1
|
Joković N, Pešić S, Vitorović J, Bogdanović A, Sharifi-Rad J, Calina D. Glucosinolates and Their Hydrolytic Derivatives: Promising Phytochemicals With Anticancer Potential. Phytother Res 2025; 39:1035-1089. [PMID: 39726346 DOI: 10.1002/ptr.8419] [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: 06/19/2024] [Revised: 10/29/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Recent research has increasingly focused on phytochemicals as promising anticancer agents, with glucosinolates (GSLs) and their hydrolytic derivatives playing a central role. These sulfur-containing compounds, found in plants of the Brassicales order, are converted by myrosinase enzymes into biologically active products, primarily isothiocyanates (ITCs) and indoles, which exhibit significant anticancer properties. Indole-3-carbinol, diindolylmethane, sulforaphane (SFN), phenethyl isothiocyanate (PEITC), benzyl isothiocyanate, and allyl isothiocyanate have shown potent anticancer effects in animal models, particularly in breast, prostate, lung, melanoma, bladder, hepatoma, and gastrointestinal cancers. Clinical studies further support the chemopreventive effects of SFN and PEITC, particularly in detoxifying carcinogens and altering biochemical markers in cancer patients. These compounds have demonstrated good bioavailability, low toxicity, and minimal adverse effects, supporting their potential therapeutic application. Their anticancer mechanisms include the modulation of reactive oxygen species, suppression of cancer-related signaling pathways, and direct interaction with tumor cell proteins. Additionally, semi-synthetic derivatives of GSLs have been developed to enhance anticancer efficacy. In conclusion, GSLs and their derivatives offer significant potential as both chemopreventive and therapeutic agents, warranting further clinical investigation to optimize their application in cancer treatment.
Collapse
Affiliation(s)
- Nataša Joković
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Strahinja Pešić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Jelena Vitorović
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Andrija Bogdanović
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador
- Department of Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
2
|
Zhang Y, Zhao X, Liu Y, Yang X. Sulforaphane and ophthalmic diseases. Food Sci Nutr 2024; 12:5296-5311. [PMID: 39139965 PMCID: PMC11317731 DOI: 10.1002/fsn3.4230] [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: 11/08/2023] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 08/15/2024] Open
Abstract
Sulforaphane (SFN) is an organosulfur compound categorized as an isothiocyanate (ITC), primarily extracted from cruciferous vegetables like broccoli and cabbage. The molecular formula of sulforaphane (SFN) is C6H11NOS2. SFN is generated by the hydrolysis of glucoraphanin (GRP) through the enzyme myrosinase, showing notable properties including anti-diabetic, anti-inflammatory, antimicrobial, anti-angiogenic, and anticancer attributes. Ongoing clinical trials are investigating its potential in diseases such as cancer, neurodegenerative diseases, diabetes-related complications, chronic kidney disease, cardiovascular disease, and liver diseases. Several animal carcinogenesis models and cell culture models have shown it to be a very effective chemopreventive agent, and the protective effects of SFN in ophthalmic diseases have been linked to multiple mechanisms. In murine models of diabetic retinopathy and age-related macular degeneration, SFN delays retinal photoreceptor cell degeneration through the Nrf2 antioxidative pathway, NF-κB pathway, AMPK pathway, and Txnip/mTOR pathway. In rabbit models of keratoconus and cataract, SFN has been shown to protect corneal and lens epithelial cells from oxidative stress injury by activating the Keap1-Nrf2-ARE pathway and the Nrf-2/HO-1 antioxidant pathway. Oral delivery or intraperitoneal injection at varying concentrations are the primary strategies for SFN intake in current preclinical studies. Challenges remain in the application of SFN in eye disorders due to its weak solubility in water and limited bioavailability because of the presence of blood-ocular barrier systems. This review comprehensively outlines recent research on SFN, elucidates its mechanisms of action, and discusses potential therapeutic benefits for eye disorders such as age-related macular degeneration (AMD), diabetic retinopathy (DR), cataracts, and other ophthalmic diseases, while also indicating directions for future clinical research to achieve efficient SFN treatment for ophthalmic diseases.
Collapse
Affiliation(s)
- Yichi Zhang
- Department of OphthalmologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Xiaojing Zhao
- Department of OphthalmologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Yang Liu
- Department of OphthalmologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Xiuxia Yang
- Department of OphthalmologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| |
Collapse
|
3
|
Camero S, Milazzo L, Vulcano F, Ceccarelli F, Pontecorvi P, Pedini F, Rossetti A, Scialis ES, Gerini G, Cece F, Pomella S, Cassandri M, Porrazzo A, Romano E, Festuccia C, Gravina GL, Ceccarelli S, Rota R, Lotti LV, Midulla F, Angeloni A, Marchese C, Marampon F, Megiorni F. Antitumour effects of SFX-01 molecule in combination with ionizing radiation in preclinical and in vivo models of rhabdomyosarcoma. BMC Cancer 2024; 24:814. [PMID: 38977944 PMCID: PMC11229215 DOI: 10.1186/s12885-024-12536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/18/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Despite a multimodal approach including surgery, chemo- and radiotherapy, the 5-year event-free survival rate for rhabdomyosarcoma (RMS), the most common soft tissue sarcoma in childhood, remains very poor for metastatic patients, mainly due to the selection and proliferation of tumour cells driving resistance mechanisms. Personalised medicine-based protocols using new drugs or targeted therapies in combination with conventional treatments have the potential to enhance the therapeutic effects, while minimizing damage to healthy tissues in a wide range of human malignancies, with several clinical trials being started. In this study, we analysed, for the first time, the antitumour activity of SFX-01, a complex of synthetic d, l-sulforaphane stabilised in alpha-cyclodextrin (Evgen Pharma plc, UK), used as single agent and in combination with irradiation, in four preclinical models of alveolar and embryonal RMS. Indeed, SFX-01 has shown promise in preclinical studies for its ability to modulate cellular pathways involved in inflammation and oxidative stress that are essential to be controlled in cancer treatment. METHODS RH30, RH4 (alveolar RMS), RD and JR1 (embryonal RMS) cell lines as well as mouse xenograft models of RMS were used to evaluate the biological and molecular effects induced by SFX-01 treatment. Flow cytometry and the modulation of key markers analysed by q-PCR and Western blot were used to assess cell proliferation, apoptosis, autophagy and production of intracellular reactive oxygen species (ROS) in RMS cells exposed to SFX-01. The ability to migrate and invade was also investigated with specific assays. The possible synergistic effects between SFX-01 and ionising radiation (IR) was studied in both the in vitro and in vivo studies. Student's t-test or two-way ANOVA were used to test the statistical significance of two or more comparisons, respectively. RESULTS SFX-01 treatment exhibited cytostatic and cytotoxic effects, mediated by G2 cell cycle arrest, apoptosis induction and suppression of autophagy. Moreover, SFX-01 was able to inhibit the formation and the proliferation of 3D tumorspheres as monotherapy and in combination with IR. Finally, SFX-01, when orally administered as single agent, displayed a pattern of efficacy at reducing the growth of tumour masses in RMS xenograft mouse models; when combined with a radiotherapy regime, it was observed to act synergistically, resulting in a more positive outcome than would be expected by adding each exposure alone. CONCLUSIONS In summary, our results provide evidence for the antitumour properties of SFX-01 in preclinical models of RMS tumours, both as a standalone treatment and in combination with irradiation. These forthcoming findings are crucial for deeper investigations of SFX-01 molecular mechanisms against RMS and for setting up clinical trials in RMS patients in order to use the SFX-01/IR co-treatment as a promising therapeutic approach, particularly in the clinical management of aggressive RMS disease.
Collapse
Affiliation(s)
- Simona Camero
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Luisa Milazzo
- Department of Oncology and Molecular Medicine, Italian National Institute of Health (ISS), Rome, Italy
| | - Francesca Vulcano
- Department of Oncology and Molecular Medicine, Italian National Institute of Health (ISS), Rome, Italy
| | - Federica Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Paola Pontecorvi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Francesca Pedini
- Department of Oncology and Molecular Medicine, Italian National Institute of Health (ISS), Rome, Italy
| | - Alessandra Rossetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elena Sofia Scialis
- Department of Innovative Technologies in Medicine and Dentistry, University "G. D'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Giulia Gerini
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Fabrizio Cece
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Silvia Pomella
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Department of Oncohematology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Matteo Cassandri
- Department of Oncohematology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Antonella Porrazzo
- Department of Oncohematology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Enrico Romano
- Department of Sense Organs, "Sapienza" University of Rome, Rome, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simona Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Rossella Rota
- Department of Oncohematology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Fabio Midulla
- Department of Maternal Infantile and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
4
|
Liu P, Zhang B, Li Y, Yuan Q. Potential mechanisms of cancer prevention and treatment by sulforaphane, a natural small molecule compound of plant-derived. Mol Med 2024; 30:94. [PMID: 38902597 PMCID: PMC11191161 DOI: 10.1186/s10020-024-00842-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Despite recent advances in tumor diagnosis and treatment technologies, the number of cancer cases and deaths worldwide continues to increase yearly, creating an urgent need to find new methods to prevent or treat cancer. Sulforaphane (SFN), as a member of the isothiocyanates (ITCs) family, which is the hydrolysis product of glucosinolates (GLs), has been shown to have significant preventive and therapeutic cancer effects in different human cancers. Early studies have shown that SFN scavenges oxygen radicals by increasing cellular defenses against oxidative damage, mainly through the induction of phase II detoxification enzymes by nuclear factor erythroid 2-related factor 2 (Nrf2). More and more studies have shown that the anticancer mechanism of SFN also includes induction of apoptotic pathway in tumor cells, inhibition of cell cycle progression, and suppression of tumor stem cells. Therefore, the application of SFN is expected to be a necessary new approach to treating cancer. In this paper, we review the multiple molecular mechanisms of SFN in cancer prevention and treatment in recent years, which can provide a new vision for cancer treatment.
Collapse
Affiliation(s)
- Pengtao Liu
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Bo Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Yuanqiang Li
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China.
| |
Collapse
|
5
|
Baralić K, Živanović J, Marić Đ, Bozic D, Grahovac L, Antonijević Miljaković E, Ćurčić M, Buha Djordjevic A, Bulat Z, Antonijević B, Đukić-Ćosić D. Sulforaphane-A Compound with Potential Health Benefits for Disease Prevention and Treatment: Insights from Pharmacological and Toxicological Experimental Studies. Antioxidants (Basel) 2024; 13:147. [PMID: 38397745 PMCID: PMC10886109 DOI: 10.3390/antiox13020147] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/31/2023] [Accepted: 01/11/2024] [Indexed: 02/25/2024] Open
Abstract
Sulforaphane (SFN), which is a hydrolysis product from glucoraphanin, a compound found in cruciferous vegetables, has been studied for its potential health benefits, particularly in disease prevention and treatment. SFN has proven to be effective in combating different types of cancer by inhibiting the proliferation of tumors and triggering apoptosis. This dual action has been demonstrated to result in a reduction in tumor size and an enhancement of survival rates in animal models. SFN has also shown antidiabetic and anti-obesity effects, improving glucose tolerance and reducing fat accumulation. SFN's ability to activate Nrf2, a transcription factor regulating oxidative stress and inflammation in cells, is a primary mechanism behind its anticancerogenic and antidiabetic effects. Its antioxidant, anti-inflammatory, and anti-apoptotic properties are also suggested to provide beneficial effects against neurodegenerative diseases. The potential health benefits of SFN have led to increased interest in its use as a dietary supplement or adjunct to chemotherapy, but there are insufficient data on its efficacy and optimal doses, as well as its safety. This review aims to present and discuss SFN's potential in treating various diseases, such as cancer, diabetes, cardiovascular diseases, obesity, and neurodegenerative diseases, focusing on its mechanisms of action. It also summarizes studies on the pharmacological and toxicological potential of SFN in in vitro and animal models and explores its protective role against toxic compounds through in vitro and animal studies.
Collapse
Affiliation(s)
- Katarina Baralić
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (J.Ž.); (Đ.M.); (D.B.); (L.G.); (E.A.M.); (M.Ć.); (A.B.D.); (Z.B.); (B.A.); (D.Đ.-Ć.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Ma C, Gu C, Lian P, Wazir J, Lu R, Ruan B, Wei L, Li L, Pu W, Peng Z, Wang W, Zong Y, Huang Z, Wang H, Lu Y, Su Z. Sulforaphane alleviates psoriasis by enhancing antioxidant defense through KEAP1-NRF2 Pathway activation and attenuating inflammatory signaling. Cell Death Dis 2023; 14:768. [PMID: 38007430 PMCID: PMC10676357 DOI: 10.1038/s41419-023-06234-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 11/27/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease that affects millions of people worldwide. Sulforaphane (SFN) has been shown to have anti-inflammatory and antioxidant properties. In this study, we investigated the effects of SFN on a mouse model of psoriasis induced by imiquimod (IMQ) and its underlying molecular mechanism. Mice treated with SFN showed significant improvement in psoriatic symptoms, including reduced erythema, scales, and cutaneous thickness. Histopathological analysis and immunohistochemical staining revealed decreased expression of K16, K17, and Ki67 in SFN-treated mice, indicating reduced abnormal differentiation of keratinocytes and cutaneous inflammation. SFN treatment also reduced the activation of STAT3 and NF-κB pathways and downregulated pro-inflammatory cytokines IL-1β, IL-6, and CCL2. In vitro experiments using HaCaT cells demonstrated that SFN inhibited IL-22 and TNF-α-induced activation of inflammatory pathways and keratinocyte proliferation. Network pharmacology analysis suggested that the KEAP1-NRF2 pathway might be involved in the protective effects of SFN on psoriasis. We observed reduced NRF2 expression in human psoriatic lesions, and subsequent experiments showed that SFN activated KEAP1-NRF2 pathway in vivo and in vitro. Importantly, NRF2-deficient mice exhibited aggravated psoriasis-like symptoms and reduced response to SFN treatment. Our findings indicate that SFN ameliorates psoriasis symptoms and inflammation through the KEAP1-NRF2 pathway, suggesting a potential therapeutic role for SFN in the treatment of psoriasis.
Collapse
Affiliation(s)
- Chujun Ma
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
- Department of Dermatology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China
| | - Chaode Gu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Panpan Lian
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Renwei Lu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Binjia Ruan
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Ziqi Peng
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Wentong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Yangyongyi Zong
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China.
| | - Yan Lu
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Zhonglan Su
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
7
|
Vivanco PG, Taboada P, Coelho A. The Southern European Atlantic Diet and Its Supplements: The Chemical Bases of Its Anticancer Properties. Nutrients 2023; 15:4274. [PMID: 37836558 PMCID: PMC10574233 DOI: 10.3390/nu15194274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Scientific evidence increasingly supports the strong link between diet and health, acknowledging that a well-balanced diet plays a crucial role in preventing chronic diseases such as obesity, diabetes, cardiovascular issues, and certain types of cancer. This perspective opens the door to developing precision diets, particularly tailored for individuals at risk of developing cancer. It encompasses a vast research area and involves the study of an expanding array of compounds with multilevel "omics" compositions, including genomics, transcriptomics, proteomics, epigenomics, miRNomics, and metabolomics. We review here the components of the Southern European Atlantic Diet (SEAD) from both a chemical and pharmacological standpoint. The information sources consulted, complemented by crystallographic data from the Protein Data Bank, establish a direct link between the SEAD and its anticancer properties. The data collected strongly suggest that SEAD offers an exceptionally healthy profile, particularly due to the presence of beneficial biomolecules in its foods. The inclusion of olive oil and paprika in this diet provides numerous health benefits, and scientific evidence supports the anticancer properties of dietary supplements with biomolecules sourced from vegetables of the brassica genus. Nonetheless, further research is warranted in this field to gain deeper insights into the potential benefits of the SEAD's bioactive compounds against cancer.
Collapse
Affiliation(s)
- Pablo García Vivanco
- Spanish Academy of Nutrition and Dietetics, 31006 Pamplona, Spain
- Nutrition and Digestive Working Group, Spanish Society of Clinical, Family, and Community Pharmacy (SEFAC), 28045 Madrid, Spain
| | - Pablo Taboada
- Department of Condensed Matter Physics, Faculty of Physics, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Institute of Materials-USC (IMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Alberto Coelho
- Institute of Materials-USC (IMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
8
|
Ashrafizadeh M, Mohan CD, Rangappa S, Zarrabi A, Hushmandi K, Kumar AP, Sethi G, Rangappa KS. Noncoding RNAs as regulators of STAT3 pathway in gastrointestinal cancers: Roles in cancer progression and therapeutic response. Med Res Rev 2023; 43:1263-1321. [PMID: 36951271 DOI: 10.1002/med.21950] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/09/2022] [Accepted: 02/28/2023] [Indexed: 03/24/2023]
Abstract
Gastrointestinal (GI) tumors (cancers of the esophagus, gastric, liver, pancreas, colon, and rectum) contribute to a large number of deaths worldwide. STAT3 is an oncogenic transcription factor that promotes the transcription of genes associated with proliferation, antiapoptosis, survival, and metastasis. STAT3 is overactivated in many human malignancies including GI tumors which accelerates tumor progression, metastasis, and drug resistance. Research in recent years demonstrated that noncoding RNAs (ncRNAs) play a major role in the regulation of many signaling pathways including the STAT3 pathway. The major types of endogenous ncRNAs that are being extensively studied in oncology are microRNAs, long noncoding RNAs, and circular RNAs. These ncRNAs can either be tumor-promoters or tumor-suppressors and each one of them imparts their activity via different mechanisms. The STAT3 pathway is also tightly modulated by ncRNAs. In this article, we have elaborated on the tumor-promoting role of STAT3 signaling in GI tumors. Subsequently, we have comprehensively discussed the oncogenic as well as tumor suppressor functions and mechanism of action of ncRNAs that are known to modulate STAT3 signaling in GI cancers.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chakrabhavi D Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, India
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri University, Nagamangala Taluk, India
| | - Ali Zarrabi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Sariyer, Turkey
| | - Kiavash Hushmandi
- Division of Epidemiology, Faculty of Veterinary Medicine, Department of Food Hygiene and Quality Control, University of Tehran, Tehran, Iran
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
9
|
Asif Ali M, Khan N, Kaleem N, Ahmad W, Alharethi SH, Alharbi B, Alhassan HH, Al-Enazi MM, Razis AFA, Modu B, Calina D, Sharifi-Rad J. Anticancer properties of sulforaphane: current insights at the molecular level. Front Oncol 2023; 13:1168321. [PMID: 37397365 PMCID: PMC10313060 DOI: 10.3389/fonc.2023.1168321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Abstract
Sulforaphane (SFN) is an isothiocyanate with multiple biomedical applications. Sulforaphane can be extracted from the plants of the genus Brassica. However, broccoli sprouts are the chief source of sulforaphane and are 20 to 50 times richer than mature broccoli as they contain 1,153 mg/100 g. SFN is a secondary metabolite that is produced as a result of the hydrolysis of glucoraphanin (a glucosinolate) by the enzyme myrosinase. This review paper aims to summarize and understand the mechanisms behind the anticancer potential of sulforaphane. The data was collected by searching PubMed/MedLine, Scopus, Web of Science, and Google Scholar. This paper concludes that sulforaphane provides cancer protection through the alteration of various epigenetic and non-epigenetic pathways. It is a potent anticancer phytochemical that is safe to consume with minimal side effects. However, there is still a need for further research regarding SFN and the development of a standard dose.
Collapse
Affiliation(s)
- Muhammad Asif Ali
- Department of Food Science and Human Nutrition, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Noohela Khan
- Department of Nutrition Sciences, Rashid Latif Medical College, Lahore, Pakistan
| | - Nabeeha Kaleem
- Department of Food Science and Human Nutrition, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Waqas Ahmad
- Department of Food Science and Human Nutrition, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudi Arabia
| | - Bandar Alharbi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail, Saudi Arabia
| | - Hassan H. Alhassan
- Department of Clinical Laboratory Science, College of Applied medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Maher M. Al-Enazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Babagana Modu
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Science, University of Maiduguri, Maiduguri, Borno, Nigeria
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | |
Collapse
|
10
|
Wu X, Zhang J, Zhang X, Xiang M, Xu Z, Cao Z. Prognostic value of miR-219-5p in relation to mortality in patients with small cell lung cancer: a retrospective, observational cohort study in China. BMJ Open 2023; 13:e064700. [PMID: 36997257 PMCID: PMC10069522 DOI: 10.1136/bmjopen-2022-064700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
OBJECTIVES Small cell lung cancer (SCLC) is a lethal human malignancy, and previous studies support the contribution of microRNA to cancer progression. The prognostic value of miR-219-5p in patients with SCLC remains unclear. This study aimed to evaluate the predictive value of miR-219-5p with respect to mortality in patients with SCLC and to incorporate miR-219-5p level into a prediction model and nomogram for mortality. DESIGN Retrospective observational cohort study. SETTING AND PARTICIPANTS Our main cohort included data from 133 patients with SCLC between 1 March 2010 and 1 June 2015 from the Suzhou Xiangcheng People's Hospital. Data from 86 patients with non-SCLC at Sichuan Cancer Hospital and the First Affiliated Hospital of Soochow University were used for external validation. OUTCOME MEASURES Tissue samples were taken during admission and stored, and miR-219-5p levels were measured at a later date. A Cox proportional hazard model was used for survival analyses and for analysing risk factors to create a nomogram for mortality prediction. The accuracy of the model was evaluated by C-index and calibration curve. RESULTS Mortality in patients with a high level of miR-219-5p (≥1.50) (n=67) was 74.6%, while mortality in the low-level group (n=66) was 100.0%. Based on univariate analysis, we included significant factors (p<0.05) in a multivariate regression model: patients with high level of miR-219-5p (HR 0.39, 95% CI 0.26-0.59, p<0.001), immunotherapy (HR 0.44, 95% CI 0.23-0.84, p<0.001) and prognostic nutritional index score >47.9 (HR=0.45, 95% CI 0.24-0.83, p=0.01) remained statistically significant factors for improved overall survival. The nomogram had good accuracy in estimating the risk, with a bootstrap-corrected C-index of 0.691. External validation indicated an area under the curve of 0.749 (0.709-0.788). CONCLUSIONS The miR-219-5p level was associated with a reduced risk of mortality in patients with SCLC. A nomogram incorporating MiR-219-5p level and clinical factors demonstrated good accuracy in estimating the risk of overall mortality. Prospective validation of the prognostic nomogram is needed.
Collapse
Affiliation(s)
- Xiangmei Wu
- Endocrinology, Suzhou Xiangcheng People's Hospital, Suzhou, Jiangsu, China
| | - Jigang Zhang
- Traumatology Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaohui Zhang
- Medicine, Respiratory, Emergency and Intensive Care Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mengqi Xiang
- Medical Oncology, Medical School of University of Electronic Science and Technology of China, Chengdu, China
| | - Zhihua Xu
- General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhijun Cao
- Urology, Suzhou Ninth People's Hospital, Suzhou, Jiangsu, China
| |
Collapse
|
11
|
Adelakun SA, Akintunde OW, Ogunlade B, Adeyeluwa BE. Histochemical and histomorphological evidence of the modulating role of 1-isothiocyanate-4-methyl sulfonyl butane on cisplatin-induced testicular-pituitary axis degeneration and cholesterol homeostasis in male Sprague-Dawley rats. Morphologie 2023; 107:80-98. [PMID: 35659716 DOI: 10.1016/j.morpho.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND This study examine the histochemical and histomorphological effect of 1-isothiocyanato-4-methyl sulfonyl butane (SFN) on cisplatin (CP) induced testicular alteration and cholesterol homeostasis. MATERIALS AND METHODS Ninety adult-male Sprague-Dawley rats were randomized into nine groups of ten (n=10) rats each. Group A (control) received normal saline, group B received a single dose of 10mg/Kg body weight (bwt) CP (i.p.), group C received 50mg/Kg bwt of SFN, group D received 100mg/Kg bwt of SFN, group E received 10mg/Kg bwt CP and 50mg/Kg bwt of SFN, group F received 10mg/Kg bwt CP and 100mg/Kg bwt of SFN, group G received 10mg/Kg bwt CP and 50mg/Kg bwt vitamin C, group H received 50mg/Kg bwt of SFN and 10mg/Kg bwt CP, group I received 100mg/Kg bwt of SFN and 10mg/Kg bwt CP. The procedure lasted for 56 days. Testicular histomorphology and histochemistry, testicular testosterone, sperm parameters, total antioxidant status (TSA), total oxidant status (TOS), oxidative stress index (OSI), and serum lipid profile were examined. RESULTS Cisplatin decrease intra-testicular testosterone, sperm quality, and expression of glycogen and increases testicular TOS and OSI, serum lipid profile, collagen, and disruption of germinal epithelium. However, the intervention of SFN reversed the effect of CP on testes' weight and volume, DSP, ESP, testosterone production, TAS, TOS, and OSI. Histoarchitectecture showing normal seminiferous tubules and even distribution of glycogen and collagen fibers. CONCLUSION Treatment with SFN ameliorate CP-induced testicular toxicity by reversing the cytotoxic mechanisms of CP.
Collapse
Affiliation(s)
- S A Adelakun
- Department of Human Anatomy, College of Health Sciences, Federal University of Technology, Akure, Nigeria; Department of Anatomy, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - O W Akintunde
- Department of Anatomy, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| | - B Ogunlade
- Department of Human Anatomy, College of Health Sciences, Federal University of Technology, Akure, Nigeria
| | - B E Adeyeluwa
- Department of Human Anatomy, College of Health Sciences, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
12
|
Zhang Y, Ai P, Chen SZ, Lei SY. Sulforaphane suppresses skin squamous cell carcinoma cells proliferation through miR-199a-5p/Sirt1/CD44ICD signaling pathway. Immunopharmacol Immunotoxicol 2023; 45:52-60. [PMID: 35947042 DOI: 10.1080/08923973.2022.2112221] [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: 01/24/2023]
Abstract
BACKGROUND The present study aimed to explore the impact of sulforaphane on the growth of sSCC cells, and the activation of miR-199a-5p/Sirt1 and CD44ICD signaling pathways. METHODS Cell viability, count, apoptosis, and invasion assays were performed in the sSCC cell line (SCC-13) in which miR-199a-5p was over-expressed or under-expressed. The expression levels of miR-199a-5p, Sirt1 and CD44ICD mRNA were measured by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Sulforaphane significantly inhibited the cell growth and invasion of SCC-13 cells, and dramatically induced cell apoptosis. Additionally, sulforaphane also greatly increased miR-199a-5p expression and suppressed Sirt1 and CD44ICD mRNA levels. Moreover, miR-199a-5p overexpression considerably down-regulated the expressions of Sirt1 and CD44ICD mRNA, and promoted the ability of sulforaphane to represses cell growth and invasion, and to induce cell apoptosis. However, miR-199a-5p underexpression has the opposite effects. CONCLUSIONS Sulforaphane appears to inhibit sCC progression by impacting its growth and invasion ability, and regulates miR-199a-5p/Sirt1 and CD44ICD signaling pathways, and may be utilized to develop a curative approach for sSCC.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Dermatology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, PR China
| | - Ping Ai
- Department of Dermatology, Minda Hospital of Hubei Minzu University, Enshi, PR China
| | - Shang-Zhou Chen
- Department of Dermatology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, PR China
| | - Shu-Ying Lei
- Department of Dermatology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, PR China
| |
Collapse
|
13
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Bradu P, Sukumar A, Patil M, Renu K, Dey A, Vellingiri B, George A, Ganesan R. Implications of cancer stem cells in diabetes and pancreatic cancer. Life Sci 2022; 312:121211. [PMID: 36414089 DOI: 10.1016/j.lfs.2022.121211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
This review provides a detailed study of pancreatic cancer (PC) and the implication of different types of cancers concerning diabetes. The combination of anti-diabetic drugs with other anti-cancer drugs and phytochemicals can help prevent and treat this disease. PC cancer stem cells (CSCs) and how they migrate and develop into malignant tumors are discussed. A detailed explanation of the different mechanisms of diabetes development, which can enhance the pancreatic CSCs' proliferation by increasing the IGF factor levels, epigenetic modifications, DNA damage, and the influence of lifestyle factors like obesity, and inflammation, has been discussed. It also explains how cancer due to diabetes is associated with high mortality rates. One of the well-known diabetic drugs, metformin, can be combined with other anti-cancer drugs and prevent the development of PC and has been taken as one of the prime focus in this review. Overall, this paper provides insight into the relationship between diabetes and PC and the methods that can be employed to diagnose this disease at an earlier stage successfully.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Pragya Bradu
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Aarthi Sukumar
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Megha Patil
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda - 151401, Punjab, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, 680005, Kerala, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24252, Republic of Korea
| |
Collapse
|
14
|
Tabnak P, Masrouri S, Mafakheri A. Natural products in suppressing glioma progression: A focus on the role of microRNAs. Phytother Res 2022; 36:1576-1599. [PMID: 35174549 DOI: 10.1002/ptr.7414] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/07/2021] [Accepted: 01/29/2022] [Indexed: 11/06/2022]
Abstract
Glioma is one of the most common malignancies of the central nervous system. Due to inadequate response to the current treatments available, glioma has been at the center of recent cancer studies searching for novel treatment strategies. This has prompted an intensive search using linkage studies and preliminary evidence to gain efficient insight into the mechanisms involved in the alleviation of the pathogenesis of glioma mediated by miRNAs, a group of noncoding RNAs that affect gene expression posttranscriptionally. Dysregulated expression of miRNAs can exacerbate the malignant features of tumor cells in glioma and other cancers. Natural products can exert anticancer effects on glioma cells by stimulating the expression levels of tumor suppressor miRNAs and repressing the expression levels of oncogenic miRNAs. In this review, we aimed to collect and analyze the literature addressing the roles of natural products in the treatment of glioma, with an emphasis on their involvement in the regulation of miRNAs.
Collapse
Affiliation(s)
- Peyman Tabnak
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Masrouri
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asrin Mafakheri
- Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
15
|
Chen Y, Tang L, Ye X, Chen Y, Shan E, Han H, Zhong C. Regulation of ZO-1 on β-catenin mediates sulforaphane suppressed colorectal cancer stem cell properties in colorectal cancer. Food Funct 2022; 13:12363-12370. [DOI: 10.1039/d2fo02932d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
ZO-1 suppresses colorectal CSCs by interacting with β-Catenin and attenuating its nuclear translocation.
Collapse
Affiliation(s)
- Yue Chen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - LvYuwei Tang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinrong Ye
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - Yimeng Chen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - Enfang Shan
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - Hongyu Han
- Department of Clinical Nutrition, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Sulforaphane: A Broccoli Bioactive Phytocompound with Cancer Preventive Potential. Cancers (Basel) 2021; 13:cancers13194796. [PMID: 34638282 PMCID: PMC8508555 DOI: 10.3390/cancers13194796] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary As of the past decade, phytochemicals have become a major target of interest in cancer chemopreventive and chemotherapeutic research. Sulforaphane (SFN) is a metabolite of the phytochemical glucoraphanin, which is found in high abundance in cruciferous vegetables, such as broccoli, watercress, Brussels sprouts, and cabbage. In both distant and recent research, SFN has been shown to have a multitude of anticancer effects, increasing the need for a comprehensive review of the literature. In this review, we critically evaluate SFN as an anticancer agent and its mechanisms of action based on an impressive number of in vitro, in vivo, and clinical studies. Abstract There is substantial and promising evidence on the health benefits of consuming broccoli and other cruciferous vegetables. The most important compound in broccoli, glucoraphanin, is metabolized to SFN by the thioglucosidase enzyme myrosinase. SFN is the major mediator of the health benefits that have been recognized for broccoli consumption. SFN represents a phytochemical of high interest as it may be useful in preventing the occurrence and/or mitigating the progression of cancer. Although several prior publications provide an excellent overview of the effect of SFN in cancer, these reports represent narrative reviews that focused mainly on SFN’s source, biosynthesis, and mechanisms of action in modulating specific pathways involved in cancer without a comprehensive review of SFN’s role or value for prevention of various human malignancies. This review evaluates the most recent state of knowledge concerning SFN’s efficacy in preventing or reversing a variety of neoplasms. In this work, we have analyzed published reports based on in vitro, in vivo, and clinical studies to determine SFN’s potential as a chemopreventive agent. Furthermore, we have discussed the current limitations and challenges associated with SFN research and suggested future research directions before broccoli-derived products, especially SFN, can be used for human cancer prevention and intervention.
Collapse
|
17
|
Iahtisham-Ul-Haq, Khan S, Awan KA, Iqbal MJ. Sulforaphane as a potential remedy against cancer: Comprehensive mechanistic review. J Food Biochem 2021; 46:e13886. [PMID: 34350614 DOI: 10.1111/jfbc.13886] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/30/2021] [Accepted: 07/14/2021] [Indexed: 12/21/2022]
Abstract
Sulforaphane belongs to the active class of isothiocyanates capable of delivering various biological benefits for health promotion and disease prevention. This compound is considered vital to curtail numerous metabolic disorders. Various studies have proven its beneficial effects against cancer prevention and its possible utilization as a therapeutic agent in cancer treatment. Understanding the mechanistic pathways and possible interactions at cellular and subcellular levels is key to design and develop cancer therapeutics for humans. In this respect, a number of mechanisms such as modulation of carcinogen metabolism & phase II enzymatic activities, cell cycle arrest, activation of Nrf2, cytotoxic, proapoptotic and apoptotic pathways have been reported to be involved in cancer prevention. This article provides sufficient information by critical analysis to understand the mechanisms involved in cancer prevention attributed to sulforaphane. Furthermore, various clinical studies have also been included for design and development of novel therapies for cancer prevention and cure. PRACTICAL APPLICATIONS: Diet and dietary components are potential tools to address various lifestyle-related disorders. Due to plenty of environmental and cellular toxicants, the chances of cancer prevalence are quite large which are worsen by adopting unhealthy lifestyles. Cancer can be treated with various therapies but those are acquiring side effects causing the patients to suffer the treatment regime. Nutraceuticals and functional foods provide safer options to prevent or delay the onset of cancer. In this regard, sulforaphane is a pivotal compound to be targeted as a potential agent for cancer treatment both in preventive and therapeutic regimes. This article provides sufficient evidence via discussing the underlying mechanisms of positive effects of sulforaphane to further the research for developing anticancer drugs that will help assuage this lethal morbidity.
Collapse
Affiliation(s)
- Iahtisham-Ul-Haq
- School of Food and Nutrition, Faculty of Allied Health Sciences, Minhaj University, Lahore, Pakistan
| | - Sipper Khan
- Institute of Agricultural Engineering, Tropics and Subtropics Group, University of Hohenheim, Stuttgart, Germany
| | - Kanza Aziz Awan
- Department of Food Science and Technology, Faculty of Life Sciences, University of Central Punjab, Lahore, Pakistan
| | | |
Collapse
|
18
|
Han S, Wang Z, Liu J, Wang HMD, Yuan Q. miR-29a-3p-dependent COL3A1 and COL5A1 expression reduction assists sulforaphane to inhibit gastric cancer progression. Biochem Pharmacol 2021; 188:114539. [PMID: 33819468 DOI: 10.1016/j.bcp.2021.114539] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 01/22/2023]
Abstract
The antitumor properties of cruciferous vegetables are mainly due to their high content of isothiocyanates, and sulforaphane (SFA) is the most well-known compound. The aim of this study was to determine the mechanism of SFA inhibiting gastric cancer (GC) progression. After verifying SFA suppressing GC growth in vivo, we utilized the GSE79973 and GSE118916 datasets to identify the GC development signatures that overlap with the RNA-seq analysis in SFA-treated AGS cells. GSEA of the RNA-seq data indicated that SFA regulation of GC progression was related to extracellular matrix and collagens; thus, we identified COL3A1 and COL5A1 as the targets of SFA, which functioned as oncogenes. We found positive correlations between COL3A1 and COL5A1 expression in GC cells, and confirmed that miR-29a-3p is the common regulator of their expression. RNA immunoprecipitation assays based on Ago2, Dicer, and exportin-5 showed that SFA could promote mature miR-29a-3p generation. We also proved that SFA inactivated the Wnt/β-catenin pathway in GC cells in a miR-29a-3p-dependent manner. Overall, SFA boosts miR-29a-3p maturation to downregulate COL3A1 and COL5A1 and inactivate the Wnt/ β -catenin pathway to suppress GC progression.
Collapse
Affiliation(s)
- Sichong Han
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Zhe Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Jining Liu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung City 402, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan; College of Food and Biological Engineering, Jimei University, Xiamen City 361021 Fujia Province, PR China; Undergraduate Program Study of Biomedical Engineering, Physics Department, Airlangga University, Surabaya City 60115, Indonesia.
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China.
| |
Collapse
|
19
|
Ashrafizadeh M, Ahmadi Z, Yaribeygi H, Sathyapalan T, Sahebkar A. Astaxanthin and Nrf2 signaling pathway: a novel target for new therapeutic approaches. Mini Rev Med Chem 2021; 22:312-321. [PMID: 33964864 DOI: 10.2174/1389557521666210505112834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/27/2021] [Accepted: 03/18/2021] [Indexed: 11/22/2022]
Abstract
Astaxanthin (AST) is a naturally occurring compound isolated from various sources such as fungi, plants, salmon, and crab. However, Haematococcus Pluvialis, a green alga, is the primary source of this beta carotenoid compound. AST has several favourable biological and pharmacological activities such as antioxidant, anti-inflammatory, anti-tumor, anti-diabetes, hepatoprotective and neuroprotective. Nevertheless, the exact molecular mechanisms of these protective effects of AST are unclear yet. The Nrf2 signaling pathway is one of the critical candidate signaling pathways that may be involved in these beneficial effects of AST. This signaling pathway is responsible for maintaining the redox balance in the physiologic state. Upon nuclear translocation, Nrf2 signaling activates antioxidant enzymes to reduce oxidative stress and protect cells against damage. In the current study, we have reviewed the effects of AST on the Nrf2 signaling pathway, which could potentially be developed as a novel therapeutic approach for the management of various diseases.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Zahra Ahmadi
- PhD student of Clinical Pathology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Habib Yaribeygi
- PhD student of Clinical Pathology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Tufekci KU, Ercan I, Isci KB, Olcum M, Tastan B, Gonul CP, Genc K, Genc S. Sulforaphane inhibits NLRP3 inflammasome activation in microglia through Nrf2-mediated miRNA alteration. Immunol Lett 2021; 233:20-30. [PMID: 33711331 DOI: 10.1016/j.imlet.2021.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 12/12/2022]
Abstract
The NLRP3 inflammasome is a multiprotein complex that activates caspase-1 and triggers the release of the proinflammatory cytokines IL-1β and IL-18 in response to diverse signals. Although inflammasome activation plays critical roles against various pathogens in host defense, overactivation of inflammasome contributes to the pathogenesis of inflammatory diseases, including acute CNS injuries and chronic neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. In the current study, we demonstrated that Sulforaphane (SFN), a dietary natural product, inhibits NLRP3 inflammasome mediated IL-1β and IL-18 secretion and pyroptosis in murine microglial cells. SFN decreased the secretion of IL-1β and IL-18, and their mRNA levels in LPS primed microglia triggered by ATP. SFN suppressed the overexpression of cleaved caspase-1 and NLRP3 protein expressions as measured by caspase activity assay and western blot, respectively. SFN also prevented caspase-1 dependent pyroptotic cell death in microglia. Our data indicate that SFN suppresses NLRP3 inflammasome via the inhibition of NF-κB nuclear translocation and Nrf2 mediated miRNAs expression modulation in murine microglia.
Collapse
Affiliation(s)
- Kemal Ugur Tufekci
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey; Vocational School of Health Services, Izmir Democracy University, Izmir, Turkey
| | - Ilkcan Ercan
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Kamer Burak Isci
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey; Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Izmir, Turkey
| | - Melis Olcum
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey
| | - Bora Tastan
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ceren Perihan Gonul
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey; Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Izmir, Turkey.
| |
Collapse
|
21
|
Li T, Pang Q, Liu Y, Bai M, Peng Y, Zhang Z. Sulforaphane protects human umbilical vein endothelial cells from oxidative stress via the miR-34a/SIRT1 axis by upregulating nuclear factor erythroid-2-related factor 2. Exp Ther Med 2021; 21:186. [PMID: 33488795 PMCID: PMC7812584 DOI: 10.3892/etm.2021.9617] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress-induced vascular endothelial cell dysfunction serves an essential role in the initiation and development of atherosclerosis. Sulforaphane (SFN), a naturally occurring antioxidant, has previously demonstrated to exert protective effects on the endothelium against oxidative stress. However, further studies are required to determine its underlying molecular mechanism prior to clinical application. Accumulating evidence suggests that alterations in the microRNA (miRNA/miR)-34a/sirtuin-1 (SIRT1) axis occur with oxidative stress. Therefore, the present study aimed to investigate if SFN exerts a protective role against oxidative stress in vascular endothelial cells through regulation of the miR-34a/SIRT1 axis. Human umbilical vein endothelial cells (HUVECs) were treated with H2O2 in the presence or absence of SFN pretreatment. Cell viability and apoptosis were analyzed using CellTiter-Blue and flow cytometry, respectively. Reverse transcription-quantitative PCR and western blot analyses were performed to determine changes in the expression levels of miR-34a and SIRT1. The expression levels of miR-34a and SIRT1 were artificially regulated following transfection with miR-34a mimic and inhibitor or SIRT1expression plasmid and small interfering RNA, respectively. Subsequently, the effect of the expression changes of miR-34 and SIRT1 on oxidative stress-induced cell injury was investigated. Dual-luciferase reporter assay was used to confirm the targeted binding of miR-34a to SIRT1. SFN was found to ameliorate cellular damage caused by H2O2 and inhibited intracellular reactive oxygen species production. In addition, miR-34a upregulation was accompanied with reduced SIRT1 expression in HUVECs, following H2O2 treatment. miR-34a was revealed to directly target SIRT1 by binding to its 3'-untranslated region. Down-regulation of miR-34a and up-regulation of SIRT1 increased the survival of HUVECs under oxidative stress. Taken together, the results of the present study suggest that SFN may protect HUVECs from oxidative stress by inducing changes in the miR-34a/SIRT1 axis via upregulation of nuclear factor erythroid-2-related factor 2 expression.
Collapse
Affiliation(s)
- Tao Li
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Qi Pang
- Department of Traditional Chinese Medicine, The Gansu Gem Flower Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Yongbin Liu
- Department of Cardiology, The Gansu Gem Flower Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Ming Bai
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yu Peng
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zheng Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
22
|
Epigenetic Regulation of NRF2/KEAP1 by Phytochemicals. Antioxidants (Basel) 2020; 9:antiox9090865. [PMID: 32938017 PMCID: PMC7555619 DOI: 10.3390/antiox9090865] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Epigenetics has provided a new dimension to our understanding of nuclear factor erythroid 2–related factor 2/Kelch-like ECH-associated protein 1 (human NRF2/KEAP1 and murine Nrf2/Keap1) signaling. Unlike the genetic changes affecting DNA sequence, the reversible nature of epigenetic alterations provides an attractive avenue for cancer interception. Thus, targeting epigenetic mechanisms in the corresponding signaling networks represents an enticing strategy for therapeutic intervention with dietary phytochemicals acting at transcriptional, post-transcriptional, and post-translational levels. This regulation involves the interplay of histone modifications and DNA methylation states in the human NFE2L2/KEAP1 and murine Nfe2l2/Keap1 genes, acetylation of lysine residues in NRF2 and Nrf2, interaction with bromodomain and extraterminal domain (BET) acetyl “reader” proteins, and non-coding RNAs such as microRNA (miRNA) and long non-coding RNA (lncRNA). Phytochemicals documented to modulate NRF2 signaling act by reversing hypermethylated states in the CpG islands of NFE2L2 or Nfe2l2, via the inhibition of DNA methyltransferases (DNMTs) and histone deacetylases (HDACs), through the induction of ten-eleven translocation (TET) enzymes, or by inducing miRNA to target the 3′-UTR of the corresponding mRNA transcripts. To date, fewer than twenty phytochemicals have been reported as NRF2 epigenetic modifiers, including curcumin, sulforaphane, resveratrol, reserpine, and ursolic acid. This opens avenues for exploring additional dietary phytochemicals that regulate the human epigenome, and the potential for novel strategies to target NRF2 signaling with a view to beneficial interception of cancer and other chronic diseases.
Collapse
|
23
|
Li J, Xu J, Cao Z, Du S, Zhang L. MiR-1231 decrease the risk of cancer-related mortality in patients combined with non-small cell lung cancer and diabetes mellitus. Cancer Cell Int 2020; 20:438. [PMID: 32939184 PMCID: PMC7487554 DOI: 10.1186/s12935-020-01525-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is a deadly human malignancy, and previous studies support the contribution of microRNAs (miRNAs) to cancer assessment. It has been reported that miR-1231 can be used as a biomarker to assess prognosis in different cancers. However, the prognostic value of miR-1231 in NSCLC patients with comorbid diabetes mellitus (DM) remains unclear. The present study evaluated the risk factors for NSCLC with DM and developed a predictive model for it. Methods A real-world study was conducted, including data from 108 patients with NSCLC combined with DM from April 1, 2010, to June 1, 2015. MiR-1231 was recorded during hospital admission. Cox-proportional hazards model was applied for survival analysis of risk factors for cancer-related mortality and to create nomograms for prediction. The accuracy of the model was evaluated by C-index and calibration curves. Results The mortality rate in the high miR-1231 level (≥ 1.775) group was 57.4%. On the basis of univariate analysis, we put factors (P < 0.05) into multivariate regression models, and high miR-1231 levels (P < 0.001, HR = 0.57), surgery (P < 0.001, HR = 0.37) and KPS score > 80 (P = 0.01, HR = 0.47) had a better prognosis and were considered as independent protective factors. These independently relevant factors were used to create nomograms to predict long-term patient survival. Nomogram showed good accuracy in risk estimation with a guide-corrected C-index of 0.691. Conclusion MiR-1231 reduced the risk of cancer-related death in patients with combined NSCLC and DM. Nomogram based on multivariate analysis showed good accuracy in estimating the overall risk of death.
Collapse
Affiliation(s)
- Jing Li
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Jialiang Xu
- Department of Cardiovascular, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhijun Cao
- Department of Urology, The Ninth People's Hospital of Suzhou, Suzhou, China
| | - Shouzuo Du
- Department of Endocrinology, Suzhou Xiangcheng People's Hospital, Suzhou, China
| | - Luyu Zhang
- Department of Endocrinology, Suzhou Xiangcheng People's Hospital, Suzhou, China
| |
Collapse
|
24
|
Li Q, Li G, Liu C, Chen N, Deng B, Xie Y. Cell Differentiation Agent-2 (CDA-2) Inhibits the Growth and Migration of Saos-2 Cells via miR-124/MAPK1. Cancer Manag Res 2020; 12:4541-4548. [PMID: 32606947 PMCID: PMC7304673 DOI: 10.2147/cmar.s248851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND CDA-2 (cell differentiation agent 2), isolated from healthy human urine, exerts antitumor effects in multiple types of cancer cells. However, its role in osteosarcoma has not been studied. METHODS The MTT assay was used to examine the cell proliferation rate. A colony formation assay was used to examine cell growth. The Transwell assay was used to examine cell migration ability. A real-time PCR assay was used to examine the expression levels of miR-124 and MAPK1. A Western blot assay was used to examine protein expression levels. MAPK1 was selected as a possible target of miR-124, and the targeting relationship was examined by a luciferase reporter assay. RESULTS We revealed that CDA-2 decreased the growth, migration and invasion ability of the osteosarcoma cell line Saos-2. Further study revealed that CDA-2 elevated the expression level of miR-124. MAPK1 was identified as a downstream target of miR-124. Knockdown of miR-124 or overexpression of MAPK1 counteracted CDA-2's effects on cell growth and invasion. CONCLUSION Our data revealed that the miR-124/MAPK1 axis mediated CDA-2's function in Saos-2 cells. CDA-2 can be used as a new treatment strategy for osteosarcoma.
Collapse
Affiliation(s)
- Quanxiu Li
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China,Correspondence: Quanxiu Li Email
| | - Guangchun Li
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Changyi Liu
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Na Chen
- Department of Pathology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Bangyu Deng
- Department of Oncology, Affiliated Tumor Hospital of Guangzhou University, Guangzhou, Guangdong, People’s Republic of China
| | - Youke Xie
- Department of Oncology, Hospital of Ruikang Affiliated to Guangxi University of Chinese Medicine, Guangxi, People’s Republic of China
| |
Collapse
|
25
|
Gong TT, Liu XD, Zhan ZP, Wu QJ. Sulforaphane enhances the cisplatin sensitivity through regulating DNA repair and accumulation of intracellular cisplatin in ovarian cancer cells. Exp Cell Res 2020; 393:112061. [PMID: 32437713 DOI: 10.1016/j.yexcr.2020.112061] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/20/2020] [Accepted: 05/08/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Cisplatin is commonly applied as anticancer agent for various cancers, including ovarian cancer. Unfortunately, the drug resistance frequently occurred which obstructing the effect of cisplatin on tumors. The goal of our research was to investigate the reversal actions and the potential mechanisms of sulforaphane (SFN) on cisplatin resistance in ovarian carcinoma. METHODS The A2780 and IGROV1 cells and their cisplatin resistance cells A2780/CP70 and IGROV1-R10 were used in this study. Cell viability was detected by CCK-8. The DNA repair was measured by comet assay. The cisplatin transporter proteins were measured with western blotting. The concentration of intracellular cisplatin was detected by HPLC. The luciferase activity assay was applied to determine the target site of miR-30a-3p on the 3'UTR of ERCC1 and ATP7A. A2780/CP70 and IGROV1-R10 xenograft mouse model were established to confirm the antineoplastic action of SFN combined with cisplatin. RESULTS SFN reversed the resistance of A2780/CP70 and IGROV1-R10 ovarian carcinoma cells to cisplatin through inducing DNA damage and accumulation of intracellular cisplatin. SFN treatment notably increased miR-30a-3p expression, which was decreased in cisplatin-resistant cells. Moreover, overexpressed miR-30a-3p enhanced the sensitivity of A2780/CP70 and IGROV1-R10 cells to cisplatin treatment, and inhibiting miR-30a-3p activity abated the reversal actions of SFN on cisplatin resistance. The luciferase assay findings showed that miR-30a-3p binds to ERCC1 and ATP7A which are the key regulators for DNA repair and cisplatin transportation. CONCLUSIONS Our findings indicated that SFN could enhance cisplatin sensitivity of ovarian carcinoma cells through up-regulating miR-30a-3p to induce DNA damage and accumulation of intracellular cisplatin.
Collapse
Affiliation(s)
- Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Dong Liu
- Department of Pharmacy, Shengjing hospital of China Medical University, Shenyang, China
| | - Zhi-Peng Zhan
- Department of Nutrition, School of Public Health, Jinzhou Medical University, Jinzhou, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
26
|
Calcabrini C, Maffei F, Turrini E, Fimognari C. Sulforaphane Potentiates Anticancer Effects of Doxorubicin and Cisplatin and Mitigates Their Toxic Effects. Front Pharmacol 2020; 11:567. [PMID: 32425794 PMCID: PMC7207042 DOI: 10.3389/fphar.2020.00567] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
The success of cancer therapy is often compromised by the narrow therapeutic index of many anticancer drugs and the occurrence of drug resistance. The association of anticancer therapies with natural compounds is an emerging strategy to improve the pharmaco-toxicological profile of cancer chemotherapy. Sulforaphane, a phytochemical found in cruciferous vegetables, targets multiple pathways involved in cancer development, as recorded in different cancers such as breast, brain, blood, colon, lung, prostate, and so forth. As examples to make the potentialities of the association chemotherapy raise, here we highlight and critically analyze the information available for two associations, each composed by a paradigmatic anticancer drug (cisplatin or doxorubicin) and sulforaphane.
Collapse
Affiliation(s)
- Cinzia Calcabrini
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Francesca Maffei
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| |
Collapse
|