconi
- grid.417520.50000 0004 1760 5276Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariantonia Carosi
- grid.417520.50000 0004 1760 5276Anatomy Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Rossella Galati
- grid.417520.50000 0004 1760 5276Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
8
|
Combination of ERK2 inhibitor VX-11e and voreloxin synergistically enhances anti-proliferative and pro-apoptotic effects in leukemia cells. Apoptosis 2020; 24:849-861. [PMID: 31482470 PMCID: PMC6823322 DOI: 10.1007/s10495-019-01564-6] [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] [Indexed: 12/14/2022]
Abstract
ERK1/2 inhibitors are new promising anticancer drugs. The aim of this study was to investigate the effect of the combination of ERK2 inhibitor VX-11e and voreloxin on MOLM-14, K562, REH and MOLT-4 leukemia cell lines. We found that VX-11e alone and in combination with voreloxin significantly decreased ERK activation in all cell lines tested. To evaluate the interactions of the drugs, cells were treated for 24 h with VX-11e or voreloxin alone and in combination at fixed ratios based on IC50 values. The combinatorial effects of both drugs were synergistic over a wide range of concentrations in MOLM-14, REH and MOLT-4 cell lines. In K562 cells, three effects were found to be additive, one antagonistic and only one synergistic. The results showed that incubation with both VX-11e and voreloxin inhibited the growth of leukemia cells, affected cell cycle and induced apoptosis. Furthermore, the molecular mechanism of these effects might be attributed to an increased expression of p21 and a decreased expression of survivin and NF-κB in all cell lines tested except from K562 cells. In conclusion, combination of VX-11e and voreloxin can exert a synergistic anticancer effect in leukemia cells.
Collapse
|
9
|
Curcumin as an Anticancer Agent in Malignant Mesothelioma: A Review. Int J Mol Sci 2020; 21:ijms21051839. [PMID: 32155978 PMCID: PMC7084180 DOI: 10.3390/ijms21051839] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
Malignant mesothelioma is an infrequent tumor that initiates from the mesothelial cells lining of body cavities. The great majority of mesotheliomas originate in the pleural cavity, while the remaining cases initiate in the peritoneal cavity, in the pericardial cavity or on the tunica vaginalis. Usually, mesotheliomas grow in a diffuse pattern and tend to enclose and compress the organs in the various body cavities. Mesothelioma incidence is increasing worldwide and still today, the prognosis is very poor, with a reported median survival of approximately one year from presentation. Thus, the development of alternative and more effective therapies is currently an urgent requirement. The aim of this review article was to describe recent findings about the anti-cancer activity of curcumin and some of its derivatives on mesotheliomas. The potential clinical implications of these findings are discussed.
Collapse
|
10
|
Fallahi H, Godini R. System-level responses to cisplatin in pro-apoptotic stages of breast cancer MCF-7 cell line. Comput Biol Chem 2019; 83:107155. [PMID: 31706153 DOI: 10.1016/j.compbiolchem.2019.107155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 07/09/2019] [Accepted: 10/23/2019] [Indexed: 01/21/2023]
Abstract
Cisplatin ceases cell division and induces apoptosis in cancer cell lines. It is well established that cisplatin alters the expression of many genes involved in several cellular processes and pathways including transcription, p53 signaling pathway, and apoptosis. However, system-wide responses to cisplatin in breast cancer cell lines have not been studied. Therefore, we have used a network analysis approach to unveil such responses at early stages of drug treatment. To do this, we have first identified those genes that are responding to cisplatin treatment in MCF-7 cell line. Network and gene ontology analyses were then employed to uncover the molecular pathways affected by cisplatin treatment. Then the results obtained from cisplatin-treated MCF7 cell line were compared to those obtained from other cancer cell lines at comparable time points. In conclusion, we found that ADCY9, GSK3B, MAPK14, NCK1, NCOA2, PIK3CA, PIK3CB, PTK2, RHOB act as hub genes in the cisplatin-responsive regulatory network at the pro-apoptotic stages. The results could be useful in finding new drugs to target these genes in order to obtain similar responses.
Collapse
Affiliation(s)
- Hossein Fallahi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, 6714115111, Iran.
| | - Rasoul Godini
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, 3800, Australia
| |
Collapse
|
11
|
Di Meo F, Aversano R, Diretto G, Demurtas OC, Villano C, Cozzolino S, Filosa S, Carputo D, Crispi S. Anti-cancer activity of grape seed semi-polar extracts in human mesothelioma cell lines. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
12
|
Di Meo F, Filosa S, Madonna M, Giello G, Di Pardo A, Maglione V, Baldi A, Crispi S. Curcumin C3 complex®/Bioperine® has antineoplastic activity in mesothelioma: an in vitro and in vivo analysis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:360. [PMID: 31419989 PMCID: PMC6698046 DOI: 10.1186/s13046-019-1368-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/08/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND A major limitation in the treatment for malignant mesothelioma is related to serious side effects caused by chemotherapeutics and to the development of cancer-resistance. Advances in cancer therapies have been reached thanks to the introduction of alternative approaches, such as the use of phytochemicals. Curcumin-C3complex®/Bioperine® is a commercially standardized extract containing a ratio-defined mixture of three curcuminoids and piperine that greatly increase its bioavailability. Interestingly, the anticancer effect of this formulation has been described in different studies and several clinical trials have been started, but to our knowledge none refers to human mesothelioma. METHODS Curcumin-C3complex®/Bioperine® anticancer effect was evaluated in vitro in different human mesothelioma cell lines analysing cell proliferation, colony-forming assay, wound healing assays, invasion assay and FACS analysis. In vivo anticancer properties were analysed in a mesothelioma xenograft mouse model in CD1 Nude mice. RESULTS Curcumin-C3complex®/Bioperine® in vitro induced growth inhibition in all mesothelioma cell lines analysed in a dose- and time-depended manner and reduced self-renewal cell migration and cell invasive ability. Cell death was due to apoptosis. The analysis of the molecular signalling pathway suggested that intrinsic apoptotic pathway is activated by this treatment. This treatment in vivo delayed the growth of the ectopic tumours in a mesothelioma xenograft mouse model. CONCLUSIONS Curcumin-C3complex®/Bioperine® treatment strongly reduces in vitro tumorigenic properties of mesothelioma cells by impairing cellular self-renewal ability, proliferative cell rate and cell migration and delays tumor growth in xenograft mouse model by reducing angiogenesis and increasing apoptosis. Considering that curcumin in vivo synergizes drug effects, its administration to treatment regimen may help to enhance drug therapeutic efficacy in mesothelioma. Our results suggest that implementation of standard pharmacological therapies with novel compounds may pave the way to develop alternative approaches to mesothelioma.
Collapse
Affiliation(s)
- Francesco Di Meo
- Institute of Biosciences and BioResources-UOS Naples CNR, via P. Castellino, 111-, 80131, Naples, Italy.,Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo via Cinthia, Naples, Italy
| | - Stefania Filosa
- Institute of Biosciences and BioResources-UOS Naples CNR, via P. Castellino, 111-, 80131, Naples, Italy.,IRCCS Neuromed, Localitá Camerelle, Pozzilli, IS, Italy
| | | | - Gerarda Giello
- Institute of Biosciences and BioResources-UOS Naples CNR, via P. Castellino, 111-, 80131, Naples, Italy
| | - Alba Di Pardo
- IRCCS Neuromed, Localitá Camerelle, Pozzilli, IS, Italy
| | | | - Alfonso Baldi
- Institute of Biosciences and BioResources-UOS Naples CNR, via P. Castellino, 111-, 80131, Naples, Italy. .,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy.
| | - Stefania Crispi
- Institute of Biosciences and BioResources-UOS Naples CNR, via P. Castellino, 111-, 80131, Naples, Italy.
| |
Collapse
|
13
|
Boccellino M, Pinto F, Ieluzzi V, Giovane A, Quagliuolo L, Fariello C, Coppola M, Carlucci A, Santini M, Ferati K, Bexheti-Ferati A, Giordano A, Di Domenico M. Proteomics analysis of human serum of patients with non-small-cell lung cancer reveals proteins as diagnostic biomarker candidates. J Cell Physiol 2019; 234:23798-23806. [PMID: 31180588 DOI: 10.1002/jcp.28948] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
Non-small-cell lung carcinomas (NSCLC) is the most common type of lung cancer and it has a poor prognosis, because overall survival after 5 years is 20-25% for all stages. Thus, it is extremely important to increase the survival rate in the early stages NSCLC by focusing on novel screening tests of cancer identifying specific biomarkers expression associated with a more accurate tumor staging and patient prognosis. In this study, we focused our attention on quantitative proteomics of three heavily glycosylated serum proteins: AMBP, α2 macroglobulin, and SERPINA1. In particular, we analyzed serum samples from 20 NSCLC lung adenocarcinoma cancer patients in early and advanced stages, and 10 healthy donors to obtain a relative quantification through the MRM analysis of these proteins that have shown to be markers of cancer development and progression. AMBP, α2 macroglobulin, and SERPINA1 were chosen because all of them possess endopeptidase inhibitor activity and play key roles in cancer. We observe a variation in the expression of these proteins linked to the stage of the disease. Therefore, we believe that proteins like α2 macroglobulin, αmicroglobulin/bikunin, and SERPINA1 could be useful biomarkers for early detection of lung cancer and in monitoring its evolution.
Collapse
Affiliation(s)
| | - Federica Pinto
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Ieluzzi
- Department of Cardio-Respiratory Disease, Thoracic Surgery Unit, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - Alfonso Giovane
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucio Quagliuolo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Chiara Fariello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Coppola
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annalisa Carlucci
- Department of Cardio-Respiratory Disease, Thoracic Surgery Unit, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - Mario Santini
- Department of Cardio-Respiratory Disease, Thoracic Surgery Unit, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - Kenan Ferati
- Department of Dentistry, Faculty of Medicine, University of Tetovo, Tetovo, FYR of Macedonia
| | | | - Antonio Giordano
- Department of Medical Biotechnology, University of Siena, Siena, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Brown A, Kumar S, Tchounwou PB. Cisplatin-Based Chemotherapy of Human Cancers. JOURNAL OF CANCER SCIENCE & THERAPY 2019; 11:97. [PMID: 32148661 PMCID: PMC7059781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cisplatin (cis-diammine-dichloro-platinum II) was initially discovered to prevent the growth of Escherichia coli and was further recognized for its anti-neoplastic and cytotoxic effects on cancer cells. Administered intravenously to humans, cisplatin is used as first-line chemotherapy treatment for patients diagnosed with various types of malignancies, such as leukemia, lymphomas, breast, testicular, ovarian, head and neck, and cervical cancers, and sarcomas. Once cisplatin enters the cell it exerts its cytotoxic effect by losing one chloride ligand, binding to DNA to form intra-strand DNA adducts, and inhibiting DNA synthesis and cell growth. The DNA lesions formed from cisplatin-induced DNA damage activate DNA repair response via NER (nuclear excision repair system) by halting cisplatin-induced cell death by activation of ATM (ataxia telangiectasia mutated) pathway. Although treatment has been shown to be effective, many patients experience relapse due to drug resistance. As a result, other platinum compounds such as oxaliplatin and carboplatin have since been used and have shown some levels of effectiveness. In this review, the clinical applications of cisplatin are discussed with a special emphasis on its use in cancer chemotherapy.
Collapse
Affiliation(s)
| | | | - Paul B Tchounwou
- Corresponding author: Paul B Tchounwou, Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD-RCMI Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box18750, Jackson, Mississippi, MS 39217, USA, Tel: +601-979-0777; Fax: +601-979-0570;
| |
Collapse
|
15
|
Di Domenico M, Pinto F, Quagliuolo L, Contaldo M, Settembre G, Romano A, Coppola M, Ferati K, Bexheti-Ferati A, Sciarra A, Nicoletti GF, Ferraro GA, Boccellino M. The Role of Oxidative Stress and Hormones in Controlling Obesity. Front Endocrinol (Lausanne) 2019; 10:540. [PMID: 31456748 PMCID: PMC6701166 DOI: 10.3389/fendo.2019.00540] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
The accumulation of adipose tissue in the body occurs because the energy introduced with food and drink exceeds that expense, but to understand why this imbalance is established and why it is maintained over time, it is important to consider the main causes and risk factors of excess weight. In this review, we will refer to the main factors linked to obesity, starting from oxidative stress to hormonal factors including the role of obesity in breast cancer. Among the many hypotheses formulated on the etiopathology of obesity, a key role can be attributed to the relationship between stress oxidative and intestinal microbiota. Multiple evidences tend to show that genetic, epigenetic, and lifestyle factors contribute to determine in the obese an imbalance of the redox balance correlated with the alteration of the intestinal microbial flora. Obesity acts negatively on the wound healing, in fact several studies indicate morbid obesity significantly increased the risk of a post-operative wound complication and infection. Currently, in the treatment of obesity, medical interventions are aimed not only at modifying caloric intake, but also to modulate and improve the composition of diet with the aim of rebalancing the microbiota-redox state axis.
Collapse
Affiliation(s)
- Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Federica Pinto
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Lucio Quagliuolo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Contaldo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuliana Settembre
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Antonio Romano
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mario Coppola
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Kenan Ferati
- Faculty of Medicine, University of Tetovo, Tetovo, Macedonia
| | | | - Antonella Sciarra
- Department of Translational Medicad Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovanni Francesco Nicoletti
- Plastic Surgery Unit, Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppe Andrea Ferraro
- Plastic Surgery Unit, Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Naples, Italy
- *Correspondence: Giuseppe Andrea Ferraro
| | | |
Collapse
|
16
|
Abdo J, Wichman CS, Dietz NE, Ciborowski P, Fleegel J, Mittal SK, Agrawal DK. Discovery of Novel and Clinically Relevant Markers in Formalin-Fixed Paraffin-Embedded Esophageal Cancer Specimen. Front Oncol 2018; 8:157. [PMID: 29868478 PMCID: PMC5954028 DOI: 10.3389/fonc.2018.00157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/25/2018] [Indexed: 12/26/2022] Open
Abstract
Due to the ineffectiveness of chemoradiation and targeted therapy in esophageal anticancer care and the subsequent low survival rates, we constructed a high throughput method to discover and investigate new markers with prognostic, diagnostic, and therapeutic clinical utility. This was accomplished by developing a quick, inexpensive, and dependable platform to simultaneously quantify thousands of proteins which subsequently revealed novel markers involved in the pathogenesis of esophageal adenocarcinoma (EAC) via discovery mass spectrometry paired with conservative biostatistics. Our method uncovered a perfect storm of tumor suppressors being downregulated, proliferation markers ramped up, and chemoresistance markers overexpressed—many of which could serve as new therapy targets for EAC. The 12 markers discovered by this method are novel regarding their involvement in the pathogenesis of EAC. The molecular oncology arena now has a dozen new proteomic targets suitable for validation and elucidation of their clinical utility via gene knockdown in cellular and animal models. This new method can be replicated and applied to other cancers or disease states for research and development and discovery-based investigations. Our findings, which serve as a proof of concept, will hopefully motivate research groups to further expound on the molecular processes involved in the aggressiveness of EAC and other solid tumor diseases, ultimately leading to improved patient management strategies.
Collapse
Affiliation(s)
- Joe Abdo
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| | - Christopher S Wichman
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Nicholas E Dietz
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Department of Pathology, CHI Health Creighton University Medical Center, College of Medicine, Omaha, NE, United States
| | - Pawel Ciborowski
- Department of Pharmacology, University of Nebraska Medical Center, Omaha, NE, United States
| | - John Fleegel
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| | - Sumeet K Mittal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, United States
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
17
|
Satoh T, Tatsuta T, Sugawara S, Hara A, Hosono M. Synergistic anti-tumor effect of bullfrog sialic acid-binding lectin and pemetrexed in malignant mesothelioma. Oncotarget 2018; 8:42466-42477. [PMID: 28476017 PMCID: PMC5522080 DOI: 10.18632/oncotarget.17198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/06/2017] [Indexed: 12/26/2022] Open
Abstract
Malignant mesothelioma is an aggressive cancer with limited therapeutic options. Sialic acid-binding lectin isolated from Rana catesbeiana oocytes (cSBL) is a multifunctional protein with anti-cancer activity. The effects of pemetrexed, cisplatin, and cSBL were evaluated in mesothelioma and normal mesothelial cell lines. We evaluated cytotoxicity, apoptosis, caspase-3 cleavage and activation, cell proliferation, cell cycle arrest, and levels of cell cycle proteins in H28 cells treated with pemetrexed, cisplatin, and cSBL alone or in combination. Treatment with cSBL alone was cytotoxic to mesothelioma cells. The anti-cancer effect of cSBL was observed in a broader range of cell lines and exhibited greater cancer cell selectivity than pemetrexed or cisplatin. Combination treatment with pemetrexed + cSBL resulted in greater dose-dependent cytotoxicity than pemetrexed + cisplatin, the standard of care in mesothelioma. The synergistic effect of pemetrexed + cSBL was mediated by the cytostatic effect of pemetrexed and the cytotoxic effect of cSBL. It thus appears that cSBL has therapeutic potential for the treatment of mesothelioma.
Collapse
Affiliation(s)
- Toshiyuki Satoh
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Miyagi 981-8558, Japan.,Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Takeo Tatsuta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Shigeki Sugawara
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Akiyoshi Hara
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Masahiro Hosono
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Aobaku, Sendai, Miyagi 981-8558, Japan
| |
Collapse
|
18
|
Ghosh S, Mukhopadhyay S, Sarkar M, Mandal A, Das V, Kumar A, Giri B. Biological evaluation of a halogenated triterpenoid, 2α-bromo-dihydrobelulonic acid as inhibitor of human topoisomerase IIα and HeLa cell proliferation. Chem Biol Interact 2017; 268:68-76. [PMID: 28254521 DOI: 10.1016/j.cbi.2017.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 02/24/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND The pentacyclic lupane-type (6-6-6-6-5 type) triterpenoid, Betulinic acid (BA) is a potent inhibitor of topoisomerases and is of immense interest as anticancer drugs. However, the compound being highly lipophilic, has limited in vivo uptake capacity. BA derivatives with halogen substituent at C-2 have improved membrane permeability and cytotoxicity against cancer cells. AIM The halogenated triterpenoid, 2α-bromo-dihydrobetulonic acid (B1) was synthesized from betulinic acid (BA) isolated from Bischofia javanica. Aim of the study was to determine whether B1 could act as a more efficient inhibitor of Topo IIα activity and HeLa cell proliferation, in comparison to BA. RESULT B1 displayed efficient inhibition of DNA relaxation activity of topoisomerase IIα and the inhibitory effect was markedly improved upon pre-incubation of the compound with enzyme. Topoisomerase IIα inhibition by B1 was relieved in presence of increasing concentrations of DNA suggesting the compound as a reversible catalytic inhibitor. Subsequent UV and fluorescence spectroscopy studies indicated that B1 interacts and intercalates with DNA at concentrations signicantly greater than that required for topoisomerase IIα inhibition. The compound showed cytotoxic activity against HeLa cells with significantly lower IC50 value (7.5 μM) as compared to that of BA (30 μM) and had very low damaging/cytotoxic effect on normal cells. Treatment of B1 impaired HeLa cell proliferation by inducing Go-G1 arrest through lowered expression of cyclin D1 and PCNA polypeptides, and enhanced expression of p21. B1 treatment also increased the accumulation of early and late apoptotic cells in a concentration dependent manner as indicated by annexin V-FITC/PI binding assay.
Collapse
Affiliation(s)
- Shilpi Ghosh
- Department of Biotechnology, University of North Bengal, Darjeeling, 734 013, West Bengal, India.
| | - Swagata Mukhopadhyay
- Department of Biotechnology, University of North Bengal, Darjeeling, 734 013, West Bengal, India
| | - Mrinmoy Sarkar
- Experimental Medicine & Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata, 700 126, India
| | - Amitava Mandal
- Natural Products and Polymer Laboratory, Department of Chemistry, University of North Bengal, Darjeeling, 734 013, West Bengal, India; Molecular Complexicity Laboratory, Department of Chemistry, Raiganj University, Raiganj, 733134, West Bengal, India
| | - Vaskar Das
- Department of Biotechnology, University of North Bengal, Darjeeling, 734 013, West Bengal, India
| | - Anoop Kumar
- Department of Biotechnology, University of North Bengal, Darjeeling, 734 013, West Bengal, India
| | - Biplab Giri
- Experimental Medicine & Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata, 700 126, India; Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India.
| |
Collapse
|
19
|
Thanee M, Loilome W, Techasen A, Sugihara E, Okazaki S, Abe S, Ueda S, Masuko T, Namwat N, Khuntikeo N, Titapun A, Pairojkul C, Saya H, Yongvanit P. CD44 variant-dependent redox status regulation in liver fluke-associated cholangiocarcinoma: A target for cholangiocarcinoma treatment. Cancer Sci 2016; 107:991-1000. [PMID: 27176078 PMCID: PMC4946726 DOI: 10.1111/cas.12967] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/04/2016] [Accepted: 05/11/2016] [Indexed: 12/13/2022] Open
Abstract
Expression of CD44, especially the variant isoforms (CD44v) of this major cancer stem cell marker, contributes to reactive oxygen species (ROS) defense through stabilizing xCT (a cystine–glutamate transporter) and promoting glutathione synthesis. This enhances cancer development and increases chemotherapy resistance. We investigate the role of CD44v in the regulation of the ROS defense system in cholangiocarcinoma (CCA). Immunohistochemical staining of CD44v and p38MAPK (a major ROS target) expression in Opisthorchis viverrini‐induced hamster CCA tissues (at 60, 90, 120, and 180 days) reveals a decreased phospho‐p38MAPK signal, whereas the CD44v signal was increased during bile duct transformation. Patients with CCA showed CD44v overexpression and negative‐phospho‐p38MAPK patients a significantly shorter survival rate than the low CD44v signal and positive‐phospho‐p38MAPK patients (P = 0.030). Knockdown of CD44 showed that xCT and glutathione levels were decreased, leading to a high level of ROS. We examined xCT‐targeted CD44v cancer stem cell therapy using sulfasalazine. Glutathione decreased and ROS increased after the treatment, leading to inhibition of cell proliferation and induction of cell death. Thus, the accumulation of CD44v leads to the suppression of p38MAPK in transforming bile duct cells. The redox status regulation of CCA cells depends on the expression of CD44v to contribute the xCT function and is a link to the poor prognosis of patients. Thus, an xCT inhibitor could inhibit cell growth and activate cell death. This suggests that an xCT‐targeting drug may improve CCA therapy by sensitization to the available drug (e.g. gemcitabine) by blocking the mechanism of the cell's ROS defensive system.
Collapse
Affiliation(s)
- Malinee Thanee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Anchalee Techasen
- Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Eiji Sugihara
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Shogo Okazaki
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Shinya Abe
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Higashiosaka, Japan.,Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Shiho Ueda
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Higashiosaka, Japan
| | - Takashi Masuko
- Cell Biology Laboratory, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Higashiosaka, Japan
| | - Nisana Namwat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Narong Khuntikeo
- Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Attapol Titapun
- Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chawalit Pairojkul
- Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Puangrat Yongvanit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
20
|
Identification of Novel Pathways in Plant Lectin-Induced Cancer Cell Apoptosis. Int J Mol Sci 2016; 17:228. [PMID: 26867193 PMCID: PMC4783960 DOI: 10.3390/ijms17020228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 01/01/2023] Open
Abstract
Plant lectins have been investigated to elucidate their complicated mechanisms due to their remarkable anticancer activities. Although plant lectins seems promising as a potential anticancer agent for further preclinical and clinical uses, further research is still urgently needed and should include more focus on molecular mechanisms. Herein, a Naïve Bayesian model was developed to predict the protein-protein interaction (PPI), and thus construct the global human PPI network. Moreover, multiple sources of biological data, such as smallest shared biological process (SSBP), domain-domain interaction (DDI), gene co-expression profiles and cross-species interolog mapping were integrated to build the core apoptotic PPI network. In addition, we further modified it into a plant lectin-induced apoptotic cell death context. Then, we identified 22 apoptotic hub proteins in mesothelioma cells according to their different microarray expressions. Subsequently, we used combinational methods to predict microRNAs (miRNAs) which could negatively regulate the abovementioned hub proteins. Together, we demonstrated the ability of our Naïve Bayesian model-based network for identifying novel plant lectin-treated cancer cell apoptotic pathways. These findings may provide new clues concerning plant lectins as potential apoptotic inducers for cancer drug discovery.
Collapse
|
21
|
Rai N, Sarkar M, Raha S. Piroxicam, a traditional non-steroidal anti-inflammatory drug (NSAID) causes apoptosis by ROS mediated Akt activation. Pharmacol Rep 2015; 67:1215-23. [DOI: 10.1016/j.pharep.2015.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/31/2015] [Accepted: 05/14/2015] [Indexed: 01/10/2023]
|
22
|
Zang X, Chen M, Zhou Y, Xiao G, Xie Y, Wang X. Identifying CDKN3 Gene Expression as a Prognostic Biomarker in Lung Adenocarcinoma via Meta-analysis. Cancer Inform 2015; 14:183-91. [PMID: 26052221 PMCID: PMC4444140 DOI: 10.4137/cin.s17287] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is among the major causes of cancer deaths, and the survival rate of lung cancer patients is extremely low. Recent studies have demonstrated that the gene CDKN3 is related to neoplasia, but in the literature severe controversy exists over whether it is involved in cancer progression or, conversely, tumor inhibition. In this study, we investigated the expression of CDKN3 and its association with prognosis in lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC) using datasets in Lung Cancer Explorer (LCE; http://qbrc.swmed.edu/lce/). We found that CDKN3 was up-regulated in ADC and SCC compared to normal tissues. We also found that CDKN3 was expressed at a higher level in SCC than in ADC, which was further validated through meta-analysis (coefficient = 2.09, 95% CI = 1.50-2.67, P < 0.0001). In addition, based on meta-analysis for the prognostic value of CDKN3, we found that higher CDKN3 expression was associated with poorer survival outcomes in ADC (HR = 1.65, 95% CI = 1.39-1.96, P < 0.0001), but not in SCC (HR = 1.10, 95% CI = 0.84-1.44, P = 0.494). Our findings indicate that CDKN3 may be a prognostic marker in ADC, though the detailed mechanism is yet to be revealed.
Collapse
Affiliation(s)
- Xiao Zang
- Quantitative Biomedical Research Center, Department of Clinical Sciences
| | - Min Chen
- Department of Mathematical Sciences, University of Texas at Dallas
| | - Yunyun Zhou
- Quantitative Biomedical Research Center, Department of Clinical Sciences
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Department of Clinical Sciences
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
| | - Yang Xie
- Quantitative Biomedical Research Center, Department of Clinical Sciences
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
| | - Xinlei Wang
- Department of Statistics, Southern Methodist University
| |
Collapse
|
23
|
Menale C, Piccolo MT, Favicchia I, Aruta MG, Baldi A, Nicolucci C, Barba V, Mita DG, Crispi S, Diano N. Efficacy of Piroxicam Plus Cisplatin-Loaded PLGA Nanoparticles in Inducing Apoptosis in Mesothelioma Cells. Pharm Res 2014; 32:362-74. [DOI: 10.1007/s11095-014-1467-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/24/2014] [Indexed: 10/24/2022]
|