1
|
Paul BM, Sundararajan VV, Raj FJ, Kannan G, Durairajan MB, Thangaraj P. In silico docking, ADMET profiling, and bio-accessibility experimentation on Breynia retusa phytocompounds and in vitro validation for anti-proliferative potencies against ovarian carcinoma. 3 Biotech 2025; 15:121. [PMID: 40225420 PMCID: PMC11981996 DOI: 10.1007/s13205-025-04276-8] [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: 01/18/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
This study aimed to assess the medicinal properties of Breynia retusa, a plant rich in phytocompounds predominantly used as an ethnomedicinal agent in Western Ghats, which appeared to be promising for therapeutic use, especially in the treatment of ovarian cancer. Herein, its cytotoxic potential on ovarian cancer cell lines SKOV-3, neurotoxicity, antioxidant activity, and molecular docking was determined to aid in explaining the mechanisms of interactions with proteins related to ovarian cancer. B . retusa methanolic extract demonstrated exuberant antioxidant activity, with 81.91% scavenging ability of DPPH radicals and efficient reduction of phosphomolybdenum (22.98 mg ascorbic acid equivalents antioxidant capacity/g extract). The extract proved to be an important anti-inflammatory agent through membrane stabilization inhibition of 83%. The cytotoxicity study against the SKOV-3 cell line indicated an IC50 value of 34.01 µg/mL and a very negligible neurotoxicity in SH-SY5Y cell lines. The GC-MS and HPLC profiling indicated many anticancer compounds in the extract such as secalciferol, methyl gallate, ricinoleic acid, gallic acid, and naringenin. The docking study showed significant interactions of secalciferol molecules with the key ovarian cancer proteins, which include IGF1 (-6.758 kcal/mol) and c-ERBB2 (-4.281 kcal/mol). Fatty acid derivatives and methyl gallate showed efficient dock scores (< -5.0 kcal/mol) with antioxidant (catalase and superoxide dismutase) enzymes and inflammatory cytokines (IL-6 and COX-1), respectively, as evidences of antioxidant and anti-inflammatory potentials. The bio-accessibility of phenolics and their antioxidant activity ranged above 90%, indicating the promising bioavailability of phytochemicals expected in vivo. Hence the current study emphasizes the anticancer potential of B. retusa phytocompounds that appeared to interact very strongly with ovarian cancer targets and confirms the dose-dependent cytotoxic and antioxidant activities of B. retusa methanolic extract. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04276-8.
Collapse
Affiliation(s)
- Benedict Mathews Paul
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Vetri Velavan Sundararajan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Francis Jegan Raj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Gowtham Kannan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Madhu Bala Durairajan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Parimelazhagan Thangaraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| |
Collapse
|
2
|
Shi Y, Gilkes DM. HIF-1 and HIF-2 in cancer: structure, regulation, and therapeutic prospects. Cell Mol Life Sci 2025; 82:44. [PMID: 39825916 PMCID: PMC11741981 DOI: 10.1007/s00018-024-05537-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/27/2024] [Accepted: 12/01/2024] [Indexed: 01/20/2025]
Abstract
Hypoxia, or a state of low tissue oxygenation, has been characterized as an important feature of solid tumors that is related to aggressive phenotypes. The cellular response to hypoxia is controlled by Hypoxia-inducible factors (HIFs), a family of transcription factors. HIFs promote the transcription of gene products that play a role in tumor progression including proliferation, angiogenesis, metastasis, and drug resistance. HIF-1 and HIF-2 are well known and widely described. Although these proteins share a high degree of homology, HIF-1 and HIF-2 have non-redundant roles in cancer. In this review, we summarize the similarities and differences between HIF-1α and HIF-2α in their structure, expression, and DNA binding. We also discuss the canonical and non-canonical regulation of HIF-1α and HIF-2α under hypoxic and normal conditions. Finally, we outline recent strategies aimed at targeting HIF-1α and/or HIF-2α.
Collapse
Affiliation(s)
- Yi Shi
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Kanika, Ahmad A, Kumar A, Rahul, Mishra RK, Ali N, Navik U, Parvez S, Khan R. Leveraging thiol-functionalized biomucoadhesive hybrid nanoliposome for local therapy of ulcerative colitis. Biomaterials 2025; 312:122747. [PMID: 39142219 DOI: 10.1016/j.biomaterials.2024.122747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/06/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
Directly administering medication to inflamed intestinal sites for treating ulcerative colitis (UC), poses significant challenges like retention time, absorption variability, side effects, drug stability, and non-specific delivery. Recent advancements in therapy to treat colitis aim to improve local drug availability that is enema therapy at the site of inflammation, thereby reducing systemic adverse effects. Nevertheless, a key limitation lies in enemas' inability to sustain medication in the colon due to rapid peristaltic movement, diarrhea, and poor local adherence. Therefore, in this work, we have developed site-specific thiolated mucoadhesive anionic nanoliposomes to overcome the limitations of conventional enema therapy. The thiolated delivery system allows prolonged residence of the delivery system at the inflamed site in the colon, confirmed by the adhesion potential of thiolated nanoliposomes using in-vitro and in-vivo models. To further provide therapeutic efficacy thiolated nanoliposomes were loaded with gallic acid (GA), a natural compound known for its antibacterial, antioxidant, and potent anti-inflammatory properties. Consequently, Gallic Acid-loaded Thiolated 2,6 DALP DMPG (GATh@APDL) demonstrates the potential for targeted adhesion to the inflamed colon, facilitated by their small size 100 nm and anionic nature. Therapeutic studies indicate that this formulation offers protective effects by mitigating colonic inflammation, downregulating the expression of NF-κB, HIF-1α, and MMP-9, and demonstrating superior efficacy compared to the free GA enema. The encapsulated GA inhibits the NF-κB expression, leading to enhanced expression of MUC2 protein, thereby promoting mucosal healing in the colon. Furthermore, GATh@APDL effectively reduces neutrophil infiltration and regulates immune cell quantification in colonic lamina propria. Our findings suggest that GATh@APDL holds promise for alleviating UC and addressing the limitations of conventional enema therapy.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab, 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N4N1, Canada
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab, 140306, India
| | - Rahul
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, Rajasthan, 302017, India
| | - Rakesh Kumar Mishra
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Ghudda, Punjab, 151401, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab, 140306, India.
| |
Collapse
|
4
|
Azevedo T, Ferreira T, Peña‐Corona SI, Cortes H, Silva‐Reis R, da Costa RMG, Faustino‐Rocha AI, Oliveira PA, Calina D, Cardoso SM, Büsselberg D, Leyva‐Gómez G, Sharifi‐Rad J, Cho WC. Natural products‐based antiangiogenic agents: New frontiers in cancer therapy. FOOD FRONTIERS 2024; 5:2423-2466. [DOI: 10.1002/fft2.466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractAngiogenesis, vital for tumor growth and metastasis, is a promising target in cancer therapy. Natural compounds offer potential as antiangiogenic agents with reduced toxicity. This review provides a comprehensive overview of natural product‐based antiangiogenic therapies, focusing on molecular mechanisms and therapeutic potential. A systematic search identified relevant articles from 2019 to 2023. Various natural compounds, including polyphenols, terpenes, alkaloids, cannabinoids, omega‐3 fatty acids, polysaccharides, proteins, and carotenoids, were investigated for their antiangiogenic properties. Challenges such as dose standardization, routes of administration, and potential side effects remain. Further studies, including in‐depth animal models and human epidemiological studies, must elucidate clinical efficacy and safety. Synergistic effects with current antiangiogenic therapies, such as bevacizumab and tyrosine kinase inhibitors, should be explored. Additionally, the potential hormone‐dependent effects of compounds like genistein highlight the need for safety evaluation. In conclusion, natural products hold promise as adjunctive therapies to conventional antineoplastic drugs in modulating angiogenesis in cancer. However, robust clinical trials are needed to validate preclinical findings and ensure safety and efficacy.
Collapse
Affiliation(s)
- Tiago Azevedo
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Tiago Ferreira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genómica Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Ciudad de México Mexico
| | - Rita Silva‐Reis
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | - Rui M. Gil da Costa
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI‐IPOP)/RISE@CI‐IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC) Porto Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering University of Porto Porto Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering University of Porto Porto Portugal
- Postgraduate Programme in Adult Health (PPGSAD), Department of Morphology Federal University of Maranhão (UFMA), UFMA University Hospital (HUUFMA) São Luís Brazil
| | - Ana I. Faustino‐Rocha
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology University of Évora Evora Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Daniela Calina
- Department of Clinical Pharmacy University of Medicine and Pharmacy of Craiova Craiova Romania
| | - Susana M. Cardoso
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | | | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Javad Sharifi‐Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo Veracruz Mexico
- Department of Medicine, College of Medicine Korea University Seoul Republic of Korea
- Facultad de Medicina Universidad del Azuay Cuenca Ecuador
| | - William C. Cho
- Department of Clinical Oncology Queen Elizabeth Hospital Kowloon Hong Kong
| |
Collapse
|
5
|
Hadidi M, Liñán-Atero R, Tarahi M, Christodoulou MC, Aghababaei F. The Potential Health Benefits of Gallic Acid: Therapeutic and Food Applications. Antioxidants (Basel) 2024; 13:1001. [PMID: 39199245 PMCID: PMC11352096 DOI: 10.3390/antiox13081001] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024] Open
Abstract
Gallic acid (GA), a phenolic acid found in fruits and vegetables, has been consumed by humans for centuries. Its extensive health benefits, such as antimicrobial, antioxidant, anticancer, anti-inflammatory, and antiviral properties, have been well-documented. GA's potent antioxidant capabilities enable it to neutralize free radicals, reduce oxidative stress, and protect cells from damage. Additionally, GA exerts anti-inflammatory effects by inhibiting inflammatory cytokines and enzymes, making it a potential therapeutic agent for inflammatory diseases. It also demonstrates anticancer properties by inhibiting cancer cell growth and promoting apoptosis. Furthermore, GA offers cardiovascular benefits, such as lowering blood pressure, decreasing cholesterol, and enhancing endothelial function, which may aid in the prevention and management of cardiovascular diseases. This review covers the chemical structure, sources, identification and quantification methods, and biological and therapeutic properties of GA, along with its applications in food. As research progresses, the future for GA appears promising, with potential uses in functional foods, pharmaceuticals, and nutraceuticals aimed at improving overall health and preventing disease. However, ongoing research and innovation are necessary to fully understand its functional benefits, address current challenges, and establish GA as a mainstay in therapeutic and nutritional interventions.
Collapse
Affiliation(s)
- Milad Hadidi
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Rafael Liñán-Atero
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, 13071 Ciudad Real, Spain;
| | - Mohammad Tarahi
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz 7144165186, Iran;
| | | | | |
Collapse
|
6
|
Antonio-Andres G, Morales-Martinez M, Jimenez-Hernandez E, Huerta-Yepez S. The Role of PTEN in Chemoresistance Mediated by the HIF-1α/YY1 Axis in Pediatric Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:7767. [PMID: 39063014 PMCID: PMC11276810 DOI: 10.3390/ijms25147767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Current chemotherapy treatment regimens have improved survival rates to approximately 80%; however, resistance development remains the primary cause of treatment failure, affecting around 20% of cases. Some studies indicate that loss of the phosphatase and tensin homolog (PTEN) leads to deregulation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway, increasing the expression of proteins involved in chemoresistance. PTEN loss results in deregulation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and induces hypoxia-inducible factor 1-alpha (HIF-1α) expression in various cancers. Additionally, it triggers upregulation of the Yin Yang 1 (YY1) transcription factor, leading to chemoresistance mediated by glycoprotein p-170 (Gp-170). The aim of this study was to investigate the role of the PTEN/NF-κB axis in YY1 regulation via HIF-1α and its involvement in ALL. A PTEN inhibitor was administered in RS4;11 cells, followed by the evaluation of PTEN, NF-κB, HIF-1α, YY1, and Gp-170 expression, along with chemoresistance assessment. PTEN, HIF-1α, and YY1 expression levels were assessed in the peripheral blood mononuclear cells (PBMC) from pediatric ALL patients. The results reveal that the inhibition of PTEN activity significantly increases the expression of pAkt and NF-κB, which is consistent with the increase in the expression of HIF-1α and YY1 in RS4;11 cells. In turn, this inhibition increases the expression of the glycoprotein Gp-170, affecting doxorubicin accumulation in the cells treated with the inhibitor. Samples from pediatric ALL patients exhibit PTEN expression and higher HIF-1α and YY1 expression compared to controls. PTEN/Akt/NF-κB axis plays a critical role in the regulation of YY1 through HIF-1α, and this mechanism contributes to Gp-170-mediated chemoresistance in pediatric ALL.
Collapse
Affiliation(s)
- Gabriela Antonio-Andres
- Oncology Disease Research Unit, Children’s Hospital of Mexico, Federico Gomez, Mexico City 06720, Mexico;
| | - Mario Morales-Martinez
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | | | - Sara Huerta-Yepez
- Oncology Disease Research Unit, Children’s Hospital of Mexico, Federico Gomez, Mexico City 06720, Mexico;
| |
Collapse
|
7
|
Xie T, Ding YH, Sang CS, Lin ZX, Dong J, Fu XA. Vitexin enhances radiosensitivity of mouse subcutaneous xenograft glioma by affecting the miR-17-5p/miR-130b-3p/PTEN/HIF-1α pathway. Strahlenther Onkol 2024; 200:535-543. [PMID: 38453699 DOI: 10.1007/s00066-024-02220-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE Vitexin can cooperate with hyperbaric oxygen to sensitize the radiotherapy of glioma by inhibiting the hypoxia-inducible factor (HIF)-1α. However, whether vitexin has a direct radiosensitization and how it affects the HIF-1α expression remain unclear. This study investigated these issues. METHODS The SU3 cells-inoculated nude mice were divided into control, radiation, and vitexin + radiation groups. The vitexin + radiation-treated mice were intraperitoneally injected with 75 mg/kg vitexin daily for 21 days. On the 3rd, 10th, and 17th days during the vitexin treatment, the radiation-treated mice were locally irradiated with 10 Gy, respectively. In vitro, the microRNA (miR)-17-5p or miR-130b-3p mimics-transfected SU3 cells were used to examine the effects of vitexin plus radiation on expression of miR-17-5p- or miR-130b-3p-induced radioresistance-related pathway proteins. The effects of vitexin on miR-17-5p and miR-130b-3p expression in SU3 cells were also evaluated. RESULTS Compared with the radiation group, the tumor volume, tumor weight, and expression of HIF-1α, vascular endothelial growth factor, and glucose transporter-1/3 proteins, miR-17-5p, and miR-130b-3p in tumor tissues in the vitexin + radiation group decreased, whereas the expression of phosphatase and tensin homolog (PTEN) protein increased. After treatment of miR-17-5p or miR-130b-3p mimics-transfected SU3 cells with vitexin plus radiation, the PTEN protein expression also increased, the HIF-1α protein expression decreased correspondingly. Moreover, vitexin decreased the miR-17-5p and miR-130b-3p expression in SU3 cells. CONCLUSION Vitexin can enhance the radiosensitivity of glioma, and its mechanism may partly be related to the attenuation of HIF-1α pathway after lowering the inhibitory effect of miR-17-5p and miR-130b-3p on PTEN.
Collapse
Affiliation(s)
- Tao Xie
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
- The Experimental Center and Department of Neurosurgery, The Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, China
| | - Yu-Hao Ding
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Chun-Sheng Sang
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Ze-Xi Lin
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Jun Dong
- The Experimental Center and Department of Neurosurgery, The Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province, China.
| | - Xi-An Fu
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
8
|
Bahreini E, Babaei M, Mohammadi F, Alizadeh-Fanalou S. Evaluation of serum pro/anti-angiogenic biomarkers in hyperglycemic rats treated with Securigera securidaca seeds, alone and in combination with Glibenclamide. J Cardiovasc Thorac Res 2024; 16:8-14. [PMID: 38584657 PMCID: PMC10997982 DOI: 10.34172/jcvtr.32960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/14/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Herbal medicines are commonly used by many people with diabetes in addition to standard treatment. Plants contain numerous known and unknown compounds that may exacerbate or ameliorate diabetes complications. Therefore, it is crucial to be aware of the side effects of these herbs before prescribing them. This study aimed to investigate the effects of hydroalcoholic extracts of Securigera securidaca (HESS) seeds alone and in combination with glibenclamide on the angiogenic/anti-angiogenic balance in streptozotocin (STZ)-induced diabetic rats. Methods Groups involved in this animal study included diabetic and healthy controls, three doses of HESS, glibenclamide, and combination therapy. Serum samples were collected and analyzed for a vascular endothelial growth factor (VEGF), fibroblast growth factor 21 (FGF21), fetal liver kinase 1 (FLK-1), soluble fms-like tyrosine kinase 1 (sFLT-1), and transforming growth factor -beta (TGF-β). Results Induction of diabetes increased VEGF, FGF21, and TGF-β serum levels and decreased circulating FLK-1 and sFLT-1 factors. Herbal extract, except TGF-β, had little effect on the above blood levels even at the highest doses. Glibenclamide was more effective than the highest dose of HESS in improving the vascular complications of diabetes. Combination therapy with the highest dose of HESS partly enhanced the glibenclamide effects. Conclusion Compared with glibenclamide as a standard chemical drug, HESS had no significant effects on the blood levels of the pro/anti-angiogenesis factor in diabetic rats. Glibenclamide attenuated the levels of the biomarkers and its effects were somewhat enhanced in combination with the highest dose of HESS.
Collapse
Affiliation(s)
- Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Babaei
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Forogh Mohammadi
- Department of Veterinary, Agriculture Faculty, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Shahin Alizadeh-Fanalou
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
9
|
Yadav NK, Yadav R. Medicinal Effects, Phytochemistry, Pharmacology of Euphorbia prostrata and Promising Molecular Mechanisms. Chin J Integr Med 2024; 30:181-192. [PMID: 36653685 DOI: 10.1007/s11655-023-3544-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 01/20/2023]
Abstract
Euphorbiaceae is a large family of dicotyledonous angiosperms with diverse genera including Euphorbia prostrata (E. prostrata). Current research has provided scientific evidence for traditional uses of E. prostrata against diverse pathological conditions such as anti-hemorrhoidal, anti-inflammatory, analgesic, wound healing, antioxidant, antibacterial, leishmanicidal, antitumor activity, and so on. The phytochemical screening has revealed the presence of glycosides, phytosterols, flavonoids, polyphenols, tannins, and anthraquinones with chemical structures elucidation of their respective compounds. The uniqueness of such multifactorial compounds present in this species endorses it as the potent therapeutic or prophylactic choice for several fatal diseases. Although ethnomedical applications served as a significant citation for pharmacology, the molecular mechanism has not been reviewed yet. The present paper provides a comprehensive review of research outcomes, pharmacology, toxicology, and molecular signaling of phytochemicals of E. prostrata species as a reference for relevant researchers. The study of bioactive compounds in crude extracts and fractions, the demonstration of primary mechanisms of pharmacology, along with the addition of toxicity, and clinical trials, should be conceded in depth. This review underlines the E. prostrata species that can be a promising phytomedicine since we are committed to excavating more intensely into their pharmacological role.
Collapse
Affiliation(s)
- Nirmala Kumari Yadav
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 122502, Haryana, India
| | - Rakesh Yadav
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
- National Forensic Sciences University, Tripura Campus, Agartala, 799001, Tripura, India.
| |
Collapse
|
10
|
Chu X, Xie F, Hou C, Zhang X, Wang S, Xie H, An C, Li Y, Zhao L, Xue P, Zhu S. Deciphering the Mechanism of Siwu Decoction Inhibiting Liver Metastasis by Integrating Network Pharmacology and In Vivo Experimental Validation. Integr Cancer Ther 2024; 23:15347354241236205. [PMID: 38462929 DOI: 10.1177/15347354241236205] [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] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Siwu Decoction (SWD) is a well-known classical TCM formula that has been shown to be effective as a basis for preventing and reducing liver metastases (LM). However, the active ingredients and potential molecular mechanisms remain unclear. OBJECTIVE This study aimed to systematically analyze the active ingredients and potential molecular mechanisms of SWD on LM and validate mechanisms involved. MATERIALS AND METHODS The active ingredients in SWD were extracted by UHPLC-MS/MS in a latest study. Protox II was retrieved to obtain toxicological parameters to detect safety. Swiss Target Prediction database was exploited to harvest SWD targets. Five databases, Gene Cards, DisGeNET, Drugbank, OMIM, and TTD, were employed to filter pathogenic targets of LM. STRING database was utilized to construct the protein-protein interaction network for therapeutic targets, followed by Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. GEPIA database and the Human Protein Atlas were taken to observe the expression of core genes and proteins. ImmuCellAI algorithm was applied to analyze the immune microenvironment and survival relevant to core genes. Molecular docking was performed to verify the affinity of SWD effective ingredients to core targets. In vivo experiments were carried out to validate the anti-LM efficacy of SWD and verify the pivotal mechanisms of action. RESULTS Eighteen main bioactive phytochemicals identified were all non-hepatotoxic. PPI network acquired 118 therapeutic targets, of which VEGFA, CASP3, STAT3, etc. were identified as core targets. KEGG analysis revealed that HIF-1 pathway and others were critical. After tandem targets and pathways, HIF-1/VEGF was regarded as the greatest potential pathway. VEGFA and HIF-1 were expressed differently in various stages of cancer and normal tissues. There was a negative regulation of immunoreactive cells by VEGFA, which was influential for prognosis. Molecular docking confirmed the tight binding to VEGFA. This study revealed the exact effect of SWD against LM, and identified significant inhibition the expression of HIF-1α, VEGF, and CD31 in the liver microenvironment. CONCLUSION This study clarified the active ingredients of SWD, the therapeutic targets of LM and potential molecular mechanisms. SWD may protect against LM through suppressing HIF-1/VEGF pathway.
Collapse
Affiliation(s)
- Xuelei Chu
- Wangjing Hospital Affiliated to China Academy of Chinese Medical Sciences, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Feiyu Xie
- Beijing University of Chinese Medicine, Beijing, China
| | - Chengzhi Hou
- Wangjing Hospital Affiliated to China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | - Sijia Wang
- Wangjing Hospital Affiliated to China Academy of Chinese Medical Sciences, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongting Xie
- Beijing University of Chinese Medicine, Beijing, China
| | - Chen An
- Wangjing Hospital Affiliated to China Academy of Chinese Medical Sciences, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Leyi Zhao
- Beijing University of Chinese Medicine, Beijing, China
| | - Peng Xue
- Wangjing Hospital Affiliated to China Academy of Chinese Medical Sciences, Beijing, China
| | - Shijie Zhu
- Wangjing Hospital Affiliated to China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Zhao H, Wang S, Williamson PTF, Ewing RM, Tang X, Wang J, Wang Y. Integrated network pharmacology and cellular assay reveal the biological mechanisms of Limonium sinense (Girard) Kuntze against Breast cancer. BMC Complement Med Ther 2023; 23:408. [PMID: 37957642 PMCID: PMC10644419 DOI: 10.1186/s12906-023-04233-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Limonium Sinense (Girard) Kuntze (L. sinense) has been widely used for the treatment of anaemia, bleeding, cancer, and other disorders in Chinese folk medicine. The aim of this study is to predict the therapeutic effects of L. sinense and investigate the potential mechanisms using integrated network pharmacology methods and in vitro cellular experiments. METHODS The active ingredients of L. sinense were collected from published literature, and the potential targets related to L. sinense were obtained from public databases. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and DisGeNET enrichment analyses were performed to explore the underlying mechanisms. Molecular docking, cellular experiments, RNA-sequencing (RNA-seq) and Gene Expression Omnibus (GEO) datasets were employed to further evaluate the findings. RESULTS A total of 15 active ingredients of L. sinense and their corresponding 389 targets were obtained. KEGG enrichment analysis revealed that the biological effects of L. sinense were primarily associated with "Pathways in cancer". DisGeNET enrichment analysis highlighted the potential role of L. sinense in the treatment of breast cancer. Apigenin within L. sinense showed promising potential against cancer. Cellular experiments demonstrated that the L. sinense ethanol extract (LSE) exhibited a significant growth inhibitory effect on multiple breast cancer cell lines in both 2D and 3D cultures. RNA-seq analysis revealed a potential impact of LSE on breast cancer. Additionally, analysis of GEO datasets verified the significant enrichment of breast cancer and several cancer-related pathways upon treatment with Apigenin in human breast cancer cells. CONCLUSION This study predicts the biological activities of L. sinense and demonstrates the inhibitory effect of LSE on breast cancer cells, highlighting the potential application of L. sinense in cancer treatment.
Collapse
Affiliation(s)
- Hualong Zhao
- School of Marine and Biological Engineering, Yancheng Teachers' University, Xiwang Road, Yancheng, 224002, PR China
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Siyuan Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Philip T F Williamson
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Rob M Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Xinhui Tang
- School of Marine and Biological Engineering, Yancheng Teachers' University, Xiwang Road, Yancheng, 224002, PR China
| | - Jialian Wang
- School of Marine and Biological Engineering, Yancheng Teachers' University, Xiwang Road, Yancheng, 224002, PR China.
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
12
|
Islam MM, Sreeharsha N, Alshabrmi FM, Asif AH, Aldhubiab B, Anwer MK, Krishnasamy R, Rehman A. From seeds to survival rates: investigating Linum usitatissimum's potential against ovarian cancer through network pharmacology. Front Pharmacol 2023; 14:1285258. [PMID: 37964873 PMCID: PMC10642394 DOI: 10.3389/fphar.2023.1285258] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Ovarian cancer is a malignant tumor that primarily forms in the ovaries. It often goes undetected until it has spread to the pelvis and abdomen, making it more challenging to treat and often fatal. Historically, natural products and their structural analogues have played a pivotal role in pharmacotherapy, especially for cancer. Numerous studies have demonstrated the therapeutic potential of Linum usitatissimum against ovarian cancer, but the specific molecular mechanisms remain elusive. This study combines data mining, network pharmacology, and molecular docking analysis to pioneer an innovative approach for ovarian cancer treatment by identifying potent phytochemicals. Findings of current study revealed that Apigenin, Vitamin E, Palmitic acid, Riboflavin, Isolariciresinol, 5-Dehydro-avenasterol, Cholesterol, Pantothenic acid, Nicotinic acid, Campesterol, Beta-Sitosterol, Stigmasterol, Daucosterol, and Vitexin suppress tumor growth by influencing AKT1, JUN, EGFR, and VEGFA. Kaplan-Meier survival analysis spotlighted AKT1, JUN, EGFR, and VEGFA as potential diagnostic and prognostic biomarkers for ovarian cancer. However, it is imperative to conduct in vivo and in vitro examinations to ascertain the pharmacokinetics and biosafety profiles, bolstering the candidacy of L. usitatissimum in ovarian cancer therapeutics.
Collapse
Affiliation(s)
- Mohammed Monirul Islam
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Bangalore, India
| | - Fahad M. Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Afzal Haq Asif
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Bandar Aldhubiab
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, China
| |
Collapse
|
13
|
Hassani S, Ghanbari F, Lotfi M, Alam W, Aschner M, Popović-Djordjević J, Shahcheraghi SH, Khan H. How gallic acid regulates molecular signaling: role in cancer drug resistance. Med Oncol 2023; 40:308. [PMID: 37755616 DOI: 10.1007/s12032-023-02178-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
Cancer is one of the deadliest and most heterogeneous diseases. Cancers often develop drug resistance, which can lead to treatment failure or recurrence. Accordingly, anticancer compounds are essential for chemotherapy-resistant cancer cells. Phenolic compounds are of interest in the development of cancer drugs due to their medicinal properties and ability to target different molecular pathways. Gallic acid (GA), as one of the main components of phenol, which is abundantly present in plant compounds such as walnut, sumac, grapes, tea leaves, oak bark, and other plant compounds, has antitumor properties. GA can prevent cancer progression, cell invasion, and metastasis by targeting molecular pathways and is an effective complement to chemotherapy drugs and combating multidrug resistance (MDR). In this review, we discuss various mechanisms related to cancer, the therapeutic potential of GA, the antitumor properties of GA in various cancers, and the targeted delivery of GA with nanocarriers.
Collapse
Affiliation(s)
- Samira Hassani
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Fahimeh Ghanbari
- Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jelena Popović-Djordjević
- Faculty of Agriculture, Department for Chemistry and Biochemistry, University of Belgrade, Nemanjina 6, 11080, Belgrade, Serbia
| | - Seyed Hossein Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
| |
Collapse
|
14
|
Masarkar N, Ray SK, Saleem Z, Mukherjee S. Potential anti-cancer activity of Moringa oleifera derived bio-active compounds targeting hypoxia-inducible factor-1 alpha in breast cancer. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 0:jcim-2023-0182. [PMID: 37712721 DOI: 10.1515/jcim-2023-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/13/2023] [Indexed: 09/16/2023]
Abstract
Breast cancer (BC) will become a highly detected malignancy in females worldwide in 2023, with over 2 million new cases. Studies have established the role of hypoxia-inducible factor-1α (HIF1α), a transcription factor that controls cellular response to hypoxic stress, and is essential for BC spread. HIF-1 is implicated in nearly every critical stage of the metastatic progression, including invasion, EMT, intravasation, extravasation, angiogenesis, and the formation of metastatic niches. HIF-1 overexpression has been associated with poor prognosis and increased mortality in BC patients. This is accomplished by controlling the expression of HIF-1 target genes involved in cell survival, angiogenesis, metabolism, and treatment resistance. Studies have indicated that inhibiting HIF-1 has an anti-cancer effect on its own and that inhibiting HIF-1-mediated signaling improves the efficacy of anti-cancer therapy. Approximately 74 % of recognized anti-cancer drugs are sourced from plant species. Studies on anti-cancer characteristics of phytochemicals derived from Moringa oleifera (MO), also known as the 'Tree of Life', have revealed a high therapeutic potential for BC. In this review, we have highlighted the various mechanisms through which bioactive compounds present in MO may modulate HIF and its regulatory genes/pathways, to prove their efficacy in treating and preventing BC.
Collapse
Affiliation(s)
- Neha Masarkar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | | | - Zirha Saleem
- Department of Biotechnology, Institute for Excellence in Higher Education, Bhopal, Madhya Pradesh, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| |
Collapse
|
15
|
Ahmad GM, Abu Serie MM, Abdel-Latif MS, Ghoneem T, Ghareeb DA, Yacout GA. Potential anti-proliferative activity of Salix mucronata and Triticum spelta plant extracts on liver and colorectal cancer cell lines. Sci Rep 2023; 13:3815. [PMID: 36882428 PMCID: PMC9992471 DOI: 10.1038/s41598-023-30845-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Cancer's etiology is linked to oxidative stress. As a result, it's vital to find effective natural antioxidant remedies. Salix mucronata and Triticum spelta plant extracts were prepared using five different solvents and examined for their cytotoxicity against liver HepG2 cancer cell line. It was found that Salix mucronata ethanolic extract is high in antioxidant mediated anti-cancer activity. The functional constituents (phenolic and flavonoids) as well as preparation of different ethanolic concentrations used to study their properties that include DPPH, oxygen, hydroxyl, nitrogen radical scavenging activities, ferric reducing power and metal chelating activities. The MTT assay was used to determine antioxidant-mediated anti-cancer activity against human liver (HepG2) and colorectal (Caco-2) cancer cells to calculate the half-maximal growth inhibitory concentration (IC50). Moreover, flow cytometry analysis was used to quantify the apoptotic effect on the treated cancer cells. Additionally, qRTPCR of p53, BCL2, Cyclin D, MMP9 and VEGF were measured. Furthermore, HPLC was used to assess the most effective ingredients of the plant extract. Salix mucronata 50% ethanol extract had the highest polyphenolic content, anti-oxidant, and anti-proliferative activity. Salix mucronata increased the number of total apoptotic cells, and caused an upregulation of p53 gene expression by more than five folds and a downregulation of gene expression level of BCL2, Cyclin D, MMP9 and VEGF by more than five folds. Consequently, that could modulate oxidative stress and improve the effectiveness of cancer therapy. Results, also, showed that Triticum spelta ethanolic extract was less effective than Salix mucronata. Therefore, Salix mucronata ethanolic extract represents promising surrogate natural therapy for apoptosis-mediated cancer and recommended for further investigation using animal model.
Collapse
Affiliation(s)
- Ghada M Ahmad
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, Egypt.
| | - Marwa M Abu Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GE-BRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, 21934, Alexandria, Egypt
| | - Mohamed S Abdel-Latif
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, Egypt
| | - Tayseer Ghoneem
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Doaa A Ghareeb
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Galila A Yacout
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Nazam N, Jabir NR, Ahmad I, Alharthy SA, Khan MS, Ayub R, Tabrez S. Phenolic Acids-Mediated Regulation of Molecular Targets in Ovarian Cancer: Current Understanding and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:274. [PMID: 37259418 PMCID: PMC9962268 DOI: 10.3390/ph16020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a global health concern with a dynamic rise in occurrence and one of the leading causes of mortality worldwide. Among different types of cancer, ovarian cancer (OC) is the seventh most diagnosed malignant tumor, while among the gynecological malignancies, it ranks third after cervical and uterine cancer and sadly bears the highest mortality and worst prognosis. First-line treatments have included a variety of cytotoxic and synthetic chemotherapeutic medicines, but they have not been particularly effective in extending OC patients' lives and are associated with side effects, recurrence risk, and drug resistance. Hence, a shift from synthetic to phytochemical-based agents is gaining popularity, and researchers are looking into alternative, cost-effective, and safer chemotherapeutic strategies. Lately, studies on the effectiveness of phenolic acids in ovarian cancer have sparked the scientific community's interest because of their high bioavailability, safety profile, lesser side effects, and cost-effectiveness. Yet this is a road less explored and critically analyzed and lacks the credibility of the novel findings. Phenolic acids are a significant class of phytochemicals usually considered in the nonflavonoid category. The current review focused on the anticancer potential of phenolic acids with a special emphasis on chemoprevention and treatment of OC. We tried to summarize results from experimental, epidemiological, and clinical studies unraveling the benefits of various phenolic acids (hydroxybenzoic acid and hydroxycinnamic acid) in chemoprevention and as anticancer agents of clinical significance.
Collapse
Affiliation(s)
- Nazia Nazam
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida 201301, Uttar Pradesh, India
| | - Nasimudeen R. Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur 613403, Tamil Nadu, India
| | - Iftikhar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Saif A. Alharthy
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rashid Ayub
- Technology and Innovation Unit, Department of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Effects of the Ethanol and Ethyl Acetate Extracts of Terminalia chebula Retz. on Proliferation, Migration, and HIF-1α and CXCR-4 Expression in MCF-7 Cells: an In Vitro Study. Appl Biochem Biotechnol 2022; 195:3327-3344. [PMID: 36585552 DOI: 10.1007/s12010-022-04301-z] [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] [Accepted: 12/16/2022] [Indexed: 01/01/2023]
Abstract
Over recent years, much attention has been devoted to the field of screening natural products and/or their novel structures because of reversing cancer progression. The current research work was intended to explore the cytotoxic activity of ethanol and ethyl acetate extracts of dried fruit of Terminalia chebula Retz. (T. chebula) in MCF-7 cell line. High-performance thin-layer chromatographic (HPTLC) method and Folin-Ciocalteu colorimetric techniques were performed. Anti-proliferative activities of T. chebula fruit extracts on the MCF-7 cell line were evaluated using MTT assay. Effects of both extracts on the migration of MCF-7 cells and the size of MCF-7-derived spheroids were also evaluated. Moreover, antioxidant properties were measured by DPPH and FRAP methods. Western blotting was used to measure the HIF-1α and CXCR-4 protein levels. Chebulagic acid, gallic acid, chebulinic acid, and ellagic acid were found as major compounds in both extracts. The total phenolic contents based on gallic acid equivalent (GAE) in the ethanol and ethyl acetate extracts of T. chebula were found to be 453.68 ± 0.31 and 495.12 ± 0.43 mg GAE/g dry weight of the extract, respectively. Both extracts exerted a significant dose- and time-dependent cytotoxicity effect on MCF-7 cells. They also had a marked negative effect on the average size of MCF-7-derived spheroids and their migration rate. None of the extracts exhibited stronger antioxidant activities than vitamin C. Furthermore, both extracts at a concentration of 125 µg/ml could meaningfully decrease the expression levels of HIF-1α and CXCR-4 in MCF-7 cells. These data represent that T. chebula may be a valuable medicinal resource in the regulation of breast cancer proliferation, growth, and metastasis.
Collapse
|
18
|
Srivastava S, Dubey AK, Madaan R, Bala R, Gupta Y, Dhiman BS, Kumar S. Emergence of nutrigenomics and dietary components as a complementary therapy in cancer prevention. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:89853-89873. [PMID: 36367649 DOI: 10.1007/s11356-022-24045-x] [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: 04/23/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
Cancer is an illness characterized by abnormal cell development and the capability to infiltrate or spread to rest of the body. A tumor is the term for this abnormal growth that develops in solid tissues like an organ, muscle, or bone and can spread to other parts of the body through the blood and lymphatic systems. Nutrition is a critical and immortal environmental component in the development of all living organisms encoding the relationship between a person's nutrition and their genes. Nutrients have the ability to modify gene expression and persuade alterations in DNA and protein molecules which is researched scientifically in nutrigenomics. These interactions have a significant impact on the pharmacokinetic properties of bioactive dietary components as well as their site of action/molecular targets. Nutrigenomics encompasses nutrigenetics, epigenetics, and transcriptomics as well as other "omic" disciplines like proteomics and metabolomics to explain the vast disparities in cancer risk among people with roughly similar life style. Clinical trials and researches have evidenced that alternation of dietary habits is potentially one of the key approaches for reducing cancer risk in an individual. In this article, we will target how nutrigenomics and functional food work as preventive therapy in reducing the risk of cancer.
Collapse
Affiliation(s)
| | - Ankit Kumar Dubey
- Institute of Scholars, Bengaluru, 577102, Karnataka, India.
- iGlobal Research and Publishing Foundation, New Delhi, 110059, India.
| | - Reecha Madaan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rajni Bala
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Yugam Gupta
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, Punjab, India
| |
Collapse
|
19
|
Xie L, Li J, Wang L, Dai Y. Engineering metal‐phenolic networks for enhancing cancer therapy by tumor microenvironment modulation. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 15:e1864. [PMID: 36333962 DOI: 10.1002/wnan.1864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022]
Abstract
The complicated tumor microenvironment (TME) is featured by low pH values, high redox status, and hypoxia, which greatly supports the genesis, development, and metastasis of tumors, leading to drug resistance and clinical failure. Moreover, a lot of immunosuppressive cells infiltrate in such TME, resulting in depressing immunotherapy. Therefore, the development of TME-responsive nanoplatforms has shown great significance in enhancing cancer therapeutics. Metal-phenolic networks (MPNs)-based nanosystems, which self-assemble via coordination of phenolic materials and metal ions, have emerged as excellent TME theranostic nanoplatforms. MPNs have unique properties including fast preparation, tunable morphologies, pH response, and biocompatibility. Besides, functionalization and surface modification can endow MPNs with specific functions for application requirements. Here, the representative engineering strategies of various polyphenols are first introduced, followed by the introduction of the engineering mechanisms of polyphenolic nanosystems, fabrication, and distinct properties of MPNs. Then, their advances in TME modulation are highlighted, such as antiangiogenesis, hypoxia relief, combination therapy sensitization, and immunosuppressive TME reversion. Finally, we will discuss the challenges and future perspectives of MPNs-based nanosystems for enhancing cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Lisi Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University Guangzhou China
| | - Jie Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
| | - Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering Southern Medical University Guangzhou Guangdong China
| | - Yunlu Dai
- Cancer Center and Institute of Translational Medicine, Faculty of Health Sciences University of Macau Macau China
- MOE Frontiers Science Center for Precision Oncology University of Macau Macau China
| |
Collapse
|
20
|
Sweidan K, Elfadel H, Sabbah DA, Bardaweel SK, Hajjo R, Anjum S, Sinoj J, Nair VA, Abu‐Gharbieh E, El‐Huneidi W. Novel Derivatives of 4,6‐Dihydroxy‐2‐Quinolone‐3‐Carboxamides as Potential PI3Kα Inhibitors. ChemistrySelect 2022. [DOI: 10.1002/slct.202202263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kamal Sweidan
- Department of Chemistry Institution The University of Jordan Amman 11942 Jordan
| | - Hussein Elfadel
- Department of Chemistry Institution The University of Jordan Amman 11942 Jordan
| | - Dima A. Sabbah
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan P.O. Box 130 Amman 11733 Jordan
| | - Sanaa K. Bardaweel
- Department of Pharmaceutical Sciences School of Pharmacy Institution The University of Jordan Amman 11942 Jordan
| | - Rima Hajjo
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan P.O. Box 130 Amman 11733 Jordan
| | - Shabana Anjum
- Sharjah Institute for Medical Research Institution University of Sharjah Sharjah 27272 United Arab Emirates
| | - Jithna Sinoj
- Sharjah Institute for Medical Research Institution University of Sharjah Sharjah 27272 United Arab Emirates
| | - Vidhya A. Nair
- Sharjah Institute for Medical Research Institution University of Sharjah Sharjah 27272 United Arab Emirates
| | - Eman Abu‐Gharbieh
- Sharjah Institute for Medical Research Institution University of Sharjah Sharjah 27272 United Arab Emirates
- College of Medicine Institution University of Sharjah Sharjah 27272 United Arab Emirates
| | - Waseem El‐Huneidi
- Sharjah Institute for Medical Research Institution University of Sharjah Sharjah 27272 United Arab Emirates
- College of Medicine Institution University of Sharjah Sharjah 27272 United Arab Emirates
| |
Collapse
|
21
|
Chen P, Ning X, Li W, Pan Y, Wang L, Li H, Fan X, Zhang J, Luo T, Wu Y, Ou C, Chen M. Fabrication of Tβ4-Exosome-releasing artificial stem cells for myocardial infarction therapy by improving coronary collateralization. Bioact Mater 2022; 14:416-429. [PMID: 35386821 PMCID: PMC8964820 DOI: 10.1016/j.bioactmat.2022.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Currently, stem cell transplantations in cardiac repair are limited owing to disadvantages, such as immunological rejection and poor cell viability. Although direct injection of exosomes can have a curative effect similar to that of stem cell transplantation, high clearance hinders its application in clinical practice. Previous reports suggested that induction of coronary collateralization can be a desired method of adjunctive therapy for someone who had missed the optimal operation time to attenuate myocardial ischemia. In this study, to mimic the paracrine and biological activity of stem cells, we developed artificial stem cells that can continuously release Tβ4-exosomes (Tβ4-ASCs) by encapsulating specific exosomes within microspheres using microfluidics technology. The results show that Tβ4-ASCs can greatly promote coronary collateralization in the periphery of the myocardial infarcted area, and its therapeutic effect is superior to that of directly injecting the exosomes. In addition, to better understand how it works, we demonstrated that the Tβ4-ASC-derived exosomes can enhance the angiogenic capacity of coronary endothelial cells (CAECs) via the miR-17-5p/PHD3/Hif-1α pathway. In brief, as artificial stem cells, Tβ4-ASCs can constantly release functional exosomes and stimulate the formation of collateral circulation after myocardial infarction, providing a feasible and alternative method for clinical revascularization. Inspired by the paracrine of stem cells, we fabricated artificial stem cells (Tβ4-ASCs) by loading engineered Tβ4-exosomes with microspheres using microfluidics technology. Tβ4-ASCs stimulate the formation of coronary collateralization in myocardial infarcted area through a slowly sustained release of engineered Tβ4-exosomes. Tβ4-ASCs improve coronary collateralization via the miR-17-5p/PHD3/Hif-1α signaling pathway.
Collapse
Affiliation(s)
- Peier Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaodong Ning
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weirun Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuxuan Pan
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Ling Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Hekai Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xianglin Fan
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jiexin Zhang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tiantian Luo
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yaobin Wu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Corresponding author.
| | - Caiwen Ou
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Corresponding author.
| | - Minsheng Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Corresponding author.
| |
Collapse
|
22
|
Gallic Acid: A Natural Phenolic Compound Exerting Antitumoral Activities in Colorectal Cancer via Interaction with G-Quadruplexes. Cancers (Basel) 2022; 14:cancers14112648. [PMID: 35681628 PMCID: PMC9179882 DOI: 10.3390/cancers14112648] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Gallic acid, a natural phenolic compound in diet, interacts with DNA G-quadruplexes both in vitro and in vivo. In particular, gallic acid targets G-quadruplexes in ribosomal DNA and CMYC oncogene, affecting gene expression. This action leads to antitumoral effects in colorectal cancer. In a patient cohort with CRC, we demonstrate that gallic acid could be explored as a therapeutic agent. Abstract Natural phenolic compounds have gained momentum for the prevention and treatment of cancer, but their antitumoral mechanism of action is not yet well understood. In the present study, we screened the antitumoral potential of several phenolic compounds in a cellular model of colorectal cancer (CRC). We selected gallic acid (GA) as a candidate in terms of potency and selectivity and extensively evaluated its biological activity. We report on the role of GA as a ligand of DNA G-quadruplexes (G4s), explaining several of its antitumoral effects, including the transcriptional inhibition of ribosomal and CMYC genes. In addition, GA shared with other established G4 ligands some effects such as cell cycle arrest, nucleolar stress, and induction of DNA damage. We further confirmed the antitumoral and G4-stabilizing properties of GA using a xenograft model of CRC. Finally, we succinctly demonstrate that GA could be explored as a therapeutic agent in a patient cohort with CRC. Our work reveals that GA, a natural bioactive compound present in the diet, affects gene expression by interaction with G4s both in vitro and in vivo and paves the way towards G4s targeting with phenolic compounds.
Collapse
|
23
|
Lee HS, Lee IH, Kang K, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. A Network Pharmacology Study to Uncover the Mechanism of FDY003 for Ovarian Cancer Treatment. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221075432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological tumors responsible for 0.21 million deaths per year worldwide. Despite the increasing interest in the use of herbal drugs for cancer treatment, their pharmacological effects in OC treatment are not understood from a systems perspective. Using network pharmacology, we determined the anti-OC potential of FDY003 from a comprehensive systems view. We observed that FDY003 suppressed the viability of human OC cells and further chemosensitized them to cytotoxic chemotherapy. Through network pharmacological and pharmacokinetic approaches, we identified 16 active ingredients in FDY003 and their 108 targets associated with OC mechanisms. Functional enrichment investigation revealed that the targets may coordinate diverse cellular behaviors of OC cells, including their growth, proliferation, survival, death, and cell cycle regulation. Furthermore, the FDY003 targets are important constituents of diverse signaling pathways implicated in OC mechanisms (eg, phosphoinositide 3-kinase [PI3K]-Akt, mitogen-activated protein kinase [MAPK], focal adhesion, hypoxia-inducible factor [HIF]-1, estrogen, tumor necrosis factor [TNF], erythroblastic leukemia viral oncogene homolog [ErbB], Janus kinase [JAK]-signal transducer and activator of transcription [STAT], and p53 signaling). In summary, our data present a comprehensive understanding of the anti-OC effects and mechanisms of action of FDY003.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| | - In-Hee Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
| | - Kyungrae Kang
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| | - Sang-In Park
- Forestheal Hospitalo, Songpa-gu, Seoul, Republic of Korea
| | - Minho Jung
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| | - Seung Gu Yang
- Kyunghee Naro Hospital, Bundang-gu, Seongnam, Republic of Korea
| | - Tae-Wook Kwon
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| | - Dae-Yeon Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| |
Collapse
|
24
|
García-Caballero M, Torres-Vargas JA, Marrero AD, Martínez-Poveda B, Medina MÁ, Quesada AR. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022; 14:pharmaceutics14020256. [PMID: 35213989 PMCID: PMC8875200 DOI: 10.3390/pharmaceutics14020256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
The number of cancer cases worldwide keeps growing unstoppably, despite the undeniable advances achieved by basic research and clinical practice. Urologic tumors, including some as prevalent as prostate, bladder or kidney tumors, are no exceptions to this rule. Moreover, the fact that many of these tumors are detected in early stages lengthens the duration of their treatment, with a significant increase in health care costs. In this scenario, prevention offers the most cost-effective long-term strategy for the global control of these diseases. Although specialized diets are not the only way to decrease the chances to develop cancer, epidemiological evidence support the role of certain plant-derived foods in the prevention of urologic cancer. In many cases, these plants are rich in antiangiogenic phytochemicals, which could be responsible for their protective or angiopreventive properties. Angiogenesis inhibition may contribute to slow down the progression of the tumor at very different stages and, for this reason, angiopreventive strategies could be implemented at different levels of chemoprevention, depending on the targeted population. In this review, epidemiological evidence supporting the role of certain plant-derived foods in urologic cancer prevention are presented, with particular emphasis on their content in bioactive phytochemicals that could be used in the angioprevention of cancer.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - José Antonio Torres-Vargas
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Ana Dácil Marrero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28019 Madrid, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
| | - Ana R. Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
- Correspondence:
| |
Collapse
|
25
|
Wang X, Du ZW, Xu TM, Wang XJ, Li W, Gao JL, Li J, Zhu H. HIF-1α Is a Rational Target for Future Ovarian Cancer Therapies. Front Oncol 2022; 11:785111. [PMID: 35004308 PMCID: PMC8739787 DOI: 10.3389/fonc.2021.785111] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/06/2021] [Indexed: 01/17/2023] Open
Abstract
Ovarian cancer is the eighth most commonly diagnosed cancer among women worldwide. Even with the development of novel drugs, nearly one-half of the patients with ovarian cancer die within five years of diagnosis. These situations indicate the need for novel therapeutic agents for ovarian cancer. Increasing evidence has shown that hypoxia-inducible factor-1α(HIF-1α) plays an important role in promoting malignant cell chemoresistance, tumour metastasis, angiogenesis, immunosuppression and intercellular interactions. The unique microenvironment, crosstalk and/or interaction between cells and other characteristics of ovarian cancer can influence therapeutic efficiency or promote the disease progression. Inhibition of the expression or activity of HIF-1α can directly or indirectly enhance the therapeutic responsiveness of tumour cells. Therefore, it is reasonable to consider HIF-1α as a potential therapeutic target for ovarian cancer. In this paper, we summarize the latest research on the role of HIF-1α and molecules which can inhibit HIF-1α expression directly or indirectly in ovarian cancer, and drug clinical trials about the HIF-1α inhibitors in ovarian cancer or other solid malignant tumours.
Collapse
Affiliation(s)
- Xin Wang
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - Zhen-Wu Du
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, China.,Research Center, The Second Hospital of Jilin University, Changchun, China
| | - Tian-Min Xu
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - Xiao-Jun Wang
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - Jia-Li Gao
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing Li
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - He Zhu
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Zhan D, Bian Z, Li H, Wang R, Fang G, Yao Q, Wu Z. Novel detection method for gallic acid: A water soluble boronic acid-based fluorescent sensor with double recognition sites. Bioorg Med Chem Lett 2021; 57:128483. [PMID: 34871766 DOI: 10.1016/j.bmcl.2021.128483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 11/02/2022]
Abstract
As one of the widespread phenols in nature, gallic acid (GA) has attracted a subject of attention due to its extensive biological properties. It is very important and significant to develop a sensitive and selective gallic acid sensor. In recent years, owing to their reversible covalent binding with Lewis bases and polyols, boronic acid compounds have been widely reported as fluorescence sensors for the identification of carbohydrates, ions and hydrogen peroxide, etc. However, boronic acid sensors for specific recognition of gallic acid have not been reported. Herein, a novel water-soluble boronic acid sensor with double recognition sites is reported. When the concentration of gallic acid added was 1.1 × 10-4 M, the fluorescence intensity of sensor 9b decreased by 80%, followed by pyrogallic acid and dopamine. However, the fluorescence of the sensor 9b combined with other analytes such as ATP, sialic acid, and uridine was basically unchanged, indicating that the sensor 9b had no ability to recognize these analytes. Also, sensor 9b has a fast response time to gallic acid at room temperature, and has a high binding constant (12355.9 ± 156.89 M-1) and low LOD (7.30 × 10-7 M). Moreover, gallic acid content of real samples was also determined, and the results showed that this method has a higher recovery rate. Therefore, sensor 9b can be used as a potential tool for detecting biologically significant gallic acid in actual samples such as food, medicine, and environmental analysis samples.
Collapse
Affiliation(s)
- Dongxue Zhan
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong, China; Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, China; Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, China
| | - Zhancun Bian
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong, China; Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, China; Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, China
| | - Haizhen Li
- Development and Planning Department, Shandong Light Industry Collective Enterprise Association, Jinan 250102, Shandong, China
| | - Ran Wang
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong, China; Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, China; Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, China
| | - Guiqian Fang
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong, China; Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, China; Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, China
| | - Qingqiang Yao
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong, China; Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, China; Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, China.
| | - Zhongyu Wu
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong, China; Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, China; Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, China.
| |
Collapse
|
27
|
Balogun TA, Ige OM, Alausa AO, Onyeani CO, Tiamiyu ZA, Omoboyowa DA, Saibu OA, Abdullateef OT. Receptor tyrosine kinases as a therapeutic target by natural compounds in cancer treatment. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00346-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Receptor tyrosine kinases (RTKs) are single-pass transmembrane proteins that play significant roles in regulating cellular processes, including cell division and growth. Overexpression and mutations of RTKs have been found in clinical manifestations of different forms of cancer. Therefore, RTKs have received considerable interest as a therapeutic biomarker in the treatment of cancer cells.
Main body of the abstract
Comprehensive data on RTKs, pharmacological and biological properties of natural compounds were systematically searched up to 2021 using relevant keywords from various databases, such as Google Scholar, PubMed, Web of Science, and Scopus. The scientific search by various standard electronic resources and databases unveils the effectiveness of medicinal plants in the treatment of various cancers. In vitro and in vivo studies suggested that bioactive compounds such as flavonoids, phenols, alkaloids, and many others can be used pharmacologically as RTKs inhibitors (RTKI) either by competing with ATP at the ATP binding site of the tyrosine kinase domain or competing for the receptor extracellular domain. Additionally, studies conducted on animal models indicated that inhibition of RTKs catalytic activity by natural compounds is one of the most effective ways to block the activation of RTKs signaling cascades, thereby hampering the proliferation of cancer cells. Furthermore, various pharmacological experiments, transcriptomic, and proteomic data also reported that cancer cells treated with different plants extracts or isolated phytochemicals exhibited better anticancer properties with minimal side effects than synthetic drugs. Clinically, natural compounds have demonstrated significant anti-proliferative effect via induction of cell apoptosis in cancer cell lines.
Short conclusion
An in-depth knowledge of the mechanism of inhibition and structural characterization of RTKs is important to the design of novel and selective RTKIs. This review focuses on the molecular mechanisms and structures of natural compounds RTKI targeting vascular endothelial growth factor, epidermal growth factor receptor, insulin receptor, and platelet-derived growth factor while also giving future directions to ameliorate the scientific burden of cancer.
Graphic abstract
Collapse
|
28
|
Onuma T, Mizutani T, Fujita Y, Yamada S, Yoshida Y. Copper content in ascitic fluid is associated with angiogenesis and progression in ovarian cancer. J Trace Elem Med Biol 2021; 68:126865. [PMID: 34601284 DOI: 10.1016/j.jtemb.2021.126865] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/24/2021] [Accepted: 09/16/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Ascites is associated with the poor prognosis of malignant tumors. The biological importance of the changes in the content of trace elements in the ascitic fluid is unknown. Herein, we analyzed trace elements in the ascitic fluid of patients with ovarian tumors and used cultured cells to determine the copper (Cu)-induced changes in gene expression in ovarian cancer. METHODS Inductively coupled plasma mass spectrometry (ICP-MS) was used to compare ascitic fluid trace element levels in patients with benign ovarian tumors (n = 22) and borderline/malignant tumors (n = 5) for primary screening. Cu levels were validated using atomic absorption spectrometry (AAS) in 88 benign, 11 borderline, and 25 malignant ovarian tumor patients. To confirm Cu-induced gene expression changes, microarray analysis was performed for Cu-treated OVCAR3, A2780, and Met5A cells. The vascular endothelial growth factor (VEGF) concentration in the cell supernatant or ascitic fluid (ovarian cancer samples) was measured using ELISA. RESULTS ICP-MS showed that Co, Ni, Cu, Zn, As, Se, and Mo levels significantly increased in patients with malignant/borderline ovarian tumors compared to those in patients with benign ovarian tumors. AAS showed that malignant ovarian tumors were independently associated with elevated levels of Cu in ascites adjusted for age, body mass index, alcohol, smoking, and supplement use (p < 0.001). Microarray analysis of both Cu-treated ovarian cancer cell lines OVCAR3 and A2780 and the mesothelial cell line Met-5A revealed the upregulation of the angiogenesis biological process. Real-time polymerase chain reaction and ELISA demonstrated that an increased Cu content significantly enhanced VEGF mRNA expression and protein secretion in OVCAR3, A2780, and Met-5A cells. VEGF levels and clinical stages of the tumors correlated with the ascitic fluid Cu content in patients with malignant ovarian tumors (correlation coefficient 0.445, 95 % confidence interval [CI]: 0.069-0.710, p = 0.023 and correlation coefficient 0.406, 95 % CI: 0.022-0.686, p = 0.040, respectively). CONCLUSION Cu levels significantly increased in patients with malignant ovarian cancer. Cu induced angiogenic effects in ovarian cancer and mesothelial cells, which affected ascites fluid production. This study clarifies the link between elevated Cu in ascites and malignant ovarian tumor progression. Strategies to decrease Cu levels in the ascitic fluid may help downregulate VEGF expression, thereby improving the prognosis of ovarian malignancies.
Collapse
Affiliation(s)
- Toshimichi Onuma
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Tetsuya Mizutani
- Department of Nursing, Faculty of Nursing and Welfare Sciences, Fukui Prefectural University, Fukui, 910-1195, Japan
| | - Yuko Fujita
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Shizuka Yamada
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan.
| |
Collapse
|
29
|
Brunty S, Clower L, Mitchell B, Fleshman T, Zgheib NB, Santanam N. Peritoneal Modulators of Endometriosis-Associated Ovarian Cancer. Front Oncol 2021; 11:793297. [PMID: 34900746 PMCID: PMC8655857 DOI: 10.3389/fonc.2021.793297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer is the 4th largest cause of cancer death in women. Approximately 10-15% of women of childbearing age suffer from endometriosis. Endometriosis is defined by the growth and presence of endometrial tissue (lesions) outside of the uterus. The women with endometriosis also have an increased presence of peritoneal fluid (PF) that comprises of inflammatory cells, growth factors, cytokines/chemokines, etc. Epidemiological studies have shown that >3% of women with endometriosis develop ovarian cancer (low-grade serous or endometrioid types). Our hypothesis is that the PF from women with endometriosis induces transformative changes in the ovarian cells, leading to ovarian cancer development. PF from women with and without endometriosis was collected after IRB approval and patient consent. IOSE (human normal ovarian epithelial cells) and TOV-21G cells (human ovarian clear cell carcinoma cell line) were treated with various volumes of PF (no endometriosis or endometriosis) for 48 or 96 h and proliferation measured. Expression levels of epigenetic regulators and FoxP3, an inflammatory tumor suppressor, were determined. A Human Cancer Inflammation and Immunity Crosstalk RT2 Profiler PCR array was used to measure changes in cancer related genes in treated cells. Results showed increased growth of TOV-21G cells treated with PF from women with endometriosis versus without endometriosis and compared to IOSE cells. Endo PF treatment induced EZH2, H3K27me3, and FoxP3. The RT2 PCR array of TOV-21G cells treated with endo PF showed upregulation of various inflammatory genes (TLRs, Myd88, etc.). These studies indicate that PF from women with endometriosis can both proliferate and transform ovarian cells and hence this microenvironment plays a major mechanistic role in the progression of endometriosis to ovarian cancer.
Collapse
Affiliation(s)
- Sarah Brunty
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Lauren Clower
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Brenda Mitchell
- Department of Obstetrics & Gynecology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Taylor Fleshman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Nadim Bou Zgheib
- Department of Obstetrics & Gynecology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| |
Collapse
|
30
|
Tuli HS, Mistry H, Kaur G, Aggarwal D, Garg VK, Mittal S, Yerer MB, Sak K, Khan MA. Gallic acid: a dietary polyphenol that exhibits anti-neoplastic activities by modulating multiple oncogenic targets. Anticancer Agents Med Chem 2021; 22:499-514. [PMID: 34802408 DOI: 10.2174/1871520621666211119085834] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/08/2021] [Accepted: 06/18/2021] [Indexed: 11/22/2022]
Abstract
Phytochemicals are being used for thousands of years to prevent dreadful malignancy. Side effects of existing allopathic treatment have also initiated intense research in the field of bioactive phytochemicals. Gallic acid, a natural polyphenolic compound, exists freely as well as in polymeric forms. The anti-cancer properties of gallic acid are indomitable by a variety of cellular pathways such as induction of programmed cell death, cell cycle apprehension, reticence of vasculature and tumor migration, and inflammation. Furthermore, gallic acid is found to show synergism with other existing chemotherapeutic drugs. Therefore, the antineoplastic role of gallic acid suggests its promising therapeutic candidature in the near future. The present review describes all these aspects of gallic acid at a single platform. In addition nanotechnology-mediated approaches are also discussed to enhance bioavailability and therapeutic efficacy.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana. India
| | - Hiral Mistry
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai 400056, Maharashtra. India
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai 400056, Maharashtra. India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana. India
| | - Vivek Kumar Garg
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University, Gharuan, Mohali - 140413, Punjab. India
| | - Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi. India
| | - Mükerrem Betül Yerer
- Erciyes University, Faculty of Pharmacy Department of Pharmacology, Erciyes University Drug Application and Research Center, 05056784551. Turkey
| | | | - Md Asaduzzaman Khan
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000. China
| |
Collapse
|
31
|
Jiang Y, Pei J, Zheng Y, Miao YJ, Duan BZ, Huang LF. Gallic Acid: A Potential Anti-Cancer Agent. Chin J Integr Med 2021; 28:661-671. [PMID: 34755289 DOI: 10.1007/s11655-021-3345-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 10/19/2022]
Abstract
Cancer is one of the most devastating diseases worldwide and definitive therapeutics for treating cancer are not yet available despite extensive research efforts. The key challenges include limiting factors connected with traditional chemotherapeutics, primarily drug resistance, low response rates, and adverse side-effects. Therefore, there is a high demand for novel anti-cancer drugs that are both potent and safe for cancer prevention and treatment. Gallic acid (GA), a natural botanic phenolic compound, can mediate various therapeutic properties that are involved in anti-inflammation, anti-obesity, and anti-cancer activities. More recently, GA has been shown to exert anti-cancer activities via several biological pathways that include migration, metastasis, apoptosis, cell cycle arrest, angiogenesis, and oncogene expression. This review discusses two aspects, one is the anti-cancer potential of GA against different types of cancer and the underlying molecular mechanisms, the other is the bibliometric analysis of GA in cancer and tumor research. The results indicated that lung cancer, prostate cancer, stomach cancer, and colon adenocarcinoma may become a hot topic in further research. Overall, this review provides evidence that GA represents a promising novel, potent, and safe anti-cancer drug candidate for treating cancer.
Collapse
Affiliation(s)
- Yuan Jiang
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.,State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,College of Pharmaceutical Science, Dali University, Dali, Yunnan Province, 671000, China
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yan Zheng
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Yu-Jing Miao
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Bao-Zhong Duan
- College of Pharmaceutical Science, Dali University, Dali, Yunnan Province, 671000, China
| | - Lin-Fang Huang
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China. .,State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
32
|
Ashrafizadeh M, Zarrabi A, Mirzaei S, Hashemi F, Samarghandian S, Zabolian A, Hushmandi K, Ang HL, Sethi G, Kumar AP, Ahn KS, Nabavi N, Khan H, Makvandi P, Varma RS. Gallic acid for cancer therapy: Molecular mechanisms and boosting efficacy by nanoscopical delivery. Food Chem Toxicol 2021; 157:112576. [PMID: 34571052 DOI: 10.1016/j.fct.2021.112576] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/23/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Cancer is the second leading cause of death worldwide. Majority of recent research efforts in the field aim to address why cancer resistance to therapy develops and how to overcome or prevent it. In line with this, novel anti-cancer compounds are desperately needed for chemoresistant cancer cells. Phytochemicals, in view of their pharmacological activities and capacity to target various molecular pathways, are of great interest in the development of therapeutics against cancer. Plant-derived-natural products have poor bioavailability which restricts their anti-tumor activity. Gallic acid (GA) is a phenolic acid exclusively found in natural sources such as gallnut, sumac, tea leaves, and oak bark. In this review, we report on the most recent research related to anti-tumor activities of GA in various cancers with a focus on its underlying molecular mechanisms and cellular pathwaysthat that lead to apoptosis and migration of cancer cells. GA down-regulates the expression of molecular pathways involved in cancer progression such as PI3K/Akt. The co-administration of GA with chemotherapeutic agents shows improvements in suppressing cancer malignancy. Various nano-vehicles such as organic- and inorganic nano-materials have been developed for targeted delivery of GA at the tumor site. Here, we suggest that nano-vehicles improve GA bioavailability and its ability for tumor suppression.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey; Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Farid Hashemi
- Phd student of pharmacology, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| | - Pooyan Makvandi
- Centre for Materials Interfaces, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy.
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
33
|
Turkez H, Tozlu OO, Arslan ME, Mardinoglu A. Safety and Efficacy Assessments to Take Antioxidants in Glioblastoma Therapy: From In Vitro Experiences to Animal and Clinical Studies. Neurochem Int 2021; 150:105168. [PMID: 34450218 DOI: 10.1016/j.neuint.2021.105168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is considered one of the most common malignant brain tumors, occurring as over 15% of all primary central nervous system and brain neoplasms. The unique and standard treatment option towards GBM involves the combination of surgical resection followed by radiotherapy (RT) and chemotherapy (CT). However, due to the aggressive nature and heterogeneity of GBMs, they remained difficult to treat. Recent findings from preclinical studies have revealed that disruption of the redox balance via using either oxidative or anti-oxidative agents in GBM presented an effective and promising therapeutic approach. A limited number of clinical trials substantially encouraged their concomitant use with RT or CT. Thus, treatment of GBMs may benefit from natural or synthetic antioxidative compounds as novel therapeutics. Despite the presence of variegated in vitro and in vivo studies focusing on safety and efficacy issues of these promising therapeutics, nowadays their translation to clinics is far from applicability due to several challenges. In this review, we briefly introduce the enzymatic and non-enzymatic antioxidant defense systems as well as potential signaling pathways related to the pathogenesis of GBM with a special interest in antioxidant mechanisms. In addition, we describe the advantages and limitations of antioxidant supplementation in GBM cases or disease models as well as growing challenges for GBM therapies with antioxidants in the future.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Ataturk University, 25240, Erzurum, Turkey
| | - Ozlem Ozdemir Tozlu
- Department of Molecular Biology and Genetics, Faculty of Science, 25250; Erzurum Technical University, Erzurum, Turkey
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Faculty of Science, 25250; Erzurum Technical University, Erzurum, Turkey
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK; Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, SE-17121, Sweden.
| |
Collapse
|
34
|
Oliva MA, Castaldo S, Rotondo R, Staffieri S, Sanchez M, Arcella A. Inhibiting effect of p-Coumaric acid on U87MG human glioblastoma cell growth. J Chemother 2021; 34:173-183. [PMID: 34424147 DOI: 10.1080/1120009x.2021.1953888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
p-Coumaric acid (pCA) is a hydroxycinnamic acid derivative commonly found in many natural products that has been extensively studied for its anticancer activity in multiple cell lines. In this report we investigated the effects of this phytochemical as adjuvant therapy to treat glioblastoma, an infaust brain tumour characterized by the acquired or innate resistance to the conventional chemotherapy temozolomide (TMZ). U87Mg glioblastoma cell growth and viability was assessed by growth rate curves and MTT assay incubating cells with 0.5 and 1 mM pCA for 24 h, 48 h and 72 h. Cell cycle analysis, performed by flow cytometry, showed that pCA led the accumulation of GBM cells in G2/M phase. Western blot analysis shows that pCA induced CDK4 cyclin-dependent kinase reduction and p53 increase, followed by induction of the CDK inhibitor p21. Furthermore, pCA treatment mediated the activation of apoptosis and the inhibition of migration of U87Mg cells. Finally, the treatment of glioblastoma cells in vitro with pCA concomitantly with the TMZ revealed a synergistic effect between the natural substance and the chemotherapy. In conclusion, our results demonstrated that pCA acts influencing the cell viability and cell cycle of U87Mg cells by promoting cell cycle arrest in G2/M phase and apoptosis.
Collapse
|
35
|
Qiao Y, Zhang Q, Wang Q, Li Y, Wang L. Filament-anchored hydrogel layer on polypropylene hernia mesh with robust anti-inflammatory effects. Acta Biomater 2021; 128:277-290. [PMID: 33866036 DOI: 10.1016/j.actbio.2021.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/18/2022]
Abstract
The efficacy of implanted polypropylene (PP) hernia meshes is often compromised by an inflammatory response. Thus, engineering an anti-inflammatory mesh has significant implications for hernioplasty. Here, we report a facile strategy to develop a filament-anchored hydrogel layer (FAHL) on PP mesh (FAHL-P). The network of FAHL, made up of chondroitin sulfate and gelatin (CG), provided a biomimetic surface with immunoregulatory properties. The use of tannic acid (TA) as a crosslinker for CG additionally enhanced its anti-inflammatory properties. In addition, the fabrication protocol ensured that the hydrogel maintained the properties of the knitted mesh and the firmly adherent FAHL during general handling (dry state) and in the simulated body environment (wet state). CG/TA-PP killed 99.99% of S. aureus and retained 73% of its original antioxidant properties after 7 d. The FAHL durably performed with a controlled release of TA for 15 d. The strong anti-inflammatory effects of FAHL-P reduced collagen deposition and increased vascularization, which promoted native tissue generation. The fabrication strategy has potential applications in hernioplasty and may provide new insights into the design of other anti-inflammatory implants. STATEMENT OF SIGNIFICANCE: A hydrogel layer with robust anti-inflammatory effects was anchored firmly on mesh filament for hernia repair. Requiring no drug loading, this chondroitin sulphate -gelatin (CG) based hydrogel itself could inhibit the immunological attack owing to the biomimetic microenvironment created by the CG. Moreover, the hydrogel's crosslinker (tannic acid) content served as an effective scavenger for reducing pro-inflammatory factors, significantly mitigating the inflammation. Interestingly, the antibacterial effect of such hydrogel layer was also observed. In terms of the synergistic outcome of the design, our mesh can remarkably attenuate inflammation and promote constructive tissue regeneration in vivo. Furthermore, considering the relatively simple and easily scaled up formulation process, our strategy may indeed have great potential in alleviating post-implantation outcomes.
Collapse
|
36
|
Habib MAH, Ismail MN. Extraction and identification of biologically important proteins from the medicinal plant God's crown (Phaleria macrocarpa). J Food Biochem 2021; 45:e13817. [PMID: 34137461 DOI: 10.1111/jfbc.13817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 11/30/2022]
Abstract
The fruit and leaf of God's crown (Phaleria macrocarpa) have been traditionally used to treat a wide variety of diseases. However, the proteins of this tropical plant are still heavily understudied. Three protein extraction methods; phenol (Phe), trichloroacetic acid (TCA)-acetone-phenol (TCA-A-Phe), and ultrasonic (Ult) were compared on the fruit and leaf of P. macrocarpa. The Phe extraction method showed the highest percentage of recovered protein after the resolubilization process for both leaf (12.24%) and fruit (30.41%) based on protein yields of the leaf (6.15 mg/g) and fruit (36.98 mg/g). Phe and TCA-A-Phe extraction methods gave well-resolved bands over a wide range of molecular weights through sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Following liquid chromatography-tandem mass spectrometry analysis, proteins identified through the Phe extraction method were 30%-35% enzymatic proteins, including oxidoreductases, transferases, hydrolases, lyases, isomerases, and ligases that possess various biological functions. PRACTICAL APPLICATIONS: Every part of God's crown plant is traditionally consumed to treat various illnesses. While plant's benefits are well known and have led to a plethora of health products, the proteome remains mostly unknown. This study compares three protein extraction methods for the leaf and fruit of P. macrocarpa and identifies their proteins thru LC-MS/MS coupled with PEAKS. These method comparisons can be a guide for works on other plants as well. In addition, the proteomics data from this study may shed light on the functional properties of these plant parts and their products.
Collapse
Affiliation(s)
- Mohd Afiq Hazlami Habib
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia (USM), Bayan Lepas, Penang, Malaysia
| | - Mohd Nazri Ismail
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia (USM), Bayan Lepas, Penang, Malaysia.,Institute For Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Bayan Lepas, Penang, Malaysia
| |
Collapse
|
37
|
Formulation and characterization of gallic acid and quercetin chitosan nanoparticles for sustained release in treating colorectal cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102523] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
38
|
Silva GAL, Araújo LB, Silva LCR, Gouveia BB, Barberino RS, Lins TLBG, Monte APO, Macedo TJS, Santos JMS, Menezes VG, Silva RLS, Matos MHT. Gallic acid promotes the in vitro development of sheep secondary isolated follicles involving the phosphatidylinositol 3-kinase pathway. Anim Reprod Sci 2021; 230:106767. [PMID: 34030069 DOI: 10.1016/j.anireprosci.2021.106767] [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: 11/20/2020] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 10/21/2022]
Abstract
This study was conducted to evaluate the effect of addition of gallic acid as the single antioxidant to the base medium for in vitro culture of sheep secondary follicles and if the phosphatidylinositol 3-kinase (PI3K) pathway is involved in the action of gallic acid. Secondary follicles were isolated and cultured for 12 days in α-MEM supplemented with bovine serum albumin (BSA), insulin, glutamine, hypoxanthine, transferrin, selenium, and ascorbic acid (control medium: α-MEM+) or in α-MEM supplemented with BSA, insulin, glutamine, hypoxanthine and different concentrations of gallic acid (25, 50 or 100 μM), thus replacing transferrin, selenium and ascorbic acid in the medium. Follicle morphology, glutathione (GSH), and mitochondrial activity, and meiotic resumption were evaluated. Furthermore, inhibition of PI3K pathway was performed by pretreatment with LY294002. After 12 days of culture, the follicle survival in a medium containing 100 μM gallic acid was similar (P > 0.05) to α-MEM+ and greater (P < 0.05) compared with other gallic acid concentrations. Antrum formation, follicle diameter, GSH, and mitochondrial activity, and meiotic resumption, however, were greater (P < 0.05) when 100 μM gallic acid was included in the α-MEM+ culture medium compared with the control medium. Furthermore, LY294002 inhibited (P < 0.05) follicle survival, development, and meiotic resumption stimulated by 100 μM gallic acid. In conclusion, concentration of 100 μM of gallic acid can be a substitute for transferrin, selenium, and ascorbic acid in the base medium during in vitro culture of sheep secondary follicles, inducing follicle development likely through the PI3K pathway.
Collapse
Affiliation(s)
- Gizele A L Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Luana B Araújo
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Larissa C R Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Bruna B Gouveia
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Thae Lanne B G Lins
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Alane P O Monte
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Taís J S Macedo
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Jamile M S Santos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Vanúzia G Menezes
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Regina L S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, 56300-990, Brazil.
| |
Collapse
|
39
|
Extracellular Vesicles from Human Umbilical Cord Mesenchymal Stem Cells Facilitate Diabetic Wound Healing Through MiR-17-5p-mediated Enhancement of Angiogenesis. Stem Cell Rev Rep 2021; 18:1025-1040. [PMID: 33942217 DOI: 10.1007/s12015-021-10176-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Endothelial dysfunction caused by persistent hyperglycemia in diabetes is responsible for impaired angiogenesis in diabetic wounds. Extracellular vehicles (EVs) are considered potential therapeutic tools to promote diabetic wound healing. The aim of this study was to investigate the effects of EVs secreted by human umbilical cord mesenchymal stem cells (hucMSC-EVs) on angiogenesis under high glucose (HG) conditions in vivo and in vitro and to explore the underlying mechanisms. In vivo, local application of hucMSC-EVs enhanced wound healing and angiogenesis. In vitro, hucMSC-EVs promoted proliferation, migration, and tube formation by inhibiting phosphatase and tensin homolog (PTEN) expression and activating the AKT/HIF-1α/VEGF pathways. MiR-17-5p was found to be highly enriched in hucMSC-EVs. In vitro, MiR-17-5p agomirs downregulated the expression of PTEN and activated the AKT/HIF-1α/VEGF pathway to promote proliferation, migration, and tube formation in HG-treated HUVECs. In vivo, miR-17-5p agomirs mimicked the effects of hucMSC-EVs on wound healing and angiogenesis, whereas miR-17-5p inhibitors reversed their effects. Our findings suggest that hucMSC-EVs have regenerative and protective effects on HG-induced endothelial cells via transfer of miR-17-5p targeting PTEN/ AKT/HIF-1α/VEGF pathway, thereby accelerating diabetic wound healing. Thus, hucMSC-EVs may be promising therapeutic candidates for improving diabetic wound angiogenesis.
Collapse
|
40
|
He Z, Liu X, Wu F, Wu S, Rankin GO, Martinez I, Rojanasakul Y, Chen YC. Gallic Acid Induces S and G2 Phase Arrest and Apoptosis in Human Ovarian Cancer Cells In Vitro. APPLIED SCIENCES (BASEL, SWITZERLAND) 2021; 11:3807. [PMID: 34386269 PMCID: PMC8356902 DOI: 10.3390/app11093807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ovarian cancer (OC) is among the top gynecologic cancers in the US with a death tally of 13,940 in the past year alone. Gallic acid (GA) is a natural compound with pharmacological benefits. In this research, the role of GA on cell proliferation, cell apoptosis, cell cycle-related protein expression was explored in OC cell lines OVCAR-3 and A2780/CP70. After 24,48 and 72 h of GA treatment, the IC50 values in OVCAR-3 cells were 22.14 ± 0.45, 20.36 ± 0.18, 15.13 ± 0.53 μM, respectively and in A2780/CP70 cells IC50 values were 33.53 ± 2.64, 27.18 ± 0.22, 22.81 ± 0.56, respectively. Hoechst 33,342 DNA staining and flow cytometry results showed 20 μM GA exposure could significantly accelerate apoptosis in both OC cell lines and the total apoptotic rate increased from 5.34%(control) to 21.42% in OVCAR-3 cells and from 8.01%(control) to 17.69% in A2780/CP70 cells. Western blot analysis revealed that GA stimulated programmed OC cell death via a p53-dependent intrinsic signaling. In addition, GA arrested cell cycle at the S or G2 phase via p53-p21-Cdc2-cyclin B pathway in the same cells. In conclusion, we provide some evidence of the efficacy of GA in ovarian cancer prevention and therapy.
Collapse
Affiliation(s)
- Zhiping He
- The Key Laboratory for Quality Improvement of Agricultural Products of Zhejiang Province, College of Agriculture and Food Science, Zhejiang A & F University, Lin’ an, Hangzhou 311300, China
- College of Health, Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Xingquan Liu
- The Key Laboratory for Quality Improvement of Agricultural Products of Zhejiang Province, College of Agriculture and Food Science, Zhejiang A & F University, Lin’ an, Hangzhou 311300, China
| | - Fenghua Wu
- The Key Laboratory for Quality Improvement of Agricultural Products of Zhejiang Province, College of Agriculture and Food Science, Zhejiang A & F University, Lin’ an, Hangzhou 311300, China
| | - Shaozhen Wu
- The Key Laboratory for Quality Improvement of Agricultural Products of Zhejiang Province, College of Agriculture and Food Science, Zhejiang A & F University, Lin’ an, Hangzhou 311300, China
| | - Gary O’Neal Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Ivan Martinez
- Department of Microbiology, Immunology & Cell Biology and WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV 26506, USA
| | - Yi Charlie Chen
- College of Health, Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
41
|
Yan W, Wang Y, Chen Y, Guo Y, Li Q, Wei X. Exosomal miR-130b-3p Promotes Progression and Tubular Formation Through Targeting PTEN in Oral Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:616306. [PMID: 33829013 PMCID: PMC8019696 DOI: 10.3389/fcell.2021.616306] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC), accounting for two-thirds of head and neck cancer, is characterized by poor prognosis and a high rate of mortality. Exosomes have emerged as potential molecule-shuttle in intercellular communication, thereby regulating the physiological processes of recipient cells. To date, the effect of exosomal microRNAs (miRNAs) on the progression of OSCC has not been fully investigated. In this study, we found that the protein, but not mRNA expression of Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) was decreased in OSCC. The results revealed that miR-130b-3p was an important negative regulator for PTEN expression. Additionally, overexpression and knockdown of miR-130b-3p enhanced and inhibited angiogenesis in human umbilical vein endothelial cells (HUVECs), respectively. Also, miR-130b-3p was transferred by exosomes to HUVECs and then promoted angiogenesis and inhibit the expression of PTEN. Furthermore, exosomal miR-130b-3p derived from OSCC cells promoted tumor growth and blood vessel formation in the xenograft mice model. Taken together, we demonstrated that exosome-mediated miR-130b-3p promoted progression and tubular formation in OSCC in vitro and in vivo. These results would provide new insight into exploring biomarkers and effective therapeutic strategies for OSCC.
Collapse
Affiliation(s)
- Wei Yan
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Yuping Wang
- Department of Stomatology of Shennongju Hospital, Huanghua, China
| | - Yong Chen
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Yanjun Guo
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Qiang Li
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaotong Wei
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
42
|
Woźniak M, Krajewski R, Makuch S, Agrawal S. Phytochemicals in Gynecological Cancer Prevention. Int J Mol Sci 2021; 22:1219. [PMID: 33530651 PMCID: PMC7865323 DOI: 10.3390/ijms22031219] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
Gynecological cancer confers an enormous burden among women worldwide. Accumulating evidence points to the role of phytochemicals in preventing cervical, endometrial, and ovarian cancer. Experimental studies emphasize the chemopreventive and therapeutic potential of plant-derived substances by inhibiting the early stages of carcinogenesis or improving the efficacy of traditional chemotherapeutic agents. Moreover, a number of epidemiological studies have investigated associations between a plant-based diet and cancer risk. This literature review summarizes the current knowledge on the phytochemicals with proven antitumor activity, emphasizing their effectiveness and mechanism of action in gynecological cancer.
Collapse
Affiliation(s)
- Marta Woźniak
- Department of Pathology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (S.M.)
| | - Rafał Krajewski
- Department and Clinic of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Sebastian Makuch
- Department of Pathology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (S.M.)
| | - Siddarth Agrawal
- Department of Pathology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (S.M.)
- Department and Clinic of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
- Department of Cancer Prevention and Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
43
|
He Z, Wu S, Lin J, Booth A, Rankin GO, Martinez I, Chen YC. Polyphenols Extracted from Chinese Hickory ( Carya cathayensis) Promote Apoptosis and Inhibit Proliferation through the p53-Dependent Intrinsic and HIF-1α-VEGF Pathways in Ovarian Cancer Cells. APPLIED SCIENCES (BASEL, SWITZERLAND) 2020; 10:8615. [PMID: 33520293 PMCID: PMC7842596 DOI: 10.3390/app10238615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ovarian cancer is the second most common gynecologic cancer with an estimated 13,940 mortalities across the United States in 2020. Natural polyphenols have been shown to double the survival time of some cancer patients due to their anticancer properties. Therefore, the effect of polyphenols extracted from Chinese hickory seed skin Carya cathayensis (CHSP) on ovarian cancer was investigated in the present study. Cell viability results showed that CHSP is more effective in inhibiting ovarian cancer cells than normal ovarian cells, with the IC50 value for inhibition of cell proliferation of Ovarian cancer cells (OVCAR-3) being 10.33 ± 0.166 μg/mL for a 24 h treatment. Flow cytometry results showed that the apoptosis rate was significantly increased to 44.21% after 24 h treatment with 20 μg/mL of CHSP. Western blot analysis showed that CHSP induced apoptosis of ovarian cancer cells through a p53-dependent intrinsic pathway. Compared with control values, levels of VEGF excreted by OVCAR-3 cancer cells were reduced to 7.87% with a 40 μg/mL CHSP treatment. Consistent with our previous reports, CHSP inhibits vascular endothelial growth factor (VEGF) secretion by regulating the HIF-1α-VEGF pathway. In addition, we also found that the inhibitory effect of CHSP on ovarian cancer is related to the up-regulation of Phosphatase and tension homolog (PTEN) and down-regulation of nuclear factor kappa-B (NF-kappa B). These findings provide some evidence of the anti-ovarian cancer properties of CHSP and support the polyphenols as potential candidates for ovarian cancer adjuvant therapy.
Collapse
Affiliation(s)
- Zhiping He
- The Key Laboratory for Quality Improvement of Agricultural Products of Zhejiang Province, College of Agriculture and Food Science, Zhejiang A & F University, Hangzhou 311300, China
| | - Shaozhen Wu
- The Key Laboratory for Quality Improvement of Agricultural Products of Zhejiang Province, College of Agriculture and Food Science, Zhejiang A & F University, Hangzhou 311300, China
| | - Ju Lin
- College of Health, Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Ashley Booth
- College of Health, Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Gary O’Neal Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Ivan Martinez
- Department of Microbiology, Immunology & Cell Biology and WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Yi Charlie Chen
- College of Health, Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
44
|
Cornebise C, Courtaut F, Taillandier-Coindard M, Valls-Fonayet J, Richard T, Monchaud D, Aires V, Delmas D. Red Wine Extract Inhibits VEGF Secretion and Its Signaling Pathway in Retinal ARPE-19 Cells to Potentially Disrupt AMD. Molecules 2020; 25:molecules25235564. [PMID: 33260857 PMCID: PMC7731402 DOI: 10.3390/molecules25235564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/26/2022] Open
Abstract
Age-related macular degeneration (AMD) is a degenerative disease of the retina where the molecular mechanism involves the production of vascular endothelial growth factor (VEGF), a factor of poor prognosis of the progression of the disease. Previous studies have shown that resveratrol, a polyphenol of grapevines, can prevent VEGF secretion induced by stress from retinal cells. Considering the fundamental role played by VEGF in development and progression of AMD, we investigate the potential effect of red wine extract (RWE) on VEGF secretion and its signaling pathway in human retinal cells ARPE-19. To examine the effect of RWE in ARPE-19, a quantitative and qualitative analysis of the RWE was performed by HPLC MS/MS. We show for the first time that RWE decreased VEGF-A secretion from ARPE-19 cells and its protein expression in concentration-dependent manner. RWE-induced alteration in VEGF-A production is associated with a down of VEGF-receptor 2 (VEGF-R2) protein expression and its phosphorylated intracytoplasmic domain. Subsequently, the activation of kinase pathway is disturbing and RWE prevents the phosphorylation of MEK and ERK 1/2 in human retinal cells ARPE-19. Finally, this study sheds light on the interest that the use of polyphenolic cocktails could represent in a prevention strategy.
Collapse
Affiliation(s)
- Clarisse Cornebise
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (C.C.); (F.C.); (M.T.-C.); (D.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Flavie Courtaut
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (C.C.); (F.C.); (M.T.-C.); (D.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Marie Taillandier-Coindard
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (C.C.); (F.C.); (M.T.-C.); (D.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Josep Valls-Fonayet
- Unité de Recherche Oenologie, EA 4577, USC 1366 INRA-ISVV, F-33882 Villenave d’Ornon, France; (J.V.-F.); (T.R.)
| | - Tristan Richard
- Unité de Recherche Oenologie, EA 4577, USC 1366 INRA-ISVV, F-33882 Villenave d’Ornon, France; (J.V.-F.); (T.R.)
| | - David Monchaud
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (C.C.); (F.C.); (M.T.-C.); (D.M.); (V.A.)
- Institut de Chimie Moléculaire (ICMUB), CNRS UMR6302, UBFC, F-21078 Dijon, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (C.C.); (F.C.); (M.T.-C.); (D.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (C.C.); (F.C.); (M.T.-C.); (D.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
- Centre Anticancéreux Georges François Leclerc, F-21000 Dijon, France
- Correspondence: ; Tel.: +33-380-39-32-26
| |
Collapse
|
45
|
Choi J, Lee DH, Jang H, Park SY, Seol JW. Naringenin exerts anticancer effects by inducing tumor cell death and inhibiting angiogenesis in malignant melanoma. Int J Med Sci 2020; 17:3049-3057. [PMID: 33173425 PMCID: PMC7646117 DOI: 10.7150/ijms.44804] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022] Open
Abstract
Malignant melanoma is one of the most deadly skin cancer, due to its aggressive proliferation and metastasis. Naringenin, abundantly present in citrus fruits, has widely studied in cancer therapy. In this study, we investigated whether naringenin also has anticancer effects against B16F10 murine and SK-MEL-28 human melanoma cells. Moreover, we assessed the effects of naringenin treatment on angiogenesis of HUVECs and ex vivo sprouting of microvessels.Naringenin inhibited tumor cell proliferation and migration in a dose-dependent manner in B16F10 and SK-MEL-28 cells, which is supported by the results that phosphorylation of ERK1/2 and JNK MAPK decreased. Furthermore, naringenin induced cell apoptosis. Western blot analysisshowed naringenin treatment significantly upregulated the protein expression of activated cas3 and PARP in B16F10 and SK-MEL-28 cells. In addition, in vitro and ex vivo angiogenesis assays demonstrated that naringenin treatment potently suppressed EC migration, tube formation, and sprouting of microvessels. RT-PCR analysis showed that naringenin treatment significantly reduced the mRNA expression of Tie2, but did not inhibit the expression of Ang2. In conclusion, present study demonstrates the anticancer effects of naringenin by its induction of tumor cell death and inhibition of angiogenesis in malignant melanoma, suggesting that naringenin has potential as a safe and effective therapeutic agent to treat melanoma.
Collapse
Affiliation(s)
- Jawun Choi
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Dae-Hyo Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Hyuk Jang
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Sang-Youel Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Jae-Won Seol
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| |
Collapse
|
46
|
Polyphenolic Composition and Anti-Melanoma Activity of White Forsythia ( Abeliophyllum distichum Nakai) Organ Extracts. PLANTS 2020; 9:plants9060757. [PMID: 32560393 PMCID: PMC7356668 DOI: 10.3390/plants9060757] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022]
Abstract
Abeliophyllum distichum Nakai, commonly called white forsythia, is a monotypic genus endemic to Korea. Although A. distichum is mainly used as an ornamental plant because of its horticultural value, recent studies have demonstrated its bioactivities, including antioxidant and anti-inflammatory activities, prompting us to investigate the potential anticancer effect of A. distichum organ extracts (leaves, fruit, and branches) against human melanoma SK-MEL-2 cells. The methanol extract of A. distichum leaves (AL) exhibited dose- and time-dependent cytotoxicities against SK-MEL-2 cells but not against HDFa human dermal fibroblasts. Based on high-performance liquid chromatography analysis, we identified 18 polyphenolic compounds from A. distichum organ extracts and suggest that differences in anticancer activity between organ extracts should be caused by different compositions of polyphenolic compounds. Additionally, the Annexin V/propidium iodide staining assay and analysis of caspase activity and expression indicated that AL induced cell death, including early and late apoptosis, as well as necrosis, by inducing the extrinsic pathway. Furthermore, we analyzed the differentially expressed genes between mock- and AL-treated cells using RNA-seq technology, suggesting that the anti-melanoma action of AL is mediated by down-regulation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Taken together, these results shed light on the potential use of A. distichum as a green resource with potent anti-melanoma activity.
Collapse
|
47
|
Cao S, Han Y, Li Q, Chen Y, Zhu D, Su Z, Guo H. Mapping Pharmacological Network of Multi-Targeting Litchi Ingredients in Cancer Therapeutics. Front Pharmacol 2020. [DOI: 10.3389/fphar.2020.00451
expr 967555229 + 995954239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
|
48
|
Cao S, Han Y, Li Q, Chen Y, Zhu D, Su Z, Guo H. Mapping Pharmacological Network of Multi-Targeting Litchi Ingredients in Cancer Therapeutics. Front Pharmacol 2020; 11:451. [PMID: 32390834 PMCID: PMC7193898 DOI: 10.3389/fphar.2020.00451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Considerable pharmacological studies have demonstrated that the extracts and ingredients from different parts (seeds, peels, pulps, and flowers) of Litchi exhibited anticancer effects by affecting the proliferation, apoptosis, autophagy, metastasis, chemotherapy and radiotherapy sensitivity, stemness, metabolism, angiogenesis, and immunity via multiple targeting. However, there is no systematical analysis on the interaction network of “multiple ingredients-multiple targets-multiple pathways” anticancer effects of Litchi. In this study, we summarized the confirmed anticancer ingredients and molecular targets of Litchi based on published articles and applied network pharmacology approach to explore the complex mechanisms underlying these effects from a perspective of system biology. The top ingredients, top targets, and top pathways of each anticancer function were identified using network pharmacology approach. Further intersecting analyses showed that Epigallocatechin gallate (EGCG), Gallic acid, Kaempferol, Luteolin, and Betulinic acid were the top ingredients which might be the key ingredients exerting anticancer function of Litchi, while BAX, BCL2, CASP3, and AKT1 were the top targets which might be the main targets underling the anticancer mechanisms of these top ingredients. These results provided references for further understanding and exploration of Litchi as therapeutics in cancer as well as the application of “Component Formula” based on Litchi’s effective ingredients.
Collapse
Affiliation(s)
- Sisi Cao
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yaoyao Han
- College of Pharmacy, Guangxi Medical University, Nanning, China.,Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| | - Qiaofeng Li
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China.,School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Yanjiang Chen
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia
| | - Dan Zhu
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Hongwei Guo
- College of Pharmacy, Guangxi Medical University, Nanning, China.,Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
49
|
Croitoru A, Ficai D, Craciun L, Ficai A, Andronescu E. Evaluation and Exploitation of Bioactive Compounds of Walnut, Juglans regia. Curr Pharm Des 2020; 25:119-131. [PMID: 30931854 DOI: 10.2174/1381612825666190329150825] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/22/2019] [Indexed: 12/12/2022]
Abstract
In the last few years, great importance has been given to natural materials (such as walnuts, peanuts, chestnuts) due to their medicinal and pharmaceutical uses induced by the presence of natural agents, including polyphenols. Juglans regia is a traditional plant that has been used since ancient times in traditional medicine for the treatment of various diseases like microbial infections, stomach ache, thyroid dysfunctions, cancer, heart diseases and sinusitis. Recently, scientific attention for the phytochemical profile of walnut by-products is increasing due to their valuable active constituents. Natural polyphenols are important compounds present in walnut with valuable properties that have been studied for the treatment of inflammation, cancer or anti-ageing effect. The use of nanocarriers as a drug delivery system is now a promising strategy to get more stable products and is easier to apply in a medical, therapeutic and pharmaceutical environment. The aim of this work was to review the latest information provided by scientific investigators regarding the nutritional value, bioactive compounds, antioxidant and antitumor activity of walnut by-product extracts. Moreover, this review provides comprehensive information on the nanoencapsulation of bioactive constituents for application in clinical medicine, particularly in cancer research.
Collapse
Affiliation(s)
- Alexa Croitoru
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh Polizu St 1-7, 011061 Bucharest, Romania
| | - Denisa Ficai
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh Polizu St 1-7, 011061 Bucharest, Romania
| | - Luminiţa Craciun
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh Polizu St 1-7, 011061 Bucharest, Romania
| | - Anton Ficai
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh Polizu St 1-7, 011061 Bucharest, Romania.,Academy of Romanian Scientists, Spl. Independenţei 54, Bucharest, Romania
| | - Ecaterina Andronescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh Polizu St 1-7, 011061 Bucharest, Romania.,Academy of Romanian Scientists, Spl. Independenţei 54, Bucharest, Romania
| |
Collapse
|
50
|
Therapeutic Potential of Plant Phenolic Acids in the Treatment of Cancer. Biomolecules 2020; 10:biom10020221. [PMID: 32028623 PMCID: PMC7072661 DOI: 10.3390/biom10020221] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/26/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
Globally, cancer is the second leading cause of death. Different conventional approaches to treat cancer include chemotherapy or radiotherapy. However, these are usually associated with various deleterious effects and numerous disadvantages in clinical practice. In addition, there are increasing concerns about drug resistance. In the continuous search for safer and more effective treatments, plant-derived natural compounds are of major interest. Plant phenolics are secondary metabolites that have gained importance as potential anti-cancer compounds. Phenolics display a great prospective as cytotoxic anti-cancer agents promoting apoptosis, reducing proliferation, and targeting various aspects of cancer (angiogenesis, growth and differentiation, and metastasis). Phenolic acids are a subclass of plant phenolics, furtherly divided into benzoic and cinnamic acids, that are associated with potent anticancer abilities in various in vitro and in vivo studies. Moreover, the therapeutic activities of phenolic acids are reinforced by their role as epigenetic regulators as well as supporters of adverse events or resistance associated with conventional anticancer therapy. Encapsulation of phyto-substances into nanocarrier systems is a challenging aspect concerning the efficiency of natural substances used in cancer treatment. A summary of phenolic acids and their effectiveness as well as phenolic-associated advances in cancer treatment will be discussed in this review.
Collapse
|