1
|
Alqabandi JA, David R, Abdel-Motal UM, ElAbd RO, Youcef-Toumi K. An innovative cellular medicine approach via the utilization of novel nanotechnology-based biomechatronic platforms as a label-free biomarker for early melanoma diagnosis. Sci Rep 2024; 14:30107. [PMID: 39627312 PMCID: PMC11615046 DOI: 10.1038/s41598-024-79154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 11/06/2024] [Indexed: 12/06/2024] Open
Abstract
Innovative cellular medicine (ICM) is an exponentially emerging field with a promising approach to combating complex and ubiquitous life-threatening diseases such as multiple sclerosis (MS), arthritis, Parkinson's disease, Alzheimer's, heart disease, and cancer. Together with the advancement of nanotechnology and bio-mechatronics, ICM revolutionizes cellular therapy in understanding the essence and nature of the disease initiated at a single-cell level. This paper focuses on the intricate nature of cancer that requires multi-disciplinary efforts to characterize it well in order to achieve the objectives of modern world contemporary medicine in the early detection of the disease at a cellular level and potentially arrest its proliferation mechanism. This justifies the multidisciplinary research backgrounds of the authors of this paper in advancing cellular medicine by bridging the gap between experimental biology and the engineering field. Thus, in pursuing this approach, two novel miniaturized and highly versatile biomechatronic platforms with dedicated operating software and microelectronics are designed, modeled, nanofabricated, and tested in numerous in vitro experiments to investigate a hypothesis and arrive at a proven theorem in carcinogenesis by interrelating cellular contractile force, membrane potential, and cellular morphology for early detection and characterization of melanoma cancer cells. The novelties that flourished within this work are manifested in sixfold: (1) developing a mathematical model that utilizes a Heaviside step function, as well as a pin-force model to compute the contractile force of a living cell, (2) deriving an expression of cell-membrane potential based on Laplace and Fourier Transform and their Inverse Transform functions by encountering Warburg diffusion impedance factor, (3) nano-fabricating novel biomechatronic platforms with associated microelectronics and customized software that extract cellular physics and mechanics, (4) developing a label-free biomarker, (5) arrive at a proved theorem in developing a mathematical expression in relating cancer cell mechanobiology to its biophysics in connection to the stage of the disease, and (6) to the first time in literature, and to the best of the authors' knowledge, discriminating different stages and morphology of cancer cell melanoma based on their cell-membrane potentials, and associated contractile forces that could introduce a new venue of cellular therapeutic modalities, preclinical early cancer diagnosis, and a novel approach in immunotherapy drug development. The proposed innovative technology-based versatile bio-mechatronic platforms shall be extended for future studies, investigating the role of electrochemical signaling of the nervous system in cancer formation that will significantly impact modern oncology by pursuing a targeted immunotherapy approach. This work also provides a robust platform for immunotherapy practitioners in extending the study of cellular biophysics in stalling neural-cancer interactions, of which the FDA-approved chimeric antigen receptor (CAR)-T cell therapies can be enhanced (genetically engineered) in a lab by improving its receptors to capture cancer antigens. This work amplifies the importance of studying neurotransmitters and electrochemical signaling molecules in shaping the immune T-cell function and its effectiveness in arresting cancer proliferation rate (mechanobiology mechanism).
Collapse
Affiliation(s)
- Jassim A Alqabandi
- Mechatronics Research Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
- Mechatronics in Medicine Laboratory, Imperial College London, London, UK.
- Department of Manufacturing Engineering Technology (Bio-Mechatronics) Department, PAAET, Kuwait, State of Kuwait.
| | - Rhiannon David
- Division of Computational and Systems Medicine (CSM), Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, UK
| | - Ussama M Abdel-Motal
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rawan O ElAbd
- McGill University Health Center, Montreal, QC, Canada
| | - Kamal Youcef-Toumi
- Mechatronics Research Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| |
Collapse
|
2
|
Sun J, Tian Z, Wu J, Li J, Wang Q, Huang S, Wang M. Pristimerin Exerts Pharmacological Effects Through Multiple Signaling Pathways: A Comprehensive Review. Drug Des Devel Ther 2024; 18:1673-1694. [PMID: 38779590 PMCID: PMC11110813 DOI: 10.2147/dddt.s460093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Pristimerin, a natural triterpenoid isolated from the plants of southern snake vine and Maidenwood in the family Weseraceae, is anti-inflammatory, insecticidal, antibacterial, and antiviral substance and has been used for its cardioprotective and antitumor effects and in osteoporosis treatment. These qualities explain Pristimerin's therapeutic effects on different types of tumors and other diseases. More and more studies have shown that pristimerin acts in a wide range of biological activities and has shown great potential in various fields of modern and Chinese medicine. While Pristimerin's wide range of pharmacological effects have been widely studied by others, our comprehensive review suggests that its mechanism of action may be through affecting fundamental cellular events, including blocking the cell cycle, inducing apoptosis and autophagy, and inhibiting cell migration and invasion, or through activating or inhibiting certain key molecules in several cell signaling pathways, including nuclear factor κB (NF-κB), phosphatidylinositol 3-kinase/protein kinase B/mammalian-targeted macromycin (PI3K/Akt/mTOR), mitogen-activated protein kinases (MAPKs), extracellular signal-regulated protein kinase 1/2 (ERK1/2), Jun amino-terminal kinase (JNK1/2/3), reactive oxygen species (ROS), wingless/integrin1 (Wnt)/β-catenin, and other signaling pathways. This paper reviews the research progress of Pristimerin's pharmacological mechanism of action in recent years to provide a theoretical basis for the molecular targeting therapy and further development and utilization of Pristimerin. It also provides insights into improved treatments and therapies for clinical patients and the need to explore pristimerin as a potential facet of treatment.
Collapse
Affiliation(s)
- Jian Sun
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Zhaochun Tian
- Science and Technology Innovation Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Jing Wu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Jiafei Li
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Qixia Wang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Shuhong Huang
- Science and Technology Innovation Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Meng Wang
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
3
|
Prabhu KS, Jessy S, Kuttikrishnan S, Mujeeb F, Mariyam Z, Habeeba U, Ahmad N, Bhat AA, Uddin S. Anticancer Potential and Molecular Targets of Pristimerin in Human Malignancies. Pharmaceuticals (Basel) 2024; 17:578. [PMID: 38794148 PMCID: PMC11123949 DOI: 10.3390/ph17050578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
The growing global burden of malignant tumors with increasing incidence and mortality rates underscores the urgent need for more effective and less toxic therapeutic options. Herbal compounds are being increasingly studied for their potential to meet these needs due to their reduced side effects and significant efficacy. Pristimerin (PS), a triterpenoid from the quinone formamide class derived from the Celastraceae and Hippocrateaceae families, has emerged as a potent anticancer agent. It exhibits broad-spectrum anti-tumor activity across various cancers such as breast, pancreatic, prostate, glioblastoma, colorectal, cervical, and lung cancers. PS modulates several key cellular processes, including apoptosis, autophagy, cell migration and invasion, angiogenesis, and resistance to chemotherapy, targeting crucial signaling pathways such as those involving NF-κB, p53, and STAT3, among others. The main objective of this review is to provide a comprehensive synthesis of the current literature on PS, emphasizing its mechanisms of action and molecular targets with the utmost clarity. It discusses the comparative advantages of PS over current cancer therapies and explores the implications for future research and clinical applications. By delineating the specific pathways and targets affected by PS, this review seeks to offer valuable insights and directions for future research in this field. The information gathered in this review could pave the way for the successful development of PS into a clinically applicable anticancer therapy.
Collapse
Affiliation(s)
- Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
| | - Serah Jessy
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
| | - Farina Mujeeb
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India;
| | - Zahwa Mariyam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
| | - Ummu Habeeba
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
| | - Nuha Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
| | - Ajaz A. Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity, and Cancer Program, Sidra Medicine, Doha 26999, Qatar;
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.J.); (S.K.); (Z.M.); (U.H.); (N.A.)
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India;
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar
| |
Collapse
|
4
|
Wang X, Zhang X, Zheng G, Dong M, Huang Z, Lin L, Yan K, Zheng J, Wang J. Mitochondria-targeted pentacyclic triterpene NIR-AIE derivatives for enhanced chemotherapeutic and chemo-photodynamic combined therapy. Eur J Med Chem 2024; 264:115975. [PMID: 38039788 DOI: 10.1016/j.ejmech.2023.115975] [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: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
Complexes formed by combining pentacyclic triterpenes (PTs) with Aggregation-Induced Emission luminogens (AIEgens), termed pentacyclic triterpene-aggregation induced emission (PT-AIEgen) complexes, merge the chemotherapeutic properties of PTs with the photocytotoxicity of AIEgens. In this study, we synthesized derivatives by connecting three types of triphenylamine (TPA) pyridinium derivatives with three common pentacyclic triterpenes. Altering the connecting group between the electron donor TPA and the electron acceptor pyridinium resulted in increased production of reactive oxygen species (ROS) by PT-AIEgens and a red-shift in their fluorescence emission spectra. Importantly, the fluorescence emission spectra of BA-3, OA-3, and UA-3 extended into the near-infrared (NIR) range, enabling NIR-AIE imaging of the sites where the derivatives aggregated. The incorporation of the pyridinium structure improved the mitochondrial targeting of PT-AIEgens, enhancing mitochondrial pathway-mediated cell apoptosis and improving the efficiency of chemotherapy (CT) and chemo-photodynamic combined therapy (CPCT) both in vivo and in vitro. Cellular fluorescence imaging demonstrated rapid cellular uptake and mitochondrial accumulation of BA-1 (-2, -3). Cell viability experiments revealed that BA-1 (-2), OA-1 (-2), and UA-1 (-2) exhibited superior CT cytotoxicity compared to their parent drugs, with BA-1 showing the most potent inhibitory effect on HeLa cells (IC50 = 1.19 μM). Furthermore, HeLa cells treated with BA-1 (1 μM), BA-2 (1.25 μM), and BA-3 (1 μM) exhibited survival rates of 2.99 % ± 0.05 % μM, 5.92 % ± 2.04 % μM, and 2.53 % ± 0.73 % μM, respectively, under white light irradiation. Mechanistic experiments revealed that derivatives induced cell apoptosis via the mitochondrial apoptosis pathway during both CT and CPCT. Remarkably, BA-1 and BA-3 in CPCT inhibited cancer cell proliferation in an in vivo melanoma mouse xenograft model. These results collectively encourage further research of PT-AIEgens as potential anticancer agents.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Xuewei Zhang
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Guoxing Zheng
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Mingming Dong
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Zhaopeng Huang
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Liyin Lin
- Central Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Kang Yan
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Jinhong Zheng
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China
| | - Jinzhi Wang
- Department of Chemistry, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, PR China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, PR China.
| |
Collapse
|
5
|
Lu Y, Zeng Z, Bao X, Wu M, Jing Z, Feng J. Pristimerin protects against pathological cardiac hypertrophy through improvement of PPARα pathway. Toxicol Appl Pharmacol 2023; 473:116572. [PMID: 37269933 DOI: 10.1016/j.taap.2023.116572] [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: 04/05/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023]
Abstract
Pristimerin (PM), serving as a biological component mainly obtained from Celastraceae and Hippocrateaceae families, has been extensively explored for its numerous pharmacological activities, especially anti-cancer activity. However, the function of PM on pathological cardiac hypertrophy is poorly understood. This work was intended to investigate the effects of PM on pressure-overload induced myocardial hypertrophy and its potential pathways. Mouse model of pathological cardiac hypertrophy was generated by transverse aortic constriction (TAC) or minipump administration of the β-adrenergic agonist ISO for 4 weeks, and PM (0.5 mg/Kg/d, i.p.) was treated for 2 weeks. PPARα-/- mice received TAC surgery were used for mechanism exploration. Moreover, neonatal rat cardiomyocytes (NRCMs) were utilized to explore the effect of PM following Angiotensin II (Ang II, 1.0 μM) administration. We found that PM attenuated pressure-overload induced cardiac dysfunction, myocardial hypertrophy and fibrosis in mice. Likewise, PM incubation dramatically reversed Ang II-mediated cardiomyocytes hypertrophy in NRCMs. RNA-Sequence showed that PM selectively contributed to improvement of PPARα/PGC1 signaling, while silencing PPARα abrogated the beneficial effects of PM on Ang II-treated NRCMs. Importantly, PM ameliorated Ang II-induced mitochondrial dysfunction and decrease in metabolic genes, whereas knockdown of PPARα eliminated these alterations in NRCMs. Similarly, PM presented limited protective effects on pressure-overload induced systolic dysfunction and myocardial hypertrophy in PPARα deficient mice. Overall, this study revealed that PM exerted protective activity against pathological cardiac hypertrophy through improvement of PPARα/PGC1 pathway.
Collapse
Affiliation(s)
- Ye Lu
- Department of Interventional Center and Vascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Zhaoxiang Zeng
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Xianhao Bao
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Mingwei Wu
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Zaiping Jing
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Jiaxuan Feng
- Department of Interventional Center and Vascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China.
| |
Collapse
|
6
|
Ambrosio FA, Costa G, Gallo Cantafio ME, Torcasio R, Trapasso F, Alcaro S, Viglietto G, Amodio N. Natural Agents as Novel Potential Source of Proteasome Inhibitors with Anti-Tumor Activity: Focus on Multiple Myeloma. Molecules 2023; 28:molecules28031438. [PMID: 36771100 PMCID: PMC9919276 DOI: 10.3390/molecules28031438] [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: 12/27/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) is an aggressive and incurable disease for most patients, characterized by periods of treatment, remission and relapse. The introduction of new classes of drugs, such as proteasome inhibitors (PIs), has improved survival outcomes in these patient populations. The proteasome is the core of the ubiquitin-proteasome system (UPS), a complex and conserved pathway involved in the control of multiple cellular processes, including cell cycle control, transcription, DNA damage repair, protein quality control and antigen presentation. To date, PIs represent the gold standard for the treatment of MM. Bortezomib was the first PI approved by the FDA, followed by next generation of PIs, namely carfilzomib and ixazomib. Natural agents play an important role in anti-tumor drug discovery, and many of them have recently been reported to inhibit the proteasome, thus representing a new potential source of anti-MM drugs. Based on the pivotal biological role of the proteasome and on PIs' significance in the management of MM, in this review we aim to briefly summarize recent evidence on natural compounds capable of inhibiting the proteasome, thus triggering anti-MM activity.
Collapse
Affiliation(s)
- Francesca Alessandra Ambrosio
- Department of Experimental and Clinical Medicine, Campus “S. Venuta”, University “Magna Græcia” of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Giosuè Costa
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Correspondence: (G.C.); (N.A.)
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Campus “S. Venuta”, University “Magna Græcia” of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Roberta Torcasio
- Department of Experimental and Clinical Medicine, Campus “S. Venuta”, University “Magna Græcia” of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy
| | - Francesco Trapasso
- Department of Experimental and Clinical Medicine, Campus “S. Venuta”, University “Magna Græcia” of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Associazione CRISEA—Centro di Ricerca e Servizi Avanzati per l’Innovazione Rurale, Loc. Condoleo, 88055 Belcastro, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Campus “S. Venuta”, University “Magna Græcia” of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Campus “S. Venuta”, University “Magna Græcia” of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Correspondence: (G.C.); (N.A.)
| |
Collapse
|
7
|
Xu K, Zhu W, Xu A, Xiong Z, Zou D, Zhao H, Jiao D, Qing Y, Jamal MA, Wei HJ, Zhao HY. Inhibition of FOXO1‑mediated autophagy promotes paclitaxel‑induced apoptosis of MDA‑MB‑231 cells. Mol Med Rep 2022; 25:72. [PMID: 35014689 PMCID: PMC8767459 DOI: 10.3892/mmr.2022.12588] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, and it often becomes resistant to paclitaxel (PTX) therapy. Autophagy plays an important cytoprotective role in PTX-induced tumor cell death, and targeting autophagy has been promising for improving the efficacy of tumor chemotherapy in recent years. The aim of the present study was to clarify the mechanism of PTX inducing autophagy in TNBC cells to provide a potential clinical chemotherapy strategy of PTX for TNBC. The present study reported that PTX induced both apoptosis and autophagy in MDA-MB-231 cells and that inhibition of autophagy promoted apoptotic cell death. Furthermore, it was found that forkhead box transcription factor O1 (FOXO1) enhanced PTX-induced autophagy through a transcriptional activation pattern in MDA-MB-231 cells, which was associated with the downstream target genes autophagy related 5, class III phosphoinositide 3-kinase vacuolar protein sorting 34, autophagy related 4B cysteine peptidase, beclin 1 and microtubule associated protein 1 light chain 3β. Knocking down FOXO1 attenuated the survival of MDA-MB-231 cells in response to PTX treatment. These findings may be beneficial for improving the treatment efficacy of PTX and to develop autophagic targeted therapy for TNBC.
Collapse
Affiliation(s)
- Kaixiang Xu
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Wanyun Zhu
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Anyong Xu
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Zhe Xiong
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Di Zou
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Heng Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Deling Jiao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Yubo Qing
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Muhammad Ameen Jamal
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Hong-Jiang Wei
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Hong-Ye Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| |
Collapse
|
8
|
XIAO-MEI C, JIN-YU Z, YAN-LANG Y, YU-WEI W, YUAN-YUAN Y, HAI-HONG X. Pristimerin improve renal fibrosis by regulating miRNA-145-5p in vitro and vivo study. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.79021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
| | - Zhang JIN-YU
- Yijishan Hospital of Wannan Medical College, China
| | | | - Wang YU-WEI
- Yijishan Hospital of Wannan Medical College, China
| | - Yu YUAN-YUAN
- Yijishan Hospital of Wannan Medical College, China
| | - Xu HAI-HONG
- Yijishan Hospital of Wannan Medical College, China
| |
Collapse
|
9
|
Delinois LJ, De León-Vélez O, Vázquez-Medina A, Vélez-Cabrera A, Marrero-Sánchez A, Nieves-Escobar C, Alfonso-Cano D, Caraballo-Rodríguez D, Rodriguez-Ortiz J, Acosta-Mercado J, Benjamín-Rivera JA, González-González K, Fernández-Adorno K, Santiago-Pagán L, Delgado-Vergara R, Torres-Ávila X, Maser-Figueroa A, Grajales-Avilés G, Miranda Méndez GI, Santiago-Pagán J, Nieves-Santiago M, Álvarez-Carrillo V, Griebenow K, Tinoco AD. Cytochrome c: Using Biological Insight toward Engineering an Optimized Anticancer Biodrug. INORGANICS 2021; 9:83. [PMID: 35978717 PMCID: PMC9380692 DOI: 10.3390/inorganics9110083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The heme protein cytochrome c (Cyt c) plays pivotal roles in cellular life and death processes. In the respiratory chain of mitochondria, it serves as an electron transfer protein, contributing to the proliferation of healthy cells. In the cell cytoplasm, it activates intrinsic apoptosis to terminate damaged cells. Insight into these mechanisms and the associated physicochemical properties and biomolecular interactions of Cyt c informs on the anticancer therapeutic potential of the protein, especially in its ability to subvert the current limitations of small molecule-based chemotherapy. In this review, we explore the development of Cyt c as an anticancer drug by identifying cancer types that would be receptive to the cytotoxicity of the protein and factors that can be finetuned to enhance its apoptotic potency. To this end, some information is obtained by characterizing known drugs that operate, in part, by triggering Cyt c induced apoptosis. The application of different smart drug delivery systems is surveyed to highlight important features for maintaining Cyt c stability and activity and improving its specificity for cancer cells and high drug payload release while recognizing the continuing limitations. This work serves to elucidate on the optimization of the strategies to translate Cyt c to the clinical market.
Collapse
Affiliation(s)
- Louis J. Delinois
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Omar De León-Vélez
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Adriana Vázquez-Medina
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Alondra Vélez-Cabrera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Amanda Marrero-Sánchez
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Daniela Alfonso-Cano
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Jael Rodriguez-Ortiz
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Jemily Acosta-Mercado
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Josué A. Benjamín-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Kiara González-González
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Kysha Fernández-Adorno
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Lisby Santiago-Pagán
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Rafael Delgado-Vergara
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Xaiomy Torres-Ávila
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Andrea Maser-Figueroa
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | | | - Javier Santiago-Pagán
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Miguel Nieves-Santiago
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Vanessa Álvarez-Carrillo
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Kai Griebenow
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| |
Collapse
|
10
|
Chen RZ, Yang F, Zhang M, Sun ZG, Zhang N. Cellular and Molecular Mechanisms of Pristimerin in Cancer Therapy: Recent Advances. Front Oncol 2021; 11:671548. [PMID: 34026649 PMCID: PMC8138054 DOI: 10.3389/fonc.2021.671548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Seeking an efficient and safe approach to eliminate tumors is a common goal of medical fields. Over these years, traditional Chinese medicine has attracted growing attention in cancer treatment due to its long history. Pristimerin is a naturally occurring quinone methide triterpenoid used in traditional Chinese medicine to treat various cancers. Recent studies have identified alterations in cellular events and molecular signaling targets of cancer cells under pristimerin treatment. Pristimerin induces cell cycle arrest, apoptosis, and autophagy to exhibit anti-proliferation effects against tumors. Pristimerin also inhibits the invasion, migration, and metastasis of tumor cells via affecting cell adhesion, cytoskeleton, epithelial-mesenchymal transition, cancer stem cells, and angiogenesis. Molecular factors and pathways are associated with the anti-cancer activities of pristimerin. Furthermore, pristimerin reverses multidrug resistance of cancer cells and exerts synergizing effects with other chemotherapeutic drugs. This review aims to discuss the anti-cancer potentials of pristimerin, emphasizing multi-targeted biological and molecular regulations in cancers. Further investigations and clinical trials are warranted to understand the advantages and disadvantages of pristimerin treatment much better.
Collapse
Affiliation(s)
- Run-Ze Chen
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Yang
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Zhang
- Department of Dermatology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Zhang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
11
|
Noh JI, Mun SK, Lim EH, Kim H, Chang DJ, Hur JS, Yee ST. Induction of Apoptosis in MDA-MB-231 Cells Treated with the Methanol Extract of Lichen Physconia hokkaidensis. J Fungi (Basel) 2021; 7:jof7030188. [PMID: 33807853 PMCID: PMC8000577 DOI: 10.3390/jof7030188] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 12/31/2022] Open
Abstract
Physconia hokkaidensis methanol extract (PHE) was studied to identify anticancer effects and reveal its mechanism of action by an analysis of cytotoxicity, cell cycles, and apoptosis biomarkers. PHE showed strong cytotoxicity in various cancer cells, including HL-60, HeLa, A549, Hep G2, AGS, MDA-MB-231, and MCF-7. Of these cell lines, the growth of MDA-MB-231 was concentration-dependently suppressed by PHE, but MCF-7 was not affected. MDA-MB-231 cells, triple-negative breast cancer (TNBC) cells, do not express estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2), whereas MCF-7 cells are ER-positive, PR-positive, and HER-2-negative breast cancer cells. The number of cells in sub-G1 phase was increased after 24 h of treatment, and annexin V/PI staining showed that the population size of apoptotic cells was increased by prolonged exposure to PHE. Moreover, PHE treatment downregulated the transcriptional levels of Bcl-2, AMPK, and p-Akt, whereas it significantly upregulated the levels of cleaved caspase-3, cleaved caspase-9, and cleaved-PARP. In conclusion, it was confirmed that the PHE exhibited selective cytotoxicity toward MDA-MB-231, not toward MCF-7, and its cytotoxic activity is based on induction of apoptosis.
Collapse
Affiliation(s)
- Ji-In Noh
- Department of Pharmacy, Sunchon National University, Jungang-Ro, Suncheon 549-742, Korea; (J.-I.N.); (S.-K.M.); (E.H.L.); (H.K.); (D.-J.C.)
| | - Seul-Ki Mun
- Department of Pharmacy, Sunchon National University, Jungang-Ro, Suncheon 549-742, Korea; (J.-I.N.); (S.-K.M.); (E.H.L.); (H.K.); (D.-J.C.)
| | - Eui Hyeon Lim
- Department of Pharmacy, Sunchon National University, Jungang-Ro, Suncheon 549-742, Korea; (J.-I.N.); (S.-K.M.); (E.H.L.); (H.K.); (D.-J.C.)
| | - Hangun Kim
- Department of Pharmacy, Sunchon National University, Jungang-Ro, Suncheon 549-742, Korea; (J.-I.N.); (S.-K.M.); (E.H.L.); (H.K.); (D.-J.C.)
| | - Dong-Jo Chang
- Department of Pharmacy, Sunchon National University, Jungang-Ro, Suncheon 549-742, Korea; (J.-I.N.); (S.-K.M.); (E.H.L.); (H.K.); (D.-J.C.)
| | - Jae-Seoun Hur
- Department of Environmental Education, Korea Lichen Research Institute, Sunchon National University, Suncheon 549-742, Korea;
| | - Sung-Tae Yee
- Department of Pharmacy, Sunchon National University, Jungang-Ro, Suncheon 549-742, Korea; (J.-I.N.); (S.-K.M.); (E.H.L.); (H.K.); (D.-J.C.)
- Correspondence: ; Tel.: +82-61-750-3752; Fax: +82-61-750-3708
| |
Collapse
|
12
|
Aranha ESP, Portilho AJDS, Bentes de Sousa L, da Silva EL, Mesquita FP, Rocha WC, Araújo da Silva FM, Lima ES, Alves APNN, Koolen HHF, Montenegro RC, Vasconcellos MCD. 22β-hydroxytingenone induces apoptosis and suppresses invasiveness of melanoma cells by inhibiting MMP-9 activity and MAPK signaling. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113605. [PMID: 33232779 DOI: 10.1016/j.jep.2020.113605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/11/2020] [Accepted: 11/17/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 22β-hydroxytingenone (22-HTG) is a quinonemethide triterpene isolated from Salacia impressifolia (Miers) A. C. Smith (family Celastraceae), which has been used in traditional medicine to treat a variety of diseases, including dengue, renal infections, rheumatism and cancer. However, the anticancer effects of 22-HTG and the underlying molecular mechanisms in melanoma cells have not yet been elucidated. AIM OF THE STUDY The present study investigated apoptosis induction and antimetastatic potencial of 22-HTG in SK-MEL-28 human melanoma cells. MATERIALS AND METHODS First, the in vitro cytotoxic activity of 22-HTG in cultured cancer cells was evaluated. Then, cell viability was determined using the trypan blue assay in melanoma cells (SK-MEL-28), which was followed by cell cycle, annexin V-FITC/propidium iodide assays (Annexin/PI), as well as assays to evaluate mitochondrial membrane potential, production of reactive oxygen species (ROS) using flow cytometry. Fluorescence microscopy using acridine orange/ethidium bromide (AO/BE) staining was also performed. RT-qPCR was carried out to evaluate the expression of BRAF, NRAS, and KRAS genes. The anti-invasiveness potential of 22-HTG was evaluated in a three-dimensional (3D) model of reconstructed human skin. RESULTS 22-HTG reduced viability of SK-MEL-28 cells and caused morphological changes, as cell shrinkage, chromatin condensation, and nuclear fragmentation. Furthermore, 22-HTG caused apoptosis, which was demonstrated by increased staining with AO/BE and Annexin/PI. The apoptosis may have been caused by mitochondrial instability without the involvement of ROS production. The expression of BRAF, NRAS, and KRAS, which are important biomarkers in melanoma development, was reduced by the 22-HTG treatment. In the reconstructed skin model, 22-HTG was able to decrease the invasion capacity of melanoma cells in the dermis. CONCLUSIONS Our data indicate that 22-HTG has anti-tumorigenic properties against melanoma cells through the induction of cell cycle arrest, apoptosis and inhibition of invasiveness potential, as observed in the 3D model. As such, the results provide new insights for future work on the utilization of 22-HTG in malignant melanoma treatment.
Collapse
Affiliation(s)
- Elenn Suzany Pereira Aranha
- Faculty of Pharmaceutical Sciences, Post Graduate Program in Biodiversity and Biotechnology of the Amazon (Bionorte), Federal University of Amazonas, Manaus, Amazonas, 69080-900, Brazil.
| | | | - Leilane Bentes de Sousa
- Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, 69080-900, Brazil.
| | - Emerson Lucena da Silva
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil.
| | - Felipe Pantoja Mesquita
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil.
| | - Waldireny C Rocha
- Health and Biotechnology Institute, Federal University of Amazonas, Coari, Amazonas, 69460-000, Brazil.
| | | | - Emerson Silva Lima
- Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, 69080-900, Brazil.
| | | | | | - Raquel Carvalho Montenegro
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil.
| | | |
Collapse
|
13
|
Natural Products as Inducers of Non-Canonical Cell Death: A Weapon against Cancer. Cancers (Basel) 2021; 13:cancers13020304. [PMID: 33467668 PMCID: PMC7830727 DOI: 10.3390/cancers13020304] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/09/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Anticancer therapeutic approaches based solely on apoptosis induction are often unsuccessful due to the activation of resistance mechanisms. The identification and characterization of compounds capable of triggering non-apoptotic, also called non-canonical cell death pathways, could represent an important strategy that may integrate or offer alternative approaches to the current anticancer therapies. In this review, we critically discuss the promotion of ferroptosis, necroptosis, and pyroptosis by natural compounds as a new anticancer strategy. Abstract Apoptosis has been considered the main mechanism induced by cancer chemotherapeutic drugs for a long time. This paradigm is currently evolving and changing, as increasing evidence pointed out that antitumor agents could trigger various non-canonical or non-apoptotic cell death types. A considerable number of antitumor drugs derive from natural sources, both in their naturally occurring form or as synthetic derivatives. Therefore, it is not surprising that several natural compounds have been explored for their ability to induce non-canonical cell death. The aim of this review is to highlight the potential antitumor effects of natural products as ferroptosis, necroptosis, or pyroptosis inducers. Natural products have proven to be promising non-canonical cell death inducers, capable of overcoming cancer cells resistance to apoptosis. However, as discussed in this review, they often lack a full characterization of their antitumor activity together with an in-depth investigation of their toxicological profile.
Collapse
|
14
|
Shu C, Yu X, Cheng S, Jing J, Hu C, Pang B. Pristimerin Suppresses Trophoblast Cell Epithelial-Mesenchymal Transition via miR-542-5p/EGFR Axis. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4659-4670. [PMID: 33173276 PMCID: PMC7646443 DOI: 10.2147/dddt.s274595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/18/2020] [Indexed: 12/31/2022]
Abstract
Background Ectopic pregnancy (EP) is an ectopic embryo implantation occurred outside the uterine cavity. Nowadays, more attention have garnered in fast and effective treatment with less side effects. Pristimerin is known as the clinical application for anti-cancer, and the effect on EP therapy is still unclear. Materials and Methods Trophoblast cell line HTR-8/SVneo was used; then, we performed cell counting kit-8 assay, wound healing assay, flow cytometry and real-time polymerase chain reaction analysis (RT-PCR) to detect the cell viability, migration ability, apoptosis and epithelial–mesenchymal transition (EMT) under pristimerin treatment. In addition, public bioinformatic database was used to discover the connection between molecular and genes. Finally, we used miRNA transfection and RT-PCR techniques to determine the underlying molecular mechanism. Results We revealed that pristimerin inhibited trophoblast cells proliferation, migration and EMT, while induced trophoblast cell apoptosis. Furthermore, expression of miR-542-5p, AGO2 and EGFR was suppressed in HTR-8/SVneo cells post pristimerin treatment, and miR-542-5p silence showed the same effect. Combing pristimerin treatment and miR-542-5p silence showed a synergistic action. Conclusion Pristimerin could be an effective treatment to block embryo implantation by miR-542-5p and EGFR down-regulation.
Collapse
Affiliation(s)
- Chang Shu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Xiaowei Yu
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Shihuan Cheng
- Department of Rehabilitation, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Jili Jing
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Cong Hu
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China.,Department of Rehabilitation, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| | - Bo Pang
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China.,Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin 130021, People's Republic of China
| |
Collapse
|
15
|
Cheng S, Zhang Z, Hu C, Xing N, Xia Y, Pang B. Pristimerin Suppressed Breast Cancer Progression via miR-542-5p/DUB3 Axis. Onco Targets Ther 2020; 13:6651-6660. [PMID: 32753899 PMCID: PMC7354954 DOI: 10.2147/ott.s257329] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer is one of the most common and malignant tumors in the world. Nowadays more attention has been garnered in pristimerin anti-cancer effects. Here, we illustrate the function and regulatory mechanism of pristimerin in breast cancer therapy. MATERIALS AND METHODS Breast cancer cell lines MCF-7, MDA-MB-231, and 4T1 were used. Cell Counting Kit-8 (CCK-8) assay was performed to evaluate proliferation viability of breast cancer cells under pristimerin treatment. Wound healing assay was used to examine the migration ability, cell cycle, and cell apoptosis detection were tested by flow cytometry. Bioinformatic analysis was used to find the underlying molecular and gene connected with pristimerin and breast cancer survival. Finally, we used transfection and real-time polymerase chain reaction analysis to confirm the mechanism. RESULTS We observed that pristimerin inhibited breast cancer cell viability, migration, and cell cycle, meanwhile induced cell apoptosis. In addition, under pristimerin treatment, miR-542-5p was up-regulated while DUB3 was down-regulated. Furthermore, bioinformatics analysis showed higher expression of DUB3 in breast cancer compared with normal tissue, also with poor prognosis. Overexpression miR-542-5p in breast cancer cells leads to a decrease in DUB3 level. The effect was obviously post pristimerin treatment and miR-542-5p overexpression. CONCLUSION Pristimerin inhibited breast cancer progression through DUB3 expression via a canonical miRNA-mediated mechanism.
Collapse
Affiliation(s)
- Shihuan Cheng
- Department of Rehabilitation, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Zhihong Zhang
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Cong Hu
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Na Xing
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Yan Xia
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Bo Pang
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| |
Collapse
|
16
|
Liu S, Dong Y, Wang Y, Hu P, Wang J, Wang RYL. Pristimerin exerts antitumor activity against MDA-MB-231 triple-negative breast cancer cells by reversing of epithelial-mesenchymal transition via downregulation of integrin β3. Biomed J 2020; 44:S84-S92. [PMID: 35652598 PMCID: PMC9038948 DOI: 10.1016/j.bj.2020.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/11/2020] [Accepted: 07/21/2020] [Indexed: 01/06/2023] Open
Abstract
Background Pristimerin, a natural flavonoid compound, has potential anti-tumor activities. These activities have been illustrated in various cancer cell lines, including MDA-MB-231 cells. MDA-MB-231 cells are a representative mesenchymal subtype of triple negative breast cancer (MES-TNBC) cell line. Currently, the main treatment for patients with advanced MES-TNBC is cytotoxic chemotherapy. We tried to examine the role and effect of pristimerin on epithelial–mesenchymal transition (EMT) in MDA-MB-231 cells. Methods The effects of pristimerin on the proliferation of MDA-MB-231 cells were investigated by cloning formation growth assay. In vitro transwell and adhesion assays were performed for cell invasion and adhesion. The expression levels of EMT markers in E-cadherin and N-cadherin were examined by western blotting. We also established overexpressed- and silenced-integrin β3 cell lines to evaluate the role of integrin β3 in mediating the EMT reversion events in MDA-MB-231 cells. Results Pristimerin inhibited cell proliferation, and its inhibitory effect was dose-dependent. We demonstrated that pristimerin reserved EMT by upregulating E-cadherin and downregulating N-cadherin expression. Meanwhile, we revealed that pristimerin inhibited mRNA and protein expression of integrin β3, which is a key heterodimeric transmembrane receptor associated with EMT. These inhibitory effects and reversion of EMT were enhanced when integrin β3 was knockdown in MDA-MB-231 cells, while the overexpression of integrin β3 attenuated these effects. In vivo studies using xenograft mouse model demonstrated that pristimerin inhibited tumor growth. Conclusions Our findings provide important insights into the effects of pristimerin on inhibiting cancer progression and EMT reversion by suppression of integrin β3.
Collapse
|
17
|
Yan F, Liao R, Silva M, Li S, Jiang Y, Peng T, Lazarovici P, Zheng W. Pristimerin-induced uveal melanoma cell death via inhibiting PI3K/Akt/FoxO3a signalling pathway. J Cell Mol Med 2020; 24:6208-6219. [PMID: 32347651 PMCID: PMC7294164 DOI: 10.1111/jcmm.15249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/02/2020] [Accepted: 03/17/2020] [Indexed: 01/13/2023] Open
Abstract
Uveal melanoma (UM) is a highly invasive intraocular malignancy with high mortality. Presently, there is no FDA‐approved standard for the treatment of metastatic UM. Pristimerin is a natural quinine methide triterpenoid compound with anti‐angiogenic, anti‐cancer and anti‐inflammatory activities. However, Pristimerin potential cytotoxic effect on UM was poorly investigated. In the present study, we found the migration and invasion of UM‐1 cells were inhibited by Pristimerin which also caused a rapid increase of ROS, decreased mitochondrial membrane potential, induced the accumulation of cells in G0/G1 phase, ending with apoptotic cell death. Pristimerin inhibited Akt and FoxO3a phosphorylation and induced nuclear accumulation of FoxO3a in UM‐1 cells, increased the expression of pro‐apoptotic proteins Bim、p27Kip1, cleaved caspase‐3, PARP and Bax, and decreased the expression of Cyclin D1 and Bcl‐2. LY294002 or Akt‐siRNA inhibited the PI3K/Akt/FoxO3a pathway and promoted the Pristimerin‐induced apoptosis, while Pristimerin effects were partially abolished in FoxO3a knockdown UM‐1 cell cultures. Taken together, present results showed that Pristimerin induced apoptotic cell death through inhibition of PI3K/Akt/FoxO3a pathway in UM‐1 cells. These findings indicate that Pristimerin may be considered as a potential chemotherapeutic agent for patients with UM.
Collapse
Affiliation(s)
- Fengxia Yan
- Faculty of Health Sciences, University of Macau, Macau, China.,School of Medical Science, Jinan University, Guangzhou, China
| | - Rifang Liao
- Faculty of Health Sciences, University of Macau, Macau, China.,Department of pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Marta Silva
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Shuai Li
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Yizhou Jiang
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Tangming Peng
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Philip Lazarovici
- Faculty of Medicine, School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
18
|
Li G, Li B, Song J, Wang N, Gao Z. Endomorphin-2 Analog Inhibits the Growth of DLD-1 and RKO Human Colon Cancer Cells by Inducing Cell Apoptosis. Med Sci Monit 2020; 26:e921251. [PMID: 32336747 PMCID: PMC7199432 DOI: 10.12659/msm.921251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background In developed countries, colon cancer is a leading cause of cancer-associated mortality. Dietary changes have resulted in an increased incidence of colon cancer in Asia. This study aimed to investigate the effects of the structural analog of endomorphin-2 (H-Tyr-Pro-Phe-Phe-NH2) on human colon cancer cells in vitro. Material/Methods Human DLD-1 and RKO colon cancer cells and CCD-18Co normal human colonic fibroblasts were treated with increasing doses of the structural analog of endomorphin-2. Cells underwent the MTT assay, fluorescence confocal flow cytometry, and Hoechst 33258 staining to investigate cell proliferation, the cell cycle, and apoptosis. Western blot was used to measure the expression levels of poly(ADP-ribose) polymerase-1 (PARP-1), cytochrome c, caspase-3, and caspase-9. The 2′,7′-dichlorofluorescein diacetate (DCFH-DA) fluorescence method measured reactive oxygen species (ROS). Results Cell proliferation of DLD-1 and RKO cells was inhibited by the endomorphin-2 analog in a dose-dependent manner, and a 100 μM dose reduced DLD-1 and RKO cell proliferation by 28% and 23%, respectively, at 72 h. Endomorphin-2 analog induced cell apoptosis and the generation of ROS, activated caspase-3 and caspase-9, and increased the levels of p53 and cytochrome c release, and down-regulated of Akt activation in DLD-1 and RKO cells in a dose-dependent manner. Treatment of the DLD-1 and RKO cells with the endomorphin-2 analog increased the expression of Bax and reduced the expression of Bcl-2. Conclusions Endomorphin-2 analog inhibited colon cancer cell proliferation, activated apoptosis, and down-regulated Akt phosphorylation of human DLD-1 and RKO colon cancer cells in vitro in a dose-dependent manner.
Collapse
Affiliation(s)
- Guanghua Li
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Bo Li
- Department of Electrocardiography, Peoples' Hospital of Zhangqiu, Jinan, Shandong, China (mainland)
| | - Jingang Song
- Department of General Surgery, Dezhou Peoples' Hospital, Dezhou, Shandong, China (mainland)
| | - Na Wang
- Department of Oncology, Peoples' Hospital of Zhangqiu, Jinan, Shandong, China (mainland)
| | - Zhuanglei Gao
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
19
|
Hayashi D, Shirai T, Terauchi R, Tsuchida S, Mizoshiri N, Mori Y, Arai Y, Mazda O, Kubo T. Pristimerin inhibits the proliferation of HT1080 fibrosarcoma cells by inducing apoptosis. Oncol Lett 2020; 19:2963-2970. [PMID: 32218852 DOI: 10.3892/ol.2020.11405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/06/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrosarcoma is a soft tissue sarcoma that is classified as a rare cancer. Therefore, no standard anti-tumor drug therapy has been established for fibrosarcoma. Although pristimerin (PM) has been reported to exert an anti-tumor effect on various types of cancer, no studies have examined the therapeutic effect of PM on soft tissue sarcoma. The purpose of the current study was to investigate the anti-tumor effect of PM on human fibrosarcoma cells (HT1080). The present study examined the cell viability, IC50 values and ability to induce apoptosis of PM in HT1080 and normal human dermal fibroblast (aHDF) cells. The effect of PM on the following signaling pathways associated with cell proliferation was also evaluated: AKT and mitogen-activated protein kinase (MAPK). Using mice subcutaneously transplanted with fibrosarcoma cells, the effect of PM treatment was investigated on tumor growth inhibition, body weight and liver and renal function. The results revealed that PM administration reduced cell viability and induced apoptosis in a dose-dependent matter. In HT1080 cells, the IC50 value of PM was 0.16 µM at 24 h and 0.13 µM at 48 h. PM treatment also decreased the levels of phosphorylated AKT, mTOR, NF-κB and phosphorylated ERK in a dose-dependent manner. In the PM injection group, the increase in tumor volume was significantly reduced and the effect on weight loss and liver and renal function were revealed to be insignificant. PM exerted little effect on normal human dermal fibroblasts and was highly effective against human fibrosarcoma cells. The results indicated that PM may be used as a potential therapeutic agent against fibrosarcoma.
Collapse
Affiliation(s)
- Daichi Hayashi
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toshiharu Shirai
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ryu Terauchi
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shinji Tsuchida
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Naoki Mizoshiri
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yuki Mori
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yuji Arai
- Department of Sports and Parasports Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toshikazu Kubo
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
20
|
Long Y, Wang D. Inhibition of Colon Cancer Cell Growth by Imidazole Through Activation of Apoptotic Pathway. Med Sci Monit 2019; 25:7597-7604. [PMID: 31597910 PMCID: PMC6798726 DOI: 10.12659/msm.917779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background This study aimed to investigate the inhibitory effect of imidazole on colon cancer cell proliferation and understand the mechanism involved. Material/Methods MTT assay and flow cytometry using Hoechst 33258 staining were used to assess cell proliferation and morphology, respectively. Changes in protein expression was determined by western blotting assay. The reactive oxygen species (ROS) production in DLD-1 cells was analyzed by flow cytometry using DCFH-DA (2′,7′-dichlorofluorescein diacetate) stain. Results DLD-1 and HCT-116 cell viability was suppressed by imidazole in a concentration-based manner. At the concentration of 36 μM, imidazole reduced DLD-1 and HCT-116 cell viability to 22% and 28%, respectively. Treatment with imidazole led to chromatin material condensation, detaching of cells, and apoptotic nuclei. In imidazole treated cells, the G1/G0 phase cell proportion increased, whereas in the S and G2/M phases the cell proportion decreased. Imidazole treatment of DLD-1 cells markedly promoted activation of caspase-3, caspase-8, and caspase-9. The level of cleaved PARP1 was also upregulated in DLD-1 cells with imidazole treatment. Treatment of DLD-1 cells with imidazole suppressed Bcl-2 and promoted Bax, p53, and cytc expression. The Akt activation was suppressed by imidazole treatment in DLD-1 cells. ROS generation in DLD-1 cells was enhanced markedly by treatment with imidazole. Conclusions The present study demonstrated that imidazole inhibited colon cancer cell viability through activation of apoptosis and cell cycle arrest by increasing the generation of ROS, caspase activation, and apoptotic protein expression. Therefore, imidazole can act as a therapeutic molecule for the treatment of colon cancer.
Collapse
Affiliation(s)
- Yaxin Long
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China (mainland)
| | - Duo Wang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
21
|
Shen Y, Chen BL, Zhang QX, Zheng YZ, Fu Q. Traditional uses, secondary metabolites, and pharmacology of Celastrus species - a review. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:111934. [PMID: 31129308 DOI: 10.1016/j.jep.2019.111934] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plants of genus Celastrus (Celastraceae) have been widely used in traditional Chinese medicine (TCM) and Indian medicine to treat cognitive dysfunction, epilepsy, insomnia, rheumatism, gout, and dyspepsia for thousands of years. AIM OF STUDY We critically summarized the current evidence on the botanic characterization and distribution, ethnopharmacology, secondary metabolites, pharmacological activities, qualitative and quantitative analysis, and toxicology of Celastrus species to provide perspectives for developing more attractive pharmaceuticals of plant origin. MATERIALS AND METHODS The relevant information on Celastrus species was gathered from worldwide accepted scientific databases via electronic search (Web of Science, SciFinder, PubMed, Elsevier, SpringerLink, Wiley Online, China Knowledge Resource Integrated, and Google Scholar). Information was also obtained from the literature and books as well as PhD and MSc dissertations. Plant names were validated by "The Plant List" (www.theplantlist.org). RESULTS Comprehensive analysis of the above mentioned databases and other sources confirmed that ethnomedical uses of plants of Celastrus genus had been recorded in China, India, and other countries in Southern Asia. The phytochemical investigation revealed the presence of β-dihydroagarofuranoids, diterpenoids, triterpenoids, tetraterpenes, phenylpropanoids, alkaloids, flavonoids, lignans, and others. The crude extracts and isolated constituents have exhibited a wide range of in vitro and in vivo pharmacological effects, including antitumor, cytotoxic, insecticidal, antimicrobial, anti-rheumatoid arthritis (RA), anti-inflammatory, anti-ageing and antioxidative, and neuroprotective activities. CONCLUSION Plants of genus Celastrus have been confirmed to show a strong potential for therapeutic and health-maintaining effects, in light of their long traditional use and the phytochemical and pharmacological studies summarized here. Currently, pharmacological studies of this genus mainly focus on Celastrus paniculatus Willd. and Celastrus orbiculatus Thunb. Therefore, more pharmacological investigations should be implemented to support traditional uses of other medicinal plants of the genus Celastrus. Moreover, studies on the toxicity, bioavailability, and pharmacokinetics, in addition to clinical trials, are indispensable for assessing the safety and efficacy of the secondary metabolites or extracts obtained from plants belonging to this genus.
Collapse
Affiliation(s)
- Yue Shen
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture Rural Affairs, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Bi-Lian Chen
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture Rural Affairs, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Qin-Xiu Zhang
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, 610072, China
| | - Yu-Zhong Zheng
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Qiang Fu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture Rural Affairs, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China.
| |
Collapse
|
22
|
Xie X, Xie S, Xie C, Fang Y, Li Z, Wang R, Jiang W. Pristimerin attenuates cell proliferation of uveal melanoma cells by inhibiting insulin-like growth factor-1 receptor and its downstream pathways. J Cell Mol Med 2019; 23:7545-7553. [PMID: 31508890 PMCID: PMC6815816 DOI: 10.1111/jcmm.14623] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/16/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022] Open
Abstract
Uveal melanoma (UM) has a high mortality rate due to liver metastasis. The insulin‐like growth factor‐1 receptor (IGF‐1R) is highly expressed in UM and has been shown to be associated with hepatic metastases. Targeting IGF signalling may be considered as a promising approach to inhibit the process of metastatic UM cells. Pristimerin (PRI) has been demonstrated to inhibit the growth of several cancer cells, but its role and underlying mechanisms in the IGF‐1‐induced UM cell proliferation are largely unknown. The present study examined the anti‐proliferative effect of PRI on UM cells and its possible role in IGF‐1R signalling transduction. MTT and clonogenic assays were used to determine the role of PRI in the proliferation of UM cells. Flow cytometry was performed to detect the effect of PRI on the cell cycle distribution of UM cells. Western blotting was carried out to assess the effects of PRI and IGF‐1 on the IGF‐1R phosphorylation and its downstream targets. The results indicated that IGF‐1 promoted the UM cell proliferation and improved the level of IGF‐1R phosphorylation, whereas PRI attenuated the effect of IGF‐1. Interestingly, PRI could not only induce the G1 phase accumulation and reduce the G2 phase induced by IGF‐1, but also could stimulate the expression of p21 and inhibit the expression of cyclin D1. Besides, PRI could attenuate the phosphorylations of Akt, mTOR and ERK1/2 induced by IGF‐1. Furthermore, the molecular docking study also demonstrated that PRI had potential inhibitory effects on IGF‐1R. Taken together, these results indicated that PRI could inhibit the proliferation of UM cells through down‐regulation of phosphorylated IGF‐1R and its downstream signalling.
Collapse
Affiliation(s)
- Xinshu Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Saisai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Changying Xie
- Affiliated Hosptial of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yuanying Fang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Zhifeng Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, China
| | - Wei Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, China
| |
Collapse
|
23
|
Hodon J, Borkova L, Pokorny J, Kazakova A, Urban M. Design and synthesis of pentacyclic triterpene conjugates and their use in medicinal research. Eur J Med Chem 2019; 182:111653. [PMID: 31499360 DOI: 10.1016/j.ejmech.2019.111653] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 01/12/2023]
Abstract
Triterpenoids are natural products from plants and many other organisms that have various biological activities, such as antitumor, antiviral, antimicrobial, and protective activities. This review covers the synthesis and biological evaluation of pentacyclic triterpene (PT) conjugates with other molecules that have been found to increase the IC50 or improve the pharmacological profile of the parent PT. Some of these molecules are designed to target specific proteins or cellular organelles, which has resulted in highly selective lead structures for drug development. Other PT conjugates are useful for investigating their mechanism of action. This concept has been very successful: 1) Many compounds, especially mitochondria-targeting PT conjugates, have reached a selective cytotoxicity at low nanomolar concentrations in cancer cells. 2) A number of PT conjugates have had high activity against HIV or the influenza virus. 3) Fluorescent PT conjugates have been able to visualize the PT in living cells, which has allowed quantification of the uptake and distribution of the PT within the cell. 4) Biotinylated PT conjugates have been used to identify target proteins, which may help to show their mechanism of action. 5) A large number of PT conjugates with polyethylene glycol (PEG), polyamines, etc. form nanometer-sized micelles that have a much better pharmacological profile than the PT alone. In summary, the connection of a PT to an appropriate modifying molecule has resulted in extremely useful semisynthetic compounds with a high potential to treat cancer or viral infections or compounds that are useful for the study of the mechanism of action of PTs at the molecular level.
Collapse
Affiliation(s)
- Jiri Hodon
- Department of Organic Chemistry, Faculty of Science, Palacky University, 17. listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Lucie Borkova
- Department of Organic Chemistry, Faculty of Science, Palacky University, 17. listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Jan Pokorny
- Department of Organic Chemistry, Faculty of Science, Palacky University, 17. listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Anna Kazakova
- Department of Organic Chemistry, Faculty of Science, Palacky University, 17. listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Milan Urban
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hnevotinská 5, 779 00, Olomouc, Czech Republic.
| |
Collapse
|
24
|
Tan Y, Hou L, Zhou Y, Mo Z. Inhibition of Colon Cancer Cell Viability and Tumor Growth by Benzoximemethyl Amine Through Apoptosis. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.716.723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
25
|
Chen X, Tang WJ, Shi JB, Liu MM, Liu XH. Therapeutic strategies for targeting telomerase in cancer. Med Res Rev 2019; 40:532-585. [PMID: 31361345 DOI: 10.1002/med.21626] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
Telomere and telomerase play important roles in abnormal cell proliferation, metastasis, stem cell maintenance, and immortalization in various cancers. Therefore, designing of drugs targeting telomerase and telomere is of great significance. Over the past two decades, considerable knowledge regarding telomere and telomerase has been accumulated, which provides theoretical support for the design of therapeutic strategies such as telomere elongation. Therefore, the development of telomere-based therapies such as nucleoside analogs, non-nucleoside small molecules, antisense technology, ribozymes, and dominant negative human telomerase reverse transcriptase are being prioritized for eradicating a majority of tumors. While the benefits of telomere-based therapies are obvious, there is a need to address the limitations of various therapeutic strategies to improve the possibility of clinical applications. In this study, current knowledge of telomere and telomerase is discussed, and therapeutic strategies based on recent research are reviewed.
Collapse
Affiliation(s)
- Xing Chen
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Wen-Jian Tang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Jing Bo Shi
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Ming Ming Liu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Xin-Hua Liu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
26
|
Sun JM, Xu HT, Zhao L, Zhang YB, Kang PC, Song ZF, Liu HS, Cui YF. Induction of cell-cycle arrest and apoptosis in human cholangiocarcinoma cells by pristimerin. J Cell Biochem 2019; 120:12002-12009. [PMID: 30825242 DOI: 10.1002/jcb.28485] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 01/24/2023]
Abstract
Pristimerin, a triterpenoid isolated from Celastraceae and Hippocrateaceae, is known to induce cytotoxicity in several cancer cell lines. However, whether pristimerin can induce apoptosis in cholangiocarcinoma cells and the underlying mechanism remain unexplored. We assessed the function of human cholangiocarcinoma QBC and RBE cell lines using various experimental methods such as the cell viability assay to elucidate the viability of cells, flow cytometry to detect the death rate of cells, and Western blot analysis to evaluate the expression of cell cycle-related proteins and autophagy-related proteins. Human cholangiocarcinoma QBC cells were transplanted to nude mice to establish an animal model, and the effect of pristimerin on tumor growth in this model was observed. QBC and RBE cell lines treated with pristimerin (0, 5, 10, and 20 μmol/L) demonstrated the induction of apoptosis in a dose-dependent manner. The cell viability assay revealed a reduction in the cell viability with an increase in the pristimerin concentration. Similarly, flow cytometry revealed a gradual increase in the cell death rate with an increase in the pristimerin concentration. In addition, pristimerin significantly lowered the expression of apoptosis-related proteins (Bcl-2, Bcl-xL, and procaspase-3), but increased the Bax expression. Furthermore, pristimerin resulted in the G0/G1 cell-cycle arrest, reducing the expression of cell cycle-related proteins (cyclin E, CDK2, and CDK4), and increased the expression of autophagy-related proteins (LC3) in QBC cell line. Treatment with pristimerin could inhibit tumor growth in the nude mouse model. Overall, this study suggests the potential effect of pristimerin on the cell-cycle arrest and apoptosis in human cholangiocarcinoma cells.
Collapse
Affiliation(s)
- Jian-Min Sun
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hai-Tao Xu
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Liang Zhao
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yu-Bao Zhang
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Peng-Cheng Kang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zeng-Fu Song
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hai-Shi Liu
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yun-Fu Cui
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
27
|
Pristimerin inhibits glioma progression by targeting AGO2 and PTPN1 expression via miR-542-5p. Biosci Rep 2019; 39:BSR20182389. [PMID: 31015365 PMCID: PMC6522714 DOI: 10.1042/bsr20182389] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/28/2019] [Accepted: 04/15/2019] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiform is the most common and malignant primary tumor of the central nervous system in adults, the high recurrence rate and poor prognosis are critical priorities. Pristimerin is a naturally occurring quinone methide triterpenoid isolated from the Celastraceae and Hippocrateaceae families. Its anticancer effects have garnered considerable attention; nonetheless, the mechanisms of action remain unknown. To predict the hub genes of pristimerin, PharmMapper and the Coremine database were used to identify 13 potential protein targets; protein-protein interaction, for which functional enrichment analyses were performed. Compound-target, target-pathway, and compound-target-pathway networks were constructed using Cytoscape. Biological process analysis first revealed that enrichment of these target genes correlated with negative regulation of symbiont growth in the host, and regulation of chronic inflammatory response to antigenic stimulus. Survival analysis in cBioPortal showed that protein tyrosine phosphatase, non-receptor type 1 (PTPN1) and Argonaute 2 (AGO2) might be involved in the carcinogenesis, invasion, or recurrence of diffuse glioma. In addition, we observed that low-dose pristimerin inhibited the viability of glioma cells, while miR-542-5p in vitro; and reduced PTPN1 expression. Notably, high-dose pristimerin induced apoptosis. Furthermore, miR-542-5p silence with siRNA in glioma cells lead to the elevation in AGO2, and decreased PTPN1 level. The effect was obviously post pristimerin treatment and miR-542-5p suppression. In conclusion, pristimerin inhibited glioma progression through AGO2 and PTPN1 expression via a canonical miRNA-mediated mechanism.
Collapse
|
28
|
Rodrigues ACBDC, Oliveira FPD, Dias RB, Sales CBS, Rocha CAG, Soares MBP, Costa EV, Silva FMAD, Rocha WC, Koolen HHF, Bezerra DP. In vitro and in vivo anti-leukemia activity of the stem bark of Salacia impressifolia (Miers) A. C. Smith (Celastraceae). JOURNAL OF ETHNOPHARMACOLOGY 2019; 231:516-524. [PMID: 30445109 DOI: 10.1016/j.jep.2018.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/18/2018] [Accepted: 11/03/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salacia impressifolia (Miers) A. C. Smith (family Celastraceae) is a traditional medicinal plant found in the Amazon Rainforest known as "miraruíra", "cipó-miraruíra" or "panu" and is traditionally used to treat dengue, flu, inflammation, pain, diabetes, male impotency, renal affections, rheumatism and cancer. AIM OF THE STUDY The aim of this study was to investigate in vitro and in vivo anti-leukemia activity of the stem bark of S. impressifolia in experimental models. MATERIALS AND METHODS The in vitro cytotoxic activity of extracts, fractions and quinonemethide triterpenes (22-hydroxytingenone, tingenone and pristimerin) from the stem bark of S. impressifolia in cultured cancer cells was determined. The in vivo antitumor activity of the ethyl acetate extract (EAE) and of its fraction (FEAE.3) from the stem bark of S. impressifolia was assessed in C.B-17 severe combined immunodeficient (SCID) mice engrafted with human promyelocytic leukemia HL-60 cells. RESULTS The extract EAE, its fraction FEAE.3, and quinonemethide triterpenes exhibited potent cytotoxicity against cancer cell lines, including in vitro anti-leukemia activity against HL-60 and K-562 cells. Moreover, extract EAE and its fraction FEAE.3 inhibited the in vivo development of HL-60 cells engrafted in C.B-17 SCID mice. Tumor mass inhibition rates were measured as 40.4% and 81.5% for the extract EAE (20 mg/kg) and for its fraction FEAE.3 (20 mg/kg), respectively. CONCLUSIONS Ethyl acetate extract and its fraction from the stem bark of S. impressifolia exhibit in vitro and in vivo anti-leukemia activity that can be attributed to their quinonemethide triterpenes. These data confirm the ethnopharmacological use of this species and may contribute to the development of a novel anticancer herbal medicine.
Collapse
Affiliation(s)
| | - Felipe P de Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil
| | - Caroline B S Sales
- Department of Biomorphology, Institute of Health Sciences, Federal University of Bahia, Salvador (UFBA), Bahia 40110-902, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil; Center of Biotechnology and Cell Therapy, Hospital São Rafael, Salvador, Bahia 41253-190, Brazil
| | - Emmanoel V Costa
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus, Amazonas 69077-000, Brazil
| | - Felipe M A da Silva
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus, Amazonas 69077-000, Brazil
| | - Waldireny C Rocha
- Health and Biotechnology Institute, Federal University of Amazonas (UFAM), Coari, Amazonas 69460-000, Brazil
| | - Hector H F Koolen
- Metabolomics and Mass Spectrometry Research Group, Amazonas State University (UEA), Manaus, Amazonas 690065-130, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia 40296-710, Brazil.
| |
Collapse
|
29
|
Zhang Y, Wang J, Hui B, Sun W, Li B, Shi F, Che S, Chai L, Song L. Pristimerin enhances the effect of cisplatin by inhibiting the miR‑23a/Akt/GSK3β signaling pathway and suppressing autophagy in lung cancer cells. Int J Mol Med 2019; 43:1382-1394. [PMID: 30664149 PMCID: PMC6365073 DOI: 10.3892/ijmm.2019.4057] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/31/2018] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is a common type of cancer with a high mortality rate in China. Cisplatin (Cis) is one of the most effective broad‑spectrum chemotherapeutic drugs for the treatment of advanced lung cancer. However, Cis resistance remains an obstacle in the treatment of advanced lung cancer. Pristimerin (Pris), a naturally occurring triterpenoid quinone compound, not only possesses anticancer properties, but also enhances chemosensitivity. Therefore, the present study aimed to investigate whether Pris can enhance the chemosensitivity of lung cancer cells to Cis and identify the underlying mechanism. A Cell Counting kit‑8 and flow cytometry were used to determine cell viability, cell cycle progression and apoptosis in A549 and NCI‑H446 cells. Western blotting was used to determine cell apoptosis‑related, cell cycle‑related and autophagy‑related proteins. The results showed that Pris inhibited cell proliferation, and induced G0/G1 arrest and cell apoptosis in A549 and NCI‑H446 cells. The western blotting revealed that Pris effectively synergized with Cis to induce cell apoptosis by inhibiting the microRNA‑23a/Akt/glycogen synthase kinase 3β signaling pathway and suppressing autophagy. In vivo xenograft experiments confirmed that Pris effectively synergized with Cis to suppress tumor growth. Collectively, these results indicate that Pris synergized with Cis and that this may be a potential therapeutic strategy to overcome lung cancer.
Collapse
Affiliation(s)
- Yingbing Zhang
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiquan Wang
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Beina Hui
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenze Sun
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Bin Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Fan Shi
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shaomin Che
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Linyan Chai
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Liping Song
- Department of Radiation Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
30
|
Wu H, Li L, Ai Z, Yin J, Chen L. Pristimerin induces apoptosis of oral squamous cell carcinoma cells via G 1 phase arrest and MAPK/Erk1/2 and Akt signaling inhibition. Oncol Lett 2019; 17:3017-3025. [PMID: 30854080 DOI: 10.3892/ol.2019.9903] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 11/12/2018] [Indexed: 01/04/2023] Open
Abstract
Pristimerin is an active compound isolated from the traditional Chinese herbs Celastraceae and Hippocrateaceae. It has been reported to exert antitumor effects under experimental and clinical conditions; however, the antitumor effects and underlying mechanisms of pristimerin in oral cancer cells have not yet been identified. In the present study, the anticancer potential of pristimerin was investigated in two oral squamous cell carcinoma (OSCC) cell lines, CAL-27 and SCC-25. Results demonstrated that pristimerin was toxic against the two cell lines, and exhibited inhibitory effects against proliferation. Furthermore, pristimerin exhibited a more potent anti-proliferative activity in CAL-27 and SCC-25 cells than the common chemotherapy drugs cisplatin and 5-fluorouracil. In addition, cell cycle distribution analysis revealed that G0/G1 phase arrest was induced following pristimerin treatment in CAL-27 and SCC-25 cells, which was strongly associated with upregulation of p21 and p27, coupled with downregulation of cyclin D1 and cyclin E. Meanwhile, pristimerin induced significant apoptosis of CAL-27 and SCC-25 cells, alongside decreased levels of caspase-3 and specific cleavage of poly (ADP-ribose) polymerase. These effects were associated with inhibition of the mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 and protein kinase B signaling pathways. With regards to these results, pristimerin may be considered a potent novel active substance for the treatment of OSCC.
Collapse
Affiliation(s)
- Haiyan Wu
- Department of Pathophysiology, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Long Li
- Department of Stomatology, Shekou People's Hospital, Shenzhen, Guangdong 518067, P.R. China
| | - Zhengdong Ai
- Department of Pathophysiology, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Jingyi Yin
- Department of Pathophysiology, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Li Chen
- Department of Pathophysiology, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
31
|
Lee Y, Na J, Lee MS, Cha EY, Sul JY, Park JB, Lee JS. Combination of pristimerin and paclitaxel additively induces autophagy in human breast cancer cells via ERK1/2 regulation. Mol Med Rep 2018; 18:4281-4288. [PMID: 30221728 PMCID: PMC6172393 DOI: 10.3892/mmr.2018.9488] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/14/2018] [Indexed: 12/25/2022] Open
Abstract
Pristimerin, a quinonemethide triterpenoid, has demonstrated anticancer activity against a number of types of cancer, including breast cancer. However, its mechanism of action remains unclear. The present study investigated the autophagy-induced anticancer efficacy of pristimerin on MDA-MB-231 human breast cancer cells. Pristimerin inhibited the growth of these cells in a concentration-dependent manner. Treatment with pristimerin dose-dependently induced an increase of light chain 3B (LC3-II), whereas autophagy inhibitor 3-methyladenine (3-MA) inhibited pristimerin-induced LC3-II accumulation and cytotoxic effects. Autophagy was also activated by paclitaxel as observed by an elevated LC3-II level. Although 24 µM paclitaxel induced autophagy without cytotoxicity, combined with pristimerin it additively induced cell growth inhibition and autophagy induction. Autophagy induction was measured with an autophagy detection kit and LC3-II levels were monitored by western blot analysis. Treatment with 3-MA inhibited LC3-II accumulation and cell death induced by a combination of paclitaxel and pristimerin. Pristimerin and paclitaxel inhibited extracellular signal-regulated kinase (ERK)1/2/p90RSK signaling, consistent with autophagy indicators, namely p62 degradation and beclin 1 expression. In addition, ERK activator ceramide C6 treatment suppressed the LC3-II levels induced by a combination of paclitaxel and pristimerin. These results suggested that exposure to pristimerin induced autophagic cell death, whereas a combination treatment of pristimerin and paclitaxel resulted in an additive effect on ERK-dependent autophagic cell death.
Collapse
Affiliation(s)
- Younju Lee
- Department of Surgery, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| | - Jinuk Na
- Department of Surgery, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| | - Myung Sun Lee
- Biomedical Research Institute, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| | - Eun Young Cha
- Biomedical Research Institute, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| | - Ji Young Sul
- Department of Surgery, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| | - Jun Beom Park
- Department of Surgery, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| | - Jin Sun Lee
- Department of Surgery, Chungnam National University Hospital, Jung‑gu, Daejeon 35015, Republic of Korea
| |
Collapse
|
32
|
Structure-Based Classification and Anti-Cancer Effects of Plant Metabolites. Int J Mol Sci 2018; 19:ijms19092651. [PMID: 30200668 PMCID: PMC6163735 DOI: 10.3390/ijms19092651] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 12/28/2022] Open
Abstract
A variety of malignant cancers affect the global human population. Although a wide variety of approaches to cancer treatment have been studied and used clinically (surgery, radiotherapy, chemotherapy, and immunotherapy), the toxic side effects of cancer therapies have a negative impact on patients and impede progress in conquering cancer. Plant metabolites are emerging as new leads for anti-cancer drug development. This review summarizes these plant metabolites with regard to their structures and the types of cancer against which they show activity, organized by the organ or tissues in which each cancer forms. This information will be helpful for understanding the current state of knowledge of the anti-cancer effects of various plant metabolites against major types of cancer for the further development of novel anti-cancer drugs.
Collapse
|
33
|
Hao X, Yuan J, Xu Y, Wang Z, Hou J, Hu T. In vitro inhibitory effects of pristimerin on human liver cytochrome P450 enzymes. Xenobiotica 2018; 48:1185-1191. [PMID: 28385095 DOI: 10.1080/00498254.2017.1316886] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
1.Pristimerin (PTM) is a biological component isolated from Chinese herbal plant Celastrus and Maytenus spp. and it possesses numerous pharmacological activities. However, whether PTM affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. 2.In this study, the inhibitory effects of PTM on the eight human liver CYP isoforms (i.e. 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19 and 2C8) were investigated in vitro using human liver microsomes (HLMs). 3.The results showed that PTM inhibited the activity of CYP1A2, 3A4 and 2C9, with IC 50 values of 21.74, 15.88 and 16.58 μM, respectively, but that other CYP isoforms were not affected. Enzyme kinetic studies showed that PTM was not only a non-competitive inhibitor of CYP3A4, but also a competitive inhibitor of CYP1A2 and 2C9, with Ki values of 7.33, 11.60 and 8.09 μM, respectively. In addition, PTM is a time-dependent inhibitor for CYP3A4 with Kinact /KI value of 0.049/11.62 μM-1 min-1. 4.The in vitro studies of PTM with CYP isoforms indicate that PTM has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP1A2, 3A4 and 2C9. Further clinical studies are needed to evaluate the significance of this interaction.
Collapse
Affiliation(s)
- Xiaoyi Hao
- a Cangzhou People's Hospital , Cangzhou , China
| | | | - Yansen Xu
- a Cangzhou People's Hospital , Cangzhou , China
| | - Zhao Wang
- a Cangzhou People's Hospital , Cangzhou , China
| | | | - Tao Hu
- a Cangzhou People's Hospital , Cangzhou , China
| |
Collapse
|
34
|
Yousef BA, Hassan HM, Zhang LY, Jiang ZZ. Pristimerin exhibits in vitro and in vivo anticancer activities through inhibition of nuclear factor-кB signaling pathway in colorectal cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 40:140-147. [PMID: 29496166 DOI: 10.1016/j.phymed.2018.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 12/23/2017] [Accepted: 01/14/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignancies associated with high mortality rate worldwide. We previously reported that pristimerin inhibits cell growth and induces apoptosis in CRC cells. HYPOTHESIS/PURPOSE To further understand the molecular mechanism by which pristimerin elicits its anticancer activities on colon cancer cells, we investigated its effect on nuclear factor-κB (NF-κB) signaling pathway. STUDY DESIGN This study consisted of both in vitro and in vivo experiments involving HCT-116 cell line and xenograft mouse model. Molecular techniques such as qRT-PCR, western blotting and immunofluorescence were used to demonstrate pristimerin in vitro effect on NF-κB signaling pathway; whereas it's in vivo activity was analyzed by western blot and immunohistochemistry on tumor tissues. RESULTS Our in vitro results on HCT-116 cells showed that pristimerin inhibited IKK phosphorylation, IкB-α degradations and IкB-α phosphorylation in both dose- and time- dependent manners, which caused suppression of NF-кB p65 phosphorylation, nuclear translocation and accumulation of NF-кB. Moreover, pristimerin was found to inhibit both constitutive activated-NF-кB and tumor necrosis factor-α (TNF-α)- and lipopolysaccharide (LPS)-induced activation of NF-кB signaling pathway. Furthermore, our in vivo results on xenograft animal model revealed that pristimerin inhibited tumor growth mainly through suppressing NF-кB activity in tumor tissues. CONCLUSION Pristimerin antitumor activities were mainly mediated through inhibition of NF-кB signaling pathway in colon tumor cells. These findings further explain that pristimerin has the therapeutic potential for targeting colon cancer.
Collapse
Affiliation(s)
- Bashir A Yousef
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Jiangsu Province, Nanjing 210009, PR China; Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan.
| | - Hozeifa M Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Jiangsu Province, Nanjing 210009, PR China; Department of Pharmacology, Faculty of Pharmacy, University of Gezira, Wad-Medani, Sudan
| | - Lu-Yong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Jiangsu Province, Nanjing 210009, PR China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, PR China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhen-Zhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Jiangsu Province, Nanjing 210009, PR China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, PR China.
| |
Collapse
|
35
|
Ganesan K, Xu B. Telomerase Inhibitors from Natural Products and Their Anticancer Potential. Int J Mol Sci 2017; 19:ijms19010013. [PMID: 29267203 PMCID: PMC5795965 DOI: 10.3390/ijms19010013] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/10/2017] [Accepted: 12/19/2017] [Indexed: 12/25/2022] Open
Abstract
Telomeres and telomerase are nowadays exploring traits on targets for anticancer therapy. Telomerase is a unique reverse transcriptase enzyme, considered as a primary factor in almost all cancer cells, which is mainly responsible to regulate the telomere length. Hence, telomerase ensures the indefinite cell proliferation during malignancy—a hallmark of cancer—and this distinctive feature has provided telomerase as the preferred target for drug development in cancer therapy. Deactivation of telomerase and telomere destabilization by natural products provides an opening to succeed new targets for cancer therapy. This review aims to provide a fundamental knowledge for research on telomere, working regulation of telomerase and its various binding proteins to inhibit the telomere/telomerase complex. In addition, the review summarizes the inhibitors of the enzyme catalytic subunit and RNA component, natural products that target telomeres, and suppression of transcriptional and post-transcriptional levels. This extensive understanding of telomerase biology will provide indispensable information for enhancing the efficiency of rational anti-cancer drug design.
Collapse
Affiliation(s)
- Kumar Ganesan
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai 519087, China.
| | - Baojun Xu
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai 519087, China.
| |
Collapse
|
36
|
Cevatemre B, Erkısa M, Aztopal N, Karakas D, Alper P, Tsimplouli C, Sereti E, Dimas K, Armutak EII, Gurevin EG, Uvez A, Mori M, Berardozzi S, Ingallina C, D'Acquarica I, Botta B, Ozpolat B, Ulukaya E. A promising natural product, pristimerin, results in cytotoxicity against breast cancer stem cells in vitro and xenografts in vivo through apoptosis and an incomplete autopaghy in breast cancer. Pharmacol Res 2017; 129:500-514. [PMID: 29197639 DOI: 10.1016/j.phrs.2017.11.027] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 12/19/2022]
Abstract
Several natural products have been suggested as effective agents for the treatment of cancer. Given the important role of CSCs (Cancer Stem Cells) in cancer, which is a trendy hypothesis, it is worth investigating the effects of pristimerin on CSCs as well as on the other malignant cells (MCF-7 and MDA-MB-231) of breast cancer. The anti-growth activity of pristimerin against MCF-7 and MCF-7s (cancer stem cell enriched population) cells was investigated by real time viability monitorization (xCELLigence System®) and ATP assay, respectively. Mode of cell death was evaluated using electron and fluorescence microscopies, western blotting (autophagy, apoptosis and ER-stress related markers) and flow cytometry (annexin-V staining, caspase 3/7 activity, BCL-2 and PI3K expressions). Pristimerin showed an anti-growth effect on cancer cells and cancer stem cells with IC50 values ranging at 0.38-1.75μM. It inhibited sphere formation at relatively lower doses (<1.56μM). Apoptosis was induced in MCF-7 and MCF-7s cells. In addition, extensive cytoplasmic vacuolation was observed, implying an incompleted autophagy as evidenced by the increase of autophagy-related proteins (p62 and LC3-II) with an unfolded protein response (UPR). Pristimerin inhibited the growth of MCF-7 and MDA-MB-231-originated xenografts in NOD.CB17-Prkdcscid/J mice. In mice, apoptosis was further confirmed by cleavage of PARP, activation of caspase 3 and/or 7 and TUNEL staining. Taken together, pristimerin shows cytotoxic activity on breast cancer both in vitro and in vivo. It seems to represent a robust promising agent for the treatment of breast cancer. Pristimerin's itself or synthetic novel derivatives should be taken into consideration for novel potent anticancer agent(s).
Collapse
Affiliation(s)
- Buse Cevatemre
- Uludag University, Faculty of Arts and Sciences, Department of Biology, Bursa, Turkey
| | - Merve Erkısa
- Uludag University, Faculty of Arts and Sciences, Department of Biology, Bursa, Turkey; Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey
| | - Nazlihan Aztopal
- Uludag University, Faculty of Arts and Sciences, Department of Biology, Bursa, Turkey; Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey
| | - Didem Karakas
- Uludag University, Faculty of Arts and Sciences, Department of Biology, Bursa, Turkey; Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey
| | - Pınar Alper
- Uludag University, Faculty of Arts and Sciences, Department of Biology, Bursa, Turkey; Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey
| | - Chrisiida Tsimplouli
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Evangelia Sereti
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Elif I Ikitimur Armutak
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University, 34320, Istanbul, Turkey
| | - Ebru Gurel Gurevin
- Department of Biology, Faculty of Science, Istanbul University, 34134, Istanbul, Turkey
| | - Ayca Uvez
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University, 34320, Istanbul, Turkey
| | - Mattia Mori
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, viale Regina Elena 291, 00161 Roma, Italy
| | - Simone Berardozzi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, viale Regina Elena 291, 00161 Roma, Italy; Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Roma, piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Cinzia Ingallina
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, viale Regina Elena 291, 00161 Roma, Italy; Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Roma, piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Ilaria D'Acquarica
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Roma, piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Roma, piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Engin Ulukaya
- Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey.
| |
Collapse
|
37
|
Mori Y, Shirai T, Terauchi R, Tsuchida S, Mizoshiri N, Hayashi D, Arai Y, Kishida T, Mazda O, Kubo T. Antitumor effects of pristimerin on human osteosarcoma cells in vitro and in vivo. Onco Targets Ther 2017; 10:5703-5710. [PMID: 29238202 PMCID: PMC5713707 DOI: 10.2147/ott.s150071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
There are very few treatments for musculoskeletal tumors, compared to other cancers; thus, novel therapeutic drugs are needed. Pristimerin (PM) is a triterpene compound isolated from plant extracts that reportedly has antitumor effects on various cancers, such as of the breast and prostate. The purpose of this study was to evaluate the antitumor effects of PM on human osteosarcoma cells. Treatment of the human osteosarcoma cell lines, MNNG and 143B, with PM led to a dose-dependent decrease in cell viability. The effects of PM on apoptosis were evaluated with the Annexin V/propidium iodide assay and analysis of caspases 3, 8, and 9 activities. Western blot analysis showed that PM caused a decrease in the expression of Akt, mTOR, and NF-κB. The volumes and weights of human osteosarcoma xenografts decreased significantly with PM treatment. The results of this study revealed that PM can inhibit human osteosarcoma growth in vitro and in vivo, and may be a novel therapeutic agent for the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuji Arai
- Department of Sports and Para-Sports Medicine
| | - Tunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | |
Collapse
|
38
|
Hui B, Zhang L, Zhou Q, Hui L. Pristimerin Inhibits LPS-Triggered Neurotoxicity in BV-2 Microglia Cells Through Modulating IRAK1/TRAF6/TAK1-Mediated NF-κB and AP-1 Signaling Pathways In Vitro. Neurotox Res 2017; 33:268-283. [PMID: 29119451 DOI: 10.1007/s12640-017-9837-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 09/24/2017] [Accepted: 10/20/2017] [Indexed: 12/25/2022]
Abstract
Microglia plays a prominent role in the brain's inflammatory response to injury or infection by migrating to affected locations and secreting inflammatory molecules. However, hyperactivated microglial is neurotoxic and plays critical roles in the pathogenesis of neurodegenerative diseases. Pristimerin, a naturally occurring triterpenoid, possesses antitumor, antioxidant, and anti-inflammatory activities. However, the effect and the molecular mechanism of pristimerin against lipopolysaccharide (LPS)-induced neurotoxicity in microglia remain to be revealed. In the present study, using BV-2 microglial cultures, we investigated whether pristimerin modifies neurotoxicity after LPS stimulation and which intracellular pathways are involved in the effect of pristimerin. Here we show that pristimerin markedly suppressed the release of Regulated on Activation, Normal T Expressed and Secreted (RANTES), transforming growth factor-β1 (TGF-β1), IL-6, tumor necrosis factor-α (TNF-α), and nitric oxide (NO). Pristimerin also significantly inhibited migration of BV-2 microglia and alleviated the death of neuron-like PC12 cell induced by the conditioned medium from LPS-activated BV-2 microglial cells. Moreover, pristimerin reduced the expression and interaction of TNF Receptor-Associated Factor 6 (TRAF6) and Interleukin-1 Receptor-Associated Kinases (IRAK1), limiting TGF-beta activating kinase 1 (TAK1) activation, and resulting in an inhibition of IKKα/β/NF-κB and MKK7/JNK/AP-1 signaling pathway in LPS-activated BV-2 microglia. Taken together, the anti-neurotoxicity action of pristimerin is mediated through the inhibition of TRAF6/IRAK1/TAK1 interaction as well as the related pathways: IKKα/β/NF-κB and MKK7/JNK/AP-1 signaling pathways. These findings may suggest that pristimerin might serve as a new therapeutic agent for treating hyperactivated microglial induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Bin Hui
- College of Pharmacy, Shanghai University of Medical & Health Sciences, Shanghai, China
| | - Liping Zhang
- Department of Emergency Medicine, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Qinhua Zhou
- College of Pharmacy, Shanghai University of Medical & Health Sciences, Shanghai, China. .,Department of Pharmacology, College of Medicine, Jiaxing University, Jiaxing, China.
| | - Ling Hui
- Center for Experimental Medicine, Lanzhou Military Command, Lanzhou General Hospital, Lanzhou, Gansu, China
| |
Collapse
|
39
|
Zhao Y, Zhang S, Wang P, Fu S, Wu D, Liu A. Seleno-short-chain chitosan induces apoptosis in human non-small-cell lung cancer A549 cells through ROS-mediated mitochondrial pathway. Cytotechnology 2017; 69:851-863. [PMID: 28421411 DOI: 10.1007/s10616-017-0098-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/09/2017] [Indexed: 01/05/2023] Open
Abstract
Seleno-short-chain chitosan (SSCC) is a synthesized chitosan derivative. In this study, antitumor activity and underlying mechanism of SSCC on human non-small-cell lung cancer A549 cells were investigated in vitro. The MTT assay showed that SSCC could inhibit cell viability in a dose- and time-dependent manner, and 200 μg/ml SSCC exhibited significantly toxic effects on A549 cells. The cell cycle assay showed that SSCC triggered S phase cell cycle arrest in a dose- and time-dependent manner, which was related to a downregulation of S phase associated cyclin A. The DAPI staining and Annexin V-FITC/PI double staining identified that the SSCC could induce A549 cells apoptosis. Further studies found that SSCC led to the generation of reactive oxygen species (ROS) and the disruption of mitochondrial membrane potential (MMP) by DCFH-DA and Rhodamin 123 staining, respectively. Meanwhile, free radical scavengers N-acetyl-L-cysteine (NAC) pretreatment confirmed that SSCC-induced A549 cells apoptosis was associated with ROS generation. Furthermore, real-time PCR and western blot assay showed that SSCC up-regulated Bax and down-regulated Bcl-2, subsequently incited the release of cytochrome c from mitochondria to cytoplasm, activated the increase of cleaved-caspase 3 and finally induced A549 cells apoptosis in vitro. In general, the present study demonstrated that SSCC induced A549 cells apoptosis via ROS-mediated mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Yana Zhao
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Shaojing Zhang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Pengfei Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Shengnan Fu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Di Wu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Anjun Liu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China.
| |
Collapse
|
40
|
Deeb D, Gao X, Liu YB, Zhang Y, Shaw J, Valeriote FA, Gautam SC. Inhibition of hTERT in pancreatic cancer cells by pristimerin involves suppression of epigenetic regulators of gene transcription. Oncol Rep 2017. [DOI: 10.3892/or.2017.5400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
41
|
Hernandes C, Pereira AMS, Severino P. Compounds From Celastraceae Targeting Cancer Pathways and Their Potential Application in Head and Neck Squamous Cell Carcinoma: A Review. Curr Genomics 2016; 18:60-74. [PMID: 28503090 PMCID: PMC5321769 DOI: 10.2174/1389202917666160803160934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/28/2015] [Accepted: 11/29/2015] [Indexed: 12/13/2022] Open
Abstract
Squamous cell carcinoma of the head and neck is one of the most common cancer types worldwide. It initiates on the epithelial lining of the upper aerodigestive tract, at most instances as a consequence of tobacco and alcohol consumption. Treatment options based on conventional therapies or targeted therapies under development have limited efficacy due to multiple genetic alterations typically found in this cancer type. Natural products derived from plants often possess biological activities that may be valuable in the development of new therapeutic agents for cancer treatment. Several genera from the family Celastraceae have been studied in this context. This review reports studies on chemical constituents isolated from species from the Celastraceae family targeting cancer mechanisms studied to date. These results are then correlated with molecular characteristics of head and neck squamous cell carcinoma in an attempt to identify constituents with potential application in the treatment of this complex disease at the molecular level.
Collapse
Affiliation(s)
- Camila Hernandes
- aAlbert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; bDepartment of Biotechnology, Universidade de Ribeirão Preto, Ribeirão Preto, Brazil
| | - Ana Maria Soares Pereira
- aAlbert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; bDepartment of Biotechnology, Universidade de Ribeirão Preto, Ribeirão Preto, Brazil
| | - Patricia Severino
- aAlbert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; bDepartment of Biotechnology, Universidade de Ribeirão Preto, Ribeirão Preto, Brazil
| |
Collapse
|
42
|
Yan YY, Wang F, Zhao XQ, Wang XK, Chen YF, Liu H, Xie Y, Fu LW. Degradation of P-glycoprotein by pristimerin contributes to overcoming ABCB1-mediated chemotherapeutic drug resistance in vitro. Oncol Rep 2016; 37:31-40. [PMID: 27840996 PMCID: PMC5355671 DOI: 10.3892/or.2016.5230] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/31/2016] [Indexed: 11/22/2022] Open
Abstract
ABCB1 (P-glycoprotein, ABCB1/MDR1) is one of the major members of the ABC transporters linked to MDR in cancer cells. In this study, we observed that pristimerin, a natural triterpenoid, potently decreased P-gp in a dose-dependent manner in both drug-resistant KBv200 and stable transfected HEK293/ABCB1 cell lines. Moreover, pristimerin also inhibited cell proliferation and induced apoptosis in both cell lines. Intriguingly, reverse transcription-PCR, real-time PCR and protein turn-over assay revealed that the decrease of P-gp was independent of mRNA level but primarily owing to its protein stability. Furthermore, immunofluorescence study with anti-P-gp antibody showed that pristimerin disturbed the subcellular distribution of P-gp with decreased location in the plasma membrane. Taken together, these data suggest that subcellular distribution of P-gp and subsequent downregulation by pristimerin contribute to overcoming ABCB1-mediated chemotherapeutic drug resistance. Our findings suggested inducing the decrease of P-gp membrane protein could be a new promising alternative therapeutic strategy in ABCB1-mediated MDR.
Collapse
Affiliation(s)
- Yan-Yan Yan
- Institute of Respiratory and Occupational Diseases, Collaborative Innovation Center for Cancer, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xiao-Qin Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xiao-Kun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Yi-Fan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Hong Liu
- Institute of Respiratory and Occupational Diseases, Collaborative Innovation Center for Cancer, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Yong Xie
- Institute of Respiratory and Occupational Diseases, Collaborative Innovation Center for Cancer, Shanxi Datong University, Datong, Shanxi 037009, P.R. China
| | - Li-Wu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
43
|
Lee SO, Kim JS, Lee MS, Lee HJ. Anti-cancer effect of pristimerin by inhibition of HIF-1α involves the SPHK-1 pathway in hypoxic prostate cancer cells. BMC Cancer 2016; 16:701. [PMID: 27581969 PMCID: PMC5007821 DOI: 10.1186/s12885-016-2730-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/19/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hypoxia is a typical character of locally advanced solid tumours. The transcription factor hypoxia-inducible factor 1α (HIF-1α) is the main regulator under the hypoxic environment. HIF-1α regulates various genes to enhance tumour progression, angiogenesis, and metastasis. Sphingosine kinase 1 (SPHK-1) is a modulator of HIF-1α. METHODS To investigate the molecular mechanisms of pristimerin in association with SPHK-1 pathways in hypoxic PC-3 cancer cells. Vascular endothelial growth factor (VEGF) production, cell cycles, and SPHK-1 activity were measured, and western blotting, an MTT assay, and an RNA interference assay were performed. RESULTS Pristimerin inhibited HIF-1α accumulation in a concentration- and-time-dependent manner in hypoxic PC-3 cells. Pristimerin suppressed the expression of HIF-1α by inhibiting SPHK-1. Moreover, inhibiting SPHK-1 with a sphingosine kinase inhibitor enhanced the suppression of HIF-1α, phosphorylation AKT, and glycogen synthase kinase-3β (GSK-3β) by pristimerin under hypoxia. Furthermore, a reactive oxygen species (ROS) scavenger enhanced the inhibition of HIF-1α and SPHK-1 by pristimerin. CONCLUSION Taken together, these findings suggest that pristimerin can exert an anti-cancer activity by inhibiting HIF-1α through the SPHK-1 pathway.
Collapse
Affiliation(s)
- Seon-Ok Lee
- Department of Cancer Preventive Material Development, Graduate School, Kyung Hee University, Seoul, Republic of Korea.,College of Korean Medicine, Kyung Hee University, 1Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Joo-Seok Kim
- College of Korean Medicine, Kyung Hee University, 1Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, Republic of Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Myoung-Sun Lee
- Department of Cancer Preventive Material Development, Graduate School, Kyung Hee University, Seoul, Republic of Korea.,College of Korean Medicine, Kyung Hee University, 1Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Hyo-Jeong Lee
- Department of Cancer Preventive Material Development, Graduate School, Kyung Hee University, Seoul, Republic of Korea. .,College of Korean Medicine, Kyung Hee University, 1Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, Republic of Korea. .,Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Pristimerin inhibits proliferation, migration and invasion, and induces apoptosis in HCT-116 colorectal cancer cells. Biomed Pharmacother 2016; 79:112-9. [DOI: 10.1016/j.biopha.2016.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/04/2016] [Accepted: 02/06/2016] [Indexed: 11/20/2022] Open
|
45
|
Xie G, Yu X, Liang H, Chen J, Tang X, Wu S, Liao C. Pristimerin overcomes adriamycin resistance in breast cancer cells through suppressing Akt signaling. Oncol Lett 2016; 11:3111-3116. [PMID: 27123073 DOI: 10.3892/ol.2016.4335] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 02/05/2016] [Indexed: 11/06/2022] Open
Abstract
Breast cancer remains a major public health problem worldwide. Chemotherapy serves an important role in the treatment of breast cancer. However, resistance to chemotherapeutic agents, in particular, multi-drug resistance (MDR), is a major cause of treatment failure in cancer. Agents that can either enhance the effects of chemotherapeutics or overcome chemoresistance are urgently needed for the treatment of breast cancer. Pristimerin, a quinonemethide triterpenoid compound isolated from Celastraceae and Hippocrateaceae, has been shown to possess antitumor, anti-inflammatory, antioxidant and insecticidal properties. The aim of the present study was to investigate whether pristimerin can override chemoresistance in MCF-7/adriamycin (ADR)-resistant human breast cancer cells. The results demonstrated that pristimerin indeed displayed potent cytocidal effect on multidrug-resistant MCF-7/ADR breast cancer cells, and that these effects occurred through the suppression of Akt signaling, which in turn led to the downregulation of antiapoptotic effectors and increased apoptosis. These findings indicate that use of pristimerin may represent a potentially promising approach for the treatment of ADR-resistant breast cancer.
Collapse
Affiliation(s)
- Gui'e Xie
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Xinpei Yu
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China; Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China
| | - Huichao Liang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Jingsong Chen
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Xuewei Tang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Shaoqing Wu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Can Liao
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| |
Collapse
|
46
|
Zhao H, Wang C, Lu B, Zhou Z, Jin Y, Wang Z, Zheng L, Liu K, Luo T, Zhu D, Chi G, Luo Y, Ge P. Pristimerin triggers AIF-dependent programmed necrosis in glioma cells via activation of JNK. Cancer Lett 2016; 374:136-148. [PMID: 26854718 DOI: 10.1016/j.canlet.2016.01.055] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/25/2015] [Accepted: 01/31/2016] [Indexed: 01/01/2023]
Abstract
Programmed necrosis is established as a new form of programmed cell death and is emerging as a new strategy of treatment for cancers. Pristimerin is a natural chemical with anti-tumor effect despite the fact that its mechanism remains poorly understood. In this study, we used glioma cell lines and mice model of xenograft glioma to investigate the effect of pristimerin on glioma and its underlying mechanism. We found that pristimerin inhibited the viabilities of glioma cells in vitro and the growth of xenograft gliomas in vivo, which was accompanied by upregulation of JNK and phosphor-JNK, nuclear accumulation of AIF, and elevation in the ratio of Bax/Bcl-2. In vitro studies showed that pristimerin induced necrosis in glioma cells, as well as mitochondrial depolarization, overproduction of ROS and reduction of GSH. Ablation of AIF level with SiRNA mitigated pristimerin-induced nuclear accumulation of AIF and prevented necrosis in glioma cells. Moreover, pharmacological inhibition of JNK with SP600125 or knockdown of its level with SiRNA reversed mitochondrial depolarization attenuated the elevation of Bax/Bcl-2 and suppressed nuclear accumulation of AIF. Further, inhibition of ROS with NAC not only rescued glioma cell necrosis but also suppressed JNK activation, mitigated Bax/Bcl-2 ratio, maintained mitochondrial membrane potential, and inhibited AIF translocation into nucleus. Therefore, we demonstrated first in this study that pristimerin triggered AIF-dependent necroptosis in glioma cells via induction of mitochondrial dysfunction by activation of JNK through overproduction of ROS. These results suggest that pristimerin has potential therapeutic effects on glioma.
Collapse
Affiliation(s)
- Hongwei Zhao
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Chen Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Bin Lu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Zijian Zhou
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Yong Jin
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Zongqi Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Linjie Zheng
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Kai Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Tianfei Luo
- Research Center of Neuroscience, First Hospital of Jilin University, Changchun 130021, China; Department of Neurology, First Hospital of Jilin University, Changchun 130021, China
| | - Dong Zhu
- Department of Orthopaedics, First Hospital of Jilin University, Changchun 130021, China
| | - Guangfan Chi
- Department of Orthopaedics, First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Yinan Luo
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun 130021, China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
47
|
Yousef BA, Guerram M, Hassan HM, Hamdi AM, Zhang LY, Jiang ZZ. Pristimerin demonstrates anticancer potential in colorectal cancer cells by inducing G1 phase arrest and apoptosis and suppressing various pro-survival signaling proteins. Oncol Rep 2015; 35:1091-100. [PMID: 26718323 DOI: 10.3892/or.2015.4457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/28/2015] [Indexed: 11/06/2022] Open
Abstract
Pristimerin is a naturally occurring triterpenoid that has a cytotoxic effect on several cancer cell lines. However, the cytotoxic effects of pristimerin as well as its molecular mechanisms of action against colorectal cancer have never been explored. In the present study, we investigated the anticancer potential of pristimerin, and examined the different signaling pathways affected by its action in three colon cancer cell lines namely HCT-116, COLO-205 and SW-620. Pristimerin was found to possess potent cytotoxic and proliferation inhibitory effects against these cell lines. Cell cycle analysis revealed G1 phase arrest, which was strongly associated with decreased expression of cyclin D1 and cyclin-dependent kinases (cdk4 and cdk6) with concomitant induction of p21. Pristimerin also induced apoptosis in a dose-dependent manner. Cell plasma membrane alterations studied by Annexin V/PI double staining, loss of mitochondrial membrane potential (ΔΨm), measurements of caspase activities and the inhibitory effect of Z-VAD-FMK (a caspase inhibitor) confirmed the apoptotic effect of pristimerin. Moreover, western blot data showed that apoptotic induction was associated with activated caspase-3 and -8, PARP-1 cleavage and modulation of the expression levels of Bcl-2 family proteins. Additionally, pristimerin treatment downregulated the phosphorylated forms of EGFR and HER2 proteins, and subsequently caused a decrease in the phosphorylated forms of Erk1/2, Akt, mTOR and NF-κB proteins. Taken together, these results suggest that pristimerin may have potential as a new targeting therapeutic strategy for the treatment of colon cancer.
Collapse
Affiliation(s)
- Bashir A Yousef
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Mounia Guerram
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Hozeifa M Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Aida M Hamdi
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Lu-Yong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Zhen-Zhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
48
|
Mejía-Manzano LA, Barba-Dávila BA, Gutierrez-Uribe JA, Escalante-Vázquez EJ, Serna-Saldívar SO. Extraction and Isolation of Antineoplastic Pristimerin from Mortonia greggii (Celastraceae). Nat Prod Commun 2015. [DOI: 10.1177/1934578x1501001130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The aim of this research was to identify, extract and isolate pristimerin in leaves, stems and roots of the Mexican plant Mortonia greggii ( Celastraceae). The principal objective was to determine the best laboratory experimental conditions for the extraction and isolation of this powerful natural anticancer agent from the root tissue. Six experimental factors in solid-liquid pristimerin extraction were analyzed: solvent systems, number of extractions, ratio of plant weight (g)/solvent volume (mL) used, time of extraction, temperature and agitation. A mathematical model was generated for pristimerin purity and yield. Ethanol, first extraction, 0.5 ratio of plant weight/solvent volume (g/mL), 0.5 h, 200 rpm and 49.7°C were optimal conditions for the extraction of this phytochemical. The degree of purification of pristimerin root extract was studied by size-exclusion chromatography (SEC) using Sephadex LH-20 reaching fractions with purification indexes (PI) greater than 2 and recoveries of 28.3%. When fractions with purification indices higher than 1 and less than 2 were accumulated, the recovery of pristimerin increased by about 73.6%. By combining the optimum extracts and SEC purification protocols, an enriched fraction containing 245.6 mg pristimerin was obtained from 100 g of root bark, representing about 14.4%, w/w, pristimerin from the total solids presented in the fraction.
Collapse
Affiliation(s)
| | | | | | - Edgardo J. Escalante-Vázquez
- Departamento de Ingeniería Industrial, Tecnológico de Monterrey. Ave. Eugenio Garza Sada 2501, Sur, 64849. Monterrey, Nuevo León, México
| | | |
Collapse
|
49
|
Dong C, Xu C, Liu H, Xu S, Gao Y, Peng J. Absorption and metabolism characteristics of pristimerin as determined by a sensitive and reliable LC–MS/MS method. Fitoterapia 2015; 106:62-7. [DOI: 10.1016/j.fitote.2015.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 01/30/2023]
|
50
|
Pereira DM, Valentão P, Correia-da-Silva G, Teixeira N, Andrade PB. Translating endoplasmic reticulum biology into the clinic: a role for ER-targeted natural products? Nat Prod Rep 2015; 32:705-22. [PMID: 25703279 DOI: 10.1039/c4np00102h] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
ER stress has been identified as a hallmark, and sometimes trigger, of several pathologies, notably cancer, inflammation and neurodegenerative diseases like Alzheimer's and Parkinson's. Among the molecules described in literature known to affect ER function, the majority are natural products, suggesting that natural molecules may constitute a significant arsenal of chemical entities for modulating this cellular target. In this review, we will start by presenting the current knowledge of ER biology and the hallmarks of ER stress, thus paving the way for presenting the natural products that have been described as being ER modulators, either stress inducers or ER protectors. The chemistry, distribution and mechanism of action of these compounds will be presented and discussed.
Collapse
Affiliation(s)
- David M Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal.
| | | | | | | | | |
Collapse
|