1
|
Luciano M, Sieberer H, Krenn PW, Dang HH, Vetter J, Neuper T, Amend D, Blöchl C, Weichenberger CX, Eglseer A, Unger MS, Andosch A, Steiner P, Neureiter D, Bauer R, Hummer L, Tesanovic S, Binder S, Elmer DP, Strandt H, Schaller S, Strunk D, Pleyer L, Greil R, Winkler S, Hartmann TN, Schmidt-Arras D, Huber CG, Aberger F, Horejs-Hoeck J. Targeting NLRP3 inhibits AML progression by inducing PERK/eIF2-mediated apoptosis. Cell Commun Signal 2024; 22:424. [PMID: 39223663 PMCID: PMC11367831 DOI: 10.1186/s12964-024-01777-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is characterized by the abnormal proliferation of myeloid precursor cells and presents significant challenges in treatment due to its heterogeneity. Recently, the NLRP3 inflammasome has emerged as a potential contributor to AML pathogenesis, although its precise mechanisms remain poorly understood. METHODS Public genome datasets were utilized to evaluate the expression of NLRP3 inflammasome-related genes (IL-1β, IL-18, ASC, and NLRP3) in AML patients compared to healthy individuals. CRISPR/Cas9 technology was employed to generate NLRP3-deficient MOLM-13 AML cells, followed by comprehensive characterization using real-time PCR, western blotting, FACS analysis, and transmission electron and immunofluorescence microscopy. Proteomic analyses were conducted to identify NLRP3-dependent alterations in protein levels, with a focus on the eIF2 kinase PERK-mediated signaling pathways. Additionally, in vivo studies were performed using a leukemic mouse model to elucidate the pathogenic role of NLRP3 in AML. RESULTS Elevated expression of NLRP3 was significantly associated with diminished overall survival in AML patients. Genetic deletion, pharmacological inhibition and silencing by RNA interference of NLRP3 led to decreased AML cell survival through the induction of apoptosis. Proteomic analyses uncovered NLRP3-dependent alterations in protein translation, characterized by enhanced eIF2α phosphorylation in NLRP3-deficient AML cells. Moreover, inhibition of PERK-mediated eIF2α phosphorylation reduced apoptosis by downregulating pro-apoptotic Bcl-2 family members. In vivo studies demonstrated reduced leukemic burden in mice engrafted with NLRP3 knockout AML cells, as evidenced by alleviated leukemic symptoms. CONCLUSION Our findings elucidate the involvement of the NLRP3/PERK/eIF2 axis as a novel driver of AML cell survival. Targeting NLRP3-induced signaling pathways, particularly through the PERK/eIF2 axis, presents a promising therapeutic strategy for AML intervention. These insights into the role of the NLRP3 inflammasome offer potential avenues for improving the prognosis and treatment outcomes of AML patients.
Collapse
Affiliation(s)
- Michela Luciano
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Helene Sieberer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Peter W Krenn
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Hieu-Hoa Dang
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Julia Vetter
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg Campus, Hagenberg, 4232, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Diana Amend
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Constantin Blöchl
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | | | - Anna Eglseer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Michael S Unger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Ancuela Andosch
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Philip Steiner
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, Linz, 4020, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
- Institute of Pathology, Paracelsus Medical University (PMU), University Hospital Salzburg (SALK), Salzburg, 5020, Austria
| | - Renate Bauer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Laura Hummer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Suzana Tesanovic
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Stephanie Binder
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Dominik P Elmer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Helen Strandt
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Susanne Schaller
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg Campus, Hagenberg, 4232, Austria
| | - Dirk Strunk
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, 5020, Austria
| | - Lisa Pleyer
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
- Salzburg Cancer Research Institute (SCRI)-LIMCR, Salzburg, 5020, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University (PMU), University Hospital Salzburg (SALK), Salzburg, 5020, Austria
| | - Richard Greil
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
- Salzburg Cancer Research Institute (SCRI)-LIMCR, Salzburg, 5020, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University (PMU), University Hospital Salzburg (SALK), Salzburg, 5020, Austria
| | - Stephan Winkler
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg Campus, Hagenberg, 4232, Austria
| | - Tanja N Hartmann
- Department of Medicine I, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Dirk Schmidt-Arras
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Christian G Huber
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria.
- Cancer Cluster Salzburg, Salzburg, 5020, Austria.
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria.
| |
Collapse
|
2
|
Chen YH, Wu JX, Yang SF, Wu YC, Hsiao YH. Molecular Mechanisms Underlying the Anticancer Properties of Pitavastatin against Cervical Cancer Cells. Int J Mol Sci 2024; 25:7915. [PMID: 39063157 PMCID: PMC11277542 DOI: 10.3390/ijms25147915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Cervical cancer ranks as the fourth most prevalent form of cancer and is a significant contributor to female mortality on a global scale. Pitavastatin is an anti-hyperlipidemic medication and has been demonstrated to exert anticancer and anti-inflammatory effects. Thus, the purpose of this study was to evaluate the anticancer effect of pitavastatin on cervical cancer and the underlying molecular mechanisms involved. The results showed that pitavastatin significantly inhibited cell viability by targeting cell-cycle arrest and apoptosis in Ca Ski, HeLa and C-33 A cells. Pitavastatin caused sub-G1- and G0/G1-phase arrest in Ca Ski and HeLa cells and sub-G1- and G2/M-phase arrest in C-33 A cells. Moreover, pitavastatin induced apoptosis via the activation of poly-ADP-ribose polymerase (PARP), Bax and cleaved caspase 3; inactivated the expression of Bcl-2; and increased mitochondrial membrane depolarization. Furthermore, pitavastatin induced apoptosis and slowed the migration of all three cervical cell lines, mediated by the PI3K/AKT and MAPK (JNK, p38 and ERK1/2) pathways. Pitavastatin markedly inhibited tumor growth in vivo in a cancer cell-originated xenograft mouse model. Overall, our results identified pitavastatin as an anticancer agent for cervical cancer, which might be expanded to clinical use in the future.
Collapse
Affiliation(s)
- Ya-Hui Chen
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan; (Y.-H.C.); (J.-X.W.)
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Jyun-Xue Wu
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan; (Y.-H.C.); (J.-X.W.)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Yun-Chia Wu
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Yi-Hsuan Hsiao
- Women’s Health Research Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan; (Y.-H.C.); (J.-X.W.)
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
3
|
Hu H, Xu W, Li Y, Wang Z, Wang S, Liu Y, Bai M, Lou Y, Yang Q. SIRT1 regulates endoplasmic reticulum stress-related organ damage. Acta Histochem 2024; 126:152134. [PMID: 38237370 DOI: 10.1016/j.acthis.2024.152134] [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: 09/13/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 02/07/2024]
Abstract
Endoplasmic reticulum (ER) stress plays a key role in the pathogenesis of several organ damages. Studies show that excessive ER stress (ERS) can destroy cellular homeostasis, causing cell damage and physiological dysfunction in various organs. In recent years, Sirtuin1 (SIRT1) has become a research hotspot on ERS. Increasing evidence suggests that SIRT1 plays a positive role in various ERS-induced organ damage via multiple mechanisms, including inhibiting cellular apoptosis and promoting autophagy. SIRT1 can also alleviate liver, heart, lung, kidney, and intestinal damage by inhibiting ERS. We discuss the possible mechanism of SIRT1, explore potential therapeutic targets of diseases, and provide a theoretical basis for treating ERS-related diseases.
Collapse
Affiliation(s)
- He Hu
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Weichao Xu
- Department of Gastroenterology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Yan Li
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhicheng Wang
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Siyue Wang
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yansheng Liu
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Minan Bai
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yingying Lou
- Department of Gastroenterology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China.
| | - Qian Yang
- Department of Gastroenterology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
4
|
Mahendran R, Selvaraj SP, Dhanapal AR, Sarasa SB, Mathias BM, Thankappan B, Femil Selta DR, Naveen P, Poorani R, Sundhar N, Pillai MM, Selvakumar R, Huang CY, Eswaran R, Angayarkanni J. Tetrahydrobiopterin from cyanide-degrading bacterium Bacillus pumilus strain SVD06 induces apoptosis in human lung adenocarcinoma cell (A549). Biotechnol Appl Biochem 2023; 70:2052-2068. [PMID: 37731306 DOI: 10.1002/bab.2509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023]
Abstract
Tetrahydrobiopterin (BH4) is an essential biological cofactor and a derivative of pterin which is considered potent anticancer agents. In continuation of our previous study on the identification of BH4 from cyanide-degrading Bacillus pumilus, the present study focuses on evaluating the anticancer properties of BH4 on A549, a human lung adenocarcinoma. Anticancer activity analysis shows that BH4 inhibited A549 cell growth after 24 h of incubation with 0.02 mg/mL. In acridine orange/ethidium bromide staining, BH4-treated A549 cells showed apoptotic morphology. BH4 treatment caused cell cycle arrest at G0/G1 phase compared to control cells. BH4 augmented p53 expression in alveolar cancer cells by downregulating MDM2 levels. There was downregulation of casp-3 and upregulation of iNOS gene in BH4-treated A549 cells. Further, docking studies indicated that BH4 had significant interactions with the above proteins affirming the apoptosis mechanism. Thus, BH4 could be considered a potential anticancer drug.
Collapse
Affiliation(s)
- Ramasamy Mahendran
- Cancer Therapeutics Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Sanjay Prasad Selvaraj
- Molecular and Biological Agricultural Science Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Anand Raj Dhanapal
- Institute of Forest Genetics and Tree Breeding (IFGTB), Forest Campus, Coimbatore, Tamil Nadu, India
| | - Sabna Bhaskaran Sarasa
- Cancer Therapeutics Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Beutline Malgija Mathias
- Computational Science Laboratory, MCC-MRF Innovation Park, Madras Christian College, Chennai, Tamil Nadu, India
| | - Bency Thankappan
- Cancer Therapeutics Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Daniel Raja Femil Selta
- Department of Biochemistry and Cancer Research Center, FASCM, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Palanivel Naveen
- Department of Chemistry, Sona College of Arts and Science, Salem, Tamil Nadu, India
| | - Rhenghachar Poorani
- Gayatri Vidya parishad Institute of Health Care and Medical Technology, Visakhapatnam, India
| | - Navaneethan Sundhar
- Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan
| | - Mamatha M Pillai
- Tissue Engineering Laboratory, PSG Institute of Advanced Studies, Coimbatore, Tamil Nadu, India
| | - Rajendran Selvakumar
- Tissue Engineering Laboratory, PSG Institute of Advanced Studies, Coimbatore, Tamil Nadu, India
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Raju Eswaran
- Department of Zoology, The Madura College, Madurai, Tamil Nadu, India
| | - Jayaraman Angayarkanni
- Cancer Therapeutics Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
5
|
Maruyama R, Sugiyama T. ER Stress Decreases Gene Expression Of Transmembrane Protein 117 Via Activation of PKR-like ER Kinase. Cell Biochem Biophys 2023; 81:459-468. [PMID: 37421592 DOI: 10.1007/s12013-023-01150-3] [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] [Accepted: 06/25/2023] [Indexed: 07/10/2023]
Abstract
Stress response is an inherent mechanism in the endoplasmic reticulum (ER). The inducers of ER cause a specific cascade of reactions, leading to gene expression. Transmembrane protein 117 (TMEM117) is in the ER and plasma membrane. In our previous study, TMEM117 protein expression was found to be decreased by an ER stress inducer. However, the mechanism underlying this decrease in TMEM117 protein expression remains unclear. This study aimed to elucidate the mechanism underlying the decrease in TMEM117 protein expression during ER stress and identify the unfolded protein response (UPR) pathway related to decreased TMEM117 protein expression. We showed that the gene expression levels of TMEM117 were decreased by ER stress inducers and were regulated by PKR-like ER kinase (PERK), indicating that TMEM117 protein expression was regulated by the signaling pathway. Surprisingly, gene knockdown of activating transcription factor 4 (ATF4) downstream of PERK did not affect the gene expression of TMEM117. These results suggest that TMEM117 protein expression during ER stress is transcriptionally regulated by PERK but not by ATF4. TMEM117 has a potential to be a new therapeutic target against ER stress-related diseases.
Collapse
Affiliation(s)
- Ryuto Maruyama
- Graduate School of Bionics, Tokyo University of Technology, 1401-1 Katakura-machi, Hachioji, Tokyo, 192-0982, Japan.
- Department of Life Science, Rikkyo University, 3-34-1 Nishi-Ikebukuro, Toshima-ku, Tokyo, 171-8501, Japan.
| | - Tomoyasu Sugiyama
- Graduate School of Bionics, Tokyo University of Technology, 1401-1 Katakura-machi, Hachioji, Tokyo, 192-0982, Japan
| |
Collapse
|
6
|
Li T, Geng Z, Zhang J, Xu L, Zhu X. BP5 alleviates endotoxemia-induced acute lung injury by activating Nrf2 via dual regulation of the Keap1-Nrf2 interaction and the Akt (Ser473)/GSK3β (Ser9)/Fyn pathway. Free Radic Biol Med 2022; 193:304-318. [PMID: 36272670 DOI: 10.1016/j.freeradbiomed.2022.10.299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Oxidative stress and inflammation play a crucial role in the pathogenesis of acute lung injury (ALI). Previously, pentapeptide bursopentin (BP5, Cys-Lys-Arg-Val-Tyr) was reported to possess significant antioxidant activity and inhibit lipopolysaccharides (LPS)-induced NF-κB activation in vitro, whereas little is known about its effects in vivo. In this study, we explored the effects of BP5 on endotoxemia-induced ALI in mice and the underlying molecular mechanisms. Our studies revealed that BP5 markedly improved survival and effectively alleviated lung injury by reducing overoxidation and excessive inflammatory response in endotoxemia mice. In LPS-stimulated mouse primary macrophages and RAW 264.7 cells, BP5 also exhibited antioxidant and anti-inflammatory properties by enhancing Nrf2 activation. Importantly, these beneficial effects were abolished by Nrf2 knockdown. To further elucidate the underlying mechanisms, we performed localized surface plasmon resonance (LSPR) assays, molecular docking, together with cell-based studies, and found that BP5 inhibited the Keap1-Nrf2 interaction to promote Nrf2 nuclear translocation and activation. Moreover, BP5-induced Nrf2 activation was shown to be accompanied by an increase in the phosphorylation of Akt (at Ser473) and GSK3β (at Ser9), and a decrease in Fyn nuclear accumulation both in vitro and in vivo. Pharmacologically inhibiting phosphorylation of Akt and GSK3β obviously enhanced Fyn nuclear accumulation in RAW 264.7 cells, which partially attenuated the promoting effect of BP5 on Nrf2 nuclear accumulation and activation. Furthermore, In Nrf2-/- mice, the protective effects of BP5 on the endotoxemia-induced ALI in WT mice were largely vanished. Our findings indicated that BP5 effectively protected endotoxemia-induced ALI against oxidative stress and inflammatory response, which are largely dependent on activation of the Nrf2 pathway. Underlying mechanisms include dual regulation of the Keap-Nrf2 interaction and the Akt (Ser473)/GSK3β (Ser9)/Fyn pathway.
Collapse
Affiliation(s)
- Tianxiang Li
- Department of Pulmonary Medicine, Zhongda Hospital, School of Medicine, South-east University, Nanjing, China.
| | - Zhirong Geng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Ju Zhang
- Wuhan Yangene Biological Technology Co, LTD, Yuechuang Center of HuaZhong Agricultural University, Wuhan, China.
| | - Lu Xu
- Department of Pulmonary Medicine, Zhongda Hospital, School of Medicine, South-east University, Nanjing, China.
| | - Xiaoli Zhu
- Department of Pulmonary Medicine, Zhongda Hospital, School of Medicine, South-east University, Nanjing, China.
| |
Collapse
|
7
|
Singh RK, Verma PK, Kumar S, Shukla A, Kumar N, Kumar S, Acharya A. Evidence that PKCα inhibition in Dalton's Lymphoma cells augments cell cycle arrest and mitochondrial-dependent apoptosis. Leuk Res 2022; 113:106772. [PMID: 35016128 DOI: 10.1016/j.leukres.2021.106772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/18/2021] [Accepted: 12/26/2021] [Indexed: 10/19/2022]
Abstract
Protein kinase Cα (PKCα), belonging to ser/thr protein kinase, perform various biological functions. Overexpression of PKCα has been observed in multiple human malignancies including lymphoma. However, the molecular pathogenesis and involvement of PKCα in Non-Hodgkin lymphoma (NHL) are not clearly understood. Hence, deciphering the role of PKCα in NHL management may provide a better therapeutic option. In the present study, we used selective pharmacological inhibitors Gö6976 and Ro320432 that potentially inhibit PKCα-mediated signaling in DL cells, resulting in the inhibition of cell growth and mitochondrial-dependent apoptosis. PKCα inhibition by these inhibitors also displays cell cycle arrest at the G1 phase and causes growth retardation of DL cells. Our results extended the mechanism of PKCα in NHL, and provided potential implications for its therapy.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Praveen Kumar Verma
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sandeep Kumar
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Alok Shukla
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Naveen Kumar
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sanjay Kumar
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Arbind Acharya
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
8
|
Arumugam P, Sampathkumar B, Perumalsamy H, Balusamy SR, Ramesh V, Sundaravadevel S. Synergistic effect of anethole and doxorubicin alleviates cell proliferation, cell cycle arrest, and ER stress and promotes ROS-mediated apoptosis in triple-negative breast cancer cells. J Biochem Mol Toxicol 2021; 35:e22928. [PMID: 34585488 DOI: 10.1002/jbt.22928] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 08/16/2021] [Accepted: 09/20/2021] [Indexed: 11/07/2022]
Abstract
The heterogeneity and poor prognosis of triple-negative breast cancer (TNBC) have limited the treatment options and made clinical management challenging. This has nurtured a major effort to discover druggable molecular targets. Currently, chemotherapy is the primary treatment strategy for this disease. Doxorubicin is the most frequently used chemotherapeutic drug for TNBC and due to the fact that chemotherapeutic drugs have a lot of side effects, we evaluated the synergistic effect of the phytocompound anethole and doxorubicin. The cytotoxic effect of anethole in combination with doxorubicin on MDA-MB-231 cells was evaluated by various parameters, including apoptosis, cell cycle analysis, DNA damage, and cell proliferation. Furthermore, mitochondrial membranepotential (MMP), endoplasmic reticulum (ER) stress, and reactive oxygen species (ROS) levels were also evaluated in the cells treated with/without anethole and doxorubicin. Expression of the apoptotic proteins was evaluated by Western blot analysis. Initial evaluation of cytotoxicity of anethole on MDA-MB-231 cells demonstrated preferential suppression of cell proliferation and when treated along with doxorubicin it showed enhanced cytotoxicity with a synergistic effect. Cell cycle analysis revealed arrest at different stages of the cell cycle, such as sub G0-G1, G0-G1, S, and G2M in various treatment groups and apoptotic cell death was subsequently evident with propidium iodide (PI) staining. The synergistic action of anethole and doxorubicin effectively induced mitochondrial membrane potential loss, which, in turn, led to a burst of ROS production, which eventually produced unfolded protein response by damaging the ER. Synergistic anticancer effect was observed on exposure of MDA-MB-231 cells to anethole and doxorubicin in inducing cell death.
Collapse
Affiliation(s)
- Poornima Arumugam
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education For Women, Coimbatore, Tamilnadu, India
| | - Banupriya Sampathkumar
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education For Women, Coimbatore, Tamilnadu, India
| | - Haribalan Perumalsamy
- Center for Next Generation Cytometry, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
| | - Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Seoul, Gwangjin-gu, Republic of Korea
| | - Vignesh Ramesh
- International Center for Clinical Research, Friedrich Alexander University, Erlangen-Nurnberb, Germany
| | - Sumathi Sundaravadevel
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education For Women, Coimbatore, Tamilnadu, India
| |
Collapse
|
9
|
Nie Z, Chen M, Wen X, Gao Y, Huang D, Cao H, Peng Y, Guo N, Ni J, Zhang S. Endoplasmic Reticulum Stress and Tumor Microenvironment in Bladder Cancer: The Missing Link. Front Cell Dev Biol 2021; 9:683940. [PMID: 34136492 PMCID: PMC8201605 DOI: 10.3389/fcell.2021.683940] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer is a common malignant tumor of the urinary system. Despite recent advances in treatments such as local or systemic immunotherapy, chemotherapy, and radiotherapy, the high metastasis and recurrence rates, especially in muscle-invasive bladder cancer (MIBC), have led to the evaluation of more targeted and personalized approaches. A fundamental understanding of the tumorigenesis of bladder cancer along with the development of therapeutics to target processes and pathways implicated in bladder cancer has provided new avenues for the management of this disease. Accumulating evidence supports that the tumor microenvironment (TME) can be shaped by and reciprocally act on tumor cells, which reprograms and regulates tumor development, metastasis, and therapeutic responses. A hostile TME, caused by intrinsic tumor attributes (e.g., hypoxia, oxidative stress, and nutrient deprivation) or external stressors (e.g., chemotherapy and radiation), disrupts the normal synthesis and folding process of proteins in the endoplasmic reticulum (ER), culminating in a harmful situation called ER stress (ERS). ERS is a series of adaptive changes mediated by unfolded protein response (UPR), which is interwoven into a network that can ultimately mediate cell proliferation, apoptosis, and autophagy, thereby endowing tumor cells with more aggressive behaviors. Moreover, recent studies revealed that ERS could also impede the efficacy of anti-cancer treatment including immunotherapy by manipulating the TME. In this review, we discuss the relationship among bladder cancer, ERS, and TME; summarize the current research progress and challenges in overcoming therapeutic resistance; and explore the concept of targeting ERS to improve bladder cancer treatment outcomes.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Xiaohong Wen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Jie Ni
- Cancer Care Center, St. George Hospital, Sydney, NSW, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| |
Collapse
|
10
|
Oladimeji O, Akinyelu J, Daniels A, Singh M. Modified Gold Nanoparticles for Efficient Delivery of Betulinic Acid to Cancer Cell Mitochondria. Int J Mol Sci 2021; 22:ijms22105072. [PMID: 34064888 PMCID: PMC8150271 DOI: 10.3390/ijms22105072] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Advances in nanomedicine have seen the adaptation of nanoparticles (NPs) for subcellular delivery for enhanced therapeutic impact and reduced side effects. The pivotal role of the mitochondria in apoptosis and their potential as a target in cancers enables selective induction of cancer cell death. In this study, we examined the mitochondrial targeted delivery of betulinic acid (BA) by the mitochondriotropic TPP+-functionalized epigallocatechin gallate (EGCG)-capped gold NPs (AuNPs), comparing the impact of polyethylene glycol (PEG) and poly-L-lysine-graft-polyethylene glycol (PLL-g-PEG) copolymer on delivery efficacy. This included the assessment of their cellular uptake, mitochondrial localization and efficacy as therapeutic delivery platforms for BA in the human Caco-2, HeLa and MCF-7 cancer cell lines. These mitochondrial-targeted nanocomplexes demonstrated significant inhibition of cancer cell growth, with targeted nanocomplexes recording IC50 values in the range of 3.12–13.2 µM compared to that of the free BA (9.74–36.31 µM) in vitro, demonstrating the merit of mitochondrial targeting. Their mechanisms of action implicated high amplitude mitochondrial depolarization, caspases 3/7 activation, with an associated arrest at the G0/G1 phase of the cell cycle. This nano-delivery system is a potentially viable platform for mitochondrial-targeted delivery of BA and highlights mitochondrial targeting as an option in cancer therapy.
Collapse
|
11
|
Jiang M, Li S, Wu J, Li W, Wen XA, Liang H, Yang F. Designing biotin-human serum albumin nanoparticles to enhance the targeting ability of binuclear ruthenium(III) compound. J Inorg Biochem 2020; 215:111318. [PMID: 33301985 DOI: 10.1016/j.jinorgbio.2020.111318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
On the one hand, to obtain a novel next-generation anticancer metal agent; on the other hand, to improve the targeting ability and decrease side effects of metal agent, we proposed to design active-targeting human serum albumin (HSA) nanoparticles (NPs) to achieve the end. Thus, we not only designed and synthesized two ruthenium (Ru) thiosemicarbazone compounds (C1 and C2) but also succeeded in constructing active Biotin-HSA NPs for Ru(III) compounds. Importantly, Biotin-HSA-C2 NPs not only possessed a stronger capacity for killing MCF-7 cells and inhibiting their migration versusC2 alone but also increased accumulation compared to non-malignant WI-38 cells. Additionally, C2 and Biotin-HSA-C2 NPs act against MCF-7 cells by the following potential mechanism: 1) arresting the cell cycle in the S phase by regulating cyclin and cyclin-dependent kinases; 2) inducing apoptosis by releasing cytochrome c to activate caspase-9/3; 3) inhibiting the expression of p-EGFR and regulating its neighboring cellular pathways, followed by the inactivation of PI3K/Akt and activation of p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Ming Jiang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi, China; School of food and biochemical engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| | - Shanhe Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi, China
| | - Junmiao Wu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi, China
| | - Wenjuan Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi, China
| | - Xiao-An Wen
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi, China
| | - Feng Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi, China.
| |
Collapse
|
12
|
Krasic J, Skara L, Ulamec M, Katusic Bojanac A, Dabelic S, Bulic-Jakus F, Jezek D, Sincic N. Teratoma Growth Retardation by HDACi Treatment of the Tumor Embryonal Source. Cancers (Basel) 2020; 12:cancers12113416. [PMID: 33217978 PMCID: PMC7698704 DOI: 10.3390/cancers12113416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Testicular germ cell tumors are the most common neoplasms in young male populations, with a rising incidence. Among them, teratomas may often be very aggressive and resistant to therapy. Our aim was to investigate the impact of two potential anti-tumor epigenetic drugs (Valproate and Trichostatin A) in a mammalian model of teratoma development from an early trilaminar mouse embryo. Both drugs applied to the embryonic tissue had a significant negative impact on the teratoma growth in a three-dimensional in vitro culture. However, Trichostatin A did not diminish some potentially dangerous features of teratomas in contrast to Valproate. This research is an original contribution to the basic knowledge of the origin and development of teratomas. Such knowledge is necessary for envisioning therapeutic strategies against human testicular tumors. Abstract Among testicular germ cell tumors, teratomas may often be very aggressive and therapy-resistant. Our aim was to investigate the impact of histone deacetylase inhibitors (HDACi) on the in vitro growth of experimental mouse teratoma by treating their embryonic source, the embryo-proper, composed only of the three germ layers. The growth of teratomas was measured for seven days, and histopathological analysis, IHC/morphometry quantification, gene enrichment analysis, and qPCR analysis on a selected panel of pluripotency and early differentiation genes followed. For the first time, within teratomas, we histopathologically assessed the undifferentiated component containing cancer stem cell-like cells (CSCLCs) and differentiated components containing numerous lymphocytes. Mitotic indices were higher than apoptotic indices in both components. Both HDACi treatments of the embryos-proper significantly reduced teratoma growth, although this could be related neither to apoptosis nor proliferation. Trichostatin A increased the amount of CSCLCs, and upregulated the mRNA expression of pluripotency/stemness genes as well as differentiation genes, e.g., T and Eomes. Valproate decreased the amount of CSCLCs, and downregulated the expressions of pluripotency/stemness and differentiation genes. In conclusion, both HDACi treatments diminished the inherent tumorigenic growth potential of the tumor embryonal source, although Trichostatin A did not diminish the potentially dangerous expression of cancer-related genes and the amount of CSCLC.
Collapse
Affiliation(s)
- Jure Krasic
- Department of Medical Biology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; (J.K.); (L.S.); (A.K.B.); (F.B.-J.)
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
| | - Lucija Skara
- Department of Medical Biology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; (J.K.); (L.S.); (A.K.B.); (F.B.-J.)
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
| | - Monika Ulamec
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre Milosrdnice University Hospital Center, 10 000 Zagreb, Croatia
- Department of Pathology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Ana Katusic Bojanac
- Department of Medical Biology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; (J.K.); (L.S.); (A.K.B.); (F.B.-J.)
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
| | - Sanja Dabelic
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10 000 Zagreb, Croatia;
| | - Floriana Bulic-Jakus
- Department of Medical Biology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; (J.K.); (L.S.); (A.K.B.); (F.B.-J.)
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
| | - Davor Jezek
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Nino Sincic
- Department of Medical Biology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; (J.K.); (L.S.); (A.K.B.); (F.B.-J.)
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia;
- Correspondence: ; Tel.: +385-1-45-66-806; Fax: +385-45-960-199
| |
Collapse
|
13
|
Ashrafizadeh M, Mohammadinejad R, Samarghandian S, Yaribeygi H, Johnston TP, Sahebkar A. Anti-Tumor Effects of Osthole on Different Malignant Tissues: A Review of Molecular Mechanisms. Anticancer Agents Med Chem 2020; 20:918-931. [PMID: 32108003 DOI: 10.2174/1871520620666200228110704] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/09/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Cancer management and/or treatment require a comprehensive understanding of the molecular and signaling pathways involved. Recently, much attention has been directed to these molecular and signaling pathways, and it has been suggested that a number of biomolecules/players involved in such pathways, such as PI3K/Akt, NF-kB, STAT, and Nrf2 contribute to the progression, invasion, proliferation, and metastasis of malignant cells. Synthetic anti-tumor agents and chemotherapeutic drugs have been a mainstay in cancer therapy and are widely used to suppress the progression and, hopefully, halt the proliferation of malignant cells. However, these agents have some undesirable side-effects and, therefore, naturally-occurring compounds with high potency and fewer side-effects are now of great interest. Osthole is a plant-derived chemical compound that can inhibit the proliferation of malignant cells and provide potent anti-cancer effects in various tissues. Therefore, in this review, we presented the main findings concerning the potential anti-tumor effects of osthole and its derivatives and described possible molecular mechanisms by which osthole may suppress malignant cell proliferation in different tissues.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | | |
Collapse
|