1
|
Singh H, Mishra AK, Mohanto S, Kumar A, Mishra A, Amin R, Darwin CR, Emran TB. A recent update on the connection between dietary phytochemicals and skin cancer: emerging understanding of the molecular mechanism. Ann Med Surg (Lond) 2024; 86:5877-5913. [PMID: 39359831 PMCID: PMC11444613 DOI: 10.1097/ms9.0000000000002392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 10/04/2024] Open
Abstract
Constant exposure to harmful substances from both inside and outside the body can mess up the body's natural ways of keeping itself in balance. This can cause severe skin damage, including basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma. However, plant-derived compounds found in fruits and vegetables have been shown to protect against skin cancer-causing free radicals and other harmful substances. It has been determined that these dietary phytochemicals are effective in preventing skin cancer and are widely available, inexpensive, and well-tolerated. Studies have shown that these phytochemicals possess anti-inflammatory, antioxidant, and antiangiogenic properties that can aid in the prevention of skin cancers. In addition, they influence crucial cellular processes such as angiogenesis and cell cycle control, which can halt the progression of skin cancer. The present paper discusses the benefits of specific dietary phytochemicals found in fruits and vegetables, as well as the signaling pathways they regulate, the molecular mechanisms involved in the prevention of skin cancer, and their drawbacks.
Collapse
Affiliation(s)
- Harpreet Singh
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh
| | | | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka
| | - Arvind Kumar
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh
| | - Amrita Mishra
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi
| | - Ruhul Amin
- Faculty of Pharmaceutical Science, Assam downtown University, Panikhaiti, Gandhinagar, Guwahati, Assam
| | | | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
2
|
Şengelen A, Önay-Uçar E. Rosmarinic acid attenuates glioblastoma cells and spheroids' growth and EMT/stem-like state by PTEN/PI3K/AKT downregulation and ERK-induced apoptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156060. [PMID: 39341126 DOI: 10.1016/j.phymed.2024.156060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Glioblastoma (GB) is a highly malignant type of brain cancer with a poor prognosis. Therapeutic strategies for GB are still limited. Rosmarinic acid (RA), a polyphenolic compound, is a promising experimental anticancer agent, but its specific protein targets for GB remain unclear. PURPOSE This study aimed to elucidate the anticancer effects of RA in 2D- and 3D-GB cells and the underlying mechanisms. METHODS 3D-tumor spheroids (mimics in vivo tumors) were obtained by the hanging-drop/agarose method. RA's anti-glioma activity on U-87MG (p53-wt/PTEN-mt) and LN229 (p53-mt/PTEN-wt) cells was evaluated through cell viability, colony-formation, migration/invasion/angiogenesis assays, fluorescence imaging, and spheroid growth analysis. The underlying mechanism of the anticancer effects of RA was investigated by Western blot and immunofluorescence analysis. The MEK inhibitor U0126 was used to block ERK phosphorylation. RESULTS RA treatments exerted anti-proliferative and pro-apoptotic effects on human GB cells. RA dose-dependently reduced angiogenesis and intracellular ROS levels, suppressed glioma growth, and migration/invasion in 2D-culture and cancer stem cell (CSC)-like 3D-spheroid culture (SPC). Repeated therapy in SPC was more effective by leading to disrupted structure than a single treatment. Treatments in SPC also suppressed epithelial-mesenchymal transition (EMT) and CSC-like properties. Strikingly, RA downregulated the SIRT1/FOXO1/NF-κB axis independently of p53 or PTEN function in both gliomas. Immunofluorescence labeling revealed decreased SIRT1 and NF-κB-p65 and increased FOXO1 and GAPDH proteins in nuclear location (associated with apoptosis). Surprisingly, RA increased p-ERK1/2 levels, but priming with U0126 abolished RA-mediated p-ERK upregulation; thus, autophagy and apoptosis induction in GB cells were prevented, and the growth of GB spheroids accelerated. Specifically, RA also inhibited the PTEN/PI3K/AKT pathway in U-87MG cells. Due to genetic differences in cells, U-87MG cells were more sensitive to RA treatments than LN229 cells. Meanwhile, our positive control drug trial results with FDA-approved temozolomide (TMZ) used in GB treatment showed that our test compound rosmarinic acid exhibited higher therapeutic effects than TMZ at lower doses. CONCLUSION Suppression of EMT, downregulation of SIRT1/FOXO1/NF-κB axis, inhibition of PTEN/PI3K/AKT signaling pathway, and ERK-induced apoptosis and autophagy were determined to be involved in stopping glioma progression. Our findings for the first time, revealed that RA may have potential therapeutic use by having multiple targets in human brain cancer with further clinical studies.
Collapse
Affiliation(s)
- Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkiye.
| | - Evren Önay-Uçar
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye
| |
Collapse
|
3
|
Wang W, Zhang Y, Huang X, Li D, Lin Q, Zhuang H, Li H. The role of the miR-30a-5p/BCL2L11 pathway in rosmarinic acid-induced apoptosis in MDA-MB-231-derived breast cancer stem-like cells. Front Pharmacol 2024; 15:1445034. [PMID: 39239646 PMCID: PMC11375422 DOI: 10.3389/fphar.2024.1445034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
Background Rosmarinic acid (RA), a natural phenolic acid, exhibits promising anti-cancer properties. The abnormal expression of microRNA (miRNA) regulates the gene expression and plays a role as an oncogenic or tumor suppressor in TNBC. However, the biological role of RA in miR-30a-5p on BCL2L11 during MDA-MB-231 induced breast cancer stem-like cells (BCSCs) progression and its regulatory mechanism have not been elucidated. Objective To investigate whether RA inhibited the silencing effect of miR-30a-5p on the BCL2L11 gene and promoted apoptosis in BCSCs. Materials and Methods We assessed the migration, colony formation, proliferation, cell cycle, and apoptosis of BCSCs after RA treatment using the wound-healing assay, colony formation assay, CCK-8 assay, and flow cytometry, respectively. The expression of mRNA and protein levels of BCL-2, Bax, BCL2L11, and P53 genes in BCSCs after RA treatment was obtained by real-time polymerase chain reaction and Western blot. Differential miRNA expression in BCSCs was analyzed by high-throughput sequencing. Targetscan was utilized to predict the targets of miR-30a-5p. The dual luciferase reporter system was used for validation of the miR-30a-5p target. Results Wound-healing assay, colony formation assay, CCK-8 assay, and cell cycle assay results showed that RA inhibited migration, colony formation and viability of BCSCs, and cell cycle arrest in the G0-G1 phase. At the highest dose of RA, we noticed cell atrophy, while the arrest rate at 100 μg/mL RA surpassed that at 200 μg/mL RA. Apoptotic cells appeared early (Membrane Associated Protein V FITC+, PI-) or late (Membrane Associated Protein V FITC+, PI+) upon administration of 200 μg/mL RA, Using high-throughput sequencing to compare the differences in miRNA expression, we detected downregulation of miR-30a-5p expression, and the results of dual luciferase reporter gene analysis indicated that BCL2L11 was a direct target of miR-30a-5p. Conclusion RA inhibited the silencing effect of miR-30a-5p on the BCL2L11 gene and enhanced apoptosis in BCSCs.
Collapse
Affiliation(s)
- Wei Wang
- School of Public Health and Health Management, Fujian Health College, Fuzhou, Fujian, China
| | - Yuefen Zhang
- Science and Technology Service Center, Fujian Health College, Fuzhou, Fujian, China
| | - Xiaomin Huang
- School of Pharmacy, Fujian Health College, Fuzhou, Fujian, China
| | - Dan Li
- School of Public Health and Health Management, Fujian Health College, Fuzhou, Fujian, China
| | - Qi Lin
- School of Public Health and Health Management, Fujian Health College, Fuzhou, Fujian, China
| | - Hailin Zhuang
- School of Public Health and Health Management, Fujian Health College, Fuzhou, Fujian, China
| | - Hong Li
- School of Public Health and Health Management, Fujian Health College, Fuzhou, Fujian, China
| |
Collapse
|
4
|
da Silva GB, Manica D, Dallagnol P, Narzetti RA, Marafon F, da Silva AP, de S Matias L, Cassol JV, Moreno M, Kempka AP, Bagatini MD. Rosmarinic acid modulates purinergic signaling and induces apoptosis in melanoma cells. Purinergic Signal 2024:10.1007/s11302-024-10040-z. [PMID: 39031243 DOI: 10.1007/s11302-024-10040-z] [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: 04/25/2024] [Accepted: 07/13/2024] [Indexed: 07/22/2024] Open
Abstract
Cancer cases have increased worldwide. Cutaneous melanoma (CM), a highly metastatic skin cancer, largely contributes to global statistical cancer death data. Research has shown that rosmarinic acid (RA) is a promising phenolic compound with antineoplastic properties. Thus, we investigated the effects of RA on apoptosis-inducing in melanoma cells, purinergic signaling modulation, and cytokine levels. We treated SK-MEL-28 cells for 24 h with different concentrations of RA and assessed the apoptosis, CD39, CD73, and A2A expression, and cytokine levels. We found RA-induced apoptosis in melanoma cells. Regarding the purinergic system, we verified that RA downregulated the expression of CD73 and A2A, specially at high concentrations of treatment. Additionally, RA increased IL-6, IL-4, IL-10, IFN-γ, and TNF-α levels. Our in vitro results confirm RA's potential to be used to induce melanoma cell apoptosis, having CD73 and A2A as targets when reversion of immune suppression is desired. Further studies in animal models and clinical trials focusing on RA's modulation of purinergic signaling in melanoma are required.
Collapse
Affiliation(s)
- Gilnei B da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Daiane Manica
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Paula Dallagnol
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Rafael A Narzetti
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Filomena Marafon
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Alana P da Silva
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Letícia de S Matias
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Joana V Cassol
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Marcelo Moreno
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Aniela P Kempka
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Margarete D Bagatini
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
5
|
Kalinowska M, Świsłocka R, Wołejko E, Jabłońska-Trypuć A, Wydro U, Kozłowski M, Koronkiewicz K, Piekut J, Lewandowski W. Structural characterization and evaluation of antimicrobial and cytotoxic activity of six plant phenolic acids. PLoS One 2024; 19:e0299372. [PMID: 38885237 PMCID: PMC11182523 DOI: 10.1371/journal.pone.0299372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/08/2024] [Indexed: 06/20/2024] Open
Abstract
Phenolic acids still gain significant attention due to their potential antimicrobial and cytotoxic properties. In this study, we have investigated the antimicrobial of six phenolic acids, namely chlorogenic, caffeic, p-coumaric, rosmarinic, gallic and tannic acids in the concentration range 0.5-500 μM, against Escherichia coli and Lactobacillus rhamnosus. The antimicrobial activity was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide colorimetric assay. Additionally, the cytotoxic effects of these phenolic acids on two cancer cell lines, the colorectal adenocarcinoma Caco-2 cell line and Dukes' type C colorectal adenocarcinoma DLD-1 cell line was examined. To further understand the molecular properties of these phenolic acids, quantum chemical calculations were performed using the Gaussian 09W program. Parameters such as ionization potential, electron affinity, electronegativity, chemical hardness, chemical softness, dipole moment, and electrophilicity index were obtained. The lipophilicity properties represented by logP parameter was also discussed. This study provides a comprehensive evaluation of the antimicrobial and cytotoxic activity of six phenolic acids, compounds deliberately selected due to their chemical structure. They are derivatives of benzoic or cinnamic acids with the increasing number of hydroxyl groups in the aromatic ring. The integration of experimental and computational methodologies provides a knowledge of the molecular characteristics of bioactive compounds and partial explanation of the relationship between the molecular structure and biological properties. This knowledge aids in guiding the development of bioactive components for use in dietary supplements, functional foods and pharmaceutical drugs.
Collapse
Affiliation(s)
- Monika Kalinowska
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Renata Świsłocka
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Elżbieta Wołejko
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Agata Jabłońska-Trypuć
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Urszula Wydro
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Maciej Kozłowski
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Kamila Koronkiewicz
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Jolanta Piekut
- Department of Agri-Food Engineering and Environmental Management, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| | - Włodzimierz Lewandowski
- Department of Chemistry, Biology and Biotechnology, Institute of Environmental Engineering and Energetics, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Bialystok, Poland
| |
Collapse
|
6
|
Xie G, Zou X, Liang Z, Zhang K, Wu D, Jin H, Wang H, Shen Q. GBF family member PfGBF3 and NAC family member PfNAC2 regulate rosmarinic acid biosynthesis under high light. PLANT PHYSIOLOGY 2024; 195:1728-1744. [PMID: 38441888 DOI: 10.1093/plphys/kiae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/12/2023] [Indexed: 06/02/2024]
Abstract
Rosmarinic acid (RA) is an important medicinal metabolite and a potent food antioxidant. We discovered that exposure to high light intensifies the accumulation of RA in the leaves of perilla (Perilla frutescens (L.) Britt). However, the molecular mechanism underlying RA synthesis in response to high light stress remains poorly understood. To address this knowledge gap, we conducted a comprehensive analysis employing transcriptomic sequencing, transcriptional activation, and genetic transformation techniques. High light treatment for 1 and 48 h resulted in the upregulation of 592 and 1,060 genes, respectively. Among these genes, three structural genes and 93 transcription factors exhibited co-expression. Notably, NAC family member PfNAC2, GBF family member PfGBF3, and cinnamate-4-hydroxylase gene PfC4H demonstrated significant co-expression and upregulation under high light stress. Transcriptional activation analysis revealed that PfGBF3 binds to and activates the PfNAC2 promoter. Additionally, both PfNAC2 and PfGBF3 bind to the PfC4H promoter, thereby positively regulating PfC4H expression. Transient overexpression of PfNAC2, PfGBF3, and PfC4H, as well as stable transgenic expression of PfNAC2, led to a substantial increase in RA accumulation in perilla. Consequently, PfGBF3 acts as a photosensitive factor that positively regulates PfNAC2 and PfC4H, while PfNAC2 also regulates PfC4H to promote RA accumulation under high light stress. The elucidation of the regulatory mechanism governing RA accumulation in perilla under high light conditions provides a foundation for developing a high-yield RA system and a model to understand light-induced metabolic accumulation.
Collapse
Affiliation(s)
- Guanwen Xie
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiuzai Zou
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zishan Liang
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ke Zhang
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Duan Wu
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Honglei Jin
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongbin Wang
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qi Shen
- School of Pharmaceutical Sciences, Institute of Medical Plant Physiology and Ecology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
7
|
Khaksar S, Kiarostami K, Ramdan M. Effect of Rosmarinic Acid on Cell Proliferation, Oxidative Stress, and Apoptosis Pathways in an Animal Model of Induced Glioblastoma Multiforme. Arch Med Res 2024; 55:103005. [PMID: 38759277 DOI: 10.1016/j.arcmed.2024.103005] [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: 12/01/2023] [Revised: 04/13/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND In brain tumors, the complexity of the pathophysiological processes such as oxidative stress, cell proliferation, angiogenesis, and apoptosis have seriously challenged the definitive treatment. Rosmarinic acid (RA), as a polyphenolic compound, has been found to prevent tumor progression in some aggressive cancers. This study was designed to evaluate the anticancer effects of RA on brain tumors. METHOD Rats were divided into six groups. Implantation of C6 glioma cells was carried out in the caudate nucleus of the right hemisphere. RA at doses of 5, 10, and 20 mg/kg (i.p.) was administered to the treatment groups for seven days. Tumor volume (by MRI imaging), locomotor ability, survival time, histological alterations (by H & E staining), expression of p53 and p21 mRNAs (by RT-PCR), activities of antioxidant enzymes (superoxide dismutase [SOD] and catalase [CAT] by assay kits), expression of caspase-3 and VEGF (by immunohistochemical analysis), and TUNEL-positive cells (by tunnel staining) were analyzed. RESULTS The results indicated that the RA at a dose of 20 mg/kg reduced the tumor volume, prolonged survival time, increased p53 and p21 mRNAs, attenuated SOD and CAT activities in tumor tissue, elevated caspase-3, and increased the number of TUNEL-positive cells. Furthermore, histological analysis revealed less invasion of tumor cells into the normal parenchyma in rats treated with RA (20 mg/kg). CONCLUSION These findings provide evidence that the ability of RA to reduce tumor volume could be related to factors that modulate oxidative stress (SOD and CAT enzymes), cell proliferation (p53 and p21), and apoptosis (caspase-3).
Collapse
Affiliation(s)
- Sepideh Khaksar
- Department of Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.
| | - Khadijeh Kiarostami
- Department of Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Mahmoud Ramdan
- Department of Biology, Faculty of Science, Al-Furat University, Deir-ez-Zor, Syrian Arab Republic
| |
Collapse
|
8
|
Khan A, Khan A, Khan MA, Malik Z, Massey S, Parveen R, Mustafa S, Shamsi A, Husain SA. Phytocompounds targeting epigenetic modulations: an assessment in cancer. Front Pharmacol 2024; 14:1273993. [PMID: 38596245 PMCID: PMC11002180 DOI: 10.3389/fphar.2023.1273993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/09/2023] [Indexed: 04/11/2024] Open
Abstract
For centuries, plants have been serving as sources of potential therapeutic agents. In recent years, there has been a growing interest in investigating the effects of plant-derived compounds on epigenetic processes, a novel and captivating Frontier in the field of epigenetics research. Epigenetic changes encompass modifications to DNA, histones, and microRNAs that can influence gene expression. Aberrant epigenetic changes can perturb key cellular processes, including cell cycle control, intercellular communication, DNA repair, inflammation, stress response, and apoptosis. Such disruptions can contribute to cancer development by altering the expression of genes involved in tumorigenesis. However, these modifications are reversible, offering a unique avenue for therapeutic intervention. Plant secondary compounds, including terpenes, phenolics, terpenoids, and sulfur-containing compounds are widely found in grains, vegetables, spices, fruits, and medicinal plants. Numerous plant-derived compounds have demonstrated the potential to target these abnormal epigenetic modifications, including apigenin (histone acetylation), berberine (DNA methylation), curcumin (histone acetylation and epi-miRs), genistein (histone acetylation and DNA methylation), lycopene (epi-miRs), quercetin (DNA methylation and epi-miRs), etc. This comprehensive review highlights these abnormal epigenetic alterations and discusses the promising efficacy of plant-derived compounds in mitigating these deleterious epigenetic signatures in human cancer. Furthermore, it addresses ongoing clinical investigations to evaluate the therapeutic potential of these phytocompounds in cancer treatment, along with their limitations and challenges.
Collapse
Affiliation(s)
- Aqsa Khan
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Asifa Khan
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Mohammad Aasif Khan
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
- Department of Radiation Oncology, The University of Texas Health Science Centre at San Antonio, San Antonio, TX, United States
| | - Zoya Malik
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Sheersh Massey
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Rabea Parveen
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Saad Mustafa
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Syed A. Husain
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| |
Collapse
|
9
|
Stavropoulou LS, Efthimiou I, Giova L, Manoli C, Sinou PS, Zografidis A, Lamari FN, Vlastos D, Dailianis S, Antonopoulou M. Phytochemical Profile and Evaluation of the Antioxidant, Cyto-Genotoxic, and Antigenotoxic Potential of Salvia verticillata Hydromethanolic Extract. PLANTS (BASEL, SWITZERLAND) 2024; 13:731. [PMID: 38475577 DOI: 10.3390/plants13050731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
This study comprises the phytochemical characterization, the evaluation of the total phenolic content (TPC) and antioxidant activity (AA), and the investigation of the cyto-genotoxic and antigenotoxic potential of hydromethanolic extract derived from Salvia verticillata L. leaves. HPLC-DAD-ESI-MS and HPLC-DAD were used for the characterization of the extract and determination of the major ingredients. Afterwards, the TPC and AA were determined. The cytotoxic and genotoxic effect of the extract on cultured human lymphocytes at concentrations of 10, 25, and 50 μg mL-1 was investigated via the Cytokinesis Block MicroNucleus (CBMN) assay. Moreover, its antigenotoxic potential against the mutagenic agent mitomycin C (MMC) was assessed using the same assay. The hydromethanolic extract comprises numerous metabolites, with rosmarinic acid being the major compound. It had a high value of TPC and exerted significant AA as shown by the results of the Ferric Reducing Antioxidant Power (FRAP) and Radical Scavenging Activity by DPPH• assays. A dose-dependent cytotoxic potential was recorded, with the highest dose (50 μg mL-1) exhibiting statistically significant cytotoxicity. None of the tested concentrations induced significant micronuclei (MN) frequencies, indicating a lack of genotoxicity. All tested concentrations reduced the MMC-mediated genotoxic effects, with the two lowest showing statistically significant antigenotoxic potential.
Collapse
Affiliation(s)
- Lamprini S Stavropoulou
- Laboratory of Pharmacognosy & Chemistry of Natural Products, Department of Pharmacy, University of Patras, GR-26504 Patras, Greece
| | - Ioanna Efthimiou
- Department of Biology, School of Natural Sciences, University of Patras, GR-26504 Patras, Greece
| | - Lambrini Giova
- Department of Biology, School of Natural Sciences, University of Patras, GR-26504 Patras, Greece
| | - Chrysoula Manoli
- Department of Biology, School of Natural Sciences, University of Patras, GR-26504 Patras, Greece
| | - Paraskevi S Sinou
- Laboratory of Pharmacognosy & Chemistry of Natural Products, Department of Pharmacy, University of Patras, GR-26504 Patras, Greece
| | - Aris Zografidis
- Laboratory of Botany, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | - Fotini N Lamari
- Laboratory of Pharmacognosy & Chemistry of Natural Products, Department of Pharmacy, University of Patras, GR-26504 Patras, Greece
| | - Dimitris Vlastos
- Department of Biology, School of Natural Sciences, University of Patras, GR-26504 Patras, Greece
| | - Stefanos Dailianis
- Department of Biology, School of Natural Sciences, University of Patras, GR-26504 Patras, Greece
| | - Maria Antonopoulou
- Department of Sustainable Agriculture, University of Patras, GR-30131 Agrinio, Greece
| |
Collapse
|
10
|
He T, Li NX, Pan ZJ, Zou ZH, Chen JC, Yu SZ, Lv F, Xie QC, Zou J. Serine/threonine kinase 36 induced epithelial-mesenchymal transition promotes docetaxel resistance in prostate cancer. Sci Rep 2024; 14:729. [PMID: 38184689 PMCID: PMC10771505 DOI: 10.1038/s41598-024-51360-9] [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: 10/13/2023] [Accepted: 01/03/2024] [Indexed: 01/08/2024] Open
Abstract
To investigate the role and potential mechanism of serine/threonine kinase 36 (STK36) in docetaxel resistance-prostate cancer (PCa). The expression of STK36 in PCa and the correlation with clinicopathological characteristics of PCa patients were analyzed using the data from different databases and tissue microarrays. To investigate the role of STK36 on cell proliferation, invasion, and migration, STK36 was overexpressed and silenced in DU-145 and PC-3 cell lines. Cell counting kit-8 (CCK8) was used to test cell proliferation. Cell invasion and migration were detected by cell wound scratch assay and trans well, respectively. The expression profile of STK36, E-Cadherin, and Vimentin was analyzed by Western blot. Cell apoptosis was detected by the TUNEL assay. STK36 expression was upregulated in PCa tissue compared with adjacent benign PCa tissue; it was higher in patients with advanced stages compared with lower stages and was significantly correlated with decreased overall survival. Up-regulation of STK36 significantly promoted the proliferation, invasion, and migration of DU-145 and PC-3 cells and compensated for the suppression caused by docetaxel treatment in vitro. A striking apoptosis inhibition could be observed when dealing with docetaxel, although the apoptosis of DU-145 and PC-3 cells was not affected by the STK36 exclusive overexpression. Besides, E-Cadherin expression was restrained while the expression levels of vimentin were all enhanced. The knockdown of STK36 reversed the above process. STK36 up-regulation could accelerate the biological behavior and docetaxel resistance of PCa by epithelial-mesenchymal transition (EMT) activation. STK36 may be potentially used as a target in PCa resolvent with docetaxel.
Collapse
Affiliation(s)
- Tao He
- Department of Emergency Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, 63 DuoBao Road, Guangzhou, Guangdong, 510150, People's Republic of China
| | - Nan-Xing Li
- Department of Emergency Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, 63 DuoBao Road, Guangzhou, Guangdong, 510150, People's Republic of China
| | - Zhao-Jun Pan
- Department of Urology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, People's Republic of China
| | - Zi-Hao Zou
- Department of Urology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, People's Republic of China
| | - Jie-Chuan Chen
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China
| | - Si-Zhe Yu
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China
| | - Fa Lv
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China
| | - Quan-Cheng Xie
- The Third Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, People's Republic of China
| | - Jun Zou
- Department of Emergency Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, 63 DuoBao Road, Guangzhou, Guangdong, 510150, People's Republic of China.
| |
Collapse
|
11
|
Sirajudeen F, Malhab LJB, Bustanji Y, Shahwan M, Alzoubi KH, Semreen MH, Taneera J, El-Huneidi W, Abu-Gharbieh E. Exploring the Potential of Rosemary Derived Compounds (Rosmarinic and Carnosic Acids) as Cancer Therapeutics: Current Knowledge and Future Perspectives. Biomol Ther (Seoul) 2024; 32:38-55. [PMID: 38148552 PMCID: PMC10762267 DOI: 10.4062/biomolther.2023.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 12/28/2023] Open
Abstract
Cancer is a global health challenge with high morbidity and mortality rates. However, conventional cancer treatment methods often have severe side effects and limited success rates. In the last decade, extensive research has been conducted to develop safe, and efficient alternative treatments that do not have the limitations of existing anticancer medicines. Plant-derived compounds have shown promise in cancer treatment for their anti-carcinogenic and anti-proliferative properties. Rosmarinic acid (RA) and carnosic acid (CA) are potent polyphenolic compounds found in rosemary (Rosmarinus officinalis) extract. They have been extensively studied for their biological properties, which include anti-diabetic, anti-inflammatory, antioxidant, and anticancer activities. In addition, RA and CA have demonstrated effective anti-proliferative properties against various cancers, making them promising targets for extensive research to develop candidate or leading compounds for cancer treatment. This review discusses and summarizes the anti-tumor effect of RA and CA against various cancers and highlights the involved biochemical and mechanistic pathways.
Collapse
Affiliation(s)
- Fazila Sirajudeen
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Lara J. Bou Malhab
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Yasser Bustanji
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
| | - Karem H. Alzoubi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Jalal Taneera
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
12
|
Bouammali H, Zraibi L, Ziani I, Merzouki M, Bourassi L, Fraj E, Challioui A, Azzaoui K, Sabbahi R, Hammouti B, Jodeh S, Hassiba M, Touzani R. Rosemary as a Potential Source of Natural Antioxidants and Anticancer Agents: A Molecular Docking Study. PLANTS (BASEL, SWITZERLAND) 2023; 13:89. [PMID: 38202397 PMCID: PMC10780489 DOI: 10.3390/plants13010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/26/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024]
Abstract
Rosmarinus officinalis L. compounds, especially its main polyphenolic compounds, carnosic acid (CA) and rosmarinic acid (RA), influence various facets of cancer biology, making them valuable assets in the ongoing fight against cancer. These two secondary metabolites exhibit formidable antioxidant properties that are a pivotal contributor against the development of cancer. Their antitumor effect has been related to diverse mechanisms. In the case of CA, it has the capacity to induce cell death of cancer cells through the rise in ROS levels within the cells, the inhibition of protein kinase AKT, the activation of autophagy-related genes (ATG) and the disrupt mitochondrial membrane potential. Regarding RA, its antitumor actions encompass apoptosis induction through caspase activation, the inhibition of cell proliferation by interrupting cell cycle progression and epigenetic regulation, antioxidative stress-induced DNA damage, and interference with angiogenesis to curtail tumor growth. To understand the molecular interaction between rosemary compounds (CA and RA) and a protein that is involved in cancer and inflammation, S100A8, we have performed a series of molecular docking analyses using the available three-dimensional structures (PDBID: 1IRJ, 1MR8, and 4GGF). The ligands showed different binding intensities in the active sites with the protein target molecules, except for CA with the 1MR8 protein.
Collapse
Affiliation(s)
- Haytham Bouammali
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| | - Linda Zraibi
- Water, Environment and Sustainable Development Laboratory (LEEDD), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco;
| | - Imane Ziani
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| | - Mohammed Merzouki
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| | - Lamiae Bourassi
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| | - Elmehdi Fraj
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| | - Allal Challioui
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| | - Khalil Azzaoui
- Laboratory of Engineering, Electrochemistry Modeling and Environment, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco;
- Euro-Mediterranean University of Fes (UEMF), Fez 30070, Morocco; (R.S.); (B.H.)
| | - Rachid Sabbahi
- Euro-Mediterranean University of Fes (UEMF), Fez 30070, Morocco; (R.S.); (B.H.)
- Higher School of Technology, Ibn Zohr University, Quartier 25 Mars, P.O. Box 3007, Laayoune 70000, Morocco
| | - Belkheir Hammouti
- Euro-Mediterranean University of Fes (UEMF), Fez 30070, Morocco; (R.S.); (B.H.)
- Laboratory of Industrial Engineering, Energy and the Environment (LI3E), SupMTI, Rabat 10000, Morocco
| | - Shehdeh Jodeh
- Department of Chemistry, An-Najah National University, Nablus P.O. Box 7, Palestine;
| | - Maryam Hassiba
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | - Rachid Touzani
- Laboratory of Applied Chemistry Environment (LCAE), Faculty of Science Oujda, University Mohammed First, Oujda 60000, Morocco; (I.Z.); (M.M.); (L.B.); (E.F.); (A.C.); (R.T.)
| |
Collapse
|
13
|
Alghareeb SA, Alfhili MA, Alsughayyir J. Rosmarinic Acid Elicits Calcium-Dependent and Sucrose-Sensitive Eryptosis and Hemolysis through p38 MAPK, CK1α, and PKC. Molecules 2023; 28:8053. [PMID: 38138543 PMCID: PMC10745317 DOI: 10.3390/molecules28248053] [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: 11/05/2023] [Revised: 12/01/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Rosmarinic acid (RA) possesses promising anticancer potential, but further development of chemotherapeutic agents is hindered by their toxicity to off-target tissue. In particular, chemotherapy-related anemia is a major obstacle in cancer therapy, which may be aggravated by hemolysis and eryptosis. This work presents a toxicity assessment of RA in human RBCs and explores associated molecular mechanisms. METHODS RBCs isolated from healthy donors were treated with anticancer concentrations of RA (10-800 μM) for 24 h at 37 °C, and hemolysis and related markers were photometrically measured. Flow cytometry was used to detect canonical markers of eryptosis, including phosphatidylserine (PS) exposure by annexin-V-FITC, intracellular Ca2+ by Fluo4/AM, cell size by FSC, and oxidative stress by H2DCFDA. Ions and pH were assessed by an ion-selective electrode, while B12 was detected by chemiluminescence. RESULTS RA elicited concentration-dependent hemolysis with AST and LDH release but rescued the cells from hypotonic lysis at sub-hemolytic concentrations. RA also significantly increased annexin-V-positive cells, which was ameliorated by extracellular Ca2+ removal and isosmotic sucrose. Furthermore, a significant increase in Fluo4-positive cells and B12 content and a decrease in FSC and extracellular pH with KCl efflux were noted upon RA treatment. Hemolysis was augmented by blocking KCl efflux and was blunted by ATP, SB203580, staurosporin, D4476, isosmotic urea, and PEG 8000. CONCLUSIONS RA stimulates Ca2+-dependent and sucrose-sensitive hemolysis and eryptosis characterized by PS exposure, Ca2+ accumulation, loss of ionic regulation, and cell shrinkage. These toxic effects were mediated through energy deprivation, p38 MAPK, protein kinase C, and casein kinase 1α.
Collapse
Affiliation(s)
| | | | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia; (S.A.A.); (M.A.A.)
| |
Collapse
|
14
|
Laschuk Herlinger A, Lovatto Michaelsen G, Sinigaglia M, Fratini L, Nogueira Debom G, Braganhol E, Brunetto de Farias C, Lunardi Brunetto A, Tesainer Brunetto A, da Cunha Jaeger M, Roesler R. Modulation of Viability, Proliferation, and Stemness by Rosmarinic Acid in Medulloblastoma Cells: Involvement of HDACs and EGFR. Neuromolecular Med 2023; 25:573-585. [PMID: 37740824 DOI: 10.1007/s12017-023-08758-x] [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: 01/24/2023] [Accepted: 08/30/2023] [Indexed: 09/25/2023]
Abstract
Medulloblastoma (MB) is a heterogeneous group of malignant pediatric brain tumors, divided into molecular groups with distinct biological features and prognoses. Currently available therapy often results in poor long-term quality of life for patients, which will be afflicted by neurological, neuropsychiatric, and emotional sequelae. Identifying novel therapeutic agents capable of targeting the tumors without jeopardizing patients' quality of life is imperative. Rosmarinic acid (RA) is a plant-derived compound whose action against a series of diseases including cancer has been investigated, with no side effects reported so far. Previous studies have not examined whether RA has effects in MB. Here, we show RA is cytotoxic against human Daoy (IC50 = 168 μM) and D283 (IC50 = 334 μM) MB cells. Exposure to RA for 48 h reduced histone deacetylase 1 (HDAC1) expression while increasing H3K9 hyperacetylation, reduced epidermal growth factor (EGFR) expression, and inhibited EGFR downstream targets extracellular-regulated kinase (ERK)1/2 and AKT in Daoy cells. These modifications were accompanied by increased expression of CDKN1A/p21, reduced expression of SOX2, and a decrease in proliferative rate. Treatment with RA also reduced cancer stem cell markers expression and neurosphere size. Taken together, our findings indicate that RA can reduce cell proliferation and stemness and induce cell cycle arrest in MB cells. Mechanisms mediating these effects may include targeting HDAC1, EGFR, and ERK signaling, and promoting p21 expression, possibly through an increase in H3K9ac and AKT deactivation. RA should be further investigated as a potential anticancer agent in experimental MB.
Collapse
Affiliation(s)
- Alice Laschuk Herlinger
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil.
| | - Gustavo Lovatto Michaelsen
- Graduate Program in Bioinformatics, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-400, Brazil
- Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Marialva Sinigaglia
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil
- Graduate Program in Bioinformatics, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-400, Brazil
- Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Lívia Fratini
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Gabriela Nogueira Debom
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, 90050-170, Brazil
| | - Elizandra Braganhol
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, 90050-170, Brazil
| | - Caroline Brunetto de Farias
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil
- Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Algemir Lunardi Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil
- Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - André Tesainer Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil
- Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Mariane da Cunha Jaeger
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil
- Children's Cancer Institute, Porto Alegre, RS, 90620-110, Brazil
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, RS, 90035-003, Brazil.
- Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
15
|
Rajaselvi ND, Jida MD, Ajeeshkumar KK, Nair SN, John P, Aziz Z, Nisha AR. Antineoplastic activity of plant-derived compounds mediated through inhibition of histone deacetylase: a review. Amino Acids 2023; 55:1803-1817. [PMID: 37389730 DOI: 10.1007/s00726-023-03298-x] [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: 04/13/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
In the combat of treating cancer recent therapeutic approaches are focused towards enzymatic targets as they occupy a pivotal participation in the cascade of oncogenesis and malignancy. There are several enzymes that modulate the epigenetic pathways and chromatin structure related to cancer mutation. Among several epigenetic mechanisms such as methylation, phosphorylation, and sumoylation, acetylation status of histones is crucial and is governed by counteracting enzymes like histone acetyl transferase (HAT) and histone deacetylases (HDAC) which have contradictory effects on the histone acetylation. HDAC inhibition induces chromatin relaxation which forms euchromatin and thereby initiates the expression of certain transcription factors attributed with apoptosis, which are mostly correlated with the expression of the p21 gene and acetylation of H3 and H4 histones. Most of the synthetic and natural HDAC inhibitors elicit antineoplastic effect through activation of various apoptotic pathways and promoting cell cycle arrest at various phases. Due to their promising chemo preventive action and low cytotoxicity against normal host cells, bioactive substances like flavonoids, alkaloids, and polyphenolic compounds from plants have recently gained importance. Even though all bioactive compounds mentioned have an HDAC inhibitory action, some of them have a direct effect and others enhance the effects of the standard well known HDAC inhibitors. In this review, the action of plant derived compounds against histone deacetylases in a variety of in vitro cancer cell lines and in vivo animal models are articulated.
Collapse
Affiliation(s)
- N Divya Rajaselvi
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, Mannuthy, Thrissur, 680 651, India
| | - M D Jida
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, Mannuthy, Thrissur, 680 651, India
| | - K K Ajeeshkumar
- Tumor Biology Lab, ICMR-National Institute of Pathology, New Delhi, India
| | - Suresh N Nair
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, Mannuthy, Thrissur, 680 651, India
| | - Preethy John
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, Pookode, Wayanad, 673 576, India
| | - Zarina Aziz
- Department of Veterinary Physiology, College of Veterinary and Animal Sciences, Mannuthy, Thrissur, 680 651, India
| | - A R Nisha
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, Mannuthy, Thrissur, 680 651, India.
| |
Collapse
|
16
|
Azhar MK, Anwar S, Hasan GM, Shamsi A, Islam A, Parvez S, Hassan MI. Comprehensive Insights into Biological Roles of Rosmarinic Acid: Implications in Diabetes, Cancer and Neurodegenerative Diseases. Nutrients 2023; 15:4297. [PMID: 37836581 PMCID: PMC10574478 DOI: 10.3390/nu15194297] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
Phytochemicals are abundantly occurring natural compounds extracted from plant sources. Rosmarinic acid (RA) is an abundant phytochemical of Lamiaceae species with various therapeutic implications for human health. In recent years, natural compounds have gained significant attention as adjuvant and complementary therapies to existing medications for various diseases. RA has gained popularity due to its anti-inflammatory and antioxidant properties and its roles in various life-threatening conditions, such as cancer, neurodegeneration, diabetes, etc. The present review aims to offer a comprehensive insight into the multifaceted therapeutic properties of RA, including its potential as an anticancer agent, neuroprotective effects, and antidiabetic potential. Based on the available evidences, RA could be considered a potential dietary component for treating various diseases, including cancer, diabetes and neurodegenerative disorders.
Collapse
Affiliation(s)
- Md. Khabeer Azhar
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India;
| | - Saleha Anwar
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India;
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia;
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 364, United Arab Emirates
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (A.I.); (M.I.H.)
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India;
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (A.I.); (M.I.H.)
| |
Collapse
|
17
|
Wang Y, Lu L, Ling C, Zhang P, Han R. Potential of Dietary HDAC2i in Breast Cancer Patients Receiving PD-1/PD-L1 Inhibitors. Nutrients 2023; 15:3984. [PMID: 37764768 PMCID: PMC10537481 DOI: 10.3390/nu15183984] [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: 08/02/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is a lethal malignancy with high morbidity and mortality but lacks effective treatments thus far. Despite the introduction of immune checkpoint inhibitors (ICIs) (including PD-1/PD-L1 inhibitors), durable and optimal clinical benefits still remain elusive for a considerable number of BC patients. To break through such a dilemma, novel ICI-based combination therapy has been explored for enhancing the therapeutic effect. Recent evidence has just pointed out that the HDAC2 inhibitor (HDAC2i), which has been proven to exhibit an anti-cancer effect, can act as a sensitizer for ICIs therapy. Simultaneously, dietary intervention, as a crucial supportive therapy, has been reported to provide ingredients containing HDAC2 inhibitory activity. Thus, the novel integration of dietary intervention with ICIs therapy may offer promising possibilities for improving treatment outcomes. In this study, we first conducted the differential expression and prognostic analyses of HDAC2 and BC patients using the GENT2 and Kaplan-Meier plotter platform. Then, we summarized the potential diet candidates for such an integrated therapeutic strategy. This article not only provides a whole new therapeutic strategy for an HDAC2i-containing diet combined with PD-1/PD-L1 inhibitors for BC treatment, but also aims to ignite enthusiasm for exploring this field.
Collapse
Affiliation(s)
- Yuqian Wang
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
| | - Changquan Ling
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Ping Zhang
- Center for Integrative Conservation, Yunnan Key Laboratory for the Conservation of Tropical Rainforests and Asian Elephants, Xishuangbanna Tropical Botanical Garden, Xishuangbanna 666303, China
| | - Rui Han
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
- Department of Oncology, The First Hospital Affiliated to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
18
|
Shoeibi A, Karimi E, Zareian M, Oskoueian E. Enhancing Healthcare Outcomes and Modulating Apoptosis- and Antioxidant-Related Genes through the Nano-Phytosomal Delivery of Phenolics Extracted from Allium ampeloprasum. Genes (Basel) 2023; 14:1547. [PMID: 37628599 PMCID: PMC10454362 DOI: 10.3390/genes14081547] [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: 05/12/2023] [Revised: 07/17/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
The application of nano drug delivery systems, particularly those utilizing natural bioactive compounds with anticancer properties, has gained significant attention. In this study, a novel nano-phytosome-loaded phenolic rich fraction (PRF) derived from Allium ampeloprasum L. was developed. The antitumor activity of the formulation was evaluated in BALB/c mice with TUBO colon carcinoma. The PRF-loaded nano-phytosome (PRF-NPs) exhibited a sphere-shaped structure (226 nm) and contained a diverse range of phenolic compounds. Animal trials conducted on TUBO tumor-bearing mice demonstrated that treatment with PRF-NPs at a dosage of 50 mg TPC/Kg/BW resulted in significant improvements in body weight and food intake, while reducing liver enzymes and lipid peroxidation. The expression of apoptosis-related genes, such as Bax and caspase-3, was upregulated, whereas Bcl2 was significantly downregulated (p < 0.05). Furthermore, the expression of GPx and SOD genes in the liver was notably increased compared to the control group. The findings suggest that the phytosomal encapsulation of the phenolic rich fraction derived from Allium ampeloprasum L. can enhance the bioavailability of natural phytochemicals and improve their antitumor properties. The development of PRF-NPs as a nano drug delivery system holds promise for effective breast cancer treatment.
Collapse
Affiliation(s)
- Ali Shoeibi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Ehsan Karimi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mohsen Zareian
- Department of Life Sciences, Chalmers University of Technology, Göteborg, Sweden
| | - Ehsan Oskoueian
- Industrial and Mineral Research Center, Arka Industrial Cluster, Mashhad, Iran
| |
Collapse
|
19
|
Ling R, Wang J, Fang Y, Yu Y, Su Y, Sun W, Li X, Tang X. HDAC-an important target for improving tumor radiotherapy resistance. Front Oncol 2023; 13:1193637. [PMID: 37503317 PMCID: PMC10368992 DOI: 10.3389/fonc.2023.1193637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Radiotherapy is an important means of tumor treatment, but radiotherapy resistance has been a difficult problem in the comprehensive treatment of clinical tumors. The mechanisms of radiotherapy resistance include the repair of sublethal damage and potentially lethal damage of tumor cells, cell repopulation, cell cycle redistribution, and reoxygenation. These processes are closely related to the regulation of epigenetic modifications. Histone deacetylases (HDACs), as important regulators of the epigenetic structure of cancer, are widely involved in the formation of tumor radiotherapy resistance by participating in DNA damage repair, cell cycle regulation, cell apoptosis, and other mechanisms. Although the important role of HDACs and their related inhibitors in tumor therapy has been reviewed, the relationship between HDACs and radiotherapy has not been systematically studied. This article systematically expounds for the first time the specific mechanism by which HDACs promote tumor radiotherapy resistance in vivo and in vitro and the clinical application prospects of HDAC inhibitors, aiming to provide a reference for HDAC-related drug development and guide the future research direction of HDAC inhibitors that improve tumor radiotherapy resistance.
Collapse
Affiliation(s)
- Rui Ling
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jingzhi Wang
- Department of Radiotherapy Oncology, Affiliated Yancheng First Hospital of Nanjing University Medical School, First People’s Hospital of Yancheng, Yancheng, China
| | - Yuan Fang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunpeng Yu
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Su
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Sun
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Tang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
20
|
da Silva GB, Manica D, da Silva AP, Marafon F, Moreno M, Bagatini MD. Rosmarinic acid decreases viability, inhibits migration and modulates expression of apoptosis-related CASP8/CASP3/NLRP3 genes in human metastatic melanoma cells. Chem Biol Interact 2023; 375:110427. [PMID: 36863647 DOI: 10.1016/j.cbi.2023.110427] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/13/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
Cutaneous melanoma is the most aggressive type of skin cancer; it is difficult to treat, and has been highlighted in recent years due to increasing numbers of cases worldwide. The use of antitumoral therapeutics for this neoplasm has been associated with severe side effects, low quality of life, and resistance. We aimed in this study to explore the effect of the phenolic compound rosmarinic acid (RA) on human metastatic melanoma cells. SK-MEL-28 melanoma cells were treated for 24 h with different concentrations of RA. In parallel, peripheral blood mononuclear cells (PBMCs) also were treated with RA under the same experimental conditions to verify the cytotoxic effect on non-tumoral cells. Then, we assessed cell viability and migration, levels of intracellular and extracellular reactive oxygen species (ROS), as well as nitric oxide (NOx), non-protein thiols (NPSH), and total thiol (PSH). Gene expression of the caspase 8, caspase 3 and NLRP3 inflammasome was evaluated by RT-qPCR. The enzymatic activity of the caspase 3 protein was assessed by a sensitive fluorescent assay. Fluorescence microscopy was employed to corroborate the effects of RA on melanoma cell viability, mitochondria transmembrane potential and apoptotic bodies formation. We found that RA potently reduces melanoma cell viability and migration after 24 h of treatment. On the other hand, it has no cytotoxic effect on non-tumoral cells. The fluorescence micrographics indicated that RA reduces transmembrane potential of mitochondria and induces apoptotic bodies formation. Moreover, RA significantly decreases intracellular and extracellular ROS levels, and increases the antioxidant defenders NPSH and PSH. A remarkable feature found in our study was that RA strongly upregulates the gene expression of the caspase 8 and caspase 3, and downregulates NLRP3 inflammasome expression. Similar to gene expression, RA greatly increases the enzymatic activity of caspase 3 protein. Taken together, we have shown for the first time that RA reduces cell viability and migration of human metastatic melanoma cells, in addition to modulates apoptosis-related gene expression. We suggest that RA may have the potential to be used in a therapeutic perspective, particularly for CM cell treatment.
Collapse
Affiliation(s)
- Gilnei Bruno da Silva
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Daiane Manica
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Alana Patrícia da Silva
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Filomena Marafon
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Marcelo Moreno
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Margarete Dulce Bagatini
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
21
|
Dong F, Qu L, Duan Z, He Y, Ma X, Fan D. Ginsenoside Rh4 inhibits breast cancer growth through targeting histone deacetylase 2 to regulate immune microenvironment and apoptosis. Bioorg Chem 2023; 135:106537. [PMID: 37043883 DOI: 10.1016/j.bioorg.2023.106537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023]
Abstract
High expression of histone deacetylase 2 (HDAC2) is recognized as a marker of invasive breast cancer (BC). HDAC2 is not only responsible for enhancing tumor cell growth, development, and anti-apoptosis, but also plays a significant role in regulating PD-L1 on the surface of tumor cells. Continuous expression of PD-L1 allows tumor cells to escape immune surveillance. There is not much research on how HDAC2 affects the immune system in breast cancer. Ginsenoside Rh4 (Rh4) is a major rare saponin in heat-treated ginseng, which is widely applied in treating and preventing various diseases because of its potent medicinal value and stable safety. However, it is unclear how Rh4 affects the tumor immune microenvironment in breast cancer. Therefore, this paper aims to investigate the effect of Rh4 on HDAC2 in breast cancer, specifically the effect of HDAC2 on apoptosis and the immune microenvironment to inhibit breast cancer growth. According to our study, ginsenoside Rh4 has been shown to significantly suppress breast cancer cell proliferation without any adverse effects. The molecular docking results of Rh4 and HDAC2 indicate a binding energy of -6.06 kcal/mol, suggesting the potential of Rh4 as a targeting modulator of HDAC2. Mechanistically, Rh4 induces apoptosis of breast cancer cells by the HDAC2-mediated caspase pathway and inhibits the HDAC2-mediated JAK/STAT pathway to regulate the immune microenvironment, which inhibits breast cancer growth. Specifically, Rh4 was shown for the first time to blockade immune checkpoints (PD-1/PD-L1) and increase levels of T-lymphocytes in the tumor. In a word, our study establishes a theoretical framework for applying Rh4 as an immune checkpoint inhibitor as part of breast cancer treatment.
Collapse
Affiliation(s)
- Fangming Dong
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Linlin Qu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Zhiguang Duan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Ying He
- Shaanxi Giant Biotechnology Co., LTD, No. 20, Zone C, Venture R&D Park, No. 69, Jinye Road, High-tech Zone, Xi'an, Shaanxi 710076, China
| | - Xiaoxuan Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China.
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
22
|
Let’s Go 3D! New Generation of Models for Evaluating Drug Response and Resistance in Prostate Cancer. Int J Mol Sci 2023; 24:ijms24065293. [PMID: 36982368 PMCID: PMC10049142 DOI: 10.3390/ijms24065293] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Prostate cancer (PC) is the third most frequently diagnosed cancer worldwide and the second most frequent in men. Several risk factors can contribute to the development of PC, and those include age, family history, and specific genetic mutations. So far, drug testing in PC, as well as in cancer research in general, has been performed on 2D cell cultures. This is mainly because of the vast benefits these models provide, including simplicity and cost effectiveness. However, it is now known that these models are exposed to much higher stiffness; lose physiological extracellular matrix on artificial plastic surfaces; and show changes in differentiation, polarization, and cell–cell communication. This leads to the loss of crucial cellular signaling pathways and changes in cell responses to stimuli when compared to in vivo conditions. Here, we emphasize the importance of a diverse collection of 3D PC models and their benefits over 2D models in drug discovery and screening from the studies done so far, outlining their benefits and limitations. We highlight the differences between the diverse types of 3D models, with the focus on tumor–stroma interactions, cell populations, and extracellular matrix composition, and we summarize various standard and novel therapies tested on 3D models of PC for the purpose of raising awareness of the possibilities for a personalized approach in PC therapy.
Collapse
|
23
|
Phenolic compounds as histone deacetylase inhibitors: binding propensity and interaction insights from molecular docking and dynamics simulations. Amino Acids 2023:10.1007/s00726-023-03249-6. [PMID: 36781452 DOI: 10.1007/s00726-023-03249-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/15/2023]
Abstract
Histone deacetylases are well-established target enzymes involved in the pathology of different diseases including cancer and neurodegenerative disorders. The approved HDAC inhibitor drugs are associated with cellular toxicities. Different phenolic compounds have been shown to possess inhibitory activities against HDACs and are, therefore, considered safer alternatives to synthetic compounds. Here, we elucidated the binding mode and calculated the binding propensity of some of the top phenolic compounds against different isoforms representing different classes of Zn2+ ion-containing HDACs using the molecular docking approach. Our data reaffirmed the activity of the studied phenolic compounds against HDACs. Binding interaction analysis suggested that these compounds can block the activity of HDACs with or without binding to the active site zinc metal ion. Furthermore, molecular dynamics (MD) simulations were carried out on the selected crystal and docking complexes of each selected HDAC isoform. Analysis of root-mean-square displacement (RMSD) showed that the phenolic compounds demonstrated a stable binding mode over 50 ns in a way that is comparable to the cocrystal ligands. Together, these findings can aid future efforts in the search for natural inhibitors of HDACs.
Collapse
|
24
|
Mi YY, Ji Y, Zhang L, Sun CY, Wei BB, Yang DJ, Wan HY, Qi XW, Wu S, Zhu LJ. A first-in-class HBO1 inhibitor WM-3835 inhibits castration-resistant prostate cancer cell growth in vitro and in vivo. Cell Death Dis 2023; 14:67. [PMID: 36709328 PMCID: PMC9884225 DOI: 10.1038/s41419-023-05606-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
The prognosis and overall survival of castration-resistant prostate cancer (CRPC) patients are poor. The search for novel and efficient anti-CRPC agents is therefore extremely important. WM-3835 is a cell-permeable, potent and first-in-class HBO1 (KAT7 or MYST2) inhibitor. Here in primary human prostate cancer cells-derived from CRPC patients, WM-3835 potently inhibited cell viability, proliferation, cell cycle progression and in vitro cell migration. The HBO1 inhibitor provoked apoptosis in the prostate cancer cells. It failed to induce significant cytotoxicity and apoptosis in primary human prostate epithelial cells. shRNA-induced silencing of HBO1 resulted in robust anti-prostate cancer cell activity as well, and adding WM-3835 failed to induce further cytotoxicity in the primary prostate cancer cells. Conversely, ectopic overexpression of HBO1 further augmented primary prostate cancer cell proliferation and migration. WM-3835 inhibited H3-H4 acetylation and downregulated several pro-cancerous genes (CCR2, MYLK, VEGFR2, and OCIAD2) in primary CRPC cells. Importantly, HBO1 mRNA and protein levels are significantly elevated in CRPC tissues and cells. In vivo, daily intraperitoneal injection of WM-3835 potently inhibited pPC-1 xenograft growth in nude mice, and no apparent toxicities detected. Moreover, intratumoral injection of HBO1 shRNA adeno-associated virus (AAV) suppressed the growth of primary prostate cancer xenografts in nude mice. H3-H4 histone acetylation and HBO1-dependent genes (CCR2, MYLK, VEGFR2, and OCIAD2) were remarkably decreased in WM-3835-treated or HBO1-silenced xenograft tissues. Together, targeting HBO1 by WM-3835 robustly inhibits CRPC cell growth.
Collapse
Affiliation(s)
- Yuan-Yuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yu Ji
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Lifeng Zhang
- Department of Urology, Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Chuan-Yu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing-Bing Wei
- Department of Urology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Dong-Jie Yang
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hong-Yuan Wan
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiao-Wei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Sheng Wu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Li-Jie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
25
|
Martins-Gomes C, Nunes FM, Silva AM. Modulation of Cell Death Pathways for Cellular Protection and Anti-Tumoral Activity: The Role of Thymus spp. Extracts and Their Bioactive Molecules. Int J Mol Sci 2023; 24:ijms24021691. [PMID: 36675206 PMCID: PMC9864824 DOI: 10.3390/ijms24021691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Natural products used for their health-promoting properties have accompanied the evolution of humanity. Nowadays, as an effort to scientifically validate the health-promoting effects described by traditional medicine, an ever-growing number of bioactivities are being described for natural products and the phytochemicals that constitute them. Among them, medicinal plants and more specifically the Thymus genus spp., arise as products already present in the diet and with high acceptance, that are a source of phytochemicals with high pharmacological value. Phenolic acids, flavonoid glycoside derivatives, and terpenoids from Thymus spp. have been described for their ability to modulate cell death and survival pathways, much-valued bioactivities in the pharmaceutical industry, that continually sought-after new formulations to prevent undesired cell death or to control cell proliferation. Among these, wound treatment, protection from endogenous/exogenous toxic molecules, or the induction of selective cell death, such as the search for new anti-tumoral agents, arise as main objectives. This review summarizes and discusses studies on Thymus spp., as well as on compounds present in their extracts, with regard to their health-promoting effects involving the modulation of cell death or survival signaling pathways. In addition, studies regarding the main bioactive molecules and their cellular molecular targets were also reviewed. Concerning cell survival and proliferation, Thymus spp. present themselves as an option for new formulations designed for wound healing and protection against chemicals-induced toxicity. However, Thymus spp. extracts and some of their compounds regulate cell death, presenting anti-tumoral activity. Therefore Thymus spp. is a rich source of compounds with nutraceutical and pharmaceutical value.
Collapse
Affiliation(s)
- Carlos Martins-Gomes
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Cell Biology and Biochemistry Lab, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Lab, UTAD Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Fernando M. Nunes
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Lab, UTAD Quinta de Prados, 5001-801 Vila Real, Portugal
- Department of Chemistry, School of Life Sciences and Environment, UTAD, 5001-801 Vila Real, Portugal
| | - Amélia M. Silva
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Cell Biology and Biochemistry Lab, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Department of Biology and Environment, School of Life Sciences and Environment, UTAD, 5001-801 Vila Real, Portugal
- Correspondence: ; Tel.: +351-259-350-921
| |
Collapse
|
26
|
Hemmati Bushehri R, Navabi P, Saeedifar AM, Keshavarzian N, Hosseini Rouzbahani N, Mosayebi G, Ghazavi A, Ghorban K, Ganji A. Integration of phytotherapy and chemotherapy: Recent advances in anticancer molecular pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:987-1000. [PMID: 37605725 PMCID: PMC10440131 DOI: 10.22038/ijbms.2023.69979.15222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/20/2023] [Indexed: 08/23/2023]
Abstract
Cancer is a disease characterized by abnormal and uncontrolled growth of cells, leading to invasion and metastasis to other tissues. Chemotherapy drugs are some of the primary treatments for cancer, which could detrimentally affect the cancer cells by various molecular mechanisms like apoptosis and cell cycle arrest. These treatment lines have always aligned with side effects and drug resistance. Due to their anticancer effects, medicinal herbs and their active derivative compounds are being profoundly used as complementary treatments for cancer. Many studies have shown that herbal ingredients exert antitumor activities and immune-modulation effects and have fewer side effects. On the other hand, combining phytotherapy and chemotherapy, with their synergistic effects, has gained much attention across the medical community. This review article discussed the therapeutic effects of essential herbal active ingredients combined with chemotherapeutic drugs in cancer therapy. To write this article, PubMed and Scopus database were searched with the keywords "Cancer," "Combination," "Herbal," "Traditional," and "Natural." After applying inclusion/exclusion criteria, 110 articles were considered. The study shows the anticancer effects of the active herbal ingredients by inducing apoptosis and cell cycle arrest in cancer cells, especially with a chemotherapeutic agent. This study also indicates that herbal compounds can reduce side effects and dosage, potentiate anticancer responses, and sensitize cancer cells to chemotherapy drugs.
Collapse
Affiliation(s)
| | - Parnian Navabi
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | | | - Nafiseh Keshavarzian
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | | | - Ghasem Mosayebi
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Ali Ghazavi
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
- Traditional and Complementary Medicine Research Center (TCMRC), Arak University of Medical Sciences, Arak, Iran
| | - Khodayar Ghorban
- Department of Immunology, Medical School, Aja University of Medical Sciences, Tehran, Iran
| | - Ali Ganji
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
27
|
Qin LT, Huang SW, Huang ZG, Dang YW, Fang YY, He J, Niu YT, Lin CX, Wu JY, Wei ZX. Clinical value and potential mechanisms of BUB1B up-regulation in nasopharyngeal carcinoma. BMC Med Genomics 2022; 15:272. [PMID: 36577966 PMCID: PMC9798722 DOI: 10.1186/s12920-022-01412-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) has insidious onset, late clinical diagnosis and high recurrence rate, which leads to poor quality of patient life. Therefore, it is necessary to further explore the pathogenesis and therapy targets of NPC. BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) was found to be up-regulated in a variety of cancers, but only two previous study showed that BUB1B was overexpressed in NPC and the sample size was small. The clinical role of BUB1B expression and its underlying mechanism in NPC require more in-depth research. Immunohistochemical samples and public RNA-seq data indicated that BUB1B protein and mRNA expression levels were up-regulated in NPC, and summary receiver operating characteristic curve indicated that BUB1B expression level had a strong ability to distinguish NPC tissues from non-NPC tissues. Gene ontology and Kyoto Encyclopedia of genes and genomes were performed and revealed that BUB1B and its related genes were mainly involved in cell cycle and DNA replication. Protein- Protein Interaction were built to interpret the BUB1B molecular mechanism. Histone deacetylase 2 (HDAC2) could be the upstream regulation factor of BUB1B, which was verified by Chromatin Immunoprecipitation Sequencing samples. In summary, BUB1B was highly expressed in NPC, and HDAC2 may affect cell cycle by regulating BUB1B to promote cancer progression.
Collapse
Affiliation(s)
- Li-Ting Qin
- grid.412594.f0000 0004 1757 2961Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Si-Wei Huang
- grid.412594.f0000 0004 1757 2961Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Zhi-Guang Huang
- grid.412594.f0000 0004 1757 2961Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Yi-Wu Dang
- grid.412594.f0000 0004 1757 2961Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Ye-Ying Fang
- grid.412594.f0000 0004 1757 2961Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Juan He
- grid.412594.f0000 0004 1757 2961Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Yi-Tong Niu
- grid.412594.f0000 0004 1757 2961Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Cai-Xing Lin
- grid.412594.f0000 0004 1757 2961Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Ji-Yun Wu
- grid.412594.f0000 0004 1757 2961Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| | - Zhu-Xin Wei
- grid.412594.f0000 0004 1757 2961Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, 6 Shuangyong Road, Nanning, 530021 People’s Republic of China
| |
Collapse
|
28
|
Chaitanya MVNL, Ramanunny AK, Babu MR, Gulati M, Vishwas S, Singh TG, Chellappan DK, Adams J, Dua K, Singh SK. Journey of Rosmarinic Acid as Biomedicine to Nano-Biomedicine for Treating Cancer: Current Strategies and Future Perspectives. Pharmaceutics 2022; 14:2401. [PMID: 36365218 PMCID: PMC9696899 DOI: 10.3390/pharmaceutics14112401] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 10/07/2023] Open
Abstract
Rosmarinic acid (RA) is a polyphenolic metabolite found in various culinary, dietary sources, and medicinal plants like Coleus scutellarioides (Linn) Benth., Lavandula angustifolia Linn., Mellisa officinalis Linn., Origanum vulgare Linn., Rosmarinus officinalis Linn., Zataria multiflora Boiss. and Zhumeria majdae Rech. F. Apart from its dietary and therapeutic values, RA is an important anticancer phytochemical owing to its multi-targeting anticancer mechanism. These properties provide a scope for RA's therapeutic uses beyond its traditional use as a dietary source. However, its oral bioavailability is limited due to its poor solubility and permeability. This impedes its efficacy in treating cancer. Indeed, in recent years, tremendous efforts have been put towards the development of nanoformulations of RA for treating cancer. However, this research is in its initial stage as bringing a nanoparticle into the market itself is associated with many issues such as stability, toxicity, and scale-up issues. Considering these pitfalls during formulation development and overcoming them would surely provide a new face to RA as a nanomedicine to treat cancer. A literature search was conducted to systematically review the various biological sources, extraction techniques, and anticancer mechanisms through which RA showed multiple therapeutic effects. Various nanocarriers of RA pertaining to its anticancer activity are also discussed in this review.
Collapse
Affiliation(s)
| | | | - Malakapogu Ravindra Babu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Dinesh Kumar Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Jon Adams
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University Technology Sydney, Ultimo, NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
29
|
Kakanezhadi A, Rezaei M, Raisi A, Dezfoulian O, Davoodi F, Ahmadvand H. Rosmarinic acid prevents post-operative abdominal adhesions in a rat model. Sci Rep 2022; 12:18593. [PMID: 36329196 PMCID: PMC9633689 DOI: 10.1038/s41598-022-22000-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
This study aims to determine the effects of rosmarinic acid which involved the mechanisms to decrease the postoperative peritoneal adhesion formation in rats. Various incisions and removing a 1 × 1 cm piece of peritoneum was used to induce the peritoneal adhesions. Experimental groups were as follows: 1-Sham group. 2-Control group: Peritoneal adhesions were induced and no treatments were performed. 3-Treatment groups: Following inducing peritoneal adhesions, animals received rosmarinic acid with 50 and 70 mg/kg dosage, respectively. Macroscopic examination of adhesions indicated that adhesion bands were reduced in both treatment groups compared to the control group. Moreover, the adhesion score was decreased in both treatment groups on day 14. Inflammation and fibroblast proliferation were both reduced in the treatment groups on day 14. TGF-β1, TNF-α, and VEGF were all evaluated by western blot and immunohistochemistry on days 3 and 14. Treatment groups reduced inflammatory cytokines on days 3 and 14. The treatment group with a 70 mg/kg dosage decreased TGF-β1 and TNF-α levels more than the other treatment group. The administration of rosmarinic acid significantly reduced MDA and increased CAT levels. In conclusion, the rosmarinic acid was effective to reduce the adhesion bands, inflammatory cytokines, angiogenesis, and oxidative stress.
Collapse
Affiliation(s)
- Ali Kakanezhadi
- grid.411406.60000 0004 1757 0173Department of Clinical Sciences, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Mehrdad Rezaei
- grid.411406.60000 0004 1757 0173Department of Clinical Sciences, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Abbas Raisi
- grid.411406.60000 0004 1757 0173Department of Clinical Sciences, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Omid Dezfoulian
- grid.411406.60000 0004 1757 0173Department of Pathobiology, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Farshid Davoodi
- grid.412763.50000 0004 0442 8645Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Hassan Ahmadvand
- grid.411950.80000 0004 0611 9280Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
30
|
Bakrim S, El Omari N, El Hachlafi N, Bakri Y, Lee LH, Bouyahya A. Dietary Phenolic Compounds as Anticancer Natural Drugs: Recent Update on Molecular Mechanisms and Clinical Trials. Foods 2022; 11:foods11213323. [PMID: 36359936 PMCID: PMC9657352 DOI: 10.3390/foods11213323] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 12/05/2022] Open
Abstract
Given the stochastic complexity of cancer diseases, the development of chemotherapeutic drugs is almost limited by problems of selectivity and side effects. Furthermore, an increasing number of protective approaches have been recently considered as the main way to limit these pathologies. Natural bioactive compounds, and particularly dietary phenolic compounds, showed major protective and therapeutic effects against different types of human cancers. Indeed, phenolic substances have functional groups that allow them to exert several anti-cancer mechanisms, such as the induction of apoptosis, autophagy, cell cycle arrest at different stages, and the inhibition of telomerase. In addition, in vivo studies show that these phenolic compounds also have anti-angiogenic effects via the inhibition of invasion and angiogenesis. Moreover, clinical studies have already highlighted certain phenolic compounds producing clinical effects alone, or in combination with drugs used in chemotherapy. In the present work, we present a major advance in research concerning the mechanisms of action of the different phenolic compounds that are contained in food medicinal plants, as well as evidence from the clinical trials that focus on them.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology, and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Naoufal El Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, Fes 30000, Morocco
| | - Youssef Bakri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya 47500, Malaysia
- Correspondence: (L.-H.L.); (A.B.)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
- Correspondence: (L.-H.L.); (A.B.)
| |
Collapse
|
31
|
Polyphenols as Potent Epigenetics Agents for Cancer. Int J Mol Sci 2022; 23:ijms231911712. [PMID: 36233012 PMCID: PMC9570183 DOI: 10.3390/ijms231911712] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 02/06/2023] Open
Abstract
Human diseases such as cancer can be caused by aberrant epigenetic regulation. Polyphenols play a major role in mammalian epigenome regulation through mechanisms and proteins that remodel chromatin. In fruits, seeds, and vegetables, as well as food supplements, polyphenols are found. Compounds such as these ones are powerful anticancer agents and antioxidants. Gallic acid, kaempferol, curcumin, quercetin, and resveratrol, among others, have potent anti-tumor effects by helping reverse epigenetic changes associated with oncogene activation and tumor suppressor gene inactivation. The role dietary polyphenols plays in restoring epigenetic alterations in cancer cells with a particular focus on DNA methylation and histone modifications was summarized. We also discussed how these natural compounds modulate gene expression at the epigenetic level and described their molecular targets in cancer. It highlights the potential of polyphenols as an alternative therapeutic approach in cancer since they modulate epigenetic activity.
Collapse
|
32
|
Zhao J, Xu L, Jin D, Xin Y, Tian L, Wang T, Zhao D, Wang Z, Wang J. Rosmarinic Acid and Related Dietary Supplements: Potential Applications in the Prevention and Treatment of Cancer. Biomolecules 2022; 12:biom12101410. [PMID: 36291619 PMCID: PMC9599057 DOI: 10.3390/biom12101410] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer constitutes a severe threat to human health and quality of life and is one of the most significant causes of morbidity and mortality worldwide. Natural dietary products have drawn substantial attention in cancer treatment and prevention due to their availability and absence of toxicity. Rosmarinic acid (RA) is known for its excellent antioxidant properties and is safe and effective in preventing and inhibiting tumors. This review summarizes recent publications on culture techniques, extraction processes, and anti-tumor applications of RA-enriched dietary supplements. We discuss techniques to improve RA bioavailability and provide a mechanistic discussion of RA regarding tumor prevention, treatment, and adjuvant therapy. RA exhibits anticancer activity by regulating oxidative stress, chronic inflammation, cell cycle, apoptosis, and metastasis. These data suggest that daily use of RA-enriched dietary supplements can contribute to tumor prevention and treatment. RA has the potential for application in anti-tumor drug development.
Collapse
Affiliation(s)
- Jiachao Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Liwei Xu
- Department of Respirology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Di Jin
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yu Xin
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Lin Tian
- Department of Respirology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Tan Wang
- Department of Respirology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Zeyu Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
- Correspondence: (Z.W.); (J.W.)
| | - Jing Wang
- Department of Respirology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
- Correspondence: (Z.W.); (J.W.)
| |
Collapse
|
33
|
Rosmarinic acid production in hairy root cultures of Salvia nemorosa L. (Lamiaceae). BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2022. [DOI: 10.1016/j.bcab.2022.102494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
Cytotoxic Effect of Rosmarinus officinalis Extract on Glioblastoma and Rhabdomyosarcoma Cell Lines. Molecules 2022; 27:molecules27196348. [PMID: 36234882 PMCID: PMC9573533 DOI: 10.3390/molecules27196348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Rosmarinus officinalis is a well-studied plant, known for its therapeutic properties. However, its biological activity against several diseases is not known in detail. The aim of this study is to present new data regarding the cytotoxic activity of a hydroethanolic extract of Rosmarinus officinalis on glioblastoma (A172) and rhabdomyosarcoma (TE671) cancer cell lines. The chemical composition of the extract is evaluated using liquid chromatography combined with time-of-flight mass spectrometry, alongside its total phenolic content and antioxidant activity. The extract showed a promising time- and dose-dependent cytotoxic activity against both cell lines. The lowest IC50 values for both cell lines were calculated at 72 h after treatment and correspond to 0.249 ± 1.09 mg/mL for TE671 cell line and 0.577 ± 0.98 mg/mL for A172 cell line. The extract presented high phenolic content, equal to 35.65 ± 0.03 mg GAE/g of dry material as well as a strong antioxidant activity. The IC50 values for the antioxidant assays were estimated at 12.8 ± 2.7 μg/mL (DPPH assay) and 6.98 ± 1.9 μg/mL (ABTS assay). The compound detected in abundance was carnosol, a phenolic diterpene, followed by the polyphenol rosmarinic acid, while the presence of phenolic compounds such as rhamnetin glucoside, hesperidin, cirsimaritin was notable. These preliminary results suggest that R. officinalis is a potential, alternative source of bioactive compounds to further examine for abilities against glioblastoma and rhabdomyosarcoma.
Collapse
|
35
|
Mahmod AI, Haif SK, Kamal A, Al-Ataby IA, Talib WH. Chemoprevention effect of the Mediterranean diet on colorectal cancer: Current studies and future prospects. Front Nutr 2022; 9:924192. [PMID: 35990343 PMCID: PMC9386380 DOI: 10.3389/fnut.2022.924192] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/18/2022] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second most deadly cancer worldwide. Nevertheless, more than 70% of CRC cases are resulted from sporadic tumorigenesis and are not inherited. Since adenoma-carcinoma development is a slow process and may take up to 20 years, diet-based chemoprevention could be an effective approach in sporadic CRC. The Mediterranean diet is an example of a healthy diet pattern that consists of a combination of nutraceuticals that prevent several chronic diseases and cancer. Many epidemiological studies have shown the correlation between adherence to the Mediterranean diet and low incidence of CRC. The goal of this review is to shed the light on the anti-inflammatory and anti-colorectal cancer potentials of the natural bioactive compounds derived from the main foods in the Mediterranean diet.
Collapse
Affiliation(s)
- Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| | - Shatha Khaled Haif
- Department of Pharmacy, Princess Sarvath Community College, Amman, Jordan
| | - Ayah Kamal
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| | - Israa A Al-Ataby
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| |
Collapse
|
36
|
Talib WH, AlHur MJ, Al.Naimat S, Ahmad RE, Al-Yasari AH, Al-Dalaeen A, Thiab S, Mahmod AI. Anticancer Effect of Spices Used in Mediterranean Diet: Preventive and Therapeutic Potentials. Front Nutr 2022; 9:905658. [PMID: 35774546 PMCID: PMC9237507 DOI: 10.3389/fnut.2022.905658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/16/2022] [Indexed: 01/18/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide, with almost 10 million cancer-related deaths worldwide in 2020, so any investigation to prevent or cure this disease is very important. Spices have been studied widely in several countries to treat different diseases. However, studies that summarize the potential anticancer effect of spices used in Mediterranean diet are very limited. This review highlighted chemo-therapeutic and chemo-preventive effect of ginger, pepper, rosemary, turmeric, black cumin and clove. Moreover, the mechanisms of action for each one of them were figured out such as anti-angiogenesis, antioxidant, altering signaling pathways, induction of cell apoptosis, and cell cycle arrest, for several types of cancer. The most widely used spice in Mediterranean diet is black pepper (Piper nigrum L). Ginger and black cumin have the highest anticancer activity by targeting multiple cancer hallmarks. Apoptosis induction is the most common pathway activated by different spices in Mediterranean diet to inhibit cancer. Studies discussed in this review may help researchers to design and test new anticancer diets enriched with selected spices that have high activities.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
- *Correspondence: Wamidh H. Talib
| | - Mallak J. AlHur
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| | - Sumaiah Al.Naimat
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| | - Rawand E. Ahmad
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| | | | - Anfal Al-Dalaeen
- Department of Clinical Nutrition and Dietetics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Samar Thiab
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| |
Collapse
|
37
|
Guan H, Luo W, Bao B, Cao Y, Cheng F, Yu S, Fan Q, Zhang L, Wu Q, Shan M. A Comprehensive Review of Rosmarinic Acid: From Phytochemistry to Pharmacology and Its New Insight. Molecules 2022; 27:3292. [PMID: 35630768 PMCID: PMC9143754 DOI: 10.3390/molecules27103292] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022] Open
Abstract
Polyphenolic acids are the widely occurring natural products in almost each herbal plant, among which rosmarinic acid (RA, C18H16O8) is well-known, and is present in over 160 species belonging to many families, especially the Lamiaceae. Aside from this herbal ingredient, dozens of its natural derivatives have also been isolated and characterized from many natural plants. In recent years, with the increasing focus on the natural products as alternative treatments, a large number of pharmacological studies have been carried out to demonstrate the various biological activities of RA such as anti-inflammation, anti-oxidation, anti-diabetes, anti-virus, anti-tumor, neuroprotection, hepatoprotection, etc. In addition, investigations concerning its biosynthesis, extraction, analysis, clinical applications, and pharmacokinetics have also been performed. Although many achievements have been made in various research aspects, there still exist some problems or issues to be answered, especially its toxicity and bioavailability. Thus, we hope that in the case of natural products, the present review can not only provide a comprehensive understanding on RA covering its miscellaneous research fields, but also highlight some of the present issues and future perspectives worth investigating later, in order to help us utilize this polyphenolic acid more efficiently, widely, and safely.
Collapse
Affiliation(s)
- Huaquan Guan
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (H.G.); (W.L.); (Q.F.)
| | - Wenbin Luo
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (H.G.); (W.L.); (Q.F.)
| | - Beihua Bao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yudan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fangfang Cheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sheng Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiaoling Fan
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (H.G.); (W.L.); (Q.F.)
| | - Li Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qinan Wu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mingqiu Shan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China; (B.B.); (Y.C.); (F.C.); (S.Y.); (L.Z.); (Q.W.)
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
38
|
Luo W, Tao Y, Chen S, Luo H, Li X, Qu S, Chen K, Zeng C. Rosmarinic Acid Ameliorates Pulmonary Ischemia/Reperfusion Injury by Activating the PI3K/Akt Signaling Pathway. Front Pharmacol 2022; 13:860944. [PMID: 35645792 PMCID: PMC9132383 DOI: 10.3389/fphar.2022.860944] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/30/2022] [Indexed: 01/01/2023] Open
Abstract
Pulmonary ischemia/reperfusion (IR) injury is the leading cause of acute lung injury, which is mainly attributed to reactive oxygen species (ROS) induced cell injuries and apoptosis. Since rosmarinic acid (RA) has been identified as an antioxidant natural ester, this natural compound might protect against pulmonary IR injury. In this study, the mice were given RA daily (50, 75, or 100 mg/kg) by gavage for 7 days before the pulmonary IR injury. We found that hypoxemia, pulmonary edema, and serum inflammation cytokines were aggravated in pulmonary IR injury. RA pretreatment (75 and 100 mg/kg) effectively reversed these parameters, while 50 mg/kg RA pretreatment was less pronounced. Our data also indicated RA pretreatment mitigated the upregulation of pro-oxidant NADPH oxidases (NOX2 and NOX4) and the downregulation of anti-oxidant superoxide dismutases (SOD1 and SOD2) upon IR injury. In vitro studies showed RA preserved the viability of anoxia/reoxygenation (AR)-treated A549 cells (a human lung epithelial cell line), and the results showed the protective effect of RA started at 5 μM concentration, reached its maximum at 15 μM, and gradually decreased at 20–25 μM. Besides, RA pretreatment (15 μM) greatly reduced the lactate dehydrogenase release levels subjected to AR treatment. Moreover, the results of our research revealed that RA eliminated ROS production and reduced alveolar epithelial cell apoptosis through activating the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) signaling pathway, which was supported by using wortmannin, because in the presence of wortmannin, the RA-mediated protection was blocked. Meanwhile, wortmannin also reversed the protective effects of RA in mice. Together, our results demonstrate the beneficial role of RA in pulmonary IR injury via PI3K/Akt-mediated anti-oxidation and anti-apoptosis, which could be a promising therapeutic intervention for pulmonary IR injury.
Collapse
Affiliation(s)
- Wenbin Luo
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Tao
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Shengnan Chen
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoping Li
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuang Qu
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Ken Chen
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, China
- *Correspondence: Ken Chen, ; Chunyu Zeng,
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, China
- *Correspondence: Ken Chen, ; Chunyu Zeng,
| |
Collapse
|
39
|
Noor S, Mohammad T, Rub MA, Raza A, Azum N, Yadav DK, Hassan MI, Asiri AM. Biomedical features and therapeutic potential of rosmarinic acid. Arch Pharm Res 2022; 45:205-228. [PMID: 35391712 PMCID: PMC8989115 DOI: 10.1007/s12272-022-01378-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/18/2022] [Indexed: 12/17/2022]
Abstract
For decades, the use of secondary metabolites of various herbs has been an attractive strategy in combating human diseases. Rosmarinic acid (RA) is a bioactive phenolic compound commonly found in plants of Lamiaceae and Boraginaceae families. RA is biosynthesized using amino acids tyrosine and phenylalanine via enzyme-catalyzed reactions. However, the chemical synthesis of RA involves an esterification reaction between caffeic acid and 3,4-dihydroxy phenyl lactic acid contributing two phenolic rings to the structure of RA. Several studies have ascertained multiple therapeutic benefits of RA in various diseases, including cancer, diabetes, inflammatory disorders, neurodegenerative disorders, and liver diseases. Many previous scientific papers indicate that RA can be used as an anti-plasmodic, anti-viral and anti-bacterial drug. In addition, due to its high anti-oxidant capacity, this natural polyphenol has recently gained attention for its possible application as a nutraceutical compound in the food industry. Here we provide state-of-the-art, flexible therapeutic potential and biomedical features of RA, its implications and multiple uses. Along with various valuable applications in safeguarding human health, this review further summarizes the therapeutic advantages of RA in various human diseases, including cancer, diabetes, neurodegenerative diseases. Furthermore, the challenges associated with the clinical applicability of RA have also been discussed.
Collapse
Affiliation(s)
- Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Malik Abdul Rub
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ali Raza
- Department of Medical Biochemistry, Jawahar Lal Nehru Medical College, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Naved Azum
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsugu, Incheon, 21924, Korea.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| | - Abdullah M Asiri
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
40
|
da Silva GB, Yamauchi MA, Zanini D, Bagatini MD. Novel possibility for cutaneous melanoma treatment by means of rosmarinic acid action on purinergic signaling. Purinergic Signal 2022; 18:61-81. [PMID: 34741236 PMCID: PMC8570242 DOI: 10.1007/s11302-021-09821-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer cases have increased significantly in Brazil and worldwide, with cutaneous melanoma (CM) being responsible for nearly 57,000 deaths in the world. Thus, this review article aims at exploring and proposed hypotheses with respect to the possibility that RA can be a promising and alternative compound to be used as an adjuvant in melanoma treatment, acting on purinergic signaling. The scarcity of articles evidencing the action of this compound in this signaling pathway requires further studies. Considering diverse evidence found in the literature, we hypothesize that RA can be an effective candidate for the treatment of CM acting as a modulating molecule of purinergic cellular pathway through P2X7 blocking, mitigating the Warburg effect, and as antagonic molecule of the P2Y12 receptor, reducing the formation of adhesive molecules that prevent adherence in tumor cells. In this way, our proposals for CM treatment based on targeting purinergic signaling permeate the integral practice, going from intracell to extracell. Undoubtedly, much is still to be discovered and elucidated about this promising compound, this paper being an interesting work baseline to support more research studies.
Collapse
Affiliation(s)
- Gilnei Bruno da Silva
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Milena Ayumi Yamauchi
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Daniela Zanini
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Margarete Dulce Bagatini
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil.
| |
Collapse
|
41
|
Arslan AKK, Paşayeva L, Tugay O. Cytotoxic evaluation and LC-MS/MS analysis of aerial parts of Eryngium kotschyi Boiss. grown in Turkey. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
42
|
Nonylphenol regulates TL1A through the AhR/HDAC2/HNF4α pathway in endothelial cells to promote the angiogenesis of colorectal cancer. Toxicol Appl Pharmacol 2021; 436:115854. [PMID: 34974051 DOI: 10.1016/j.taap.2021.115854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 12/17/2021] [Accepted: 12/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most malignant cancers worldwide. Nonylphenol (NP) is an endocrine-disruptor chemical and plays an important role in the development of cancers. However, the effects of NP on CRC remain unclear. In this study, we aimed to investigate the potential mechanisms of NP in the pathogenesis of CRC. METHODS The levels of AhR, TL1A and HDAC2 in CRC tissues and endothelial cells were assessed by RT-qPCR or western blot. CHIP and dual luciferase reporter assays were used to confirm the interaction between AhR and HDAC2, or HNF4α and TL1A. The CCK8, would healing and tube formation assays were conducted to evaluate the proliferation, migration and angiogenesis of HUVECs. Western blot determined HNF4α protein and HNF4α acetylation levels. The secreted TL1A protein was detected by ELISA. The angiogenesis-related factor CD31 was tested by IHC. RESULTS The expression level of AhR was significantly up-regulated in CRC tissues and endothelial cells. Moreover, NP activated the AhR pathway mediated colorectal endothelial cell angiogenesis and proliferation, while TL1A overexpression resisted these effects caused by NP. Besides, NP was found to modulate HNF4α deacetylation through AhR/HDAC2 to inhibit TL1A. Furthermore, in vivo experiments proved that NP regulated CRC growth and angiogenesis via AhR/HDAC2/HNF4α/TL1A axis. CONCLUSION This study revealed that NP promoted CRC growth and angiogenesis through AhR/HDAC2/HNF4α/TL1A pathway and could be a new therapeutic target for CRC treatment.
Collapse
|
43
|
Wang K, Zhang M, Wang J, Sun P, Luo J, Jin H, Li R, Pan C, Lu L. A Systematic Analysis Identifies Key Regulators Involved in Cell Proliferation and Potential Drugs for the Treatment of Human Lung Adenocarcinoma. Front Oncol 2021; 11:737152. [PMID: 34650921 PMCID: PMC8505978 DOI: 10.3389/fonc.2021.737152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/06/2021] [Indexed: 11/23/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is one of the most common and malignant cancer types. Abnormal cell proliferation, exemplified by cell cycle and cell division dysregulation, is one of the most prominent hallmarks of cancer and is responsible for recurrence, metastasis, and resistance to cancer therapy. However, LUAD-specific gene regulation and clinical significance remain obscure. Here, by using both tissues and cells from LUAD and normal lung samples, 434 increased and 828 decreased genes of biological significance were detected, including 127 cell cycle-associated genes (95 increased and 32 decreased), 66 cell division-associated genes (56 increased and 10 decreased), and 81 cell proliferation-associated genes (34 increased and 47 decreased). Among them, 12 increased genes (TPX2, CENPF, BUB1, PLK1, KIF2C, AURKB, CDKN3, BUB1B, HMGA2, CDK1, ASPM, and CKS1B) and 2 decreased genes (TACC1 and MYH10) were associated with all the three above processes. Importantly, 2 (CDKN3 and CKS1B) out of the 11 increased genes (except HMGA2) are previously uncharacterized ones in LUAD and can potentially be prognostic markers. Moreover, PLK1 could be a promising therapeutic target for LUAD. Besides, protein–protein interaction network analysis showed that CDK1 and CDC20 were the hub genes, which might play crucial roles in cell proliferation of LUAD. Furthermore, transcriptional regulatory network analysis suggested that the transcription factor E2F1 could be a key regulator in controlling cell proliferation of LUAD via expression modulation of most cell cycle-, cell division-, and cell proliferation-related DEGs. Finally, trichostatin A, hycanthone, vorinostat, and mebeverine were identified as four potential therapeutic agents for LUAD. This work revealed key regulators contributing to cell proliferation in human LUAD and identified four potential therapeutic agents for treatment strategy.
Collapse
Affiliation(s)
- Kai Wang
- Clinical Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Man Zhang
- Department of Radiology, Xiangyang Hospital of Traditional Chinese Medicine, Hubei University of Traditional Chinese Medicine, Xiangyang, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Pan Sun
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jizhuang Luo
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haizhen Jin
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Li
- Clinical Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,China Hospital Development Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Changqing Pan
- General Surgery Department, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Liming Lu
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Karaboğa Arslan AK, Paşayeva L, Esen MA, Tugay O. Synergistic Growth Inhibitory Effects of Eryngium kotschyi Extracts with Conventional Cytotoxic Agents: Cisplatin and Doxorubicin Against Human Endometrium Cancer Cells. Curr Pharm Biotechnol 2021; 23:740-748. [PMID: 34445948 DOI: 10.2174/1389201022666210826160307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/19/2021] [Accepted: 03/02/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Endometrial cancer is one of the most common types of cancer. For this reason, various studies have been carried out on its treatment and the effects of natural products on this disease. OBJECTIVE This study aimed to examine the growth inhibitory effects of Eryngium kotschyi Boiss. ethyl acetate [EKE] and butanol [EKB] obtained from the main methanol [EKM] extract from the aerial parts on human endometrium carcinoma [RL95-2] cells and their synergistic effect with cisplatin or doxorubicin. METHODS RL95-2 cells were treated with E. kotschyi extracts either alone or in combination with cisplatin or doxorubicin. The effects on cell growth were determined using the MTT assay and real-time cell analysis xCELLigence. RESULTS The extracts demonstrated growth inhibitory activity, with a certain degree of selectivity against the RL95-2 cell line. Synergistic effects of EKE/cisplatin or doxorubicin at different concentration levels were demonstrated in RL95-2 cells. In some instances, the EKE/doxorubicin combinations resulted in antagonistic effects. The reduction level of cell viability was different and specific to each combination for the RL95-2 cell line. CONCLUSION The growth inhibitory activity of cisplatin or doxorubicin, as a single agent, may be modified by combinations of the extracts and be synergistically enhanced in some cases. A significant synergistic effect of EKE on the RL95-2 cell line with cisplatin and doxorubicin was observed. This cytotoxic effect can be investigated in terms of molecular mechanisms. This study is the first of its kind in the literature. The mechanisms involved in this interaction between chemotherapeutic drugs and plant extracts remain unclear and should be further evaluated.
Collapse
Affiliation(s)
| | - Leyla Paşayeva
- Department of Pharmacognosy, Faculty of Pharmacy, Erciyes University, Kayseri 38039. Turkey
| | - Merve Ayşe Esen
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri 38039. Turkey
| | - Osman Tugay
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Selçuk University, Konya 42450. Turkey
| |
Collapse
|
45
|
Hai R, He L, Shu G, Yin G. Characterization of Histone Deacetylase Mechanisms in Cancer Development. Front Oncol 2021; 11:700947. [PMID: 34395273 PMCID: PMC8360675 DOI: 10.3389/fonc.2021.700947] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/05/2021] [Indexed: 02/01/2023] Open
Abstract
Over decades of studies, accumulating evidence has suggested that epigenetic dysregulation is a hallmark of tumours. Post-translational modifications of histones are involved in tumour pathogenesis and development mainly by influencing a broad range of physiological processes. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) are pivotal epigenetic modulators that regulate dynamic processes in the acetylation of histones at lysine residues, thereby influencing transcription of oncogenes and tumour suppressor genes. Moreover, HDACs mediate the deacetylation process of many nonhistone proteins and thus orchestrate a host of pathological processes, such as tumour pathogenesis. In this review, we elucidate the functions of HDACs in cancer.
Collapse
Affiliation(s)
- Rihan Hai
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.,School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liuer He
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.,School of Basic Medical Sciences, Central South University, Changsha, China
| | - Guang Shu
- School of Basic Medical Sciences, Central South University, Changsha, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
46
|
Shetty MG, Pai P, Deaver RE, Satyamoorthy K, Babitha KS. Histone deacetylase 2 selective inhibitors: A versatile therapeutic strategy as next generation drug target in cancer therapy. Pharmacol Res 2021; 170:105695. [PMID: 34082029 DOI: 10.1016/j.phrs.2021.105695] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Acetylation and deacetylation of histone and several non-histone proteins are the two important processes amongst the different modes of epigenetic modulation that are involved in regulating cancer initiation and development. Abnormal expression of histone deacetylases (HDACs) is often reported in various types of cancers. Few pan HDAC inhibitors have been approved for use as therapeutic interventions for cancer treatment including vorinostat, belinostat and panobinostat. However, not all the HDAC isoforms are abnormally expressed in certain cancers, such as in the case of, ovarian cancer where overexpression of HDAC1-3, lung cancer where overexpression of HDAC 1 and 3 and gastric cancer where overexpression of HDAC2 is seen. Therefore, pan-inhibition of HDAC is not an efficient way to combat cancer via HDAC inhibition. Hence, isoform-selective HDAC inhibition can be one of the best therapeutic strategies in the treatment of cancer. In this context since aberrant expression of HDAC2 largely contributes to cancer progression by silencing pro-apoptotic protein expressions such as NOXA and APAF1 (caspase 9-activating proteins) and inactivation of tumor suppressor p53, HDAC2 specific inhibitors may help to develop not only the direct targets but also indirect targets that are crucial for tumor development. However, to develop a HDAC2 specific and potent inhibitor, extensive knowledge of its structure and specific functions is essential. The present review updates details on the structural features, physiological functions, and roles of HDAC2 in different types of cancer, emphasizing the challenges and status of the development of HDAC2 selective inhibitors against various types of cancer.
Collapse
Affiliation(s)
| | - Padmini Pai
- Department of Biophysics, Manipal School of Life Sciences, MAHE, Manipal, India
| | - Renita Esther Deaver
- Department of Biotechnology, Manipal School of Life Sciences, MAHE, Manipal, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, MAHE, Manipal, India
| | | |
Collapse
|
47
|
Liu YR, Wang JQ, Huang ZG, Chen RN, Cao X, Zhu DC, Yu HX, Wang XR, Zhou HY, Xia Q, Li J. Histone deacetylase‑2: A potential regulator and therapeutic target in liver disease (Review). Int J Mol Med 2021; 48:131. [PMID: 34013366 PMCID: PMC8136123 DOI: 10.3892/ijmm.2021.4964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Histone acetyltransferases are responsible for histone acetylation, while histone deacetylases (HDACs) counteract histone acetylation. An unbalanced dynamic between histone acetylation and deacetylation may lead to aberrant chromatin landscape and chromosomal function. HDAC2, a member of class I HDAC family, serves a crucial role in the modulation of cell signaling, immune response and gene expression. HDAC2 has emerged as a promising therapeutic target for liver disease by regulating gene transcription, chromatin remodeling, signal transduction and nuclear reprogramming, thus receiving attention from researchers and clinicians. The present review introduces biological information of HDAC2 and its physiological and biochemical functions. Secondly, the functional roles of HDAC2 in liver disease are discussed in terms of hepatocyte apoptosis and proliferation, liver regeneration, hepatocellular carcinoma, liver fibrosis and non-alcoholic steatohepatitis. Moreover, abnormal expression of HDAC2 may be involved in the pathogenesis of liver disease, and its expression levels and pharmacological activity may represent potential biomarkers of liver disease. Finally, research on selective HDAC2 inhibitors and non-coding RNAs relevant to HDAC2 expression in liver disease is also reviewed. The aim of the present review was to improve understanding of the multifunctional role and potential regulatory mechanism of HDAC2 in liver disease.
Collapse
Affiliation(s)
- Ya-Ru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jie-Quan Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Zhao-Gang Huang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ruo-Nan Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xi Cao
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Dong-Chun Zhu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Hai-Xia Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xiu-Rong Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Hai-Yun Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jun Li
- The Key Laboratory of Anti‑inflammatory Immune Medicines, School of Pharmacy, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
48
|
Network Pharmacology and Molecular Docking Suggest the Mechanism for Biological Activity of Rosmarinic Acid. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5190808. [PMID: 33936238 PMCID: PMC8055417 DOI: 10.1155/2021/5190808] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/24/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022]
Abstract
Rosmarinic acid (RosA) is a natural phenolic acid compound, which is mainly extracted from Labiatae and Arnebia. At present, there is no systematic analysis of its mechanism. Therefore, we used the method of network pharmacology to analyze the mechanism of RosA. In our study, PubChem database was used to search for the chemical formula and the Chemical Abstracts Service (CAS) number of RosA. Then, the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) was used to evaluate the pharmacodynamics of RosA, and the Comparative Toxicogenomics Database (CTD) was used to identify the potential target genes of RosA. In addition, the Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of target genes were carried out by using the web-based gene set analysis toolkit (WebGestalt). At the same time, we uploaded the targets to the STRING database to obtain the protein interaction network. Then, we carried out a molecular docking about targets and RosA. Finally, we used Cytoscape to establish a visual protein-protein interaction network and drug-target-pathway network and analyze these networks. Our data showed that RosA has good biological activity and drug utilization. There are 55 target genes that have been identified. Then, the bioinformatics analysis and network analysis found that these target genes are closely related to inflammatory response, tumor occurrence and development, and other biological processes. These results demonstrated that RosA can act on a variety of proteins and pathways to form a systematic pharmacological network, which has good value in drug development and utilization.
Collapse
|
49
|
Makaremi S, Ganji A, Ghazavi A, Mosayebi G. Inhibition of tumor growth in CT-26 colorectal cancer-bearing mice with alcoholic extracts of Curcuma longa and Rosmarinus officinalis. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2020.101006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Go RE, Kim CW, Lee SM, Lee HK, Choi KC. Fenhexamid induces cancer growth and survival via estrogen receptor-dependent and PI3K-dependent pathways in breast cancer models. Food Chem Toxicol 2021; 149:112000. [PMID: 33484789 DOI: 10.1016/j.fct.2021.112000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/15/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Fenhexamid (Fen), a fungicide used to treat gray mold of fruits and vegetables, is reported to function as an endocrine disrupting chemical via the estrogen receptors (ER), despite low-toxicity of the pesticide. In this study, we elucidated that the disrupting effects of Fen are exerted via the ER and phosphatidylinositol 3-kinase (PI3K) pathways in breast cancer models. The WST assay, live cell monitoring, cell cycle analysis, colony formation assay, apoptotic analysis by JC-1 dyeing, and Western blot analysis were applied in ER positive MCF-7 and ER negative MDA-MB-231 breast cancer cells, after exposure to 17β-estradiol (E2), Fen, ICI 182,780 (ICI; an ER antagonist) and/or Pictilisib (Pic; a PI3K inhibitor). Exposure to E2 and Fen induced the cell growth and survival ability of MCF-7 cells by increasing the S-phase cells and regulating the cell cycle-related proteins (Cyclin D1 and E1, p21 and p27). In addition, E2 and Fen treatment resulted in elevated levels of the survival-related proteins (Survivin and PCNA), and inhibited apoptosis by increasing the mitochondrial membrane potential and regulating the apoptosis-related proteins (BAX, BCL-2, and Caspase-9). These changes were reversed to the same level as the control group when exposed to their respective inhibitors, thereby indicating that the changes are exerted via the ER and PI3K pathways. In particular, co-treatment with these inhibitors induced greater inhibition than single treatment. Conversely, no alterations were observed in the ER-negative MDA-MB-231 breast cancer cells. Taken together, these results indicate that Fen promotes the growth of breast cancer cells via the ER and/or PI3K pathways, similar to the E2 mechanism. Although a relatively safe pesticide, Fen possibly exerts its influence as an endocrine disrupting chemical in ER-positive breast cancer cells via the ER and PI3K pathways.
Collapse
Affiliation(s)
- Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Cho-Won Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sung-Moo Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|