1
|
Hemlata, Sharma S, Vasudeva N, Hooda T. Neuroprotective effects of oleanolic acid against secondary cascades of traumatic brain injury in mice. BRAIN DISORDERS 2024; 16:100173. [DOI: 10.1016/j.dscb.2024.100173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
2
|
Zeng Y, Wang Z, Zhang J, Jian W, Fu Q. Antitumour activity of oleanolic acid: A systematic review and meta‑analysis. Oncol Lett 2024; 28:582. [PMID: 39421313 PMCID: PMC11484195 DOI: 10.3892/ol.2024.14715] [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: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Oleanolic acid (OA), a compound known for its potent antitumour properties, has been the subject of investigations in both cell and animal models. Although OA has good biological activity, its low water solubility and bioavailability limit its therapeutic use, and therefore translating the potential of OA into the clinical oncology setting remains challenging. The present systematic review and meta-analysis utilized evidence from animal model studies to gain insights into the antitumour mechanisms of OA to address the gap in understanding, and to provide guidance for future research directions and potential clinical applications. The guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses were applied in the present study and a comprehensive search was conducted across the PubMed/MEDLINE, Web of Science, Cochrane Library and Embase databases, with a cut-off date of June 30, 2023. The primary focus was on randomized controlled trials that used animal models to assess the antitumour effects of OA. The methodological quality appraisal was conducted using the Systematic Review Centre for Laboratory Animal Experimentation risk of bias tool, and tumour volume and weight served as the principal outcome measures. Data were analysed using the RevMan (version 5.3) and Stata SE11 software packages, with an assessment of heterogeneity conducted using the I2 statistical test, sensitivity analysis conducted using the leave-one-out approach, and evaluation of publication bias performed using Egger's test and funnel plot analysis. The present study demonstrated a significant inhibitory effect of OA intervention on tumour growth and a decrease in tumour weight in animal models. Despite the broad spectrum of antitumour effects exhibited by OA, further investigations are warranted to optimize the dosage and administration routes of OA to maximize its efficacy in clinical cancer treatment.
Collapse
Affiliation(s)
- Ying Zeng
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zhonglian Wang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jing Zhang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Wei Jian
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Qiaofen Fu
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
3
|
Similie D, Minda D, Bora L, Kroškins V, Lugiņina J, Turks M, Dehelean CA, Danciu C. An Update on Pentacyclic Triterpenoids Ursolic and Oleanolic Acids and Related Derivatives as Anticancer Candidates. Antioxidants (Basel) 2024; 13:952. [PMID: 39199198 PMCID: PMC11351203 DOI: 10.3390/antiox13080952] [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: 07/10/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 09/01/2024] Open
Abstract
Cancer is a global health problem, with the incidence rate estimated to reach 40% of the population by 2030. Although there are currently several therapeutic methods, none of them guarantee complete healing. Plant-derived natural products show high therapeutic potential in the management of various types of cancer, with some of them already being used in current practice. Among different classes of phytocompounds, pentacyclic triterpenoids have been in the spotlight of research on this topic. Ursolic acid (UA) and its structural isomer, oleanolic acid (OA), represent compounds intensively studied and tested in vitro and in vivo for their anticancer and chemopreventive properties. Since natural compounds can rarely be used in practice as such due to their characteristic physico-chemical properties, to tackle this problem, their derivatization has been attempted, obtaining compounds with improved solubility, absorption, stability, effectiveness, and reduced toxicity. This review presents various UA and OA derivatives that have been synthesized and evaluated in recent studies for their anticancer potential. It can be observed that the most frequent structural transformations were carried out at the C-3, C-28, or both positions simultaneously. It has been demonstrated that conjugation with heterocycles or cinnamic acid, derivatization as hydrazide, or transforming OH groups into esters or amides increases anticancer efficacy.
Collapse
Affiliation(s)
- Diana Similie
- Department of Pharmacognosy, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (D.S.); (L.B.); (C.D.)
- Research and Processing Center of Medicinal and Aromatic Plants, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Daliana Minda
- Department of Pharmacognosy, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (D.S.); (L.B.); (C.D.)
- Research and Processing Center of Medicinal and Aromatic Plants, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Larisa Bora
- Department of Pharmacognosy, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (D.S.); (L.B.); (C.D.)
- Research and Processing Center of Medicinal and Aromatic Plants, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Vladislavs Kroškins
- Institute of Chemistry and Chemical Technology, Faculty of Natural Sciences and Technology, Riga Technical University, Paula Valdena Str. 3, LV-1048 Riga, Latvia; (V.K.); (J.L.); (M.T.)
| | - Jevgeņija Lugiņina
- Institute of Chemistry and Chemical Technology, Faculty of Natural Sciences and Technology, Riga Technical University, Paula Valdena Str. 3, LV-1048 Riga, Latvia; (V.K.); (J.L.); (M.T.)
| | - Māris Turks
- Institute of Chemistry and Chemical Technology, Faculty of Natural Sciences and Technology, Riga Technical University, Paula Valdena Str. 3, LV-1048 Riga, Latvia; (V.K.); (J.L.); (M.T.)
| | - Cristina Adriana Dehelean
- Research and Processing Center of Medicinal and Aromatic Plants, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Corina Danciu
- Department of Pharmacognosy, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (D.S.); (L.B.); (C.D.)
- Research and Processing Center of Medicinal and Aromatic Plants, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| |
Collapse
|
4
|
Jannuzzi AT, Yilmaz Goler AM, Biswas A, Mondal S, Basavanakatti VN, Yıldırım H, Yıldız M, Bayrak N, Jayaprakash V, TuYuN AF. Prospects for Prostate Cancer Chemotherapy: Cytotoxic Evaluation and Mechanistic Insights of Quinolinequinones with ADME/PK Profile. Biomedicines 2024; 12:1241. [PMID: 38927448 PMCID: PMC11200585 DOI: 10.3390/biomedicines12061241] [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: 03/08/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024] Open
Abstract
The evaluation of in vitro biological activity of several previously reported quinolinequinones (AQQ1-5) against 60 human cancer cell lines (NCI-60) used by the National Cancer Institute's Developmental Therapeutics Program (DTP) contributed to our earlier research on possible anticancer and/or antibacterial agents. Of interest, NCI-60 screening revealed that two quinolinequinones (AQQ1 and AQQ2) significantly reduced the proliferation of several cancer genotypes. Following the administration of a single dose and five additional doses, all quinolinequinones demonstrated a significant inhibitory effect on the growth of leukemia and other cancer cell lines. Hence, a series of subsequent in vitro biological assessments were performed to further understand the mechanistic impact of the compounds. In MTT assays, it was found that AQQ1 and AQQ2 exhibited higher efficacy against DU-145 cells (IC50 4.18 µM and 4.17 µM, respectively) compared to MDA-MB-231 (IC50 8.27 and 13.33 µM, respectively) and HCT-116 cells (IC50 5.83 and 9.18 µM, respectively). Additionally, AQQ1 demonstrated greater activity in this context. Further investigations revealed that AQQ1 inhibited DU-145 cell growth and migration dose-dependently. Remarkably, arrest of the DU-145 cell cycle at G0/G1 phase and ROS elevation were observed. Pharmacokinetic (PK) studies revealed that AQQ1 has better PK parameters than AQQ2 with %F of 9.83 in rat. Considering the data obtained with human liver microsomal stability studies, AQQ1 should have a better PK profile in human subjects. In silico studies (molecular dynamics) with three kinases (CDK2, CDK4, and MAPK) leading to cell cycle arrest at G0/G1 identified MAPK as a probable target for AQQ1. Taken together, our results showed that AQQ1 could be a potential chemotherapeutic lead molecule for prostate cancer.
Collapse
Affiliation(s)
- Ayse Tarbin Jannuzzi
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, İstanbul University, 34116 İstanbul, Türkiye;
| | - Ayse Mine Yilmaz Goler
- Department of Biochemistry, School of Medicine/Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34854 İstanbul, Türkiye;
| | - Abanish Biswas
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India; (A.B.); (V.J.)
| | - Subodh Mondal
- Bioanalysis, Eurofins Advinus BioPharma Services India Pvt Ltd., Bengaluru 560058, Karnataka, India;
| | | | - Hatice Yıldırım
- Department of Chemistry, Engineering Faculty, Istanbul University-Cerrahpasa, Avcılar, 34320 İstanbul, Türkiye;
| | - Mahmut Yıldız
- Department of Chemistry, Gebze Technical University, Gebze, 41400 Kocaeli, Türkiye;
| | - Nilüfer Bayrak
- Department of Chemistry, Faculty of Science, Istanbul University, Fatih, 34134 İstanbul, Türkiye;
| | - Venkatesan Jayaprakash
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India; (A.B.); (V.J.)
| | - Amaç Fatih TuYuN
- Department of Chemistry, Faculty of Science, Istanbul University, Fatih, 34134 İstanbul, Türkiye;
| |
Collapse
|
5
|
Golubnitschaja O, Kapinova A, Sargheini N, Bojkova B, Kapalla M, Heinrich L, Gkika E, Kubatka P. Mini-encyclopedia of mitochondria-relevant nutraceuticals protecting health in primary and secondary care-clinically relevant 3PM innovation. EPMA J 2024; 15:163-205. [PMID: 38841620 PMCID: PMC11148002 DOI: 10.1007/s13167-024-00358-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 06/07/2024]
Abstract
Despite their subordination in humans, to a great extent, mitochondria maintain their independent status but tightly cooperate with the "host" on protecting the joint life quality and minimizing health risks. Under oxidative stress conditions, healthy mitochondria promptly increase mitophagy level to remove damaged "fellows" rejuvenating the mitochondrial population and sending fragments of mtDNA as SOS signals to all systems in the human body. As long as metabolic pathways are under systemic control and well-concerted together, adaptive mechanisms become triggered increasing systemic protection, activating antioxidant defense and repair machinery. Contextually, all attributes of mitochondrial patho-/physiology are instrumental for predictive medical approach and cost-effective treatments tailored to individualized patient profiles in primary (to protect vulnerable individuals again the health-to-disease transition) and secondary (to protect affected individuals again disease progression) care. Nutraceuticals are naturally occurring bioactive compounds demonstrating health-promoting, illness-preventing, and other health-related benefits. Keeping in mind health-promoting properties of nutraceuticals along with their great therapeutic potential and safety profile, there is a permanently growing demand on the application of mitochondria-relevant nutraceuticals. Application of nutraceuticals is beneficial only if meeting needs at individual level. Therefore, health risk assessment and creation of individualized patient profiles are of pivotal importance followed by adapted nutraceutical sets meeting individual needs. Based on the scientific evidence available for mitochondria-relevant nutraceuticals, this article presents examples of frequent medical conditions, which require protective measures targeted on mitochondria as a holistic approach following advanced concepts of predictive, preventive, and personalized medicine (PPPM/3PM) in primary and secondary care.
Collapse
Affiliation(s)
- Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Andrea Kapinova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Nafiseh Sargheini
- Max Planck Institute for Plant Breeding Research, Carl-Von-Linne-Weg 10, 50829 Cologne, Germany
| | - Bianka Bojkova
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P. J. Šafárik University in Košice, 040 01 Košice, Slovakia
| | - Marko Kapalla
- Negentropic Systems, Ružomberok, Slovakia
- PPPM Centre, s.r.o., Ruzomberok, Slovakia
| | - Luisa Heinrich
- Institute of General Medicine, University of Leipzig, Leipzig, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Peter Kubatka
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
6
|
Jabbari S, Zakaria ZA, Ahmadimoghaddam D, Mohammadi S. The oral administration of Lotus corniculatus L. attenuates acute and chronic pain models in male rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117181. [PMID: 37734474 DOI: 10.1016/j.jep.2023.117181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lotus corniculatus L. (Fabaceae) traditionally used in Persian folk medicine to heal peritoneal inflammation and back pain. AIM OF THE STUDY To explore the antinociceptive (acute pain) and anti-neuropathic (chronic pain) activities of Lotus corniculatus leaves essential oil (LCEO) in addition to uncovering the possible mechanisms of antinociception. MATERIALS AND METHODS LCEO as well as the pure oleanolic acid (OA) compound, were assayed for their effects on acute (formalin induced paw licking test or FIPT) and chronic (cervical contusion injury models on the fifth cervical vertebra or CCS; 14-day intervals) pain. The possible involvements of NO-cGMP-K+ channel, TRPV, dopamine, cannabinoid, PPAR, adrenergic, and opioid mechanisms in the antinociceptive activity of LCEO have studied by formalin test. The levels of p53 and inflammatory markers were measured using a streptavidin biotin immune peroxidase complex and ELISA methods, respectively. RESULTS The LCEO and OA exerted antinociceptive activity in the first-phase of FIPT. Pretreatment with antagonists of TRPV1, dopamine D2, cannabinoid type1 and 2, and NO-cGMP-K+ channel blockers (glibenclamide, L-NAME and methylene blue) attenuated the antinociceptive effect of LCEO in FIPT. In addition, LCEO and OA meaningfully reduced hyperalgesia (days 6-14) and mechanical allodynia (days 2-14) in the CCS model. LCEO suppressed the apoptotic marker (p53) in CCS model and also ameliorated IL-2, TNF-α, and IL-1 in the spinal cord. CONCLUSION Finally, LCEO inhibited acute (possibly via the modulation of opioid, TRPV, dopamine, cannabinoid mechanisms as well as NO-cGMP-K+ channel) and chronic pain (via suppressing apoptotic and inflammatory markers) in male rats. The results also suggest that OA has analgesic activity against acute and chronic pain conditions.
Collapse
Affiliation(s)
- Sajjad Jabbari
- Department of Biology, Faculty of Sciences, Islamic Azad University, Tehran North Branch, Tehran, Iran.
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia.
| | - Davoud Ahmadimoghaddam
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Saeed Mohammadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
7
|
Esmaeili H, Nasrollahzadeh Sabet M, Mosaed R, Chamanara M, Hadi S, Hazrati E, Farhadi A, Heidari MF, Behroozi J. Oleanolic acid increases the anticancer potency of doxorubicin in pancreatic cancer cells. J Biochem Mol Toxicol 2023; 37:e23426. [PMID: 37345903 DOI: 10.1002/jbt.23426] [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: 11/18/2022] [Revised: 02/20/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
Combination therapy is a novel cancer therapy approach that combines two or more chemotherapy drugs. This treatment modality enhances the efficacy of chemotherapy by targeting key pathways in an additive or synergistic manner. Therefore, we investigated the efficacy of combination therapy by widely used chemotherapy drug doxorubicin (DOX) and oleanolic acid (OA) to induction of apoptosis for pancreatic cancer (PC) therapy. The effects of DOX, OA, and their combination (DOX-OA) were investigated on proliferation and viability of PC cell line (PANC-1) by MTT assay. Moreover, migration and invasion of the cancer cells were evaluated by trans-well migration assay and wound healing assay. Flow cytometry and DAPI (4',6-diamidino-2-phenylindole) staining were employed to investigate apoptosis quantification and qualification of the treated cancer cells. Finally, mRNA expression of apoptosis-related genes was assessed by quantitative real-time polymerase chain reaction. Our results demonstrated that the proliferation and metastasis potential of PC cells significantly decreased after treatment by DOX, OA, and DOX-OA. Moreover, we observed an increase in apoptosis percentage in the treated cancer cells. The apoptosis-related gene expression was modified to increase the apoptosis rate in all of the treatment groups. However, the anticancer potency of DOX-OA combination was significantly more than that of DOX and OA treatments alone. Our study suggested that DOX-OA combination exerts more profound anticancer effects against PC cell lines than DOX or OA monotherapy. This approach may increase the efficiency of chemotherapy and reduce unintended side effects by lowering the prescribed dose of DOX.
Collapse
Affiliation(s)
- Hosein Esmaeili
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mehrdad Nasrollahzadeh Sabet
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Department of Clinical Pharmacy, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Department of Clinical Pharmacy, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Saeid Hadi
- Department of Health, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ebrahim Hazrati
- Department of Anesthesiology and Critical Care, AJA University of Medical Sciences, Tehran, Iran
| | - Arezoo Farhadi
- Department of Genetics and Biotechnology, Faculty of Life Science, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Mohammad Foad Heidari
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Javad Behroozi
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Avcı CB, Sogutlu F, Pinar Ozates N, Shademan B, Gunduz C. Enhanced Anti-cancer Potency Using a Combination of Oleanolic Acid and Maslinic Acid to Control Treatment Resistance in Breast Cancer. Adv Pharm Bull 2023; 13:611-620. [PMID: 37646060 PMCID: PMC10460813 DOI: 10.34172/apb.2023.057] [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: 02/26/2022] [Revised: 07/11/2022] [Accepted: 09/09/2022] [Indexed: 09/01/2023] Open
Abstract
Purpose The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/ mTOR) pathway is a complex intracellular metabolic pathway that leads to cell growth and tumor proliferation and plays a key role in drug resistance in breast cancer. Therefore, the anti-cancer effects of oleanolic acid (OA), maslinic acid (MA), and their combination were investigated to improve the performance of the treatment strategy. Methods We investigated the effect of OA and MA on cell viability using the WST-1 method. The synergistic effect of the combination was analyzed by isobologram analysis. In addition, the effects of the two compounds, individually and in combination, on apoptosis, autophagy, and the cell cycle were investigated in MCF7 cells. In addition, changes in the expression of PI3K/AKT/mTOR genes involved in apoptosis, cell cycle and metabolism were determined by quantitative RT-PCR. Results MA, OA, and a combination of both caused G0/G1 arrest. Apoptosis also increased in all treated groups. The autophagosomal LC3-II formation was induced 1.74-fold in the MA-treated group and 3.25-fold in the MA-OA-treated group. The combination treatment resulted in increased expression of genes such as GSK3B, PTEN, CDKN1B and FOXO3 and decreased expression of IGF1, PRKCB and AKT3 genes. Conclusion The results showed that the combination of these two substances showed the highest synergistic effect at the lowest dose and using MA-OA caused cancer cells to undergo apoptosis. The use of combination drugs may reduce the resistance of cancer cells to treatment.
Collapse
Affiliation(s)
- Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | | | | | | | | |
Collapse
|
9
|
Kim YS, Lee JC, Lee M, Oh HJ, An WG, Sung ES. Discovering Potential Anti-Oral Squamous Cell Carcinoma Mechanisms from Kochiae Fructus Using Network-Based Pharmacology Analysis and Experimental Validation. Life (Basel) 2023; 13:1300. [PMID: 37374083 DOI: 10.3390/life13061300] [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: 03/24/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The natural product Kochiae Fructus (KF) is the ripe fruit of Kochia scoparia (L.) Schrad and is renowned for its anti-inflammatory, anticancer, anti-fungal, and anti-pruritic effects. This study examined the anticancer effect of components of KF to assess its potential as an adjuvant for cancer treatment. Network-based pharmacological and docking analyses of KF found associations with oral squamous cell carcinoma. The molecular docking of oleanolic acid (OA) with LC3 and SQSTM1 had high binding scores, and hydrogen binding with amino acids of the receptors suggests that OA is involved in autophagy, rather than the apoptosis pathway. For experimental validation, we exposed SCC-15 squamous carcinoma cells derived from a human tongue lesion to KF extract (KFE), OA, and cisplatin. The KFE caused SCC-15 cell death, and induced an accumulation of the autophagy marker proteins LC3 and p62/SQSTM1. The novelty of this study lies in the discovery that the change in autophagy protein levels can be related to the regulatory death of SCC-15 cells. These findings suggest that KF is a promising candidate for future studies to provide insight into the role of autophagy in cancer cells and advance our understanding of cancer prevention and treatment.
Collapse
Affiliation(s)
- Youn-Sook Kim
- Research Institute for Longevity and Well-Being, Pusan National University, Busan 46241, Republic of Korea
| | - Jin-Choon Lee
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Pusan National University and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Minhyung Lee
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Pusan National University and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Hae-Jin Oh
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Pusan National University and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Won G An
- Department of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Eui-Suk Sung
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Pusan National University and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
10
|
Gao CX, Tang CH, Wu TJ, Hu Y, Peng YL, Liu ML, Liu QW, Chen HF, Yang ZH, Zheng X. Anticancer activity of oleanolic acid and its derivatives modified at A-ring and C-28 position. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022:1-14. [PMID: 36151896 DOI: 10.1080/10286020.2022.2120863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 06/16/2023]
Abstract
Oleanolic acid (OA) is a five-ring triterpenoid compound, which is widely present in plants. Due to a wide range of pharmacological activities, oleanolic acid has attracted more and more attention. However, oleanolic acid is insoluble in water and has low bioavailability, which limits its clinical application. In this review, we focus on summarizing the anti-cancer activity and mechanism of the A ring or C-28 carboxyl modified derivatives of OA since 2015, to determine the strength of its anti-cancer effectiveness and evaluate whether it could be used as a clinical anti-cancer drug.
Collapse
Affiliation(s)
- Cong-Xi Gao
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Cai-Hong Tang
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Ting-Juan Wu
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Yue Hu
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Ya-Ling Peng
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Mei-Ling Liu
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Qian-Wen Liu
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Hong-Fei Chen
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Ze-Hua Yang
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| | - Xing Zheng
- Group of Lead Compound, Department of Pharmacy, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medicinal School, University of South China, Hengyang 421001, China
| |
Collapse
|
11
|
Mioc M, Milan A, Malița D, Mioc A, Prodea A, Racoviceanu R, Ghiulai R, Cristea A, Căruntu F, Șoica C. Recent Advances Regarding the Molecular Mechanisms of Triterpenic Acids: A Review (Part I). Int J Mol Sci 2022; 23:ijms23147740. [PMID: 35887090 PMCID: PMC9322890 DOI: 10.3390/ijms23147740] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Triterpenic acids are phytocompounds with a widespread range of biological activities that have been the subject of numerous in vitro and in vivo studies. However, their underlying mechanisms of action in various pathologies are not completely elucidated. The current review aims to summarize the most recent literature, published in the last five years, regarding the mechanism of action of three triterpenic acids (asiatic acid, oleanolic acid, and ursolic acid), corelated with different biological activities such as anticancer, anti-inflammatory, antidiabetic, cardioprotective, neuroprotective, hepatoprotective, and antimicrobial. All three discussed compounds share several mechanisms of action, such as the targeted modulation of the PI3K/AKT, Nrf2, NF-kB, EMT, and JAK/STAT3 signaling pathways, while other mechanisms that proved to only be specific for a part of the triterpenic acids discussed, such as the modulation of Notch, Hippo, and MALAT1/miR-206/PTGS1 signaling pathway, were highlighted as well. This paper stands as the first part in our literature study on the topic, which will be followed by a second part focusing on other triterpenic acids of therapeutic value.
Collapse
Affiliation(s)
- Marius Mioc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
| | - Andreea Milan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
| | - Daniel Malița
- Department of Radiology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Correspondence: (D.M.); (A.M.); Tel.: +40-256-494-604 (D.M. & A.M.)
| | - Alexandra Mioc
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
- Department of Anatomy, Physiology, Pathophysiology, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
- Correspondence: (D.M.); (A.M.); Tel.: +40-256-494-604 (D.M. & A.M.)
| | - Alexandra Prodea
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
| | - Roxana Racoviceanu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
| | - Roxana Ghiulai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
| | - Andreea Cristea
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
| | - Florina Căruntu
- Department of Medical Semiology II, Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Street, 300041 Timisoara, Romania;
| | - Codruța Șoica
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania; (M.M.); (A.M.); (A.P.); (R.R.); (R.G.); (A.C.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania
| |
Collapse
|
12
|
Signaling pathways of Periplaneta americana peptide resist H2O2-induced apoptosis in pig-ovary granulosa cells through FoxO1. Theriogenology 2022; 183:108-119. [DOI: 10.1016/j.theriogenology.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 01/12/2023]
|
13
|
Banarase NB, Kaur CD. Whole whey stabilized oleanolic acid nanosuspension: Formulation and evaluation study. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|
14
|
Fofana S, Ouédraogo M, Esposito RC, Ouedraogo WP, Delporte C, Van Antwerpen P, Mathieu V, Guissou IP. Systematic Review of Potential Anticancerous Activities of Erythrina senegalensis DC (Fabaceae). PLANTS (BASEL, SWITZERLAND) 2021; 11:plants11010019. [PMID: 35009024 PMCID: PMC8747466 DOI: 10.3390/plants11010019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 05/04/2023]
Abstract
The objective of this study was to carry out a systematic review of the substances isolated from the African medicinal plant Erythrina senegalensis, focusing on compounds harboring activities against cancer models detailed in depth herein at both in vitro and in vivo preclinical levels. The review was conducted through Pubmed and Google Scholar. Nineteen out of the forty-two secondary metabolites isolated to date from E. senegalensis displayed interesting in vitro and/or in vivo antitumor activities. They belonged to alkaloid (Erysodine), triterpenes (Erythrodiol, maniladiol, oleanolic acid), prenylated isoflavonoids (senegalensin, erysenegalensein E, erysenegalensein M, alpinumisoflavone, derrone, warangalone), flavonoids (erythrisenegalone, senegalensein, lupinifolin, carpachromene) and pterocarpans (erybraedine A, erybraedine C, phaseollin). Among the isoflavonoids called "erysenegalensein", only erysenealenseins E and M have been tested for their anticancerous properties and turned out to be cytotoxic. Although the stem bark is the most frequently used part of the plant, all pterocarpans were isolated from roots and all alkaloids from seeds. The mechanisms of action of its metabolites include apoptosis, pyroptosis, autophagy and mitophagy via the modulation of cytoplasmic proteins, miRNA and enzymes involved in critical pathways deregulated in cancer. Alpinumisoflavone and oleanolic acid were studied in a broad spectrum of cancer models both in vitro and in preclinical models in vivo with promising results. Other metabolites, including carpachromen, phaseollin, erybraedin A, erysenegalensein M and maniladiol need to be further investigated, as they display potent in vitro effects.
Collapse
Affiliation(s)
- Souleymane Fofana
- Laboratory of Drug Science, Higher Institute of Health Sciences (INSSA), Nazi BONI University, Bobo-Dioulasso P.O. Box 1091, Burkina Faso;
| | - Moussa Ouédraogo
- Laboratory of Drug Development (LADME), Training and Research Unit, Health Sciences, Joseph KI-ZERBO University, Ouagadougou P.O. Box 7021, Burkina Faso; (M.O.); (W.P.O.)
| | - Rafaèle Calvo Esposito
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- Protein Chemistry Unit, Department of General Chemistry I, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Windbedema Prisca Ouedraogo
- Laboratory of Drug Development (LADME), Training and Research Unit, Health Sciences, Joseph KI-ZERBO University, Ouagadougou P.O. Box 7021, Burkina Faso; (M.O.); (W.P.O.)
| | - Cédric Delporte
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Universite’ Libre de Bruxelles (ULB), 1050 Brussels, Belgium; (C.D.); (P.V.A.)
| | - Pierre Van Antwerpen
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Universite’ Libre de Bruxelles (ULB), 1050 Brussels, Belgium; (C.D.); (P.V.A.)
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- ULB Cancer Research Center, Université Libre de Bruxelles (ULB), 1050 Bruxelles, Belgium
- Correspondence: (V.M.); (I.P.G.); Tel.: +32-478-31-73-88 (V.M.)
| | - Innocent Pierre Guissou
- Faculty of Health Sciences, Saint Thomas d’Aquin University, Ouagadougou P.O. Box 10212, Burkina Faso
- Correspondence: (V.M.); (I.P.G.); Tel.: +32-478-31-73-88 (V.M.)
| |
Collapse
|
15
|
Tang ZY, Li Y, Tang YT, Ma XD, Tang ZY. Anticancer activity of oleanolic acid and its derivatives: Recent advances in evidence, target profiling and mechanisms of action. Biomed Pharmacother 2021; 145:112397. [PMID: 34798468 DOI: 10.1016/j.biopha.2021.112397] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Oleanolic acid (OA, 3 β - hydroxyoleanolic acid-12-en-28-oic acid) is a pentacyclic triterpenoid present in many plants. As a new framework for development of semi synthetic triterpenoids, OA is of great significance in the discovery of anticancer drugs. Some of these derivatives, such as CDDO (2-cyano-3,12-dioxooleana-1, 9 (11)-dien-28-oic acid) have been verified in clinical trials, while other derivatives studied previously, such as SZC014, SZC015 and SZC017 (OA derivatives respectively), are also candidate drugs for cancer treatment. This paper reviews the preclinical studies, literature evidence, target analysis and anticancer mechanism of OA and its derivatives. The mechanism of action of its derivatives mainly includes anti-cancer cell proliferation, inducing tumor cell apoptosis, inducing autophagy, regulating cell cycle regulatory proteins, inhibiting vascular endothelial growth, anti angiogenesis, inhibiting tumor cell migration and invasion. In recent years, the molecular mechanism of OA and its derivatives has been elucidated. These effects seem to be mediated by the alterations in a variety of signaling pathways induced by OA and its derivatives. In conclusion, OA and its derivatives are considered as important candidate drugs for the treatment of cancer, indicating that OA and its derivatives have the potential to be used as anticancer drugs in practice.
Collapse
Affiliation(s)
- Zhong-Yuan Tang
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, Jilin, PR China
| | - Yang Li
- Pharmacology Department, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Yu-Ting Tang
- Pharmacology Department, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Xiao-Dong Ma
- Pharmacology Department, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Ze-Yao Tang
- Pharmacology Department, Dalian Medical University, Dalian, Liaoning 116044, PR China.
| |
Collapse
|
16
|
Nurcahyanti ADR, Jap A, Lady J, Prismawan D, Sharopov F, Daoud R, Wink M, Sobeh M. Function of selected natural antidiabetic compounds with potential against cancer via modulation of the PI3K/AKT/mTOR cascade. Biomed Pharmacother 2021; 144:112138. [PMID: 34750026 DOI: 10.1016/j.biopha.2021.112138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with growing global incidence, as 387 million people were diagnosed in 2014 with an expected projection of 642 million in 2040. Several complications are associated with DM including heart attack, stroke, kidney failure, blindness, and cancer. The latter is the second leading cause of death worldwide accounting for one in every six deaths, with liver, pancreas, and endometrium cancers are the most abundant among patients with diabetes. Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway plays a vital role in developing a wide array of pathological disorders, among them diabetes and cancer. Natural secondary metabolites that counteract the deleterious effects of reactive oxygen species (ROS) and modulate PI3K/Akt/mTOR pathway could be a promising approach in cancer therapy. Here, 717 medicinal plants with antidiabetic activities were highlighted along with 357 bioactive compounds responsible for the antidiabetic activity. Also, 43 individual plant compounds with potential antidiabetic activities against cancer via the modulation of PI3K/Akt/mTOR cascade were identified. Taken together, the available data give an insight of the potential of repurposing medicinal plants and/or the individual secondary metabolites with antidiabetic activities for cancer therapy.
Collapse
Affiliation(s)
- Agustina Dwi Retno Nurcahyanti
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia.
| | - Adeline Jap
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Jullietta Lady
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Deka Prismawan
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, 14440 Jakarta, Indonesia
| | - Farukh Sharopov
- Chinese-Tajik Innovation Center for Natural Products, National Academy of Sciences of Tajikistan, Ayni str. 299/2, 734063, Dushanbe, Tajikistan
| | - Rachid Daoud
- African Genome Center, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, Ben Guerir 43150, Morocco
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Mansour Sobeh
- AgroBiosciences Research, Mohammed VI Polytechnic University, Lot 660-Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
| |
Collapse
|
17
|
Ghosh A, Panda CK. Role of Pentacyclic Triterpenoid Acids in the Treatment of Bladder Cancer. Mini Rev Med Chem 2021; 22:1331-1340. [PMID: 34719363 DOI: 10.2174/1389557521666211022145052] [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: 12/30/2020] [Revised: 05/27/2021] [Accepted: 08/04/2021] [Indexed: 11/22/2022]
Abstract
Bladder cancer carries a poor prognosis and has proven resistance to chemotherapy. Pentacyclic Triterpenoid Acids (PTAs) are natural bioactive compounds that have a well-known impact on cancer research because of their cytotoxic and chemopreventive activities. This review focuses on bladder cancer which can no longer be successfully treated by DNA damaging drugs. Unlike most of the existing drugs against bladder cancer, PTAs are non-toxic to normal cells. Collecting findings from both in vitro and in vivo studies, it has been concluded that PTAs may serve as promising agents in future bladder cancer therapy. In this review, the roles of various PTAs in bladder cancer have been explored, and their mechanisms of action in the treatment of bladder cancer have been described. Specific PTAs have been shortlisted from each of the chief skeletons of pentacyclic triterpenoids, which could be effective against bladder cancer because of their mode of action. This review thereby throws light on the multi targets and mechanisms of PTAs, which are responsible for their selective anticancer effects and provides guidelines for further research and development of new natural antitumor compounds.
Collapse
Affiliation(s)
- Anindita Ghosh
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata. India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata. India
| |
Collapse
|
18
|
Oleanolic Acid's Semisynthetic Derivatives HIMOXOL and Br-HIMOLID Show Proautophagic Potential and Inhibit Migration of HER2-Positive Breast Cancer Cells In Vitro. Int J Mol Sci 2021; 22:ijms222011273. [PMID: 34681931 PMCID: PMC8538366 DOI: 10.3390/ijms222011273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 01/23/2023] Open
Abstract
Approximately 20–30% of the diagnosed breast cancers overexpress the human epidermal growth factor receptor 2 (HER2). This type of cancer is associated with a more aggressive phenotype; thus, there is a need for the discovery of new compounds that would improve the survival in HER2-positive breast cancer patients. It seems that one of the most promising therapeutic cancer strategies could be based on the biological activity of pentacyclic triterpenes’ derivatives and the best-known representative of this group, oleanolic acid (OA). The biological activity of oleanolic acid and its two semisynthetic derivatives, methyl 3-hydroxyimino-11-oxoolean-12-en-28-oate (HIMOXOL) and 12α-bromo-3-hydroxyimonoolean-28→13-olide (Br-HIMOLID), was assessed in SK-BR-3 breast cancer cells (HER2-positive). Viability tests, cell cycle assessment, evaluation of apoptosis, autophagy, and adhesion/migration processes were performed using MTT, clonogenic, cytofluorometry, Western blot, and qPCR. Both derivatives revealed higher cytotoxicity in studied breast cancer cells than the maternal compound, OA. They also decreased cell viability, induced autophagy, and (when applied in sub-cytotoxic concentrations) decreased the migration of SK-BR-3 cells.This study is the first to report the cytostatic, proautophagic (mTOR/LC3/SQSTM/BECN1 pathway), and anti-migratory (integrin β1/FAK/paxillin pathway) activities of HIMOXOL and Br-HIMOLID in HER2-positive breast cancer cells.
Collapse
|
19
|
Iloun P, Hooshmandi E, Gheibi S, Kashfi K, Ghasemi R, Ahmadiani A. Roles and Interaction of the MAPK Signaling Cascade in Aβ25-35-Induced Neurotoxicity Using an Isolated Primary Hippocampal Cell Culture System. Cell Mol Neurobiol 2021; 41:1497-1507. [PMID: 32601776 DOI: 10.1007/s10571-020-00912-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/22/2020] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is characterized with increased formation of amyloid-β (Aβ) in the brain. Aβ peptide toxicity is associated with disturbances of several intracellular signaling pathways such as mitogen activated protein kinases (MAPKs). The aim of this study was to investigate the role of MAPKs and their interactions in Aβ-induced neurotoxicity using isolated hippocampal neurons from the rat. Primary hippocampal cells were cultured in neurobasal medium for 4 days. Cells were treated with Aβ25-35 and/or MAPKs inhibitors for 24 h. Cell viability was determined by an MTT assay and phosphorylated levels of P38, JNK, and ERK were measured by Western blots. Aβ treatment (10-40 µM) significantly decreased hippocampal cell viability in a dose-dependent manner. Inhibition of P38 and ERK did not restore cell viability, while JNK inhibition potentiated the Aβ-induced neurotoxicity. Compared to the controls, Aβ treatment increased levels of phosphorylated JNK, ERK, and c-Jun, while it had no effect on levels of phosphorylated P38. In addition, P38 inhibition led to decreased expression levels of phosphorylated ERK; inhibition of JNK resulted in decreased expression of c-Jun; and inhibition of ERK, decreased phosphorylated levels of JNK. These results strongly suggest that P38, ERK, and JNK are not independently involved in Aβ-induced toxicity in the hippocampal cells. In AD, which is a multifactorial disease, inhibiting a single member of the MAPK signaling pathway, does not seem to be sufficient to mitigate Aβ-induced toxicity and thus their interactions with each other or potentially with different signaling pathways should be taken into account.
Collapse
Affiliation(s)
- Parisa Iloun
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Velenjak, Chamran Exp. Way, P.O. Box 19615-1178, Tehran, Iran
| | - Sevda Gheibi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | - Rasoul Ghasemi
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Velenjak, Chamran Exp. Way, P.O. Box 19615-1178, Tehran, Iran.
| |
Collapse
|
20
|
Woo JS, Yoo ES, Kim SH, Lee JH, Han SH, Jung SH, Jung GH, Jung JY. Anticancer effects of oleanolic acid on human melanoma cells. Chem Biol Interact 2021; 347:109619. [PMID: 34364837 DOI: 10.1016/j.cbi.2021.109619] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/27/2021] [Accepted: 08/04/2021] [Indexed: 11/23/2022]
Abstract
Owing to the ineffectiveness of the currently used therapies against melanoma, there has been a shift in focus toward alternative therapies involving the use of natural compounds. This study assessed the anticancer effects of oleanolic acid (OA) and its ability to induce apoptosis in A375SM and A375P melanoma cells in vivo. Compared to the control group, viability of A375P and A375SM cells decreased following OA treatment. In OA-treated A375SM and A375P cells, 4',6-diamidino-2-phenylindole staining showed an increase in the apoptotic body, and flow cytometry revealed increased number of apoptotic cells compared to that in the control group. OA-treated A375SM cells exhibited an increased expression of the apoptotic proteins, cleaved poly (ADP-ribose) polymerase (PARP) and B-cell lymphoma (Bcl)-2-associated X protein (Bax) as well as decreased expression of the antiapoptotic protein Bcl-2 compared to that in the control group. In OA-treated A375P cells, expression patterns of cleaved PARP and Bcl-2 were similar to those in OA-treated A375SM cells; however, no difference was reported in the expression of Bax compared to that in the control group. Additionally, OA-treated melanoma cells showed decreased expression of phospho-nuclear factor-κB (p-NF-κB), phospho-inhibitor of nuclear factor-κBα (p-IκBα), and phospho-IκB kinase αβ than that in the control group. Moreover, immunohistochemistry showed a comparatively decreased level of p-NF-κB in the OA-treated group than that in the control group. Xenograft analysis confirmed the in vivo anticancer effects of OA against A375SM cells. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay revealed an increased number of TUNEL-positive cells in the OA-treated group compared to that in the control group. In conclusion, the study results suggest that OA induces apoptosis of A375SM and A375P cells in vitro and apoptosis of A375SM cells in vivo. Furthermore, the in vitro and in vivo anticancer effects were mediated by the NF-κB pathway.
Collapse
Affiliation(s)
- Joong-Seok Woo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Eun-Seon Yoo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Sung-Hyun Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Jae-Han Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - So-Hee Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Gi-Hwan Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea.
| |
Collapse
|
21
|
Gupta N. A Review on Recent Developments in the Anticancer Potential of Oleanolic acid and its analogs (2017-2020). Mini Rev Med Chem 2021; 22:600-616. [PMID: 35135459 DOI: 10.2174/1389557521666210810153627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Oleanolic acid (OA) is a pentacyclic triterpenoid class of natural product known to possess a broad range of biological activities, specifically, anticancer. Considering the anticancer potential of OA, a large number of analogs have been prepared by several researchers through modifications at C-3, C-12 and C-28 -COOH to develop the potent anticancer agents with improved cytotoxicity and pharmaceutical properties. Some of the synthesized derivatives have been assessed in clinical trials also. This review summarizes the most recent synthetic and biological efforts in the development of oleanolic acid and its analogs during the period 2017-2020. Reports published during this period revealed that both OA and its analogs possess a remarkable potential for the development of effective anticancer agents along with several others such as anti-inflammatory, anti-viral, anti-microbial and anti-diabetic agents.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Pharmaceutical Sciences, MM College of Pharmacy, M. M. University, Mullana, Ambala, Haryana. India
| |
Collapse
|
22
|
Tumor-suppressing effect of bartogenic acid in ovarian (SKOV-3) xenograft mouse model. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1815-1826. [PMID: 34255109 DOI: 10.1007/s00210-021-02088-0] [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: 10/19/2020] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Bartogenic acid (BA), a natural pentacyclic triterpenoid, proved to have chemomodulatory, anticancer, antidiabetic, anti-arthritic, and anti-inflammatory activity. Based on structure-activity relationship (SAR) approaches, BA has close structural resemblance to oleanolic acid and ursolic acid. These two pentacyclic triterpenoids are well accepted with respect to their therapeutic value in various ailments including anti-cancer activity. The aim of this study is to evaluate the efficacy of BA as a possible antitumor agent, along with its safety in SKOV-3 ovarian cancer. In vitro cytotoxicity of BA and paclitaxel on human ovarian cancer cells (SKOV-3) was assessed using MTT assay. Antitumor potential of BA alone, standard anticancer drug (paclitaxel) alone, and BA in combination with paclitaxel were evaluated in SKOV-3 xenografted SCID mice. Immunohistochemical analysis of NF-κB was performed and analyzed in SKOV-3 tumors. BA alone and BA in combination with paclitaxel significantly inhibited the tumor growth. IC50 of BA was found to be 15.72 μM. Similarly, paclitaxel showed significant antitumor effect with IC50 of 3.234 μM. Treatments of paclitaxel, BA, and combination of BA with paclitaxel were well tolerated during treatment period. Immunohistochemical analysis of NF-κB in SKOV-3 tumors treated with BA in combination with paclitaxel revealed antitumor effect in terms of inhibition of NF-κB. Our results suggested that BA exhibits promising antitumor effect in the restriction of SKOV-3 cells and tumors with considerable safety.
Collapse
|
23
|
Kim GJ, Lee KJ, Choi JW, An JH. Drug Evaluation Based on a Multi-Channel Cell Chip with a Horizontal Co-Culture. Int J Mol Sci 2021; 22:6997. [PMID: 34209790 PMCID: PMC8269340 DOI: 10.3390/ijms22136997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/11/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022] Open
Abstract
We developed a multi-channel cell chip containing a three-dimensional (3D) scaffold for horizontal co-culture and drug toxicity screening in multi-organ culture (human glioblastoma, cervical cancer, normal liver cells, and normal lung cells). The polydimethylsiloxane (PDMS) multi-channel cell chip (PMCCC) was based on fused deposition modeling (FDM) technology. The architecture of the PMCCC was an open-type cell chip and did not require a pump or syringe. We investigated cell proliferation and cytotoxicity by conducting 3-(4,5-dimethylthiazol-2-yl)-2,5-dphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays and analysis of oleanolic acid (OA)-treated multi-channel cell chips. The results of the MTT and LDH assays showed that OA treatment in the multi-channel cell chip of four cell lines enhanced chemoresistance of cells compared with that in the 2D culture. Furthermore, we demonstrated the feasibility of the application of our multi-channel cell chip in various analysis methods through Annexin V-fluorescein isothiocyanate/propidium iodide staining, which is not used for conventional cell chips. Taken together, the results demonstrated that the PMCCC may be used as a new 3D platform because it enables simultaneous drug screening in multiple cells by single point injection and allows analysis of various biological processes.
Collapse
Affiliation(s)
- Gyeong-Ji Kim
- Department of Food and Nutrition, KC University, Seoul 07661, Korea;
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea
| | - Kwon-Jai Lee
- College of H-LAC, Daejeon University, Daejeon 34520, Korea;
| | - Jeong-Woo Choi
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea
| | - Jeung Hee An
- Department of Food and Nutrition, KC University, Seoul 07661, Korea;
| |
Collapse
|
24
|
Isaković-Vidović S, Dariš B, Knez Ž, Vidović K, Oprić D, Ferk P. Antiproliferative Activity of Selected Triterpene Acids from Rosemary on Metastatic Melanoma Cell Line WM-266-4. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Natural products and their derivatives, particularly secondary metabolites, have been recognized for many years as an important source of therapeutic agents. In this context, pentacyclic triterpene acids including betulinic acid (BA), oleanolic acid (OA), and ursolic acid (UA) are highly valuable triterpenic acids because of their wide range of biological activities.
AIM: Therefore, the aim of our study was to investigate any potential effect of BA, UA, and OA on human melanoma WM-266-4 cells’ proliferation activity.
METHODS: BA, UA, and OA have been prepared in dimethyl sulfoxide in concentration range from 0.002 to 200 μM separately or in selected combination (UA+OA ratio 1:1 or 3.5:1), while cells in cell culture medium served as controls. The rapid colorimetric MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was used to measure proliferation activity of the metastatic melanoma cell line WM-266-4 after being exposed to selected concentrations of BA, UA, OA, or UA+OA and during different time periods. Student’s t-test was used for single statistical comparisons. Data were analyzed using IBM SPSS 25.0 (IBM Corp., Armonk, NY). To account for multiple comparisons bias, p < 0.001 was considered statistically significant.
RESULTS: Our results showed decreased cell proliferation activity after 4 h of incubation of WM-266-4 cells with BA, UA, OA, and UA+OA. The highest inhibitory effect was noted when cells were incubated with selected triterpenic acids and both combinations of UA+OA during the incubation period of 48 h. When compared to control cells, concentration of 2 μM was the lowest concentration of BA that showed a significant decrease of the cells’ proliferation activity regardless the incubation period (4 h, 24 h, and 48 h) (p < 0.001).
CONCLUSION: Our encouraging results could be a good starting point for further studies on possible use of BA, UA, and OA in prevention and treatment of metastatic melanoma.
Collapse
|
25
|
Zhou X, Liu H, Zhang M, Li C, Li G. Spectrum-effect relationship between UPLC fingerprints and anti-lung cancer effect of Panax ginseng. PHYTOCHEMICAL ANALYSIS : PCA 2021; 32:339-346. [PMID: 32808367 PMCID: PMC8048684 DOI: 10.1002/pca.2980] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/21/2020] [Accepted: 07/27/2020] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Lung cancer has the highest mortality rate among the various types of cancer. Panax ginseng (C. A. Mey). is a popular anti-cancer herbal supplement. The quality control of ginseng is crucial to ensure its clinical efficacy. This study aimed to establish new quality control methods for ginseng and to identify its main active components responsible for lung cancer treatment. METHODS Ultra-high-performance liquid chromatography (UPLC) was used to establish fingerprints of 18 batches of ginseng. CCK-8 test was performed to evaluate the inhibitory activity of ginseng on Lewis lung cancer (LLC) cells. The spectrum-effect relationship analysis of ginseng was assessed by canonical correlation analysis (CCA) and bioactivity validation. KEY FINDINGS Six common peaks were identified and the variation coefficients were determined. The 18 batches of ginseng inhibited the proliferation of LLC cells to different degrees, showing different half maximal inhibitory concentration (IC50 ) values. Spectrum-effect relationship analysis showed that ginsenoside Ro is the main anti-proliferative constituent of LLC cell. CONCLUSIONS Spectrum-effect relationship is suitable for quality control of ginseng used for lung cancer. It is also effective in discovering the active ingredients related to the clinical efficacy of traditional Chinese medicine.
Collapse
Affiliation(s)
- Xiaowei Zhou
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haiyang Liu
- Tonghua Institute for Food and Drug ControlTonghuaChina
| | - Mingyu Zhang
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chunyu Li
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Guohui Li
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
26
|
Gudoityte E, Arandarcikaite O, Mazeikiene I, Bendokas V, Liobikas J. Ursolic and Oleanolic Acids: Plant Metabolites with Neuroprotective Potential. Int J Mol Sci 2021; 22:4599. [PMID: 33925641 PMCID: PMC8124962 DOI: 10.3390/ijms22094599] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ursolic and oleanolic acids are secondary plant metabolites that are known to be involved in the plant defence system against water loss and pathogens. Nowadays these triterpenoids are also regarded as potential pharmaceutical compounds and there is mounting experimental data that either purified compounds or triterpenoid-enriched plant extracts exert various beneficial effects, including anti-oxidative, anti-inflammatory and anticancer, on model systems of both human or animal origin. Some of those effects have been linked to the ability of ursolic and oleanolic acids to modulate intracellular antioxidant systems and also inflammation and cell death-related pathways. Therefore, our aim was to review current studies on the distribution of ursolic and oleanolic acids in plants, bioavailability and pharmacokinetic properties of these triterpenoids and their derivatives, and to discuss their neuroprotective effects in vitro and in vivo.
Collapse
Affiliation(s)
- Evelina Gudoityte
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (E.G.); (O.A.)
- Celignis Limited, Unit 11 Holland Road, Plassey Technology Park Castletroy, County Limerick, Ireland
| | - Odeta Arandarcikaite
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (E.G.); (O.A.)
| | - Ingrida Mazeikiene
- Lithuanian Research Centre for Agriculture and Forestry, Institute of Horticulture, Akademija, LT-58344 Kedainiai Distr., Lithuania;
| | - Vidmantas Bendokas
- Lithuanian Research Centre for Agriculture and Forestry, Institute of Horticulture, Akademija, LT-58344 Kedainiai Distr., Lithuania;
| | - Julius Liobikas
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (E.G.); (O.A.)
- Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The olive tree (Olea europaea L.) has featured as a significant part of medicinal history, used to treat a variety of ailments within folk medicine. The Mediterranean diet, which is rich in olive products, is testament to Olea europaeas positive effects on health, associated with reduced incidences of cancer and cardiovascular disease. This review aims to summarise the current literature regarding the therapeutic potential of Olea europaea products in cancer, detailing the possible compounds responsible for its chemotherapeutic effects. RECENT FINDINGS Much of the existing research has focused on the use of cell culture models of disease, demonstrating Olea europaea extracts, and specific compounds within these extracts, have efficacy in a range of in vitro and in vivo cancer models. The source of Olea europaeas cytotoxicity is yet to be fully defined; however, compounds such as oleuropein and verbascoside have independent cytotoxic effects on animal models of cancer. Initial results from animal models are promising but need to be translated to a clinical setting. Treatments utilising these compounds are likely to be well tolerated and represent a promising direction for future research.
Collapse
Affiliation(s)
- Chrystalla Antoniou
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Jonathon Hull
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
28
|
Chen X, Zhang Y, Zhang S, Wang A, Du Q, Wang Z. Oleanolic acid inhibits osteosarcoma cell proliferation and invasion by suppressing the SOX9/Wnt1 signaling pathway. Exp Ther Med 2021; 21:443. [PMID: 33747179 PMCID: PMC7967867 DOI: 10.3892/etm.2021.9883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 09/24/2020] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma is the most common primary bone malignancy in children and adolescents. Inhibition of SOX9/Wnt1-mediated signaling might suppress osteosarcoma metastasis, and oleanolic acid (OA) might decrease the activity of the SOX9/Wnt1 signaling pathway. The aim of the present study was to determine the role of OA in osteosarcoma cell proliferation and invasion. Osteosarcoma cell lines (KHOS and U2OS) and an osteoblastic cell line (hFOB1.19) were used for cell viability, proliferation and invasion analysis. The data suggested that OA significantly inhibited cell viability on days 3, 4 and 5 compared with the control (Ctrl) group in both U2OS and KHOS cells. Cell proliferation in the OA-treated group was significantly decreased compared with the Ctrl group in the osteosarcoma cell lines. Analysis of the cell cycle indicated that OA significantly reduced the percentage of U2OS and KHOS cells in the S phase compared with the Ctrl group. The wound healing assay results indicated that the OA group displayed significantly decreased cell re-colonization of the wound at 48 h compared with the Ctrl group. The Transwell chamber assay results also indicated that cell invasion was significantly inhibited by OA compared with the Ctrl group. Furthermore, OA significantly increased osteosarcoma cell apoptosis compared with the Ctrl group. Similarly, the protein expression levels of SOX9 and Wnt1 were significantly decreased in OA-treated U2OS and KHOS cells compared with Ctrl cells. OA-mediated downregulation of Wnt1 expression was reversed following SOX9 small interfering RNA transfection. Collectively, the results indicated that OA inhibited SOX9/Wnt1-associated osteosarcoma cell proliferation, migration and invasion.
Collapse
Affiliation(s)
- Xianming Chen
- Department of Osteopathic Medicine, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Yulin Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400030, P.R. China
| | - Sen Zhang
- Department of Osteopathic Medicine, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Aiming Wang
- Department of Osteopathic Medicine, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Quanyin Du
- Department of Osteopathic Medicine, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Ziming Wang
- Department of Osteopathic Medicine, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
29
|
Morgan HJN, Delgado AQ, Saldanha LL, Camaforte NADP, Dokkedal AL, Bosqueiro JR. Vochysia tucanorum Mart. butanol fraction presents antitumoral activity in vivo and prevents the installation of cachexia in solid Ehrlich tumor model. BMC Complement Med Ther 2021; 21:20. [PMID: 33413302 PMCID: PMC7791751 DOI: 10.1186/s12906-020-03190-1] [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: 07/29/2020] [Accepted: 12/15/2020] [Indexed: 11/25/2022] Open
Abstract
Background Cancer is a multifactorial disease caused by uncontrolled proliferation of cells. About 50–80% of cancer patients develop cachexia, a complex metabolic syndrome associated with an increase of mortality and morbidity. However, there are no effective therapies in medical clinic for cancer cachexia. Vochysia tucanorum Mart. is a common three of the Brazilian “Cerrado”. The butanolic fraction of V. tucanorum (Fr-BuVt), very rich in triterpenes with various biological activities, might be interesting in being tested in cancer cachexia syndrome. Hence, the present study was undertaken to investigate the antitumoral activity of Fr-BuVt and its potential against cachexia development. Methods Ehrlich tumor was used as model of cancer cachexia. Ascitic Ehrlich tumor cells were collected, processed and inoculated subcutaneously in saline solution (1 × 107/100 μl; ≥95% viability) for the obtention of solid Ehrlich carcinoma. After inoculation, solid Ehrlich carcinoma-bearing mice were treated by 14 consecutive days by gavage with Fr-BuVt (200 mg/kg). Body weight and tumor volume were measure during the treatment period. Tumors were removed, weighed and properly processed to measure the content and phosphorylation levels of key-proteins involved to apoptotic and proliferation process by Western Blot. Muscles and adipose tissues were removed for weighed. Serum was collected to cytokines levels and energetic blood markers measurements. Results The treatment with the Fr-BuVt (200 mg/kg, 14 days) decreased the solid Ehrlich tumor volume and weight besides increased the expression of the pro-apoptotic proteins caspase-3 and BAX, but also decreased the expression of the proteins involved in proliferation NFκB, mTOR and ERK. In addition, our data shows that the administration of Fr-BuVt was able to prevent the installation of cancer cachexia in Ehrlich carcinoma-bearing mice, since prevented the loss of body weight, as well as the loss of muscle and adipose tissue. Moreover, an improvement in some blood parameters such as decrease in cytokines TNF-α and IL-6 levels is observed. Conclusions The study revealed that Fr-BuVt has antitumoral activity and prevent installation of cancer cachexia in Ehrlich model. Therefore, Fr-BuVt may represent an alternative treatment for cancer cachexia. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-020-03190-1.
Collapse
Affiliation(s)
- Henrique Jorge Novaes Morgan
- Laboratory of Metabolism Control, Ribeirão Preto Medical School, Department of Physiology, University of São Paulo, Ribeirão Preto, State of São Paulo, Brazil
| | - Aislan Quintiliano Delgado
- Laboratory of Endocrine Pancreas Physiology, Faculty of Science, Department of Physical Education, São Paulo State University, Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, São Paulo, Postal Code: 17033-360, Brazil
| | - Luiz Leonardo Saldanha
- Laboratory of Natural Products Chemistry, Faculty of Science, Department of Biological Sciences, São Paulo State University, Bauru, State of São Paulo, Brazil
| | - Nathalia Aparecida De Paula Camaforte
- Laboratory of Endocrine Pancreas Physiology, Faculty of Science, Department of Physical Education, São Paulo State University, Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, São Paulo, Postal Code: 17033-360, Brazil
| | - Anne Lígia Dokkedal
- Laboratory of Natural Products Chemistry, Faculty of Science, Department of Biological Sciences, São Paulo State University, Bauru, State of São Paulo, Brazil
| | - José Roberto Bosqueiro
- Laboratory of Endocrine Pancreas Physiology, Faculty of Science, Department of Physical Education, São Paulo State University, Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, São Paulo, Postal Code: 17033-360, Brazil.
| |
Collapse
|
30
|
Wang J, Zhao H, Qiao W, Cheng J, Han Y, Yang X. Nanomedicine-Cum-Carrier by Co-Assembly of Natural Small Products for Synergistic Enhanced Antitumor with Tissues Protective Actions. ACS APPLIED MATERIALS & INTERFACES 2020; 12:42537-42550. [PMID: 32852938 DOI: 10.1021/acsami.0c12641] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The application of natural small products with self-assembly characteristics in a drug-delivery system is attractive for biomedical applications because of its inherent biological safety and pharmacological activity, and there is no complex structural modification process. However, drug carriers with pharmacological effects have not been developed enough. Here, we report a pure natural nanomedicine-cum-carrier (NMC) drug delivery system. The NMC is formed by the direct co-assembly of two small molecular natural compounds through noncovalent interaction, and a molecular dynamics model for predicting the co-assembly of two small molecular compounds was established. The representative co-assembled NMC (oleanolic acid and glycyrrhetinic acid) not only shows excellent stability, high drug loading, and sustained release characteristics but also the co-assembled NMC formed by two small molecular compounds has a synergistic antitumor effect (CI < 0.7). After drug loading, the antitumor effect is further improved. In addition, this NMC highlights the unique advantages of active natural products in biosafety and health benefits. Compared with free drugs, it can reduce the liver damage caused by chemotherapy drugs through upregulating key antioxidant pathways. Compared to nonpharmacologically active drug delivery systems, it can reduce the risk of nanotoxicity. Taken together, this co-assembly drug-carrier system overcomes the shortcomings that pharmacologically active compounds cannot be directly applied, enhances the pharmacological activity of bioactive drug carriers, improves the antitumor efficacy, and slows down the side effects induced by chemotherapy drugs and the additional toxicity caused by long-term use of non-bioactive nanocarriers.
Collapse
Affiliation(s)
- Jiacheng Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, No.92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang 150001, P.R.China
| | - Haitian Zhao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, No.92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang 150001, P.R.China
| | - Wenshu Qiao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, No.92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang 150001, P.R.China
| | - Jianjun Cheng
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, No.92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang 150001, P.R.China
| | - Ying Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, No.92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang 150001, P.R.China
| | - Xin Yang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, No.92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang 150001, P.R.China
| |
Collapse
|
31
|
Lu X, Li Y, Yang W, Tao M, Dai Y, Xu J, Xu Q. Inhibition of NF-κB is required for oleanolic acid to downregulate PD-L1 by promoting DNA demethylation in gastric cancer cells. J Biochem Mol Toxicol 2020; 35:e22621. [PMID: 32894642 DOI: 10.1002/jbt.22621] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/10/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
Gastric cancer is one of the most common causes of cancer-related death worldwide. Immunotherapy via programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) blockade has shown benefits for gastric cancer. Epigenetic DNA methylation critically regulates cancer immune checkpoints. We investigated how the natural compound oleanolic acid (OA) affected PD-L1 expression in gastric cancer cells. Interleukin-1β (IL-1β) at 20 ng/mL was used to stimulate human gastric cancer MKN-45 cells. IL-1β significantly increased PD-L1 expression, which was abolished by OA. Next, OA-treated MKN-45 cells were co-cultured with activated and PD-1-overexpressing Jurkat T cells. OA restored IL-2 levels in the co-culture system and increased T cell killing toward MKN-45 cells. Overexpression of PD-L1 eliminated OA-enhanced T cell killing capacity; however, PD-1 blocking antibody abrogated the cytotoxicity of T cells. Moreover, OA abolished IL-1β-increased DNA demethylase activity in MKN-45 cells. DNA methyltransferase inhibitor 5-azacytidine rescued OA-reduced PD-L1 expression; whereas DNA demethylation inhibitor gemcitabine inhibited PD-L1 expression, and, in combination with OA, provided more potent inhibitory effects. Furthermore, OA selectively reduced the expression of DNA demethylase TET3 in IL-1β-treated MKN-45 cells, and overexpression of TET3 restored OA-reduced PD-L1 expression. Finally, OA disrupted nuclear factor κB (NF-κB) signaling IL-1β-treated MKN-45 cells, and overexpression of NF-κB restored OA downregulation of TET3 and PD-L1. The cytotoxicity of T cells toward MKN-45 cells was also weakened by NF-κB overexpression. Altogether, OA blocked the IL-1β/NF-κB/TET3 axis in gastric cancer cells, leading to DNA hypomethylation and downregulation of PD-L1. Our discoveries suggested OA as an epigenetic modulator for immunotherapy or an adjuvant therapy against gastric cancer.
Collapse
Affiliation(s)
- Xirong Lu
- Department of Spleen and Stomach and Hepatology, The Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China.,Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Yuyi Li
- Department of Spleen and Stomach and Hepatology, The Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China.,Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Wei Yang
- Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Minghao Tao
- Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Yanmiao Dai
- Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Jinkang Xu
- Department of Spleen and Stomach and Hepatology, The Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China.,Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Qianfei Xu
- Department of Spleen and Stomach and Hepatology, The Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China.,Department of Spleen and Stomach and Hepatology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
32
|
Macașoi I, Pavel IZ, Moacă AE, Avram Ș, David VL, Coricovac D, Mioc A, Spandidos DA, Tsatsakis A, Șoica C, Dumitrașcu V, Dehelean C. Mechanistic investigations of antitumor activity of a Rhodamine B‑oleanolic acid derivative bioconjugate. Oncol Rep 2020; 44:1169-1183. [PMID: 32705265 PMCID: PMC7388574 DOI: 10.3892/or.2020.7666] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer remains a major health problem worldwide due to its high mortality rate. New therapeutic options highlight the importance of discovering new compounds that target the tumor microenvironment, interrupt angiogenesis and act selectively. The present study assessed the antitumor effect and investigated the mechanism of action of a rhodamine B-conjugated oleanolic acid derivative (RhodOA). Consequently, the compound was tested on different human tumor cell lines (A375 melanoma, A549 lung adenocarcinoma and MDA-MB-231 breast adenocarcinoma) and on a non-tumor cell line HaCaT human keratinocyte. RhodOA produced a dose-dependent decrease in tumor cell viability especially in the melanoma cells while affecting the keratinocytes less. In melanoma cells, RhodOA reduced cell migration and produced condensation of cell nuclei and of actin fibers. Furthermore, an impairment in melanoma cell mitochondrial function was observed, while the mitochondrial function of keratinocytes was left intact. In the in ovo chorioallantoic membrane model, RhodOA elicited antiangiogenic effect, without showing irritation effect on the membrane. The study provides information on the selective antitumor effect of the derivative and its ability to inhibit cellular respiration, therefore RhodOA can be classified as ‘MITOCAN’.
Collapse
Affiliation(s)
- Ioana Macașoi
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ioana Zinuca Pavel
- Department of Pharmacognosy, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Alina Elena Moacă
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ștefana Avram
- Department of Pharmacognosy, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Vlad Laurențiu David
- Department of Pediatric Surgery and Orthopedics, Faculty of Medicine, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dorina Coricovac
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Alexandra Mioc
- Department of Anatomy, Physiology and Pathophysiology, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 70013 Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Codruța Șoica
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Victor Dumitrașcu
- Department of Pharmacology, Faculty of Medicine, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cristina Dehelean
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, 'Victor Babes', University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
33
|
Cheon SY, Jin BR, Kim HJ, An HJ. Oleanolic Acid Ameliorates Benign Prostatic Hyperplasia by Regulating PCNA-Dependent Cell Cycle Progression In Vivo and In Vitro. JOURNAL OF NATURAL PRODUCTS 2020; 83:1183-1189. [PMID: 32191471 DOI: 10.1021/acs.jnatprod.9b01210] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oleanolic acid (OA) is a natural, biologically active pentacyclic triterpenoid found in Cornus officinalis. Although C. officinalis and OA have antiproliferative actions, the effects and mechanisms of OA in benign prostatic hyperplasia (BPH) are unclear. We examined the effect of OA in an animal model of testosterone-induced BPH. Male rats were injected with testosterone propionate with or without OA. The inhibitory effect of OA on BPH-1 cells was determined in vitro. Rats with BPH exhibited outstanding BPH symptoms, including prostatic enlargement, upregulated dihydrotestosterone and 5α-reductase 2 levels, and histological changes. Compared with the BPH group, the OA group showed fewer pathological alterations and regular androgen events. OA inhibited prostate cell proliferation by downregulating the expression of proliferating cell nuclear antigen (PCNA) and cell cycle markers in BPH-induced animals. This indicated that OA has superior therapeutic effect in the BPH animal model than finasteride. In vitro studies demonstrated upregulation of PCNA and cell cycle proteins, whereas OA clearly reduced this upregulation. Thus, OA may inhibit the development of BPH by targeting cell cycle progression markers. These suggest that OA is a potential agent for BPH treatment.
Collapse
Affiliation(s)
- Se-Yun Cheon
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Bo-Ram Jin
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
| | - Hyo-Jung Kim
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
| |
Collapse
|
34
|
Ebadollahi SH, Pouramir M, Zabihi E, Golpour M, Aghajanpour-Mir M. The Effect of Arbutin on The Expression of Tumor Suppressor P53, BAX/BCL-2 Ratio and Oxidative Stress Induced by Tert-Butyl Hydroperoxide in Fibroblast and LNcap Cell Lines. CELL JOURNAL 2020; 22:532-541. [PMID: 32347047 PMCID: PMC7211292 DOI: 10.22074/cellj.2021.6902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/29/2019] [Indexed: 01/17/2023]
Abstract
Objective Arbutin (p-hydroxyphenyl-β-D-glucopyranoside) possesses beneficial functions including antioxidant, antiinflammatory, and anti-tumoral activities. Due to the important role of oxidative stress and apoptosis in the successful treatment of cancer, understanding mechanisms that lead to apoptosis in cancer cells, is essential. The purpose of the current study was to evaluate the effect of arbutin on tert-butyl hydroperoxide (t-BHP)-induced oxidative stress and the related mechanisms in fibroblast and Lymph Node Carcinoma of the Prostate (LNCaP) cells. Materials and Methods In this experimental study, the LNCaP and fibroblast cell lines were pre-treated with arbutin (50, 250 and 1000 μM). After 24 hours, t-BHP (30 and 35 μM) was added to the cells. Viability was measured (at 24 and 48 hours) using MTT assay. The antioxidant effect of arbutin was measured by FRAP assay. The mRNA expression of P53 and BAX/BCL-2 ratio were measured using quantitative polymerase chain reaction (PCR). The percentage of apoptotic or necrotic cells was determined using a double staining annexin V fluorescein isothiocyanate (FITC) apoptosis detection kit. Results Arbutin pre-treatment increased the total antioxidative power and cell viability in the MTT assay and reduced BAX/BCL-2 ratio, P53 mRNA expression and necrosis in fibroblasts exposed to the oxidative agent (P<0.001). In addition, our results showed that arbutin can decrease cell viability, induce apoptosis and increase BAX/BCL-2 ratio in LNCaP cells at some specific concentrations (P<0.001). Conclusion Arbutin as a potential functional β-D-glucopyranoside has strong ability to selectively protect fibroblasts against t-BHP-induced cell damage and induce apoptosis in LNCaP cells.
Collapse
Affiliation(s)
- S Hima Ebadollahi
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.,Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mahdi Pouramir
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran. Electronic Address: .,Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Monireh Golpour
- Cellular and Molecular Biology Research Center, Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohsen Aghajanpour-Mir
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
35
|
Wang JL, Ren CH, Feng J, Ou CH, Liu L. Oleanolic acid inhibits mouse spinal cord injury through suppressing inflammation and apoptosis via the blockage of p38 and JNK MAPKs. Biomed Pharmacother 2020; 123:109752. [PMID: 31924596 DOI: 10.1016/j.biopha.2019.109752] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/07/2019] [Accepted: 11/29/2019] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury (SCI) is reported as a devastating disease, leading to tissue loss and neurologic dysfunction. However, there is no effective therapeutic strategy for SCI treatment. Oleanolic acid (OA), as a triterpenoid, has anti-oxidant, anti-inflammatory, and anti-apoptotic activities. However, its regulatory effects on SCI have little to be elucidated, as well as the underlying molecular mechanisms. In this study, we attempted to explore the role of OA in SCI progression. Behavior tests suggested that OA treatments markedly alleviated motor function in SCI mice. Evans blue contents up-regulated in spinal cords of SCI mice were significantly reduced by OA in a dose-dependent manner, demonstrating the improved blood-spinal cord barrier. Moreover, we found that OA treatments significantly reduced the apoptotic cell death in spinal cord samples of SCI mice through decreasing the expression of cleaved Caspase-3. In addition, pro-inflammatory response in SCI mice was significantly attenuated by OA treatments. Furthermore, SCI mice exhibited higher activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) signaling pathways, but these effects were clearly blocked in SCI mice with OA treatments, as evidenced by the down-regulated phosphorylation of p38, c-Jun-NH 2 terminal kinase (JNK), IκB kinase α (IKKα), inhibitor of nuclear factor κB-α (IκBα) and NF-κB. The protective effects of OA against SCI were confirmed in lipopolysaccharide (LPS)-stimulated mouse neurons mainly through the suppression of apoptosis and inflammatory response, which were tightly associated with the blockage of p38 and JNK activation. Together, our data demonstrated that OA treatments could dose-dependently ameliorate spinal cord damage through impeding p38- and JNK-regulated apoptosis and inflammation, and therefore OA might be served as an effective therapeutic agent for SCI treatment.
Collapse
Affiliation(s)
- Jiang-Lin Wang
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Chang-He Ren
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Ce-Hua Ou
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China.
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China.
| |
Collapse
|
36
|
Shu Y, He D, Li W, Wang M, Zhao S, Liu L, Cao Z, Liu R, Huang Y, Li H, Yang X, Lu C, Liu Y. Hepatoprotective Effect of Citrus aurantium L. Against APAP-induced Liver Injury by Regulating Liver Lipid Metabolism and Apoptosis. Int J Biol Sci 2020; 16:752-765. [PMID: 32071546 PMCID: PMC7019131 DOI: 10.7150/ijbs.40612] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022] Open
Abstract
Acetaminophen (APAP) refers to a medication used to manage pain and fever symptoms, but it always causes liver injury when overdosed. Zhishi, dried young fruit of Citrus aurantium L., is a famous Citrus herbal medicine in Asian countries which is rich in dietary phenolic substances. In this study, the mechanism of Zhishi protected against APAP-induced liver injury was studied more deeply by metabolomic strategy and pharmacological study. The metabolomics results demonstrated that Zhishi can prevent the APAP-induced liver injury model by regulating liver metabolic disorders in glycerophospholipid metabolism, fatty acid biosynthesis and glycerolipid metabolism. Moreover, it is confirmed that Zhishi blocked apoptosis of APAP-induced BRL-3A cell by simultaneously regulating p53 up-regulated apoptosis regulator (PUMA), AMPK-SIRT1 and JNK1 signaling pathways. Our findings indicated that Zhishi exhibited a hepaprotective effect against APAP-induced liver necrosis by inhibiting the PUMA and reversing disorder of liver lipid metabolism which could assist in improving the clinical outcomes of chemical-induced liver injury.
Collapse
Affiliation(s)
- Yisong Shu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dan He
- Patent Examination Cooperation (Tianjin) Center of the Patent Office, Tianjin, 300304, China
| | - Wen Li
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Menglei Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Siyu Zhao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Linlin Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhiwen Cao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rui Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yujuan Huang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hui Li
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xueqing Yang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanyan Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
37
|
The role of JNK in prostate cancer progression and therapeutic strategies. Biomed Pharmacother 2020; 121:109679. [DOI: 10.1016/j.biopha.2019.109679] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 12/31/2022] Open
|
38
|
Zheng SW, Xiao SY, Wang J, Hou W, Wang YP. Inhibitory Effects of Ginsenoside Ro on the Growth of B16F10 Melanoma via Its Metabolites. Molecules 2019; 24:E2985. [PMID: 31426477 PMCID: PMC6721120 DOI: 10.3390/molecules24162985] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022] Open
Abstract
Ginsenoside Ro (Ro), a major saponin derived and isolated from Panax ginseng C.A. Meyer, exerts multiple biological activities. However, the anti-tumour efficacy of Ro remains unclear because of its poor in vitro effects. In this study, we confirmed that Ro has no anti-tumour activity in vitro. We explored the anti-tumour activity of Ro in vivo in B16F10 tumour-bearing mice. The results revealed that Ro considerably suppressed tumour growth with no significant side effects on immune organs and body weight. Zingibroside R1, chikusetsusaponin IVa, and calenduloside E, three metabolites of Ro, were detected in the plasma of Ro-treated tumour-bearing mice and showed excellent anti-tumour effects as well as anti-angiogenic activity. The results suggest that the metabolites play important roles in the anti-tumour efficacy of Ro in vivo. Additionally, the haemolysis test demonstrated that Ro has good biocompatibility. Taken together, the findings of this study demonstrate that Ro markedly suppresses the tumour growth of B16F10-transplanted tumours in vivo, and its anti-tumour effects are based on the biological activity of its metabolites. The anti-tumour efficacy of these metabolites is due, at least in part, to its anti-angiogenic activity.
Collapse
Affiliation(s)
- Si-Wen Zheng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Sheng-Yuan Xiao
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Jia Wang
- School of Pharmaceutical Sciences Changchun University of Chinese Medicine, Changchun 130117, China
| | - Wei Hou
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Ying-Ping Wang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China.
| |
Collapse
|
39
|
Khan MW, Zhao P, Khan A, Raza F, Raza SM, Sarfraz M, Chen Y, Li M, Yang T, Ma X, Xiang G. Synergism of cisplatin-oleanolic acid co-loaded calcium carbonate nanoparticles on hepatocellular carcinoma cells for enhanced apoptosis and reduced hepatotoxicity. Int J Nanomedicine 2019; 14:3753-3771. [PMID: 31239661 PMCID: PMC6554709 DOI: 10.2147/ijn.s196651] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Cisplatin (CDDP), a widely used chemotherapeutic agent against hepatocellular carcinoma (HCC), faces severe resistance and hepatotoxicity problems which can be alleviated through combination therapy. Purpose: The objective of this study was to develop a pH-dependent calcium carbonate nano-delivery system for the combination therapy of CDDP with oleanolic acid (OA). Methods: A microemulsion method was employed to generate lipid coated cisplatin/oleanolic acid calcium carbonate nanoparticles (CDDP/OA-LCC NPs), and the loading concentration of CDDP and OA was measured by atomic absorption spectroscopy and HPLC respectively.Transmission electron microscopy (TEM) was used to examine the nanoparticles morphology while its pH dependent release characteristics were investigated through in vitro release study. Cellular uptake was examined through a fluorescence microscopy. Apoptotic assays and western blot analysis were conducted to explore the synergistic apoptotic effect of OA on CDDP against HCC cells. The hepatoprotective of OA for CDDP was evaluated through H&E staining. Results: TEM analysis revealed nanoparticles spherical shape with an average particle size of 206±15 nm, and the overall entrapment efficiency was 63.70%±3.9%. In vitro drug release study confirmed the pH-dependent property of the formulation, with the maximum CDDP release of 70%±4.6% at pH 5.5, in contrast to 28%±4.1% CDDP release at pH 7.4. Annexin V-FITC/PI assay and cell cycle analysis confirmed that CDDP and OA synergistically promoted greater HepG2 cells apoptosis for the CDDP/OA-LCC NPs as compared to their individual free drug solutions and NPs-treated groups. Western blot analysis also proved that CDDP/OA-LCC NPs induced the apoptosis by enhancing the proapoptotic protein expressions through downregulating P13K/AKT/mTOR pathway and upregulating p53 proapoptotic pathway. OA helped CDDP to overcome the resistance by downregulating the expression of proteins like XIAP, Bcl-2 via NF-κB pathway. OA also significantly alleviated CDDP-induced hepatotoxicity as evident from the decreased alanine transaminase, aspartate transaminase levels and histochemical evaluation. The possible mechanism may be related to the Nrf-2 induction via its antioxidant mechanism to maintain the redox balance and reduction in CYP2E1 activity which can lead to ROS-mediated oxidative stress. Conclusion: These results suggest that CDDP/OA-LCC NPs have promising applications for co-delivering CDDP and OA to synergize their anti-tumor activity against HCC and to utilize OA’s protective effect against CDDP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Muhammad Waseem Khan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Pengxuan Zhao
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Asifullah Khan
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Faisal Raza
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Shahid Masood Raza
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Muhammad Sarfraz
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475001/475004, People's Republic of China
| | - Yan Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Minsi Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Tan Yang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Xiang Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, People's Republic of China
| |
Collapse
|
40
|
Wang R, Yang W, Fan Y, Dehaen W, Li Y, Li H, Wang W, Zheng Q, Huai Q. Design and synthesis of the novel oleanolic acid-cinnamic acid ester derivatives and glycyrrhetinic acid-cinnamic acid ester derivatives with cytotoxic properties. Bioorg Chem 2019; 88:102951. [PMID: 31054427 DOI: 10.1016/j.bioorg.2019.102951] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022]
Abstract
Oleanolic acid (OA) and glycyrrhetinic acid (GA) are natural products with anticancer effects. Cinnamic acid (CA) and its derivatives also exhibited certain anticancer activity. In order to improve the anticancer activity of OA and GA, we designed and synthesized a series of novel OA-CA ester derivatives and GA-CA ester derivatives by using molecular hybridization approach. The 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was used to assess their in vitro cytotoxicity on three cell lines (HeLa (cervical cancer), MCF-7 (breast cancer) and L-O2 (a normal hepatic cell)). Among the evaluated compounds, 3o presented the strongest selective cytotoxicity on HeLa cells (IC50 = 1.35 μM) and showed no inhibitory activity against MCF-7 cells (IC50 > 100 μM) and L-O2 cells (IC50 > 100 μM), and 3e presented the strongest selective inhibition of the MCF-7 cells (IC50 = 1.79 μM). What's more, compound 2d also showed very strong selective inhibitory activity against HeLa cells (IC50 = 1.55 μM). The further research using Hoechst 33342, AO/EB dual-staining, flow cytometric analysis and DCFH-DA fluorescent dye staining assay presented that 2d and 3o could induce HeLa cells apoptosis and autophagy.
Collapse
Affiliation(s)
- Rui Wang
- Marine College, Shandong University, Weihai 264209, China; Molecular Design and Synthesis, Department of Chemistry, KU Leuven, Celestijnenlaan 200F, B-3001 Heverlee, Belgium
| | - Wei Yang
- Marine College, Shandong University, Weihai 264209, China
| | - Yiqing Fan
- Marine College, Shandong University, Weihai 264209, China
| | - Wim Dehaen
- Molecular Design and Synthesis, Department of Chemistry, KU Leuven, Celestijnenlaan 200F, B-3001 Heverlee, Belgium
| | - Yang Li
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Liaocheng, China
| | - Huijing Li
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Weihai 264209, China.
| | - Wei Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Qingxuan Zheng
- Marine College, Shandong University, Weihai 264209, China
| | - Qiyong Huai
- Marine College, Shandong University, Weihai 264209, China.
| |
Collapse
|
41
|
Lechner JF, Stoner GD. Red Beetroot and Betalains as Cancer Chemopreventative Agents. Molecules 2019; 24:E1602. [PMID: 31018549 PMCID: PMC6515411 DOI: 10.3390/molecules24081602] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/21/2022] Open
Abstract
Carcinogenesis is the process whereby a normal cell is transformed into a neoplastic cell. This action involves several steps starting with initiation and followed by promotion and progression. Driving these stages are oxidative stress and inflammation, which in turn encompasses a myriad of aberrant gene expressions, both within the transforming cell population and the cells within the surrounding lesion. Chemoprevention of cancer with bioreactive foods or their extracted/purified components occurs via normalizing these inappropriate gene activities. Various foods/agents have been shown to affect different gene expressions. In this review, we discuss whereby the chemoprevention activities of the red beetroot itself may disrupt carcinogenesis and the activities of the water-soluble betalains extracted from the plant.
Collapse
Affiliation(s)
- John F Lechner
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA.
| | - Gary D Stoner
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA.
| |
Collapse
|
42
|
de Oliveira JR, Camargo SEA, de Oliveira LD. Rosmarinus officinalis L. (rosemary) as therapeutic and prophylactic agent. J Biomed Sci 2019; 26:5. [PMID: 30621719 PMCID: PMC6325740 DOI: 10.1186/s12929-019-0499-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 01/02/2019] [Indexed: 12/22/2022] Open
Abstract
Rosmarinus officinalis L. (rosemary) is a medicinal plant native to the Mediterranean region and cultivated around the world. Besides the therapeutic purpose, it is commonly used as a condiment and food preservative. R. officinalis L. is constituted by bioactive molecules, the phytocompounds, responsible for implement several pharmacological activities, such as anti-inflammatory, antioxidant, antimicrobial, antiproliferative, antitumor and protective, inhibitory and attenuating activities. Thus, in vivo and in vitro studies were presented in this Review, approaching the therapeutic and prophylactic effects of R. officinalis L. on some physiological disorders caused by biochemical, chemical or biological agents. In this way, methodology, mechanisms, results, and conclusions were described. The main objective of this study was showing that plant products could be equivalent to the available medicines.
Collapse
Affiliation(s)
- Jonatas Rafael de Oliveira
- Departamento de Biociências e Diagnóstico Bucal, Instituto de Ciência e Tecnologia, Universidade Estadual Paulista (UNESP), Av. Engenheiro Francisco José Longo, 777 - Jardim São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil.
| | | | - Luciane Dias de Oliveira
- Departamento de Biociências e Diagnóstico Bucal, Instituto de Ciência e Tecnologia, Universidade Estadual Paulista (UNESP), Av. Engenheiro Francisco José Longo, 777 - Jardim São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| |
Collapse
|