1
|
Fan Y, Wang X, Liu Y, Tang H, Li C. Therapeutic effects of focused ultrasound on vulvar squamous intraepithelial lesions in rat. Int J Hyperthermia 2024; 41:2401417. [PMID: 39255969 DOI: 10.1080/02656736.2024.2401417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
OBJECTIVE In this study, we established a Sprague-Dawley rat model of vulvar squamous intraepithelial lesions and investigated the impact of focused ultrasound on the expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and mutant type p53 (mtp53) in the vulvar skin of rats with low-grade squamous intraepithelial lesions (LSIL). MATERIALS AND METHODS The vulvar skin of 60 rats was treated with dimethylbenzanthracene (DMBA) and mechanical irritation three times a week for 14 weeks. Rats with LSIL were randomly allocated into the experimental group or the control group. The experimental group was treated with focused ultrasound, while the control group received sham treatment. RESULTS After 14 weeks treatment of DMBA combined with mechanical irritation, LSIL were observed in 44 (73.33%) rats, and high-grade squamous intraepithelial lesions (HSIL) were observed in 14 (23.33%) rats. 90.91% (20/22) of rats showed normal pathology and 9.09% (2/22) of rats exhibited LSIL in the experimental group at four weeks after focused ultrasound treatment. 22.73% (5/22) of rats exhibited LSIL, 77.27% (17/22) of rats progressed to HSIL in the control group. Compared with the control-group rats, the levels of HIF-1α, VEGF and mtp53 were significantly decreased in experimental-group rats (p < 0.05). CONCLUSIONS These results indicate that DMBA combined with mechanical irritation can induce vulvar squamous intraepithelial lesion in SD rats. Focused ultrasound can treat LSIL safely and effectively, prevent the progression of vulvar lesions, and improve the microenvironment of vulvar tissues by decreasing the localized expression of HIF-1α, VEGF, and mtp53 in rats.
Collapse
Affiliation(s)
- Yijin Fan
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Xi Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Yao Liu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Laboratory Medicine, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huajun Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Chengzhi Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Farombi EO, Ajayi BO, Ajeigbe OF, Maruf OR, Anyebe DA, Opafunso IT, Adedara IA. Mechanistic exploration of 6-shogaol's preventive effects on azoxymethane and dextran sulfate sodium -induced colorectal cancer: involvement of cell proliferation, apoptosis, carcinoembryonic antigen, wingless-related integration site signaling, and oxido-inflammation. Toxicol Mech Methods 2024:1-10. [PMID: 39034841 DOI: 10.1080/15376516.2024.2381798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Colorectal cancer (CRC) poses a significant global health burden, being the third most prevalent cancer and the second most significant contributor to cancer-related deaths worldwide. Preventive strategies are crucial to combat this rising incidence. 6-shogaol, derived from ginger, has shown promise in preventing and treating various cancers. This study investigated the preventive effects of 6-shogaol on azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced CRC in mice. Forty male BALB/c mice were randomly divided into control, 6-shogaol, AOM + DSS, and 6-shogaol + AOM + DSS. Mice in the control group received corn oil for 16 weeks, while those in the 6-Shogaol group were administered 20 mg/kg of 6-shogaol for 16 weeks. The AOM + DSS group received a single intraperitoneal dose (ip) of 10 mg/kg of AOM, followed by three cycles of 2.5% DSS in drinking water. The 6-shogaol + AOM + DSS group received both 6-shogaol for 16 weeks and a single ip of 10 mg/kg of AOM, followed by three cycles of 2.5% DSS in drinking water. The AOM + DSS-treated mice exhibited reduced food consumption, colon weight, and colon length, along with increased tumor formation. Co-administration of 6-shogaol effectively reversed these changes, inhibiting CRC development. Histopathological analysis revealed protective effects of 6-shogaol against colonic insults and modulation of inflammatory responses. 6-shogaol significantly reduced Carcinoembryonic antigen and Kiel 67 levels, indicating inhibition of tumor cell proliferation. Mechanistically, 6-shogaol promoted apoptosis by upregulating protein 53 and caspase-3 expression, and it effectively restored the balance of the Wingless-related integration site signaling pathway by regulating β-catenin and adenomatous polyposis coli levels. Moreover, 6-shogaol demonstrated anti-inflammatory effects, reducing myeloperoxidase, Tumor necrosis factor alpha, and cyclooxygenase-2 levels in AOM/DSS-treated mice. Additionally, 6-shogaol restored redox homeostasis by reducing lipid peroxidation and nitrosative stress and enhancing antioxidant enzyme activities. The findings suggest that 6-shogaol inhibits cell proliferation, induces apoptosis, regulates Wnt signaling, suppresses inflammation, and restores redox homeostasis, providing comprehensive insights into its potential therapeutic benefits for CRC.
Collapse
Affiliation(s)
- Ebenezer Olatunde Farombi
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Biochemistry, College of Natural and Applied Sciences, Chrisland University, Abeokuta, Nigeria
| | - Babajide Oluwaseun Ajayi
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Oncopreventives and Systems Oncology Research Laboratory, Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Olufunke Florence Ajeigbe
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Opeyemi Rabiat Maruf
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Daniel Abu Anyebe
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ifeoluwa Tobi Opafunso
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Isaac Adegboyega Adedara
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
3
|
Yang C, Chen W, Ye B, Nie K. An overview of 6-shogaol: new insights into its pharmacological properties and potential therapeutic activities. Food Funct 2024; 15:7252-7270. [PMID: 38287779 DOI: 10.1039/d3fo04753a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
Ginger (Zingiber officinale Roscoe) has traditionally been used as a cooking spice and herbal medicine for treating nausea and vomiting. More recently, ginger was found to effectively reduce the risk of diseases such as gastroenteritis, migraine, gonarthritis, etc., due to its various bioactive compounds. 6-Shogaol, the pungent phenolic substance in ginger, is the most pharmacologically active among such compounds. The aim of the present study was to review the pharmacological characteristic of 6-shogaol, including the properties of anti-inflammatory, antioxidant and antitumour, and its corresponding molecular mechanism. With its multiple mechanisms, 6-shogaol is considered a beneficial natural compound, and therefore, this review will shed some light on the therapeutic role of 6-shogaol and provide a theoretical basis for the development and clinical application of 6-shogaol.
Collapse
Affiliation(s)
- Chenglu Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Weijian Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Binbin Ye
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
4
|
Figueroa‐González G, Quintas‐Granados LI, Reyes‐Hernández OD, Caballero‐Florán IH, Peña‐Corona SI, Cortés H, Leyva‐Gómez G, Habtemariam S, Sharifi‐Rad J. Review of the anticancer properties of 6-shogaol: Mechanisms of action in cancer cells and future research opportunities. Food Sci Nutr 2024; 12:4513-4533. [PMID: 39055196 PMCID: PMC11266911 DOI: 10.1002/fsn3.4129] [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: 09/04/2023] [Revised: 02/05/2024] [Accepted: 03/13/2024] [Indexed: 07/27/2024] Open
Abstract
Cancer is a major global health challenge that affects every nation and accounts for a large portion of the worldwide disease burden. Furthermore, cancer cases will rise significantly in the next few decades. The Food and Drug Administration has approved more than 600 drugs for treating diverse types of cancer. However, many conventional anticancer medications cause side effects, and drug resistance develops as the treatment proceeds with a concomitant impact on patients' quality of life. Thus, exploring natural products with antitumor properties and nontoxic action mechanisms is essential. Ginger (Zingiber officinale Roscoe) rhizome has a long history of use in traditional medicine, and it contains biologically active compounds, gingerols and shogaols. The main ginger shogaol is 6-shogaol, whose concentration dramatically increases during the processing of ginger, primarily due to the heat-induced conversion of 6-gingerol. Some studies have demonstrated that 6-shogaol possesses biological and pharmacological properties, such as antioxidant, anti-inflammatory, and anticancer activities. The mechanism of action of 6-shogaol as an anticancer drug includes induction of paraptosis, induction of apoptosis, increase in the production of reactive oxygen species, induction of autophagy, and the inhibition of AKT/mTOR signaling. Despite this knowledge, the mechanism of action of 6-shogaol is not fully understood, and the scientific data on its therapeutic dose, safety, and toxicity are not entirely described. This review article examines the potential of 6-shogaol as an anticancer drug, addressing the limitations of current medications; it covers 6-shogaol's attributes, mechanism of action in cancer cells, and opportunities for future research.
Collapse
Affiliation(s)
- Gabriela Figueroa‐González
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores ZaragozaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Laura Itzel Quintas‐Granados
- Colegio de Ciencias y Humanidades, Plantel CuautepecUniversidad Autónoma de la Ciudad de MéxicoCiudad de MéxicoMexico
| | - Octavio Daniel Reyes‐Hernández
- Laboratorio de Biología Molecular del Cáncer, UMIEZ, Facultad de Estudios Superiores ZaragozaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Isaac H. Caballero‐Florán
- Departamento de Farmacia, Facultad de QuímicaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de QuímicaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de GenómicaInstituto Nacional de Rehabilitación Luis Guillermo Ibarra IbarraCiudad de MexicoMexico
| | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de QuímicaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UKCentral Avenue, Chatham‐MaritimeKentME4 4TBUK
| | | |
Collapse
|
5
|
Nivetha S, Asha KRT, Srinivasan S, Murali R, Kanagalakshmi A. p-Coumaric acid pronounced protective effect against potassium bromate-induced hepatic damage in Swiss albino mice. Cell Biochem Funct 2024; 42:e4076. [PMID: 38895919 DOI: 10.1002/cbf.4076] [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: 03/06/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Potassium bromate (KBrO3) is a common dietary additive, pharmaceutical ingredient, and significant by-product of water disinfection. p-coumaric acid (PCA) is a naturally occurring nutritional polyphenolic molecule with anti-inflammatory and antioxidant activities. The goal of the current investigation was to examine the protective effects of p-coumaric acid against the liver damage caused by KBrO3. The five groups of animals-control, KBrO3 (100 mg/kg bw), treatment with KBrO3 along with Silymarin (100 mg/kg bw), KBrO3, followed by PCA (100 mg/bw, and 200 mg/kg bw) were randomly assigned to the animals. Mice were slaughtered, and blood and liver tissues were taken for assessment of the serum biochemical analysis for markers of liver function (alanine transaminase, aspartate transaminase, alkaline phosphatase, albumin, and protein), lipid markers and antioxidant markers (TBARS), glutathione peroxidase [GSH-Px], glutathione (GSH), and markers of hepatic oxidative stress (CAT), (SOD), as well as histological H&E stain, immunohistochemical stain iNOS, and COX-2 as markers of inflammatory cytokines. PCA protects against acute liver failure by preventing the augmentation of blood biochemical markers and lipid profiles. In mice liver tissues, KBrO3 increases lipid indicators and depletes antioxidants, leading to an increase in JNK, ERK, and p38 phosphorylation. Additionally, PCA inhibited the production of pro-inflammatory cytokines and reduced the histological alterations in KBrO3-induced hepatotoxicity. Notably, PCA effectively mitigated KBrO3-induced hepatic damage by obstructing the TNF-α/NF-kB-mediated inflammatory process signaling system. Additionally, in KBrO3-induced mice, PCA increased the intensities of hepatic glutathione (GSH), SOD, GSH-Px, catalase, and GSH activities. Collectively, we demonstrate the molecular evidence that PCA eliminated cellular inflammatory conditions, mitochondrial oxidative stress, and the TNF-α/NF-κB signaling process, thereby preventing KBrO3-induced hepatocyte damage.
Collapse
Affiliation(s)
- Selvaraj Nivetha
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
- Department of Biochemistry, Government Arts College, Paramakudi, India
| | | | - Subramani Srinivasan
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
- Department of Biochemistry, Government Arts College for Women, Krishnagiri, India
| | - Raju Murali
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
- Department of Biochemistry, Government Arts College for Women, Krishnagiri, India
| | - Ambothi Kanagalakshmi
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
- Department of Biochemistry, Government Arts College for Women, Krishnagiri, India
| |
Collapse
|
6
|
Wang K, Lai W, Min T, Wei J, Bai Y, Cao H, Guo J, Su Z. The Effect of Enteric-Derived Lipopolysaccharides on Obesity. Int J Mol Sci 2024; 25:4305. [PMID: 38673890 PMCID: PMC11050189 DOI: 10.3390/ijms25084305] [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: 03/11/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Endotoxin is a general term for toxic substances in Gram-negative bacteria, whose damaging effects are mainly derived from the lipopolysaccharides (LPS) in the cell walls of Gram-negative bacteria, and is a strong pyrogen. Obesity is a chronic, low-grade inflammatory condition, and LPS are thought to trigger and exacerbate it. The gut flora is the largest source of LPS in the body, and it is increasingly believed that altered intestinal microorganisms can play an essential role in the pathology of different diseases. Today, the complex axis linking gut flora to inflammatory states and adiposity has not been well elucidated. This review summarises the evidence for an interconnection between LPS, obesity, and gut flora, further expanding our understanding of LPS as a mediator of low-grade inflammatory disease and contributing to lessening the effects of obesity and related metabolic disorders. As well as providing targets associated with LPS, obesity, and gut flora, it is hoped that interventions that combine targets with gut flora address the individual differences in gut flora treatment.
Collapse
Affiliation(s)
- Kai Wang
- Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (K.W.); (W.L.); (T.M.); (J.W.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Weiwen Lai
- Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (K.W.); (W.L.); (T.M.); (J.W.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tianqi Min
- Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (K.W.); (W.L.); (T.M.); (J.W.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jintao Wei
- Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (K.W.); (W.L.); (T.M.); (J.W.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China;
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China;
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (K.W.); (W.L.); (T.M.); (J.W.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
7
|
Pan S, Li Y, Zhang J. 6-Shogaol prevents benzo (A) pyrene-exposed lung carcinogenesis via modulating PRDX1-associated oxidative stress, inflammation, and proliferation in mouse models. ENVIRONMENTAL TOXICOLOGY 2024; 39:75-84. [PMID: 37638803 DOI: 10.1002/tox.23946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023]
Abstract
In this study, we have investigated the chemopreventive role of 6-shogaol (6-SGL) on benzopyrene (BaP) exposed lung carcinogenesis by modulating PRDX1-associated oxidative stress, inflammation, and proliferation in Swiss albino mouse models. Mice were exposed to BaP (50 mg/kg b.wt) orally twice a week for four consecutive weeks and maintained for 16 weeks, respectively. 6-SGL (30 mg/kg b.wt) were orally administered to mouse 1 h before BaP exposure for 16 weeks. After the experiment's termination, 6-SGL (30 mg/kg b.wt) prevented the loss in body weight, increased lung weight, and the total number of tumors in the mice. Moreover, we observed that 6-SGL treatment reverted the activity of BaP-induced lipid peroxidation and antioxidants in mice. Also, 6-SGL impeded the phosphorylation of MAPK family proteins such as Erk1, p38, and Jnk1 in BaP-exposed mice. PRDX1 is an essential antioxidant protein that scavenges toxic radicals and enhances several antioxidant proteins. Overexpression of PRDX1 substantially inhibits MAPKs, proliferation, and inflammation signaling axis. Hence, PRDX1 is thought to be a novel targeting protein for preventing BaP-induced lung cancer. In this study, we have obtained the 6-SGL treatment in a mouse model that reverted BaP-induced depletion of PRDX1 expression. Moreover, pretreatment of 6-SGL (30 mg/kg b.wt) significantly inhibited enhanced proinflammatory cytokines (TNF-α, IL-6, IL-β1, IL-10) and proliferative markers (Cyclin-D1, Cyclin-D2, and PCNA) in BaP-exposed mice. The histopathological studies also confirmed that 6-SGL effectively protected the cells with less damage. Thus, the study demonstrated that 6-SGL could be a potential phytochemical and act as a chemopreventive agent in BaP-induced lung cancer by enhancing PRDX1 expression.
Collapse
Affiliation(s)
- Shuang Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yaming Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jinzhao Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
8
|
Jia Y, Li X, Meng X, Lei J, Xia Y, Yu L. Anticancer perspective of 6-shogaol: anticancer properties, mechanism of action, synergism and delivery system. Chin Med 2023; 18:138. [PMID: 37875983 PMCID: PMC10594701 DOI: 10.1186/s13020-023-00839-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
Cancer is a malignant disease that has plagued human beings all the time, but the treatment effect of commonly used anticancer drugs in clinical practice is not ideal by reason of their drug tolerance and Strong adverse reactions to patients. Therefore, it is imperative to find effective and low-toxic anticancer drugs. Many research works have shown that natural products in Chinese herbal medicine have great anticancer potential, such as 6-shogaol, a monomer composition obtained from Chinese herbal ginger, which has been confirmed by numerous in vitro or vivo studies to be an excellent anti-cancer active substance. In addition, most notably, 6-shogaol has different selectivity for normal and cancer cells during treatment, which makes it valuable for further research and clinical development. Therefore, this review focus on the anti-cancer attributes, the mechanism and the regulation of related signaling pathways of 6-shogaol. In addition, its synergy with commonly used anticancer drugs, potential drug delivery systems and prospects for future research are discussed. This is the first review to comprehensively summarize the anti-cancer mechanism of 6-shogaol, hoping to provide a theoretical basis and guiding significance for future anti-cancer research and clinical development of 6-shogaol.
Collapse
Affiliation(s)
- Yaoxia Jia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Xing Li
- Jianyang Chinese Medicine Hospital, Chengdu, 641400, China
| | - Xiangqi Meng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Jinjie Lei
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Yangmiao Xia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Lingying Yu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China.
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.
| |
Collapse
|
9
|
Thiruvengadam R, Kim JH. Therapeutic strategy for oncovirus-mediated oral cancer: A comprehensive review. Biomed Pharmacother 2023; 165:115035. [PMID: 37364477 DOI: 10.1016/j.biopha.2023.115035] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/02/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Oral cancer is a neoplastic disorder of the oral cavities, including the lips, tongue, buccal mucosa, and lower and upper gums. Oral cancer assessment entails a multistep process that requires deep knowledge of the molecular networks involved in its progression and development. Preventive measures including public awareness of risk factors and improving public behaviors are necessary, and screening techniques should be encouraged to enable early detection of malignant lesions. Herpes simplex virus (HSV), human papillomavirus (HPV), Epstein-Barr virus (EBV), and Kaposi sarcoma-associated herpesvirus (KSHV) are associated with other premalignant and carcinogenic conditions leading to oral cancer. Oncogenic viruses induce chromosomal rearrangements; activate signal transduction pathways via growth factor receptors, cytoplasmic protein kinases, and DNA binding transcription factors; modulate cell cycle proteins, and inhibit apoptotic pathways. In this review, we present an up-to-date overview on the use of nanomaterials for regulating viral proteins and oral cancer as well as the role of phytocompounds on oral cancer. The targets linking oncoviral proteins and oral carcinogenesis were also discussed.
Collapse
Affiliation(s)
- Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
10
|
Zong X, Ding Q, Liu X, Liu Q, Song S, Yan X, Zhang Y. Preventive Effect of 6-shogaol on D-galactosamine Induced Hepatotoxicity Through NF-?B/MAPK Signaling Pathway in Rats. Physiol Res 2023; 72:445-454. [PMID: 37795887 PMCID: PMC10634558 DOI: 10.33549/physiolres.935092] [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: 03/03/2023] [Accepted: 04/19/2023] [Indexed: 01/05/2024] Open
Abstract
This analysis aims to see whether 6-shogaol could protect rats against D-galactosamine (D-GalN)-induced Hepatotoxicity. The Wistar rats were divided into four groups (n=6). Group 1 received a standard diet, Group 2 received an oral administration of 6-shogaol (20 mg/kg b.wt), Group 3 received an intraperitoneal injection of D-GalN (400 mg/kg b.wt) on 21st day, and Group 4 received an oral administration of 6-shogaol (20mg/kg b.wt) for 21 days and D-GalN (400 mg/kg b.wt) injection only on 21st day. The hepatic marker enzymes activity, lipid peroxidative markers level increased significantly and antioxidant activity/level significantly reduced in D-GalN-induced rats. 6-shogaol Pretreatment effectively improves the above changes in D-GalN-induced rats. Further, inflammatory marker expression and MAPK signaling molecules were downregulated by 6-shogaol. These findings showed that 6-shogaol exerts hepatoprotective effects via the enhanced antioxidant system and attenuated the inflammation and MAPK signaling pathway in D-GalN-induced rats.
Collapse
Affiliation(s)
- X Zong
- Department of Clinical Laboratory, Baoding First Central Hospital, Baoding, Hebei province, China.
| | | | | | | | | | | | | |
Collapse
|
11
|
Zhou X, Zeng M, Huang F, Qin G, Song Z, Liu F. The potential role of plant secondary metabolites on antifungal and immunomodulatory effect. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12601-5. [PMID: 37272939 DOI: 10.1007/s00253-023-12601-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/13/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023]
Abstract
With the widespread use of antibiotic drugs worldwide and the global increase in the number of immunodeficient patients, fungal infections have become a serious threat to global public health security. Moreover, the evolution of fungal resistance to existing antifungal drugs is on the rise. To address these issues, the development of new antifungal drugs or fungal inhibitors needs to be targeted urgently. Plant secondary metabolites are characterized by a wide variety of chemical structures, low price, high availability, high antimicrobial activity, and few side effects. Therefore, plant secondary metabolites may be important resources for the identification and development of novel antifungal drugs. However, there are few studies to summarize those contents. In this review, the antifungal modes of action of plant secondary metabolites toward different types of fungi and fungal infections are covered, as well as highlighting immunomodulatory effects on the human body. This review of the literature should lay the foundation for research into new antifungal drugs and the discovery of new targets. KEY POINTS: • Immunocompromised patients who are infected the drug-resistant fungi are increasing. • Plant secondary metabolites toward various fungal targets are covered. • Plant secondary metabolites with immunomodulatory effect are verified in vivo.
Collapse
Affiliation(s)
- Xue Zhou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Meng Zeng
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Fujiao Huang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, People's Republic of China.
- Molecular Biotechnology Platform, Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - Fangyan Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
12
|
Li J, Wang L, Sun Y, Wang Z, Qian Y, Duraisamy V, Antary TMA. Zerumbone-induced reactive oxygen species-mediated oxidative stress re-sensitizes breast cancer cells to paclitaxel. Biotechnol Appl Biochem 2023; 70:28-37. [PMID: 35240000 DOI: 10.1002/bab.2326] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 01/21/2022] [Indexed: 12/24/2022]
Abstract
Chemotherapy is an effective approach for cancer therapy when plant-derived sensitizers are combined with chemotherapeutics. Zerumbone, a natural phytochemical, has been documented to have various pharmacological roles. Here, we evaluated the chemosensitization potential of zerumbone in a breast cancer cell line in vitro. Zerumbone-induced cytotoxicity in MCF-7 cells was assessed by 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT)-based metabolic analysis. Reactive oxygen species (ROS)-mediated mitochondrial membrane alterations, DNA damage, and apoptotic morphological changes were measured by fluorescence microscopy methods. A biochemical assay was employed to analyze Thiobarbituric acid reactive substances (TBARS) and antioxidant levels. Apoptotic marker expression levels were investigated by immunoblotting. MTT assay revealed that zerumbone significantly enhanced paclitaxel (PTX)-induced cell death in breast cancer cells in a concentration-dependent manner. Furthermore, our study demonstrated that zerumbone (15 μM) significantly enhanced ROS when combined with PTX (1 μM) treatment. Additionally, we observed that zerumbone enhanced the impairment of mitochondrial membrane potential and oxidative DNA damage, thereby inducing apoptosis in combination with PTX. Western blot analysis indicated that zerumbone significantly upregulated BAX, caspase-7, and caspase-9 expression and decreased BCL-2 expression, thereby inducing proapoptotic protein-mediated cell death combined with PTX. The prooxidant properties of zerumbone potentially resensitize breast cancer cells to PTX by enhancing intracellular ROS-mediated oxidative stress.
Collapse
Affiliation(s)
- Jutao Li
- Breast and Thyroid Surgery Ward 1, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, Liaoning, China
| | - Lingying Wang
- Department of Thoracic Surgery, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei, China
| | - Yuxin Sun
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, Liaoning, China
| | - Zhe Wang
- Department of Pharmacy Medical Guarantee Center, PLA General Hospital in The Fourth Medical Center, Beijing, China
| | - Ye Qian
- Department of Oncology, Affiliated Hai 'an Hospital of Nantong University, Haian, Jiangsu, 226600, China
| | | | - Tawfiq M Al Antary
- Pesticide and Economic Entomology, Faculty of Agriculture, the University of Jordan, Amman, Jordan
| |
Collapse
|
13
|
Chen M, Tong C, Wu Q, Zhong Z, He Q, Zeng L, Xiao L. 6-Shogaol Inhibits the Cell Migration of Colon Cancer by Suppressing the EMT Process Through the IKKβ/NF-κB/Snail Pathway. Integr Cancer Ther 2023; 22:15347354231172732. [PMID: 37157810 DOI: 10.1177/15347354231172732] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
6-Shogaol from ginger has anti-inflammatory, anti-oxidation and anti-cancer effects. Aim of the Study: To study the effects and possible mechanisms of 6-Shogaol on inhibiting the migration of colon cancer cells Caco2 and HCT116 and prove the effects on proliferation and apoptosis. Materials and methods: The cells were treated with 6-Shogaol at the concentrations of 20, 40, 60, 80, and 100 µM, the cytotoxicity was tested by Colony formation assays and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and the Western blot was used to evaluate IKKβ/NF-κB/Snail pathway and EMT-related proteins. In addition, in order to eliminate the interference of proliferation inhibition on the experiment, Caco2 cells were treated with 6-Shogaol at the concentrations of 0, 40, and 80 µM, HCT116 cells were treated with 6-Shogaol at the concentrations of 0, 20, and 40 µM, apoptosis was measured by Annex V/PI staining, and migration was measured by Wound healing assays and Transwell test. Results: 6-Shogaol significantly inhibited the growth of cells. The maximum inhibitory concentration of half of them was 86.63 µM in Caco2 cells and 45.25 µM in HCT116 cells. At 80 µM and 40 µM concentrations, 6-Shogaol significantly promoted apoptosis of colon cancer Caco2 cells and HCT116 cells, and also significantly inhibited cell migration (P < .05). In addition, Western blot analysis showed that at 80 µM dose of 6-Shogaol significantly reduced MMP-2, N-cadherin, IKKβ, P-NF-κB and Snail expression in Caco2 cells (P < .05). 40 µM dose of 6-Shogaol significantly reduced VEGF, IKKβ, and P-NF-κB expression, and MMP-2, N-cadherin and Snail was significantly decreased at 60 µM of 6-Shogaol in HCT116 cells(P < .05). However, there was no significant change in E-cadherin in Caco2 cells, and the expression of E-cadherin protein in HCT116 cells decreased. Conclusion: This study proposes and confirms that 6-Shogaol can significantly inhibit the migration of colon cancer cells Caco2 and HCT116, and its mechanism may be produced by inhibiting EMT through IKKβ/NF-κB/Snail signaling pathway. It was also confirmed that 6-Shogaol inhibited the proliferation and promoted apoptosis of Caco2 and HCT116 cells.
Collapse
Affiliation(s)
- Min Chen
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR,, China
- The Fifth Affiliated (Zhuhai) Hospital of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Chiin Tong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR,, China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR,, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong University of Technology, Guangzhou, Guangdong, China
- Zhuhai MUST Science and Technology Research Institute, Zhuhai, Guangdong, China
| | - Zhenghong Zhong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR,, China
| | - Qida He
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR,, China
| | - Li Zeng
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR,, China
| | - Lu Xiao
- Zhuhai Campus, Zunyi Medical University, Zhuhai, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
14
|
Multifaceted Pharmacological Potentials of Curcumin, Genistein, and Tanshinone IIA through Proteomic Approaches: An In-Depth Review. Cancers (Basel) 2022; 15:cancers15010249. [PMID: 36612248 PMCID: PMC9818426 DOI: 10.3390/cancers15010249] [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: 09/08/2022] [Revised: 12/03/2022] [Accepted: 12/12/2022] [Indexed: 01/03/2023] Open
Abstract
Phytochemicals possess various intriguing pharmacological properties against diverse pathological conditions. Extensive studies are on-going to understand the structural/functional properties of phytochemicals as well as the molecular mechanisms of their therapeutic function against various disease conditions. Phytochemicals such as curcumin (Cur), genistein (Gen), and tanshinone-IIA (Tan IIA) have multifaceted therapeutic potentials and various efforts are in progress to understand the molecular dynamics of their function with different tools and technologies. Cur is an active lipophilic polyphenol with pleiotropic function, and it has been shown to possess various intriguing properties including antioxidant, anti-inflammatory, anti-microbial, anticancer, and anti-genotoxic properties besides others beneficial properties. Similarly, Gen (an isoflavone) exhibits a wide range of vital functions including antioxidant, anti-inflammatory, pro-apoptotic, anti-proliferative, anti-angiogenic activities etc. In addition, Tan IIA, a lipophilic compound, possesses antioxidant, anti-angiogenic, anti-inflammatory, anticancer activities, and so on. Over the last few decades, the field of proteomics has garnered great momentum mainly attributed to the recent advancement in mass spectrometry (MS) techniques. It is envisaged that the proteomics technology has considerably contributed to the biomedical research endeavors lately. Interestingly, they have also been explored as a reliable approach to understand the molecular intricacies related to phytochemical-based therapeutic interventions. The present review provides an overview of the proteomics studies performed to unravel the underlying molecular intricacies of various phytochemicals such as Cur, Gen, and Tan IIA. This in-depth study will help the researchers in better understanding of the pharmacological potential of the phytochemicals at the proteomics level. Certainly, this review will be highly instrumental in catalyzing the translational shift from phytochemical-based biomedical research to clinical practice in the near future.
Collapse
|
15
|
Health benefits of bioactive components in pungent spices mediated via the involvement of TRPV1 channel. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
16
|
Bao T, Feng L, Cho S, Yu H, Jin W, Dai L, Zhang J, Bai L, Fu M, Chen Y. RNA-Seq Reveals Protective Mechanisms of Mongolian Medicine Molor-Dabos-4 on Acute Indomethacin-Induced Gastric Ulcers in Rats. Genes (Basel) 2022; 13:genes13101740. [PMID: 36292625 PMCID: PMC9602025 DOI: 10.3390/genes13101740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to apply transcriptomics to determine how Molor-Dabos-4 (MD-4) protects healthy rats against indomethacin (IND)-induced gastric ulcers and to identify the mechanism behind this protective effect. Rats were pretreated with MD-4 (0.3, 1.5, or 3 g/kg per day) for 21 days before inducing gastric ulcers by oral administration with indomethacin (30 mg/kg). Unulcerated and untreated healthy rats were used as controls. Effects of the treatment were assessed based on the ulcer index, histological and pathological examinations, and indicators of inflammation, which were determined by enzyme-linked immunosorbent assay. Transcriptomic analysis was performed for identifying potential pharmacological mechanisms. Eventually, after identifying potential target genes, the latter were validated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). After pretreatment with MD-4, gastric ulcers, along with other histopathological features, were reduced. MD-4 significantly (p < 0.05) increased the superoxide dismutase (SOD) levels in ulcers and reduced pepsin, TNF-α, and IL-6 levels. RNA-seq analysis identified a number of target genes on which MD-4 could potentially act. Many of these genes were involved in pathways that were linked to anti-inflammatory and antioxidant responses, and other protective mechanisms for the gastric mucosa. qRT-PCR showed that altered expression of the selected genes, such as Srm, Ryr-1, Eno3, Prkag3, and Eef1a2, was consistent with the transcriptome results. MD-4 exerts protective effects against IND-induced gastric ulcers by reducing inflammatory cytokines and pepsin and increasing the expression of SOD levels. Downregulation of Srm, Ryr-1, Eno3, Prkag3, and Eef1a2 genes involved in regulating arginine and proline metabolism, calcium signaling pathway, HIF-1 signaling pathway, oxytocin signaling pathway, and legionellosis are possibly involved in MD-4-mediated protection against gastric ulcers.
Collapse
Affiliation(s)
- Terigele Bao
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Lan Feng
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Sungbo Cho
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Hongzhen Yu
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Wenjie Jin
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Lili Dai
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Junqing Zhang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Laxinamujila Bai
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Minghai Fu
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
- Correspondence: (M.F.); (Y.C.)
| | - Yongsheng Chen
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
- Correspondence: (M.F.); (Y.C.)
| |
Collapse
|
17
|
Wang W, Gu W, He C, Zhang T, Shen Y, Pu Y. Bioactive components of Banxia Xiexin Decoction for the treatment of gastrointestinal diseases based on flavor-oriented analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115085. [PMID: 35150814 DOI: 10.1016/j.jep.2022.115085] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin Decoction (BXD) was first recorded in a Chinese medical classic, Treatise on Febrile Diseases and Miscellaneous Diseases, which was written in the Eastern Han dynasty of China. This ancient prescription consists of seven kinds of Chinese herbal medicine, namely, Pinellia ternata, Rhizoma Coptidis, Radix scutellariae, Rhizoma Zingiberis, Ginseng, Jujube, and Radix Glycyrrhizaepreparata. In clinic practice, its original application in China mainly has focused on the treatment of chronic gastritis for several hundred years. BXD is also effective in treating other gastrointestinal diseases (GIDs) in modern medical application. Despite available literature support and clinical experience, the treatment mechanisms or their relationships with the bioactive compounds in BXD responsible for its pharmacological actions, still need further explorations in more diversified channels. According to the analysis based on the five-flavor theory of TCM, BXD is traditionally viewed as the most representative prescription for pungent-dispersion, bitter-purgation and sweet-tonification. Consequently, based on the flavor-oriented analysis, the compositive herbs in BXD can be divided into three flavor groups, namely, the pungent, bitter, and sweet groups, each of which has specific active ingredients that are possibly relevant to GID treatment. AIM OF THE REVIEW This paper summarized recent literatures on BXD and its bioactive components used in GID treatment, and provided the pharmacological or chemical basis for the further exploration of the ancient prescription and the relative components. METHOD ology: Relevant literature was collected from various electronic databases such as Pubmed, Web of Science, and China National Knowledge Infrastructure (CNKI). Citations were based on peer-reviewed articles published in English or Chinese during the last decade. RESULTS Multiple components were found in the pungent, bitter, and sweet groups in BXD. The corresponding bioactive components include gingerol, shogaol, stigmasterol, and β-sitosterol in the pungent group; berberine, palmatine, coptisine, baicalein, and baicalin in the bitter group; and ginsenosides, polysaccharides, liquiritin, and glycyrrhetinic acid in the sweet group. These components have been found directly or indirectly responsible for the remarkable effects of BXD on GID. CONCLUSION This review provided some valuable reference to further clarify BXD treatment for GID and their possible material basis, based on the perspective of the flavor-oriented analysis.
Collapse
Affiliation(s)
- Weiwei Wang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weiliang Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao He
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yao Shen
- Shanghai Center of Biomedicine Development, Shanghai, 201203, China.
| | - Yiqiong Pu
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
18
|
Bischoff-Kont I, Primke T, Niebergall LS, Zech T, Fürst R. Ginger Constituent 6-Shogaol Inhibits Inflammation- and Angiogenesis-Related Cell Functions in Primary Human Endothelial Cells. Front Pharmacol 2022; 13:844767. [PMID: 35281937 PMCID: PMC8914105 DOI: 10.3389/fphar.2022.844767] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
Rhizomes from Zingiber officinale Roscoe are traditionally used for the treatment of a plethora of pathophysiological conditions such as diarrhea, nausea, or rheumatoid arthritis. While 6-gingerol is the pungent principle in fresh ginger, in dried rhizomes, 6-gingerol is dehydrated to 6-shogaol. 6-Shogaol has been demonstrated to exhibit anticancer, antioxidative, and anti-inflammatory actions more effectively than 6-gingerol due to the presence of an electrophilic Michael acceptor moiety. In vitro, 6-shogaol exhibits anti-inflammatory actions in a variety of cell types, including leukocytes. Our study focused on the effects of 6-shogaol on activated endothelial cells. We found that 6-shogaol significantly reduced the adhesion of leukocytes onto lipopolysaccharide (LPS)-activated human umbilical vein endothelial cells (HUVECs), resulting in a significantly reduced transmigration of THP-1 cells through an endothelial cell monolayer. Analyzing the mediators of endothelial cell–leukocyte interactions, we found that 30 µM of 6-shogaol blocked the LPS-triggered mRNA and protein expression of cell adhesion molecules. In concert with this, our study demonstrates that the LPS-induced nuclear factor κB (NFκB) promoter activity was significantly reduced upon treatment with 6-shogaol. Interestingly, the nuclear translocation of p65 was slightly decreased, and protein levels of the LPS receptor Toll-like receptor 4 remained unimpaired. Analyzing the impact of 6-shogaol on angiogenesis-related cell functions in vitro, we found that 6-shogaol attenuated the proliferation as well as the directed and undirected migration of HUVECs. Of note, 6-shogaol also strongly reduced the chemotactic migration of endothelial cells in the direction of a serum gradient. Moreover, 30 µM of 6-shogaol blocked the formation of vascular endothelial growth factor (VEGF)-induced endothelial sprouts from HUVEC spheroids and from murine aortic rings. Importantly, this study shows for the first time that 6-shogaol exhibits a vascular-disruptive impact on angiogenic sprouts from murine aortae. Our study demonstrates that the main bioactive ingredient in dried ginger, 6-shogaol, exhibits beneficial characteristics as an inhibitor of inflammation- and angiogenesis-related processes in vascular endothelial cells.
Collapse
Affiliation(s)
- Iris Bischoff-Kont
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Tobias Primke
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Lea S. Niebergall
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Zech
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt, Germany
- *Correspondence: Robert Fürst,
| |
Collapse
|
19
|
Dey S, Singh AK, Singh AK, Rawat K, Banerjee J, Agnihotri V, Upadhaya D. Critical pathways of oral squamous cell carcinoma: molecular biomarker and therapeutic intervention. Med Oncol 2022; 39:30. [DOI: 10.1007/s12032-021-01633-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
|
20
|
Cai Y, Sheng Z, Wang J. Xanthorrhizol inhibits non-small cell carcinoma (A549) cell growth and promotes apoptosis through modulation of PI3K/AKT and NF-κB signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:120-130. [PMID: 34664399 DOI: 10.1002/tox.23383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/12/2021] [Accepted: 09/25/2021] [Indexed: 06/13/2023]
Abstract
Xanthorrhizol (XNT) is a sesquiterpenoid agent isolated from Curcuma xanthorrhiza; It is known to exhibit various pharmacological activities including anti-cancer. We investigated the anti-cell proliferative and proapoptotic effects of XNT on Non-small cell carcinoma (A549) cells were analyzed by the generation of reactive oxygen species (ROS), alteration of mitochondrial membrane potential (ΔΨm), oxidative DNA damage, and apoptosis morphological changes were explored by Hoechst and AO/EtBr staining. Our study demonstrated that XNT treatment significantly reduced the viability of A549 cells in a concentration-dependent manner. We observed that XNT-induced oxidative stress-mediated apoptotic cell death by increasing intracellular ROS generation, depleting antioxidant levels, enhancing lipid peroxidation, increased apoptotic morphological changes, and % of DNA damage on human lung cancer cells. Furthermore, we observed that the XNT induce apoptosis through inhibits phosphorylation of PI3K, AKTand inhibit NF-κBp65 transcriptional signaling activity. In addition, XNT treatment alters the ΔΨm, thereby induces apoptosis was closely coordinated with the induction of pro-apoptotic markers Bax, Bad, caspase- 3, 9 and cytochrome c, and suppression of anti-apoptotic (Bcl-2, Bcl-XL) protein expression. According to our results, XNT-inducing apoptosis in A549 cells by causing oxidative damage and modulating apoptotic signaling events. Finally, XNT-induced apoptotic cell death was confirmed by the TUNEL assay. Therefore, XNT might be used as a chemotherapeutic agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Yong Cai
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhaoying Sheng
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiying Wang
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Kang H, Sun Y, Hu X, Liu L. Gigantol inhibits proliferation and enhanced oxidative stress-mediated apoptosis through modulating of Wnt/β-catenin signaling pathway in HeLa cells. J Biochem Mol Toxicol 2021; 36:e22944. [PMID: 34729850 DOI: 10.1002/jbt.22944] [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: 11/19/2020] [Revised: 08/10/2021] [Accepted: 10/18/2021] [Indexed: 11/07/2022]
Abstract
Cervical cancer is one of the leading malignant cancers that is the fourth prominent cause of malignancy-related mortality in women globally. There is a predominant validation to a beneficial target in Wnt/β-catenin signaling in cervical carcinogenesis as they are very much deregulated in cancer. Previous studies reported Gigantol (GG) showed suppressive properties on the Wnt/β-catenin pathway in other tumor cells, but no evidence is available regarding GG suppressing Wnt/β-catenin signaling cervical tumor cells. Hence, the current research was planned to examine the suppressive effects of GG on HeLa cells and investigate the mechanism of action. HeLa cells were treated by GG in various doses and then appraising cell viability, oxidant/antioxidant levels, ∆ѰM status, reactive oxygen species (ROS) generation, apoptosis, and cell proliferation via Wnt/β-catenin signaling. We observed that GG noticeably inhibits cell proliferation, increased ROS generation, lipid peroxidation, mitochondrial membrane depolarization (∆ѰM), and increased apoptotic morphological changes of nuclear fragmentation and condensation. Moreover, GG effectively enhances proapoptotic, decreased ∆ѰM and antioxidant amounts, and mitigated Wnt/β-catenin signaling. Concisely, these findings proved that activating apoptosis and suppression of cell proliferation in GG treated HeLa cells was documented by the alleviation of Wnt/β-catenin signaling. Therefore, this study suggested that GG might develop a therapeutic effect against cervical carcinogenesis.
Collapse
Affiliation(s)
- Huanan Kang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yiming Sun
- Department of Andrology, Heilongjiang Provincial Hospital of Traditional Chinese Medicine, Harbin, China
| | - Xijiao Hu
- Second Department of Gynecology, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Li Liu
- Department of Hysteroscopy, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
22
|
Kunnumakkara AB, Rana V, Parama D, Banik K, Girisa S, Henamayee S, Thakur KK, Dutta U, Garodia P, Gupta SC, Aggarwal BB. COVID-19, cytokines, inflammation, and spices: How are they related? Life Sci 2021; 284:119201. [PMID: 33607159 PMCID: PMC7884924 DOI: 10.1016/j.lfs.2021.119201] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/20/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cytokine storm is the exaggerated immune response often observed in viral infections. It is also intimately linked with the progression of COVID-19 disease as well as associated complications and mortality. Therefore, targeting the cytokine storm might help in reducing COVID-19-associated health complications. The number of COVID-19 associated deaths (as of January 15, 2021; https://www.worldometers.info/coronavirus/) in the USA is high (1199/million) as compared to countries like India (110/million). Although the reason behind this is not clear, spices may have some role in explaining this difference. Spices and herbs are used in different traditional medicines, especially in countries such as India to treat various chronic diseases due to their potent antioxidant and anti-inflammatory properties. AIM To evaluate the literature available on the anti-inflammatory properties of spices which might prove beneficial in the prevention and treatment of COVID-19 associated cytokine storm. METHOD A detailed literature search has been conducted on PubMed for collecting information pertaining to the COVID-19; the history, origin, key structural features, and mechanism of infection of SARS-CoV-2; the repurposed drugs in use for the management of COVID-19, and the anti-inflammatory role of spices to combat COVID-19 associated cytokine storm. KEY FINDINGS The literature search resulted in numerous in vitro, in vivo and clinical trials that have reported the potency of spices to exert anti-inflammatory effects by regulating crucial molecular targets for inflammation. SIGNIFICANCE As spices are derived from Mother Nature and are inexpensive, they are relatively safer to consume. Therefore, their anti-inflammatory property can be exploited to combat the cytokine storm in COVID-19 patients. This review thus focuses on the current knowledge on the role of spices for the treatment of COVID-19 through suppression of inflammation-linked cytokine storm.
Collapse
Affiliation(s)
- Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sahu Henamayee
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Uma Dutta
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | | | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | | |
Collapse
|
23
|
Jafarzadeh A, Jafarzadeh S, Nemati M. Therapeutic potential of ginger against COVID-19: Is there enough evidence? JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2021. [PMCID: PMC8492833 DOI: 10.1016/j.jtcms.2021.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In addition to the respiratory system, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strikes other systems, including the digestive, circulatory, urogenital, and even the central nervous system, as its receptor angiotensin-converting enzyme 2 (ACE2) is expressed in various organs, such as lungs, intestine, heart, esophagus, kidneys, bladder, testis, liver, and brain. Different mechanisms, in particular, massive virus replication, extensive apoptosis and necrosis of the lung-related epithelial and endothelial cells, vascular leakage, hyper-inflammatory responses, overproduction of pro-inflammatory mediators, cytokine storm, oxidative stress, downregulation of ACE2, and impairment of the renin-angiotensin system contribute to the COVID-19 pathogenesis. Currently, COVID-19 is a global pandemic with no specific anti-viral treatment. The favorable capabilities of the ginger were indicated in patients suffering from osteoarthritis, neurodegenerative disorders, rheumatoid arthritis, type 2 diabetes, respiratory distress, liver diseases and primary dysmenorrheal. Ginger or its compounds exhibited strong anti-inflammatory and anti-oxidative influences in numerous animal models. This review provides evidence regarding the potential effects of ginger against SARS-CoV-2 infection and highlights its antiviral, anti-inflammatory, antioxidative, and immunomodulatory impacts in an attempt to consider this plant as an alternative therapeutic agent for COVID-19 treatment.
Collapse
|
24
|
Shang H, Jang X, Shi L, Ma Y. Lycorine inhibits cell proliferation and induced oxidative stress-mediated apoptosis via regulation of the JAK/STAT3 signaling pathway in HT-3 cells. J Biochem Mol Toxicol 2021; 35:e22882. [PMID: 34558146 DOI: 10.1002/jbt.22882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/18/2021] [Accepted: 08/02/2021] [Indexed: 11/09/2022]
Abstract
Human cervical cancer is the fourth most common carcinoma in women in the world. The JAK/STAT3 signaling pathways crucially regulate cell growth and apoptosis. It is a significant target signaling pathway for the development of novel antitumor medicine. This study intended to explore whether lycorine could prevent HT-3 proliferation and induce apoptosis by targeting the JAK/STAT3 signaling cascade. The HT-3 cells were treated with various lycorine dosages and we analyzed cell growth, lipid peroxidation, antioxidants, mitochondrial membrane potential (ΔΨm), DNA damage, apoptosis markers by different in vitro methodologies. Our results revealed that lycorine substantially reserved cell growth via decreased antioxidants, augmented reactive oxygen species (ROS) generation which leads to loss of ΔΨm, increased nuclear crumbling and chromatin condensation, thus resulting in representative increased apoptotic cell death. Furthermore, we analyzed that the molecular mechanical action of lycorine considerably repressed JAK1/STAT3 transactional activation and decrease its downstream molecules Bcl-2, and enhances the expressional activity of Bax, cytochrome c, caspase 3 and 9 in HT-3 cells. Finally, the fact that N-acetylcysteine inhibits lycorine-induced ROS-mediated apoptosis was confirmed in HT-3 cells. Thus, the results indicate that lycorine efficiently enhances apoptosis and inhibits HT-3 cell proliferation. These outcomes collectively proposed that lycorine could be a beneficial chemotherapeutic agent for treating and managing human cervical carcinoma.
Collapse
Affiliation(s)
- Hui Shang
- Department of Obstetrics & Gynecology, Central Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province, China
| | - Xuena Jang
- Department of Obstetrics and Gynecology, Qingdao Longtian Jinqiu Women's and Children's Hospital, Qingdao, Shandong Province, China
| | - Lingyun Shi
- Department of Obstetrics & Gynecology, Qingdao Ninth People's Hospital, Qingdao, Shandong Province, China
| | - Yifei Ma
- Department of Obstetrics & Gynecology, Central Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province, China
| |
Collapse
|
25
|
Prakash S, Radha, Kumar M, Kumari N, Thakur M, Rathour S, Pundir A, Sharma AK, Bangar SP, Dhumal S, Singh S, Thiyagarajan A, Sharma A, Sharma M, Changan S, Sasi M, Senapathy M, Pradhan PC, Garg NK, Ilakiya T, Nitin M, Abdel-Daim MM, Puri S, Natta S, Dey A, Amarowicz R, Mekhemar M. Plant-Based Antioxidant Extracts and Compounds in the Management of Oral Cancer. Antioxidants (Basel) 2021; 10:1358. [PMID: 34572990 PMCID: PMC8466097 DOI: 10.3390/antiox10091358] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 12/11/2022] Open
Abstract
Oral cancer continues to be a leading cause of death worldwide, and its prevalence is particularly high in developing countries, where people chew tobacco and betel nut on a regular basis. Radiation-, chemo-, targeted-, immuno-, and hormone-based therapies along with surgery are commonly used as part of a treatment plan. However, these treatments frequently result in various unwanted short- to long-term side effects. As a result, there is an urgent need to develop treatment options for oral cancer that have little or no adverse effects. Numerous bioactive compounds derived from various plants have recently attracted attention as therapeutic options for cancer treatment. Antioxidants found in medicinal plants, such as vitamins E, C, and A, reduce damage to the mucosa by neutralizing free radicals found in various oral mucosal lesions. Phytochemicals found in medicinal plants have the potential to modulate cellular signalling pathways that alter the cellular defence mechanisms to protect normal cells from reactive oxygen species (ROS) and induce apoptosis in cancer cells. This review aims to provide a comprehensive overview of various medicinal plants and phytoconstituents that have shown the potential to be used as oral cancer therapeutics.
Collapse
Affiliation(s)
- Suraj Prakash
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India; (S.P.); (N.K.); (M.T.); (S.R.); (S.P.)
| | - Radha
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India; (S.P.); (N.K.); (M.T.); (S.R.); (S.P.)
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR—Central Institute for Research on Cotton Technology, Mumbai 400019, India
| | - Neeraj Kumari
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India; (S.P.); (N.K.); (M.T.); (S.R.); (S.P.)
| | - Mamta Thakur
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India; (S.P.); (N.K.); (M.T.); (S.R.); (S.P.)
| | - Sonia Rathour
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India; (S.P.); (N.K.); (M.T.); (S.R.); (S.P.)
| | - Ashok Pundir
- School of Mechanical and Civil Engineering, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India;
| | - Abhishek Kumar Sharma
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India;
| | - Sneh Punia Bangar
- Department of Food, Nutrition, & Packaging Sciences, Clemson University, Clemson, SC 29634, USA;
| | - Sangram Dhumal
- Division of Horticulture, RCSM College of Agriculture, Kolhapur 416004, India;
| | - Surinder Singh
- Dr. S.S. Bhatnagar University Institute of Chemical Engineering and Technology, Panjab University, Chandigarh 160014, India;
| | - Anitha Thiyagarajan
- Department of Postharvest Technology, Horticultural College and Research Institute, Periyakulam 625604, India;
| | - Anshu Sharma
- Department of Food Science and Technology, Dr. Y.S. Parmar University of Horticulture and Forestry, Nauni 173230, India;
| | - Munisha Sharma
- Sri Shankara Cancer Hospital and Research Centre, Bengaluru 560004, India;
| | - Sushil Changan
- Division of Crop Physiology, Biochemistry and Post-Harvest Technology, ICAR-Central Potato Research Institute, Shimla 171001, India;
| | - Minnu Sasi
- Division of Biochemistry, ICAR—Indian Agricultural Research Institute, New Delhi 110012, India;
| | - Marisennayya Senapathy
- Department of Rural Development and Agricultural Extension, College of Agriculture, Wolaita Sodo University, Wolaita Sodo, SNNPR, Ethiopia;
| | - Prakash Chandra Pradhan
- Division of Agricultural Chemicals, ICAR—Indian Agricultural Research Institute, New Delhi 110012, India;
| | - Nitin Kumar Garg
- Division of Biochemistry, Sri Karan Narendra Agriculture University, Jobner 303329, India;
| | - Tamilselvan Ilakiya
- Department of Vegetable Science, Tamil Nadu Agricultural University, Coimbatore 641003, India;
| | - Mukesh Nitin
- Department of Tech. Biosciences, Digianalix, South Samaj Street, Tharpakhna, Ranchi 834001, India;
| | - Mohamed M. Abdel-Daim
- Pharmacy Program, Department of Phamaceutical Sciences, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Sunil Puri
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India; (S.P.); (N.K.); (M.T.); (S.R.); (S.P.)
| | - Suman Natta
- ICAR—National Research Centre for Orchids, Pakyong 737106, India;
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India;
| | - Ryszard Amarowicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland;
| | - Mohamed Mekhemar
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrecht’s University, 24105 Kiel, Germany
| |
Collapse
|
26
|
Zhang Z, Pan Y, Zhao Y, Ren M, Li Y, Lu G, Wu K, He S. Delphinidin modulates JAK/STAT3 and MAPKinase signaling to induce apoptosis in HCT116 cells. ENVIRONMENTAL TOXICOLOGY 2021; 36:1557-1566. [PMID: 33955636 DOI: 10.1002/tox.23152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/06/2021] [Accepted: 04/11/2021] [Indexed: 06/12/2023]
Abstract
Delphinidin is an anthocyanin that belongs to the group of flavonoids that exert numerous biological activities. However, the molecular mechanisms underlying the anticancer effects of delphinidin remain poorly understood. In our study we analyzed delphinidin modulate STAT-3 and MAPKinase signaling thereby inhbits cell proliferation and promote apoptosis. Our study demonstrated that delphinidin treatment significantly reduced the viability of human colon cancer HCT116 in a concentration-dependent manner. We noticed that delphinidin effectively induced oxidative stress-mediated apoptosis by generating intracellular ROS, decreasing antioxidant levels, inducing lipid peroxidation, and single-strand break on colon cancer cells. In this study, we observed that delphinidin treatment alters the mitochondrial membrane potential, thereby induces apoptosis was closely associated with the induction of pro-apoptotic Bax, Caspase- 3,8 & 9, cytochrome C, and inhibition of anti-apoptotic protein expression. Studies on STAT-3 and MAPKinase signaling showed delphinidin inhibited the phosphorylation of these transcription factors' activity. Inhibition of STAT-3, p38, and ERK1/2 phosphorylation and modulation pro-apoptotic protein expression might be responsible for the anticancer activity of delphinidin in colon cancer cells.
Collapse
Affiliation(s)
- Zhiyong Zhang
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yan Pan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Zhao
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mudan Ren
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yarui Li
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guifang Lu
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuixiang He
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
27
|
Bischoff-Kont I, Fürst R. Benefits of Ginger and Its Constituent 6-Shogaol in Inhibiting Inflammatory Processes. Pharmaceuticals (Basel) 2021; 14:ph14060571. [PMID: 34203813 PMCID: PMC8232759 DOI: 10.3390/ph14060571] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Ginger (Zingiber officinale Roscoe) is widely used as medicinal plant. According to the Committee on Herbal Medicinal Products (HMPC), dried powdered ginger rhizome can be applied for the prevention of nausea and vomiting in motion sickness (well-established use). Beyond this, a plethora of pre-clinical studies demonstrated anti-cancer, anti-oxidative, or anti-inflammatory actions. 6-Shogaol is formed from 6-gingerol by dehydration and represents one of the main bioactive principles in dried ginger rhizomes. 6-Shogaol is characterized by a Michael acceptor moiety being reactive with nucleophiles. This review intends to compile important findings on the actions of 6-shogaol as an anti-inflammatory compound: in vivo, 6-shogaol inhibited leukocyte infiltration into inflamed tissue accompanied with reduction of edema swelling. In vitro and in vivo, 6-shogaol reduced inflammatory mediator systems such as COX-2 or iNOS, affected NFκB and MAPK signaling, and increased levels of cytoprotective HO-1. Interestingly, certain in vitro studies provided deeper mechanistic insights demonstrating the involvement of PPAR-γ, JNK/Nrf2, p38/HO-1, and NFκB in the anti-inflammatory actions of the compound. Although these studies provide promising evidence that 6-shogaol can be classified as an anti-inflammatory substance, the exact mechanism of action remains to be elucidated. Moreover, conclusive clinical data for anti-inflammatory actions of 6-shogaol are largely lacking.
Collapse
Affiliation(s)
- Iris Bischoff-Kont
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, 60438 Frankfurt, Germany;
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, 60438 Frankfurt, Germany;
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), 60325 Frankfurt, Germany
- Correspondence:
| |
Collapse
|
28
|
Xu P, Xiao J, Chi S. Piperlongumine attenuates oxidative stress, inflammatory, and apoptosis through modulating the GLUT-2/4 and AKT signaling pathway in streptozotocin-induced diabetic rats. J Biochem Mol Toxicol 2021; 35:1-12. [PMID: 33724628 DOI: 10.1002/jbt.22763] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/14/2020] [Accepted: 03/02/2021] [Indexed: 01/14/2023]
Abstract
The current study was done to measure the role of piperlongumine (PL) on hyperglycemia interrelated oxidative stress-mediated inflammation and apoptosis, inflammatory stress, and the diabetic insulin receptor substrate 2 (IRS2), protein kinase B (AKT), and glucose transporter 2 (GLUT-2)/4 signaling pathway in streptozotocin (STZ)-persuaded diabetic animals. Diabetes was initiated in experimental animals via a single dose intraperitoneal inoculation of STZ. Diabetic rats revealed an augmented blood-glucose level with drastically diminished plasma-insulin status. The functions of antioxidants were diminished with enhanced lipid peroxidation, conjugated dienes, and protein carbonyls noticed in diabetic rats' plasma and pancreatic tissues. An elevation of nuclear factor-κB (NF-κB), tumor necrosis factor-α, and interleukin-6 proteins was noticed in pancreatic tissues as well as IRS2, AKT, GLUT-2, and GLUT-4 marker expressions were quantified in the hepatic tissue of control and diabetic rats. Oral administration of PL for 30 days drastically lowered glucose and higher insulin status in STZ-induced diabetic rats. Impressively, PL oral supplementation considerably restored the antioxidant levels and reduced inflammation and diabetic marker expressions in STZ-diabetic rats. These results were supported through a histological study. Moreover, PL also augmented the level of B-cell lymphoma 2 and diminished the level of Bcl-2-associated X protein in STZ-treated rat's hepatic tissues. Thus, we concluded that PL excellently rescued pancreatic β cells through mitigating hyperglycemia via dynamic insulin secretion, activating antioxidants, and inhibiting inflammation and apoptosis in the pancreatic and hepatic tissue of diabetic rats.
Collapse
Affiliation(s)
- Ping Xu
- Department of Endocrinology and Metabolism, Shenzhen People's Hospital (Second Clinical Medical Collage of Jinan University), Shenzhen, Guangdong, China
| | - Juan Xiao
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Shuixia Chi
- Department of Traditional Chinese Medicine, Xianyang Central Hospital, Xianyang, China
| |
Collapse
|
29
|
Zivarpour P, Nikkhah E, Maleki Dana P, Asemi Z, Hallajzadeh J. Molecular and biological functions of gingerol as a natural effective therapeutic drug for cervical cancer. J Ovarian Res 2021; 14:43. [PMID: 33706784 PMCID: PMC7953815 DOI: 10.1186/s13048-021-00789-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Cervical cancer is one of the most common and important gynecological cancers, which has a global concern with an increasing number of patients and mortality rates. Today, most women in the world who suffer from cervical cancer are developing advanced stages of the disease. Smoking and even exposure to secondhand smoke, infections caused by the human papillomavirus, immune system dysfunction and high-risk individual-social behaviors are among the most important predisposing factors for this type of cancer. In addition, papilloma virus infection plays a more prominent role in cervical cancer. Surgery, chemotherapy or radical hysterectomy, and radiotherapy are effective treatments for this condition, the side effects of these methods endanger a person's quality of life and cause other problems in other parts of the body. Studies show that herbal medicines, including taxol, camptothecin and combretastatins, have been shown to be effective in treating cervical cancer. Ginger (Zingiber officinale, Zingiberaceae) is one of the plants with valuable compounds such as gingerols, paradols and shogoals, which is a rich source of antioxidants, anti-cancer and anti-inflammatory agents. Numerous studies have reported the therapeutic effects of this plant through various pathways in cervical cancer. In this article, we look at the signaling mechanisms and pathways in which ginger is used to treat cervical cancer.
Collapse
Affiliation(s)
- Parinaz Zivarpour
- Department of Biological sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Elhameh Nikkhah
- Medicinal Plants Research Cent Maragheh University of Medical Sciences, Maragheh, Iran
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
30
|
Ooi SL, Campbell R, Pak SC, Golombick T, Manoharan A, Ramakrishna R, Badmaev V, Schloss J. Is 6-Shogaol an Effective Phytochemical for Patients With Lower-risk Myelodysplastic Syndrome? A Narrative Review. Integr Cancer Ther 2021; 20:15347354211065038. [PMID: 34930049 PMCID: PMC8728773 DOI: 10.1177/15347354211065038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/02/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022] Open
Abstract
Myelodysplastic syndrome (MDS) evolves due to genomic instability, dysregulated signaling pathways, and overproduction of inflammatory markers. Reactive oxygen species contribute to the inflammatory response, which causes gene damage, cellular remodeling, and fibrosis. MDS can be a debilitating condition, and management options in patients with MDS aim to improve cytopenias, delay disease progression, and enhance quality of life. High serum ferritin levels, a source of iron for reactive oxygen species production, correlate with a higher risk of progression to acute myeloid leukemia, and iron overload is compounded by blood transfusions given to improve anemia. 6-shogaol is a natural phenolic compound formed when ginger is exposed to heat and/or acidic conditions, and it has been shown to possess anti-tumor activity against leukemia cell lines and antioxidant effects. This narrative review assessed the potential benefits of this phytochemical in lower-risk MDS patients through examining the current evidence on the pharmacological and therapeutic properties of ginger and 6-shogaol.
Collapse
Affiliation(s)
| | - Ron Campbell
- Charles Sturt University, Bathurst,
NSW, Australia
- The Oaks Medical Practice, The Oaks,
NSW, Australia
| | | | | | - Arumugam Manoharan
- Southern Sydney Haematology, Kogarah,
NSW, Australia
- University of Wollongong Australia,
Wollongong NSW, Australia
| | - Raj Ramakrishna
- Southern Sydney Haematology, Kogarah,
NSW, Australia
- University of Wollongong Australia,
Wollongong NSW, Australia
| | | | | |
Collapse
|
31
|
Sun Y, Ren J, Wang F. [6]-Gingerol impedes 7,12-dimethylbenz(a)anthracene-induced inflammation and cell proliferation-associated hamster buccal pouch carcinogenesis through modulating Nrf2 signaling events. J Biochem Mol Toxicol 2020; 35:e22689. [PMID: 33347680 DOI: 10.1002/jbt.22689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/22/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
The present study examines the chemopreventive role of [6]-gingerol, an active component of ginger, on 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinogenesis models. The HBP has been developed with an addition of 0.5% of DMBA to the HBP area three times per week, up to the end of the 16th experimental week. At the end of the experiment, we noticed 100% tumor incidence and precancerous lesions, such as dysplasia, hyperplasia, keratosis, and well-differentiated squamous cell carcinoma, in DMBA-induced HBP. Furthermore, we observed that [6]-gingerol inhibited the increased thiobarbituric acid-reactive substances and decreased antioxidant levels in DMBA-induced hamsters. Moreover, [6]-gingerol inhibits DMBA-exposed over expression of inflammatory markers (inducible nitric oxide synthase, interleukin [IL]-1β, IL-6, cyclooxygenase-2, and tumor necrosis factor-α) and cell proliferation markers (cyclin D1, proliferating cell nuclear antigen); induces proapoptotic markers in HBP. Nuclear factor erythroid-2-related factor-2 (Nrf2) is a major antioxidant transcription factor, which regulates the antioxidant gene-dependent scavenge of tumor proliferation and apoptosis. Overexpression of Nrf2 signaling plays a pivotal role and can be a novel target in preventing carcinogenesis. In this study, [6]-gingerol restores the DMBA-induced depletion of Nrf2 signaling and thereby prevents buccal pouch carcinogenesis in hamsters. These results point out that [6]-gingerol impedes the responses of inflammatory and cell proliferation-associated progression of cancer through the action of Nrf2 signaling.
Collapse
Affiliation(s)
- Yugang Sun
- Oral and maxillofacial surgery, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Jinmin Ren
- Health Management Center, Binzhou Municipal Hospital of Traditional Chinese Medicine, Binzhou, Shandong, China
| | - Fang Wang
- Department of Oncology, The Second People Hospital of Dezhou, Dezhou, Shandong, China
| |
Collapse
|
32
|
Hu X, Wang L, Zhang L, Zhang T. β-Elemene inhibits 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-induced skin tumorigenesis through suppression of NF-κB-associated signaling events in the mouse skin model. J Biochem Mol Toxicol 2020; 34:e22550. [PMID: 32662567 DOI: 10.1002/jbt.22550] [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: 12/25/2019] [Revised: 03/19/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022]
Abstract
β-Elemene (1-methyl-1-vinyl-2,4-diisopropenyl-cyclohexane), a natural sesquiterpene-derived curcumae radix, exhibits a variety of pharmacologic properties including anticancer. However, the molecular action of β-elemene in chemical-induced skin carcinogenesis remains unclear. Therefore, the present study executes to investigate a possible effect of β-elemene in the 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA)-promoted skin tumor model. The experimental mice were subjected to execute two-stage skin carcinogenesis and it has been initiated by the addition of DMBA on the dorsal portion of the mouse skin. One week after, for chemical carcinogen of mice, topical exposure of DMBA has been induced following with TPA (5 nmol) in acetone (200 μL) given weekly twice for 20 weeks respectively. After completion of the experimental period, we noticed that 100% of tumor incidence, histopathological changes, decreased lipid peroxidation (LPO), and decreased antioxidant levels in DMBA/TPA-promoted skin carcinogenesis. Furthermore, enhanced activity of inflammatory protein markers (nuclear factor [NF]-κB, tumor necrosis factor-α, interleukin-6, cyclooxygenase-2, and nitric oxide synthase) and cell-proliferative messenger RNA markers (PCNA, cyclin D1), and increased antiapoptotic protein Bcl-2; decreased proapoptotic protein marker events Bax and caspase 3 and 9 expressions were noticed in DMBA/TPA promoted skin tissue. In this study, we noticed that β-elemene noticeably reversed the histopathological changes and antioxidant levels in tumor-bearing mice. Conversely, β-elemene effectively inhibits inflammation, cell proliferation events, and enhances proapoptotic factors, by suppression of NF-κB transcriptional activation in DMBA/TPA animals. Thus, we concluded that β-elemene prevents DMBA/TPA promoted skin carcinogenesis through its antioxidant and abate inflammation markers and cell-proliferative markers also activating proapoptotic molecules.
Collapse
Affiliation(s)
- Xing Hu
- Department of Dermatology, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Liang Wang
- Department of Spine and Joint, The First People's Hospital of Pingyuan County, Dezhou, Shandong, China
| | - Lili Zhang
- Department of Oncology and Hematology, The First People's Hospital of Pingyuan County, Dezhou, Shandong, China
| | - Tao Zhang
- TCM Internal Medicine, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
33
|
Ahmad R, Khan MA, Srivastava A, Gupta A, Srivastava A, Jafri TR, Siddiqui Z, Chaubey S, Khan T, Srivastava AK. Anticancer Potential of Dietary Natural Products: A Comprehensive Review. Anticancer Agents Med Chem 2020; 20:122-236. [DOI: 10.2174/1871520619666191015103712] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
Nature is a rich source of natural drug-like compounds with minimal side effects. Phytochemicals
better known as “Natural Products” are found abundantly in a number of plants. Since time immemorial, spices
have been widely used in Indian cuisine as flavoring and coloring agents. Most of these spices and condiments
are derived from various biodiversity hotspots in India (which contribute 75% of global spice production) and
form the crux of India’s multidiverse and multicultural cuisine. Apart from their aroma, flavor and taste, these
spices and condiments are known to possess several medicinal properties also. Most of these spices are mentioned
in the Ayurveda, the indigenous system of medicine. The antimicrobial, antioxidant, antiproliferative,
antihypertensive and antidiabetic properties of several of these natural products are well documented in
Ayurveda. These phytoconstituemts are known to act as functional immunoboosters, immunomodulators as well
as anti-inflammatory agents. As anticancer agents, their mechanistic action involves cancer cell death via induction
of apoptosis, necrosis and autophagy. The present review provides a comprehensive and collective update
on the potential of 66 commonly used spices as well as their bioactive constituents as anticancer agents. The
review also provides an in-depth update of all major in vitro, in vivo, clinical and pharmacological studies done
on these spices with special emphasis on the potential of these spices and their bioactive constituents as potential
functional foods for prevention, treatment and management of cancer.
Collapse
Affiliation(s)
- Rumana Ahmad
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Mohsin A. Khan
- Chancellor, Era University, Sarfarazganj, Hardoi Road, Lucknow-226003, UP, India
| | - A.N. Srivastava
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Anamika Gupta
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Aditi Srivastava
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tanvir R. Jafri
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Zainab Siddiqui
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Sunaina Chaubey
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tahmeena Khan
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow 226026, UP, India
| | - Arvind K. Srivastava
- Department of Food and Nutrition, Era University, Sarfarazganj, Lucknow-226003, UP, India
| |
Collapse
|
34
|
Han Y, Li X, Zhang X, Gao Y, Qi R, Cai R, Qi Y. Isodeoxyelephantopin, a sesquiterpene lactone from Elephantopus scaber Linn., inhibits pro-inflammatory mediators' production through both NF-κB and AP-1 pathways in LPS-activated macrophages. Int Immunopharmacol 2020; 84:106528. [PMID: 32335480 DOI: 10.1016/j.intimp.2020.106528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/11/2020] [Accepted: 04/17/2020] [Indexed: 12/19/2022]
Abstract
Isodeoxyelephantopin (IDET) has been identified as an anti-tumor natural constituent whose anti-tumor activity and mechanism have been widely investigated. Since the occurrence and development of cancer usually accompany with inflammation, and tumor signaling shares many components with inflammation signaling, the agents with anti-tumor activity are likely to possess anti-inflammation potential. Thus, the current study aims to demonstrate the anti-inflammatory activity along with the underlying mechanism of IDET in lipopolysaccharide (LPS)-primed macrophages. By using Griess method and ELISA, we found that in both bone marrow derived macrophages and alveolar macrophage cell line, IDET, at relatively low concentrations (0.75, 1.5 and 3 μM), could inhibit LPS-induced expression of various pro-inflammatory mediators including nitric oxide (NO) generated by inducible nitric oxide synthase (iNOS), interleukin (IL)-6, monocyte chemotactic protein-1 (MCP-1) and IL-1β. Meanwhile, in activated MH-S cells, the inhibitory action of IDET on mRNA expression levels of these cytokines was also detected using qPCR. Mechanistically, the effects of IDET on two key inflammatory signalings, nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) pathways, were determined in LPS-activated MH-S cells by reporter gene along with western blot assays. On the one hand, IDET suppressed NF-κB signaling via down-regulating phosphorylation and degradation of inhibitor of NF-κB (IκB)-α and the subsequent p65 translocation. On the other hand, IDET dampened AP-1 signaling through attenuating phosphorylation of both c-jun N-terminal kinase 1/2 (JNK1/2) and extracellular signal regulated kinase 1/2 (ERK1/2). Our study indicates that IDET might be a promising constituent from the anti-inflammatory herb Elephantopus scaber Linn. in mitigating inflammatory conditions, especially respiratory inflammation.
Collapse
Affiliation(s)
- Yixin Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ximeng Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xiaoyu Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ruijuan Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Runlan Cai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
35
|
Sabitha R, Nishi K, Gunasekaran VP, Agilan B, David E, Annamalai G, Vinothkumar R, Perumal M, Subbiah L, Ganeshan M. p-Coumaric acid attenuates alcohol exposed hepatic injury through MAPKs, apoptosis and Nrf2 signaling in experimental models. Chem Biol Interact 2020; 321:109044. [PMID: 32151596 DOI: 10.1016/j.cbi.2020.109044] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 02/26/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023]
Abstract
Overconsumption of alcohol could lead to severe liver injury that connects with oxidative stress, apoptosis, and inflammatory response. Previously, we proved that p-coumaric acid prevents ethanol induced reproductive toxicity; however, p-coumaric acid (PCA) on ethanol mediated hepatotoxicity has not been examined yet. In our work, we sought to study the potential of PCA in contradiction of ethanol induced hepatoxicity which linking with MAPKs, apoptosis, oxidative stress, and Nrf2 signaling. Foremost, we found that PCA could protect ethanol induced both L-02 and HepG2 hepatic cells by inhibiting cytotoxicity, ROS production, mitochondrial depolarization, and nuclear fragmentation. Also, in vivo experiments showed that the ethanol increasing the lipid markers (TBARS, CD) and depletes the antioxidants thereby increased phosphorylation of JNK, ERK, and p38 in rat liver tissues. Interestingly, PCA treatments inhibit ethanol exposed lipid markers and depletion of antioxidants, which directs the inhibition of MAPKs activation in rat liver tissues. We also noticed that the PCA protected ethanol induced apoptosis and liver markers by inhibiting the expression of Bax, caspases; AST, ALT, ALS, and LDH in liver tissue. Overall, the ameliorative consequence of PCA on ethanol induced oxidative stress and apoptosis was achieved by suppressing the expression of CYP2E1 and overexpressing Nrf2 and its target protein HO-1 in rat liver tissue. As a result, PCA was marked to be an effective antioxidant with notable hepatoprotection by inhibiting MAPKs and apoptosis signaling via enhancing Nrf2 signaling.
Collapse
Affiliation(s)
- Ramakrishnan Sabitha
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Kumari Nishi
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | | | - Balupillai Agilan
- Department of Biotechnology, Thiruvalluvar University, Serkadu, 632115, Vellore, Tamil Nadu, India; Department of Biochemistry and Biotechnology, Annnamalai University, Annamalainagar, 608 002, Tamil Nadu, India
| | - Ernest David
- Department of Biotechnology, Thiruvalluvar University, Serkadu, 632115, Vellore, Tamil Nadu, India
| | - Govindhan Annamalai
- Department of Biochemistry and Biotechnology, Annnamalai University, Annamalainagar, 608 002, Tamil Nadu, India
| | - Rajamanickam Vinothkumar
- Key Laboratory of Imaging Diagnosis and Minimally Invasive and Intervention Research, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Malliga Perumal
- Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Latha Subbiah
- Department of Pharmaceutical Technology, Anna University, Bharathidasan Institute of Technology Campus, Tiruchirappalli, 620 024, Tamil Nadu, India
| | - Mathan Ganeshan
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India.
| |
Collapse
|
36
|
Su P, Veeraraghavan VP, Krishna Mohan S, Lu W. A ginger derivative, zingerone-a phenolic compound-induces ROS-mediated apoptosis in colon cancer cells (HCT-116). J Biochem Mol Toxicol 2019; 33:e22403. [PMID: 31714660 DOI: 10.1002/jbt.22403] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/30/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022]
Abstract
Zingerone (ZO), an active phenolic agent derived from Zingiber officinale (Ginger), has many pharmacological properties such as antioxidant, antiangiogenic, and antitumor. However, its potential value in cancer and the mechanism by which ZO wields its therapeutic effects remain obscure. Therefore, in this current study, we explored the effects of ZO on suppressing cell proliferation and enhancing apoptosis in colon cancer cells (HCT116). Our results indicated that ZO significantly enhances the production of reactive oxygen species, lipid peroxidation (thiobarbituric acid reactive substance [TBARS]), and loss of cell viability; and reduces mitochondrial membrane potential and antioxidant levels (SOD, CAT, and GSH) in ZO-treated HCT116 cells in a dose-dependent (2.5, 5, and 10 µM) manner. Furthermore, ZO induces oxidative stress-mediated apoptosis as evidenced by apoptotic morphological changes predicted by AO/EtBr, Hoechst staining and further confirmed by comet assay. Moreover, immunoblotting techniques showed that ZO treatment effectively enhances Bax, caspase-9, and caspase-3 expressions and decreases the expression of Bcl-2 in colon cancer cells. Together, our results evidenced that the antitumor effects of ZO reduce cell proliferation and stimulate apoptosis through modulating pro- and antiapoptotic molecular events in HCT116 colon cancer cells. Therefore, based on our findings, ZO may be used as a therapeutic agent for the treatment of colon cancer.
Collapse
Affiliation(s)
- Ping Su
- Department of Anorectal, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi Province, China
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Surapaneni Krishna Mohan
- Department of Medical Biochemistry, College of Applied Medical Sciences-Jubail (CAMSJ), Imam Abdulrahman Bin Faisal University, Al Jubail, Kingdom of Saudi Arabia (KSA)
| | - Wang Lu
- Department of Gastroenterology, The Second Affiliated Hospital of Air Force Medical University of People's Liberation Army, Xi'an, Shaanxi, China
| |
Collapse
|
37
|
Rossa C, D'Silva NJ. Immune-relevant aspects of murine models of head and neck cancer. Oncogene 2019; 38:3973-3988. [PMID: 30696955 PMCID: PMC6533118 DOI: 10.1038/s41388-019-0686-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/26/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
Head and neck cancers (HNCs) cause significant mortality and morbidity. There have been few advances in therapeutic management of HNC in the past 4 to 5 decades, which support the need for studies focusing on HNC biology. In recent years, increased recognition of the relevance of the host response in cancer progression has led to novel therapeutic strategies and putative biomarkers of tumor aggressiveness. However, tumor-immune interactions are highly complex and vary with cancer type. Pre-clinical, in vivo models represent an important and necessary step in understanding biological processes involved in development, progression and treatment of HNC. Rodents (mice, rats, hamsters) are the most frequently used animal models in HNC research. The relevance and utility of information generated by studies in murine models is unquestionable, but it is also limited in application to tumor-immune interactions. In this review, we present information regarding the immune-specific characteristics of the murine models most commonly used in HNC research, including immunocompromised and immunocompetent animals. The particular characteristics of xenograft, chemically induced, syngeneic, transgenic, and humanized models are discussed in order to provide context and insight for researchers interested in the in vivo study of tumor-immune interactions in HNC.
Collapse
Affiliation(s)
- Carlos Rossa
- Department of Diagnosis and Surgery, UNESP-State University of Sao Paulo, School of Dentistry at Araraquara, Araraquara - SP, Brazil. .,Department of Periodontics and Oral Medicine, School of Dentistry, Ann Arbor, MI, 48109, USA.
| | - Nisha J D'Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, Ann Arbor, MI, 48109, USA. .,Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
38
|
范 艺, 唐 华, 刘 瑶, 李 成. [Establishment of a rat model of dimethylbenzanthracene-induced vulvar squamous intraepithelial lesions]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1318-1324. [PMID: 30514679 PMCID: PMC6744125 DOI: 10.12122/j.issn.1673-4254.2018.11.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish a SD rat model of vulvar squamous intraepithelial lesions. METHODS Seventy female SD rats were randomized into 4 groups, namely the blank control group (n=10), mechanical irritation group (n=10), acetone solution group (n=10), and mechanical irritation with DMBA acetone solution group (n=40, model group), and the corresponding treatments were administered 3 times a week for 14 weeks. The changes of the vulvar skin of the rats were observed regularly until the 18th week. The expression of mutant p53 (mtp53) and vascular endothelial growth factor (VEGF) proteins were detected using immunohistochemistry and Western blotting, and the expressions of mtp53 and VEGF mRNA were detected with qRT- PCR in the blank control group and model group. RESULTS No significant differences were found in the morphological or histopathological changes of the skin among the blank control group, mechanical irritation group and acetone solution group. In the model group, low-grade squamous intraepithelial lesions (LSIL) occurred in 28 rats (70%) and high-grade squamous intraepithelial lesions (HSIL) in 11 rats (27.5%) at 14 weeks, with a success rate of 97.5% in inducing vulvar squamous intraepithelial lesions. Compared with the blank control group, the rats in the model group showed significantly increased expressions of mtp53 and VEGF at both the protein level (P < 0.05) and the mRNA level (P < 0.05). CONCLUSIONS DMBA in acetone solution combined with mechanical irritation can induce vulvar squamous intraepithelial lesions in female SD rats.
Collapse
Affiliation(s)
- 艺巾 范
- />重庆医科大学生物医学工程学院//省部共建国家重点实验室培育基地—重庆市超声医学工程重点实验室//重 庆市生物医学工程学重点实验室//重庆市微无创医学协同创新中心,重庆 400016College of Biomedical Engineering, Chongqing Medical University/State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology/Chongqing Key Laboratory of Biomedical Engineering/ Chongqing Collaborative Innovation Center for Minimally Invasive and Noninvasive Medicine, Chongqing 400016, China
| | - 华均 唐
- />重庆医科大学生物医学工程学院//省部共建国家重点实验室培育基地—重庆市超声医学工程重点实验室//重 庆市生物医学工程学重点实验室//重庆市微无创医学协同创新中心,重庆 400016College of Biomedical Engineering, Chongqing Medical University/State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology/Chongqing Key Laboratory of Biomedical Engineering/ Chongqing Collaborative Innovation Center for Minimally Invasive and Noninvasive Medicine, Chongqing 400016, China
| | - 瑶 刘
- />重庆医科大学生物医学工程学院//省部共建国家重点实验室培育基地—重庆市超声医学工程重点实验室//重 庆市生物医学工程学重点实验室//重庆市微无创医学协同创新中心,重庆 400016College of Biomedical Engineering, Chongqing Medical University/State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology/Chongqing Key Laboratory of Biomedical Engineering/ Chongqing Collaborative Innovation Center for Minimally Invasive and Noninvasive Medicine, Chongqing 400016, China
| | - 成志 李
- />重庆医科大学生物医学工程学院//省部共建国家重点实验室培育基地—重庆市超声医学工程重点实验室//重 庆市生物医学工程学重点实验室//重庆市微无创医学协同创新中心,重庆 400016College of Biomedical Engineering, Chongqing Medical University/State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology/Chongqing Key Laboratory of Biomedical Engineering/ Chongqing Collaborative Innovation Center for Minimally Invasive and Noninvasive Medicine, Chongqing 400016, China
| |
Collapse
|
39
|
Jafarzadeh A, Nemati M. Therapeutic potentials of ginger for treatment of Multiple sclerosis: A review with emphasis on its immunomodulatory, anti-inflammatory and anti-oxidative properties. J Neuroimmunol 2018; 324:54-75. [PMID: 30243185 DOI: 10.1016/j.jneuroim.2018.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/24/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is characterized by chronic inflammatory response-induced demyelination of the neurons and degeneration of the axons within the central nervous system (CNS). A complex network of immunopathological-, inflammatory- and oxidative parameters involve in the development and advancement of MS. The anti-inflammatory, immunomodulatory and anti-oxidative characteristics of the ginger and several of its components have been indicated in some of experimental and clinical investigations. The possible therapeutic potentials of ginger and its ingredients in the treatment of MS may exert mainly through the regulation of the Th1-, Th2-, Th9-, Th17-, Th22- and Treg cell-related immune responses, down-regulation of the B cell-related immune responses, modulation of the macrophages-related responses, modulation of the production of pro- and anti-inflammatory cytokines, down-regulation of the arachidonic acid-derived mediators, interfering with the toll like receptor-related signaling pathways, suppression of the inflammasomes, down-regulation of the oxidative stress, reduction of the adhesion molecules expression, and down-regulation of the expression of the chemokines and chemokine receptors. This review aimed to provide a comprehensive knowledge regarding the immunomodulatory-, anti-inflammatory and anti-oxidative properties of ginger and its components, and highlight novel insights into the possible therapeutic potentials of this plant for treatment of MS. The review encourages more investigations to consider the therapeutic potentials of ginger and its effective components for managing of MS.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Department of Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
40
|
Wang Q, Yang Q, Cao X, Wei Q, Firempong CK, Guo M, Shi F, Xu X, Deng W, Yu J. Enhanced oral bioavailability and anti-gout activity of [6]-shogaol-loaded solid lipid nanoparticles. Int J Pharm 2018; 550:24-34. [DOI: 10.1016/j.ijpharm.2018.08.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/29/2018] [Accepted: 08/14/2018] [Indexed: 10/28/2022]
|