1
|
The In Vitro Effect of Psoralen on Glioma Based on Network Pharmacology and Potential Target Research. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1952891. [PMID: 36065261 PMCID: PMC9440786 DOI: 10.1155/2022/1952891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022]
Abstract
Glioma is an aggressive tumor, currently there is no satisfactory management available. Psoralen, as a natural product, has been found to have an effect of treating cancer in recent years, but its effect on glioma has not been explored. In this study, we investigated the in vitro inhibition effect and potential targets of psoralen on glioma through network pharmacology and in vitro glioma treatment experiments. First, we used network pharmacology to preliminarily predict the 21 core genes of psoralen in the treatment of glioma, including PIK3CA, PIK3CB, PIK3CG, and JAK2. The CCK-8 method was used to detect the effect of psoralen on the proliferation of glioma U87 and U251 cells, and the results showed that psoralen could significantly inhibit the proliferation of U87 and U251 cells. The flow cytometry was used to detect the apoptosis and cell cycle changes, and it was found that psoralen could significantly promote the early apoptosis of U87 and U251 cells and had a significant cycle arrest effect on the two cells. The cell scratch test showed that psoralen could significantly inhibit the migration of U87 and U251 cells. The relative expression levels of PIK3CA, PIK3CB, PIK3CG, and JAK2 were analyzed by Real-time Quantitative polymerase chain reaction (QT-PCR), and the results showed that psoralen could inhibit the gene expression of PIK3CA, PIK3CB, PIK3CG, and JAK2. Later, Western blotting (WB) experiments showed that psoralen could inhibit the protein expressions of PI3K and JAK2. This study has preliminarily explored and verified the antiglioma effect of psoralen in the form of inhibiting cell proliferation and migration, promoting cell apoptosis and organizing cell cycle in vitro. And may play a role by inhibiting the expression of PIK3CA, PIK3CB, PIK3CG, JAK2 gene and PI3K, JAK2 protein, psoralen has become a potential antiglioma drug.
Collapse
|
2
|
Wu A, Lu J, Zhong G, Lu L, Qu Y, Zhang C. Xanthotoxin (8-methoxypsoralen): A review of its chemistry, pharmacology, pharmacokinetics, and toxicity. Phytother Res 2022; 36:3805-3832. [PMID: 35913174 DOI: 10.1002/ptr.7577] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/30/2022] [Accepted: 07/14/2022] [Indexed: 11/11/2022]
Abstract
Xanthotoxin (XAT) is a natural furanocoumarins, a bioactive psoralen isolated from the fruit of the Rutaceae plant Pepper, which has received increasing attention in recent years due to its wide source and low cost. By collecting and compiling literature on XAT, the results show that XAT exhibits significant activity in the treatment of various diseases, including neuroprotection, skin repair, osteoprotection, organ protection, anticancer, antiinflammatory, antioxidative stress and antibacterial. In this paper, we review the pharmacological activity and potential molecular mechanisms of XAT for the treatment of related diseases. The data suggest that XAT can mechanistically induce ROS production and promote apoptosis through mitochondrial or endoplasmic reticulum pathways, regulate NF-κB, MAPK, JAK/STAT, Nrf2/HO-1, MAPK, AKT/mTOR, and ERK1/2 signaling pathways to exert pharmacological effects. In addition, the pharmacokinetics properties and toxicity of XAT are discussed in this paper, further elucidating the relationship between structure and efficacy. It is worth noting that data from clinical studies of XAT are still scarce, limiting the use of XAT in the clinic, and in the future, more in-depth studies are needed to determine the clinical efficacy of XAT.
Collapse
Affiliation(s)
- Anxin Wu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Jing Lu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Guofeng Zhong
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ling Lu
- Chengdu University of Technology, Chengdu, PR China
| | - Yan Qu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Chen Zhang
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| |
Collapse
|
3
|
da Silva SVS, Barboza OM, Souza JT, Soares ÉN, dos Santos CC, Pacheco LV, Santos IP, Magalhães TBDS, Soares MBP, Guimarães ET, Meira CS, Costa SL, da Silva VDA, de Santana LLB, de Freitas Santos Júnior A. Structural Design, Synthesis and Antioxidant, Antileishmania, Anti-Inflammatory and Anticancer Activities of a Novel Quercetin Acetylated Derivative. Molecules 2021; 26:molecules26226923. [PMID: 34834016 PMCID: PMC8623808 DOI: 10.3390/molecules26226923] [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: 10/19/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 02/01/2023] Open
Abstract
Quercetin (Q) is a bioflavonoid with biological potential; however, poor solubility in water, extensive enzymatic metabolism and a reduced bioavailability limit its biopharmacological use. The aim of this study was to perform structural modification in Q by acetylation, thus, obtaining the quercetin pentaacetate (Q5) analogue, in order to investigate the biological potentials (antioxidant, antileishmania, anti-inflammatory and cytotoxicity activities) in cell cultures. Q5 was characterized by FTIR, 1H and 13C NMR spectra. The antioxidant potential was evaluated against the radical ABTS•+. The anti-inflammatory potential was evaluated by measuring the pro-inflammatory cytokine tumor necrosis factor (TNF) and the production of nitric oxide (NO) in peritoneal macrophages from BALB/c mice. Cytotoxicity tests were performed using the AlamarBlue method in cancer cells HepG2 (human hepatocarcinoma), HL-60 (promyelocytic leukemia) and MCR-5 (healthy human lung fibroblasts) as well as the MTT method for C6 cell cultures (rat glioma). Q and Q5 showed antioxidant activity of 29% and 18%, respectively, which is justified by the replacement of hydroxyls by acetyl groups. Q and Q5 showed concentration-dependent reductions in NO and TNF production (p < 0.05); Q and Q5 showed higher activity at concentrations > 40µM when compared to dexamethasone (20 µM). For the HL-60 lineage, Q5 demonstrated selectivity, inducing death in cancer cells, when compared to the healthy cell line MRC-5 (IC50 > 80 µM). Finally, the cytotoxic superiority of Q5 was verified (IC50 = 11 µM), which, at 50 µM for 24 h, induced changes in the morphology of C6 glioma cells characterized by a round body shape (not yet reported in the literature). The analogue Q5 had potential biological effects and may be promising for further investigations against other cell cultures, particularly neural ones.
Collapse
Affiliation(s)
- Saul Vislei Simões da Silva
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
| | - Orlando Maia Barboza
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
| | - Jéssica Teles Souza
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia, Salvador 40231-300, BA, Brazil; (J.T.S.); (É.N.S.); (C.C.d.S.); (S.L.C.); (V.D.A.d.S.)
| | - Érica Novaes Soares
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia, Salvador 40231-300, BA, Brazil; (J.T.S.); (É.N.S.); (C.C.d.S.); (S.L.C.); (V.D.A.d.S.)
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia, Salvador 40231-300, BA, Brazil; (J.T.S.); (É.N.S.); (C.C.d.S.); (S.L.C.); (V.D.A.d.S.)
| | - Luciano Vasconcellos Pacheco
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
- Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, BA, Brazil; (I.P.S.); (M.B.P.S.)
| | | | - Tatiana Barbosa dos Santos Magalhães
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, BA, Brazil; (I.P.S.); (M.B.P.S.)
- SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, BA, Brazil
| | - Elisalva Teixeira Guimarães
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
- Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, BA, Brazil; (I.P.S.); (M.B.P.S.)
| | - Cássio Santana Meira
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
- Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, BA, Brazil; (I.P.S.); (M.B.P.S.)
- SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, BA, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia, Salvador 40231-300, BA, Brazil; (J.T.S.); (É.N.S.); (C.C.d.S.); (S.L.C.); (V.D.A.d.S.)
| | - Victor Diógenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia, Salvador 40231-300, BA, Brazil; (J.T.S.); (É.N.S.); (C.C.d.S.); (S.L.C.); (V.D.A.d.S.)
| | - Lourenço Luís Botelho de Santana
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
| | - Aníbal de Freitas Santos Júnior
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil; (S.V.S.d.S.); (O.M.B.); (L.V.P.); (T.B.d.S.M.); (E.T.G.); (C.S.M.); (L.L.B.d.S.)
- Correspondence: or ; Tel.: +55-71-3117-5313
| |
Collapse
|
4
|
Li J, Zhang Z, Qiu J, Huang X. 8-Methoxypsoralen has Anti-inflammatory and Antioxidant Roles in Osteoarthritis Through SIRT1/NF-κB Pathway. Front Pharmacol 2021; 12:692424. [PMID: 34552480 PMCID: PMC8450503 DOI: 10.3389/fphar.2021.692424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is mainly manifested by joint pain, stiffness and mobility disorder, which is the main cause of pain and disability in middle-aged and elderly people. In this study, we aimed to explore the role and mechanism of 8-Methoxypsoralen (8-MOP) in the OA model both in vitro and in vivo. The rat chondrocytes were treated with IL-1β, and the proliferation, apoptosis, inflammatory reactions and oxidative stress responses were determined after treatment with different concentrations of 8-MOP. Real-time quantitative polymerase chain reaction (qRT-PCR) and/or Western blot were implemented to check the AMPK/SIRT1/NF-κB expression in chondrocytes. The NF-κB activity was determined by dual luciferase experiment. The pain threshold of OA rat model dealt with 8-MOP and/or the SIRT1 inhibitor EX527 was measured. Our results revealed that 8-MOP evidently reduced IL-1β-mediated apoptosis and inhibition of proliferation, and mitigated the expression of inflammatory cytokines and oxidative stress factors in chondrocytes. Additionally, 8-MOP promoted phosphorylated level of AMPKα, enhanced SIRT1 expression and inhibited the phosphorylation of NF-κB. After treatment with EX527, 8-MOP-mediated protective effects on chondrocytes were mostly reversed. In vivo, 8-MOP obviously improved the pain threshold in the OA rat model and reduced the injury and apoptosis of chondrocytes in the joints. In addition, 8-MOP relieved inflammatory and oxidative stress responses in the articular cartilage via enhancing SIRT1 and repressing NF-κB activation. After the treatment with EX527, the 8-MOP-mediated protective effects were distinctly weakened. In summary, our study testified that 8-MOP alleviates pain, inflammatory and oxidative stress responses in OA rats through the SIRT1/NF-κB pathway, which is expected to become a new reagent for clinical treatment of OA.
Collapse
Affiliation(s)
- Jichao Li
- The Third Department of Knee Injury, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Zeng Zhang
- Zhengzhou Orthopedic Hospital, Zhengzhou, China
| | - Jinan Qiu
- The Third Department of Knee Injury, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Xiaohan Huang
- The Third Department of Knee Injury, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| |
Collapse
|
5
|
Carvalho FV, Fonseca Santana L, Diogenes A da Silva V, Costa SL, Zambotti-Villelae L, Colepicolo P, Ferraz CG, Ribeiro PR. Combination of a multiplatform metabolite profiling approach and chemometrics as a powerful strategy to identify bioactive metabolites in Lepidium meyenii (Peruvian maca). Food Chem 2021; 364:130453. [PMID: 34186480 DOI: 10.1016/j.foodchem.2021.130453] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/28/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023]
Abstract
Lepidium meyenii is an edible plant that has been used as a nutritional supplement worldwide due to its medicinal properties. However, most of the studies have focused on the pharmacological activities of the extracts rather than their chemical composition. Herein, we used a combination of a multiplatform metabolite profiling approach and chemometrics to identify bioactive metabolites in L. meyenii. Extracts obtained with ethyl acetate and ethanol showed the promising antioxidant, anti-glioma and antibacterial activities. Sixty metabolites were identified by HPLC-MS, whereas fifteen were identified by GC-MS. Partial least squares discriminant analysis (PLS-DA), hierarchical cluster analysis (HCA), and Variable Importance in Projection (VIP) successfully discriminated extracts obtained in different organic solvents from in natura dry roots and commercial product samples of L. meyenii. Additionally, correlation analysis allowed us to pinpoint potential candidates responsible for each biological activity tested for the extracts, which could be extrapolate for other food-related species.
Collapse
Affiliation(s)
- Fernanda V Carvalho
- Metabolomics Research Group, Instituto de Química, Universidade Federal da Bahia, Rua Barão de Jeremoabo s/n, 40170-115 Salvador, Brazil
| | - Lucia Fonseca Santana
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Victor Diogenes A da Silva
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Silvia L Costa
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Leonardo Zambotti-Villelae
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-000 São Paulo, Brazil
| | - Pio Colepicolo
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, 05508-000 São Paulo, Brazil
| | - Caline G Ferraz
- Metabolomics Research Group, Instituto de Química, Universidade Federal da Bahia, Rua Barão de Jeremoabo s/n, 40170-115 Salvador, Brazil
| | - Paulo R Ribeiro
- Metabolomics Research Group, Instituto de Química, Universidade Federal da Bahia, Rua Barão de Jeremoabo s/n, 40170-115 Salvador, Brazil.
| |
Collapse
|
6
|
Zhang T, Rao J, Li W, Wang K, Qiu F. Mechanism-based inactivation of cytochrome P450 enzymes by natural products based on metabolic activation. Drug Metab Rev 2020; 52:501-530. [PMID: 33043714 DOI: 10.1080/03602532.2020.1828910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cytochrome P450 enzymes (P450 enzymes) are the most common and important phase I metabolic enzymes and are responsible for the majority of the metabolism of clinical drugs and other xenobiotics. Drug-drug interactions (DDIs) can occur when the activities of P450 enzymes are inhibited. In particular, irreversible inhibition of P450 enzymes may lead to severe adverse interactions, compared to reversible inhibition. Many natural products have been shown to be irreversible inhibitors of P450 enzymes. The risks for intake of naturally occurring irreversible P450 enzyme inhibitors have been rising due to the rapid growth of the global consumption of natural products. Irreversible inhibition is usually called mechanism-based inactivation, which is time-, concentration- and NADPH- dependent. Generally, the formation of electrophilic intermediates is fundamental for the inactivation of P450 enzymes. This review comprehensively classifies natural P450 enzyme inactivators, including terpenoids, phenylpropanoids, flavonoids, alkaloids, and quinones obtained from herbs or foods. Moreover, the structure - activity correlations according to the IC50 (or Ki) values reported in the literature as well as the underlying mechanisms based on metabolic activation are highlighted in depth.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Jinqiu Rao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Wei Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | - Kai Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| |
Collapse
|
7
|
Pan J, Chen T, Ma K, Wang S, Yang C, Cui G. A negative feedback loop of H19/miR‐675/VDR mediates therapeutic effect of cucurmin in the treatment of glioma. J Cell Physiol 2019; 235:2171-2182. [PMID: 31468534 DOI: 10.1002/jcp.29127] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Jie‐Xiang Pan
- Department of Neurosurgery The First Hospital Affiliated to Army Medical University (Southwest Hospital) Chongqing China
| | - Tu‐Nan Chen
- Department of Neurosurgery The First Hospital Affiliated to Army Medical University (Southwest Hospital) Chongqing China
| | - Kang Ma
- Department of Neurosurgery The First Hospital Affiliated to Army Medical University (Southwest Hospital) Chongqing China
| | - Shi Wang
- Department of Neurosurgery The First Hospital Affiliated to Army Medical University (Southwest Hospital) Chongqing China
| | - Chuan‐Yan Yang
- Department of Neurosurgery The First Hospital Affiliated to Army Medical University (Southwest Hospital) Chongqing China
| | - Gao‐Yu Cui
- Department of Neurosurgery The First Hospital Affiliated to Army Medical University (Southwest Hospital) Chongqing China
| |
Collapse
|
8
|
Long-term exposure to MST-312 leads to telomerase reverse transcriptase overexpression in MCF-7 breast cancer cells. Anticancer Drugs 2017; 28:750-756. [PMID: 28520570 DOI: 10.1097/cad.0000000000000508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Telomerase is an enzyme responsible for telomere maintenance in almost all human cancer cells, but generally not expressed in somatic ones. Therefore, antitelomerase therapy is a potentially revolutionary therapeutic strategy, and the antitumor activity of telomerase inhibitors (TI) has been studied extensively recently, mainly for breast cancer. However, the effects expected from treatment with TI will appear only after many cell divisions, but the effects of this long-term approach are unknown. In this work, the consequences of 3120 h exposure of human breast cancer cells to TI MST-312 were investigated. MCF-7 cells were treated with MST-312 at a subtoxic concentration for a long time, and then cell morphology, viability, senescence, and proliferation were analyzed by phase-contrast microscopy, MTT assay, β-galactosidase test, and the trypan blue exclusion assay, respectively. Also, chromosomal stability was evaluated by classical cytogenetic analysis. The average length of telomeres and telomerase reverse transcriptase expression were accessed by real-time PCR and real-time RT-PCR, respectively. The MST-312 showed cytotoxic action and promoted telomere erosion, senescence, and chromosome aberrations, as expected, but in a small proportion. Nevertheless, the proliferation rate of the culture was not affected. As the main effect, the chronic exposure led to cell adaptation by overexpression of telomerase in response to the inhibitor, which is a potential cause of therapeutic failure and may be associated with a poor prognosis. In conclusion, despite the high therapeutic potential of TIs such as MST-312, the molecular outcomes of long-term exposure of tumors on these drugs have to be evaluated when considering their clinical application, especially for breast cancer treatment.
Collapse
|
9
|
Cao Z, Bai Y, Liu C, Dou C, Li J, Xiang J, Zhao C, Xie Z, Xiang Q, Dong S. Hypertrophic differentiation of mesenchymal stem cells is suppressed by xanthotoxin via the p38‑MAPK/HDAC4 pathway. Mol Med Rep 2017; 16:2740-2746. [PMID: 28677757 PMCID: PMC5548016 DOI: 10.3892/mmr.2017.6886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 06/08/2017] [Indexed: 12/20/2022] Open
Abstract
Chondrocyte hypertrophy is a physiological process in endochondral ossification. However, the hypertrophic-like alterations of chondrocytes at the articular surface may result in osteoarthritis (OA). In addition, the generation of fibrocartilage with a decreased biological function in tissue engineered cartilage, has been attributed to chondrocyte hypertrophy. Therefore, suppressing chondrocyte hypertrophy in OA and the associated regeneration of non-active cartilage is of primary concern. The present study examined the effects of xanthotoxin (XAT), which is classified as a furanocoumarin, on chondrocyte hypertrophic differentiation of mesenchymal stem cells. Following XAT treatment, the expression levels of genes associated with chondrocyte hypertrophy were detected via immunohistochemistry, western blotting and reverse transcription-quantitative polymerase chain reaction. The results revealed that XAT inhibited the expression of various chondrocyte hypertrophic markers, including runt related transcription factor 2 (Runx2), matrix metalloproteinase 13 and collagen type X α1 chain. Further exploration indicated that XAT reduced the activation of p38-mitogen activated protein kinase and then increased the expression of histone deacetylase 4 to suppress Runx2. The findings indicated that XAT maintained the chondrocyte phenotype in regenerated cartilage and therefore may exhibit promise as a potential drug for the treatment of OA in the future.
Collapse
Affiliation(s)
- Zhen Cao
- Department of Anatomy, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yun Bai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Chuan Liu
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ce Dou
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jianmei Li
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Junyu Xiang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Chunrong Zhao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhao Xie
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Qiang Xiang
- Department of Emergency, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|