1
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
2
|
Zheng Y, Li X, Huang G, Li Q, Yao X, Zhang W, Zhang Y, Meng X, Li Q, Huang G. Shuang Bailian mixture enhanced the anti-cancer effect of cisplatin by regulating PI3K-Akt-Bcl 2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119462. [PMID: 39923956 DOI: 10.1016/j.jep.2025.119462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/01/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Esophageal cancer (EC) is a prevalent malignant tumor with the characteristics of poor prognosis and high mortality in the clinic. Cisplatin (CP), a first-line chemotherapeutic agent, faces the challenge of drug resistance, which hampers its clinical efficacy. In recent decades, traditional Chinese medicine has earned increasing attention for its potential as an adjunct to chemotherapy. Shuang Bailian mixture (SBLM) could significantly enhances the anti-cancer effect of CP in our clinical practices. However, the underlying mechanism of SBLM still needs further investigation. AIM OF THIS STUDY To investigate the underlying mechanisms that SBLM enhanced the anti-cancer effect of CP. MATERIALS AND METHODS This study used network pharmacology and molecular docking to obtain the potential active targets of SBLM. Moreover, CCK-8 assay, wound healing, colony formation assays, flow cytometry and Western blotting were used to evaluate the properties of SBLM enhancing the anti-cancer effect of CP. Furthermore, tumor xenograft in nude mice model was used to further determine the potential mechanism that SBLM enhanced the anti-cancer effect of CP. RESULTS Network pharmacology analysis showed that the PI3K-Akt-Bcl 2 signaling pathway served as the primary target of SBLM. Moreover, SBLM could significantly improve the anti-cancer effect of CP, including inhibited cell proliferation, suppressed colony formation and invasion, and induced cell cycle arrest and apoptosis. Furthermore, SLBM + CP could significantly reduce the tumor sizes on tumor-bearing mice when compared to SBLM and CP treatment. Mechanistically, SLBM could enhance CP effect by inducing cancer cells apoptosis via PI3K-Akt-Bcl 2 signaling pathway. CONCLUSION Our study revealed the underlying mechanisms that SBLM enhanced the anti-cancer effect of CP through inhibiting the PI3K/Akt/Bcl-2 signaling pathway to induce the cells apoptosis. These results suggest that SBLM could serve as a promising adjuvant in chemotherapeutic regimens for EC.
Collapse
Affiliation(s)
- Yilin Zheng
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China.
| | - Xiaoyan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Guohai Huang
- Blood Purification Center, Shantou Central Hospital, Shantou, Guangdong, China.
| | - Qingrui Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xiaohua Yao
- Tropical Agriculture and Forestry College, Guangdong AIB Polytechnic College, Guangzhou, China.
| | - Weijin Zhang
- Department of Rheumatology and Immunology, Shantou Central Hospital, Shantou, China.
| | - Yuanfeng Zhang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Qingnan Li
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China; Department of Pharmacy, Shantou Central Hospital, Shantou, China.
| | - Guoxin Huang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China.
| |
Collapse
|
3
|
El-Gammal HL, Omar F, Hyder A. Ferulic acid protects rat offspring from maternal high-fat, high-fructose diet-induced toxicity and developmental retardation through a direct effect on pancreatic islets. Food Chem Toxicol 2025; 197:115265. [PMID: 39832708 DOI: 10.1016/j.fct.2025.115265] [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: 08/28/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Maternal obesity predisposes offspring to type 2 diabetes (T2D) through a direct chronic effect of lipids on pancreatic β-cell neogenesis. β-cells produce FABP3 to bind and metabolize fatty acids. Ferulic acid (FA) is a natural product that may inhibit fatty acids' binding to FABP3, preventing their toxicity. It is aimed to evaluate the consequences of maternal feeding on high-fat, high fructose diet (HFFD) and the role of FA on the offspring. Four-week-old female rats were fed HFFD for 9 weeks prior to and throughout gestation and lactation to develop T2D. A group of them received 50 mg/kg FA daily. Offspring were sampled on gestational day 18 (GD18), and postnatal days (PND) 3 and 30. HFFD increased offspring's blood glucose, insulin, Homa-IR, HbA1c, triglycerides, cholesterol, intrahepatic and intra-insular lipid droplets. The mechanism of islet inflammation and apoptosis, detected by Il-1b and cleaved caspase3, involved the nuclear translocation of NFκB p65. Maternal HFFD caused developmental retardations in offspring's ovaries, testes, kidney and liver. Coupling FA treatment with the maternal HFFD maintained normoglycemia, lipidemia, and healthy islets, and prevented developmental retardations. FA administration to T2D mothers revealed positive effects on the offspring that is related to its direct protective effect on pancreatic β-cells.
Collapse
Affiliation(s)
- Hekmat L El-Gammal
- Zoology Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt
| | - Fatma Omar
- Zoology Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt
| | - Ayman Hyder
- Zoology Department, Faculty of Science, Damietta University, New Damietta, 34517, Egypt.
| |
Collapse
|
4
|
Properzi S, Stracci F, Rosi M, Lupi C, Villarini A, Gili A. Can a diet rich in Brassicaceae help control Helicobacter pylori infection? A systematic review. Front Med (Lausanne) 2024; 11:1454902. [PMID: 39741515 PMCID: PMC11685009 DOI: 10.3389/fmed.2024.1454902] [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: 07/02/2024] [Accepted: 11/11/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Helicobacter pylori (Hp) infection is highly prevalent globally and poses a significant public health challenge due to its link with chronic gastritis, peptic ulcers, and gastric malignancies. Hp's persistence within the gastric environment, particularly in case of infection with virulent strains, triggers chronic inflammatory responses and mucosal damage. Antibiotic therapy is the primary approach for Hp eradication, but antibiotic resistance and adverse effects hinder treatment efficacy. Emerging evidence suggests that Brassicaceae-derived metabolites could serve as adjunctive therapy for Hp infection, offering potential antimicrobial and anti-inflammatory benefits. Methods A systematic literature review was conducted following PRISMA guidelines to assess the impact of Brassicaceae-rich diets on Hp infection control. Searches were performed in MEDLINE PubMed, Web of Science, and the Cochrane Library until 18 October 2023, without language or date restrictions. Eligible studies meeting PICOS criteria were included, encompassing populations infected with Hp or Hp-infected human cell cultures, interventions involving Brassicaceae consumption or its bioactive molecules, and outcomes related to Hp infection control, antibiotic therapy interactions, reduction of antibiotic side effects, and inflammation mitigation. Animal studies, cell line experiments, reviews unrelated to the research objectives, and studies on Hp-related gastric cancer were excluded. Results Available evidence indicates that Brassicaceae consumption exhibits the potential to reduce Hp colonization but achieving complete eradication of the pathogen remains challenging. Conflicting results regarding the efficacy of broccoli in Hp treatment emerge, with certain investigations suggesting limited effectiveness. Other studies point to a potential for heightened eradication rates when combined with standard triple therapy. Furthermore, promising outcomes are observed with broccoli extract supplements, indicating their role in mitigating Hp-induced gastric mucosal damage. In fact, it is noteworthy that sulforaphane and its derivatives manifest notable reductions in pro-inflammatory markers, indicative of their anti-inflammatory properties. Adverse events associated with antibiotic therapy seem unaffected by sulforaphane derivatives or probiotics. However, individual responses to these treatments vary, underscoring the unpredictability of their efficacy in ameliorating antibiotic therapy-related side effects. Conclusion Our systematic review highlights the potential of Brassicaceae-rich diets as adjunctive therapy for Hp infection, offering synergistic interactions with antibiotics and possibly mitigating antibiotic side effects and inflammation. Further research, particularly well-designed randomized trials, is warranted to elucidate the therapeutic efficacy and optimal utilization of Brassicaceae-derived metabolites in managing human Hp-related diseases.
Collapse
Affiliation(s)
- Sara Properzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Fabrizio Stracci
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Margherita Rosi
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Chiara Lupi
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Anna Villarini
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Alessio Gili
- Department of Life Sciences, Health and Health Professions, Link Campus University, Rome, Italy
| |
Collapse
|
5
|
Carvalho EPD, Pessoa ADS, Iano FG, Ribeiro L, Leme B, Borges LF, Sanches MLR, Ximenes VF, Oliveira RCD. Antitumor effect of bromo-naphthoquinone associated with tannic acid in triple negative breast cancer cells. Int J Biochem Cell Biol 2024; 177:106697. [PMID: 39566654 DOI: 10.1016/j.biocel.2024.106697] [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: 08/06/2024] [Revised: 11/01/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive type of tumor that tends to recur in women. It is characterized by the absence of hormonal receptors, making it challenging to diagnosis and treatment. In this study, we investigated the anti-tumor effects of a pro-oxidant naphthoquinone derivative called bromo-naphthoquinone (BrNQ) isolated and combined with the antioxidant tannic acid (TA) in order to improve treatment. We used tumor cell lines MDA-MB-231 and HCC-70, as well as normal breast cells, HB4a, as control. Initially, viability assays conducted within 72 hours showed that the combination of compounds had a synergistic and notable cytotoxic effect on the tumor cells. The increased cytotoxicity appeared to be linked to changes in the cellular redox status, as indicated by a significant rise in reactive oxygen species (ROS) and though alterations in the level of thiol. The treatment also induced apoptosis, inhibited proliferation, and reduced migration, particularly in the MDA-MB-231 cell line. Furthermore, relevant changes were detected in the expression of Bcl-2, BAX, FAS, and BIRC-5, while no significant alteration in the expression of NOXs was observed. In conclusion, our findings suggested that the combination of BrNQ and TA though the ability to change redox status in tumor cells could act as a potential adjuvant treatment modality for improve prognosis in TNBC.
Collapse
Affiliation(s)
| | - Adriano de Souza Pessoa
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | - Flávia Godoy Iano
- Department of Preventive and Restorative Pediatric Dentistry, Araçatuba School of Dentistry, São Paulo State University, Brazil
| | - Laura Ribeiro
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | - Bianca Leme
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | - Luis Francisco Borges
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Brazil
| | | | - Valdecir Farias Ximenes
- Department of Chemistry, São Paulo State University (UNESP), School of Sciences, Bauru, SP, Brazil
| | | |
Collapse
|
6
|
Gan K, Shi W, Liu X, Ding W, Qiu Y, Luo X. Emerging functions of lycopene in the management of digestive premalignant lesions. Front Pharmacol 2024; 15:1478170. [PMID: 39484161 PMCID: PMC11524905 DOI: 10.3389/fphar.2024.1478170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Common digestive precancerous lesions, including oral potentially malignant disorders (OPMDs), gastric ulcers and colorectal adenoma, harbor high risk of cancerous transformation. Early intervention of these lesions is significant to prevent carcinogenesis and improve patients' prognosis. Lycopene, a carotenoid predominantly accumulated in tomatoes, is clinically recommended with its cis structure; as lycopene harbors the most potent antioxidative effects among carotenoids, its chemopreventive effects on the premalignant lesions is noted. Despite several reviews have assessed lycopene's efficacy for OPMDs, emerging studies have reported varying efficacy for digestive precancerous lesion with no comprehensive summary. Therefore, this review initially evaluates the efficacy and underlying mechanisms of lycopene for management of digestive precancerous lesions. According to the included studies, lycopene may show high promise in the management of digestive precancerous lesions, such as relieving mouth opening and burning sensation of oral submucous fibrosis (OSF), presenting potentially equivalent efficacy on managing oral lichen planus (OLP) as steroids and alleviating gastrointestinal precancers' symptoms, meanwhile lowering colon cancer risk. Moreover, its mechanisms for managing digestive precancerous lesions are concretely summarized, including anti-oxidative stress effects, anti-inflammatory response and regulation of cell proliferation and apoptosis, especially its modifications on TLR4/TRIF/NF-κB signaling pathway and p53-dependent cell cycle control and apoptosis. More studies are warranted to confirm its long-term efficacy and preventive role against malignant transformation of digestive precancerous lesions as evidence is insufficient.
Collapse
Affiliation(s)
- Kerui Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wenjin Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiangfei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wei Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yan Qiu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Liu Y, Yin R, Tian Y, Xu S, Meng X. Curcumin nanopreparations: recent advance in preparation and application. Biomed Mater 2024; 19:052009. [PMID: 39189065 DOI: 10.1088/1748-605x/ad6dc7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
Curcumin is a natural polyphenolic compound extracted from turmeric with antibacterial, antioxidant, antitumor, preventive and therapeutic neurological disorders and a variety of bioactivities, which is widely used in the field of food and medicine. However, the drawbacks of curcumin such as poor aqueous solubility and stability have limited the practical application of curcumin. To overcome these defects and enhance its functional properties, various nanoscale systems (liposomes, polymer nanoparticles, protein nanoparticles, solid lipid nanoparticles, metal nanoparticles, etc) have been extensively employed for curcumin encapsulation and delivery. Despite the rapid development of curcumin nanoformulations, there is a lack of comprehensive reviews on their preparation and properties. This review provides an overview of the construction of curcumin nano-delivery systems, mechanisms of action, nanocarrier preparation methods and the applications of curcumin nanocarriers in the food and pharmaceutical fields to provide a theoretical basis and technological support for the efficient bio-utilization, product development and early clinical application of curcumin.
Collapse
Affiliation(s)
- Yan Liu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Rui Yin
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Yuan Tian
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Shujun Xu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Xin Meng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| |
Collapse
|
8
|
Huanjie Z, Bukhari I, Fazhan L, Wen H, Wang J, Wanqing W, Yuming F, Youcai T, AlJowaie RM, Aziz IM, Xiufeng C, Yang M, Pengyuan Z. P53-associated lncRNAs regulate immune functions and RNA-modifiers in gastric cancer. Heliyon 2024; 10:e35228. [PMID: 39166030 PMCID: PMC11334848 DOI: 10.1016/j.heliyon.2024.e35228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
TP53, a guardian of the genome, suppresses or enhances tumors through various regulatory pathways. However, the role of p53-related long non-coding RNAs (lncRNAs) in immune regulation of tumor microenvironment and prognosis of gastric cancer (GC) is so far unelucidated. We analyzed the role of TP53-associated lncRNAs (obtained from the TP53LNC-DB database) in immune regulation, immune cell infiltration and RNA modification in gastric cancer. Firstly, using multivariate COX regression analysis, we identified eight lncRNAs related to the prognosis of GC. Furthermore, based on the expression of the lncRNA signature and risk score, the GC patients were divided into high-risk and low-risk groups. We found that M2-macrophages have significantly higher infiltration in the high-risk group. Similarly, significant differences in immune function (APC_co_stimulation, CCR, and checkpoint) and m6A modification (FTO, ZC3H13, YTHDC1, and RBM15), and m5C modification (NOP2 and TET1) between both groups were also observed. These signature lncRNAs were also positively associated with oxidative stress-related genes (MPO, MAPK14, HMOX1, and APP). Additionally, we found that high expression of GAS5 and low expression of MALAT1 in Helicobacter pylori (H-pylori) positive GC patients. Finally, GC patients in the low-risk group showed higher resistance to immunotherapy while patients in the high-risk group were more sensitive to various chemotherapy drugs. Based on these findings, we conclude that p53-associated lncRNAs signature could potentially predict the immune status and overall survival, and may also be used for risk management and planning immunotherapy for gastric cancer patients.
Collapse
Affiliation(s)
- Zhao Huanjie
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
| | - Ihtisham Bukhari
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
| | - Li Fazhan
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
| | - Huijuan Wen
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
| | - Jingyun Wang
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
| | - Wu Wanqing
- Department of Gastrointestinal Surgery, the Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
| | - Fu Yuming
- Department of Gastrointestinal Surgery, the Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
| | - Tang Youcai
- Department of Pediatrics, the Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
| | - Reem M. AlJowaie
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ibrahim M. Aziz
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Chu Xiufeng
- Department of Oncology, the Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
| | - Mi Yang
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhongyuan, 450001, Zhengzhou, Henan China, China
- Institute of Rehabilitation Medicine, Henan Academy of Innovations in Medical Sciences, Zhengzhou, Henan, China
| | - Zheng Pengyuan
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, ErQi 450052, Zhengzhou, Henan, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, ErQi, 450052, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhongyuan, 450001, Zhengzhou, Henan China, China
- Institute of Rehabilitation Medicine, Henan Academy of Innovations in Medical Sciences, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Bhuker S, Kaur A, Rajauria K, Tuli HS, Saini AK, Saini RV, Gupta M. Allicin: a promising modulator of apoptosis and survival signaling in cancer. Med Oncol 2024; 41:210. [PMID: 39060753 DOI: 10.1007/s12032-024-02459-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
According to the World Health Organization, cancer is the foremost cause of mortality globally. Various phytochemicals from natural sources have been extensively studied for their anticancer properties. Allicin, a powerful organosulfur compound derived from garlic, exhibits anticancer, antioxidant, anti-inflammatory, antifungal, and antibacterial properties. This review aims to update and evaluate the chemistry, composition, mechanisms of action, and pharmacokinetics Allicin. Allicin has garnered significant attention for its potential role in modulating Fas-FasL, Bcl2-Bax, PI3K-Akt-mTOR, autophagy, and miRNA pathways. At the molecular level, allicin induces the release of cytochrome c from the mitochondria and enhances the activation of caspases-3, -8, and -9. This is accompanied by the simultaneous upregulation of Bax and Fas expression in tumor cells. Allicin can inhibit excessive autophagy by activating the PI3K/Akt/mTOR and MAPK/ERK/mTOR signaling pathways. Allicin-loaded nano-formulations efficiently induce apoptosis in cancer cells while minimizing toxicity to normal cells. Safety and clinical aspects are meticulously scrutinized, providing insights into the tolerability and adverse effects associated with allicin administration, along with an overview of current clinical trials evaluating its therapeutic potential. In conclusion, this review underscores the promising prospects of allicin as a dietary-derived medicinal compound for cancer therapy. It emphasizes the need for further research to elucidate its precise mechanisms of action, optimize delivery strategies, and validate its efficacy in clinical settings.
Collapse
Affiliation(s)
- Sunaina Bhuker
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Avneet Kaur
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Kanitha Rajauria
- SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Tamil Nadu, 603203, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Adesh K Saini
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
- Central Research Laboratory, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Reena V Saini
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India.
- Central Research Laboratory, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India.
- Central Research Laboratory and Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India.
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| |
Collapse
|
10
|
Jin X, Wang C, Chen C, Hai S, Rahman SU, Zhao C, Huang W, Feng S, Wang X. Allicin attenuates the oxidative damage induced by Aflatoxin B 1 in dairy cow hepatocytes via the Nrf2 signalling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116527. [PMID: 38833978 DOI: 10.1016/j.ecoenv.2024.116527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Aflatoxin B1 (AFB1) is known to inhibit growth, and inflict hepatic damage by interfering with protein synthesis. Allicin, has been acknowledged as an efficacious antioxidant capable of shielding the liver from oxidative harm. This study aimed to examine the damage caused by AFB1 on bovine hepatic cells and the protective role of allicin against AFB1-induced cytotoxicity. In this study, cells were pretreated with allicin before the addition of AFB1 for co-cultivation. Our findings indicate that AFB1 compromises cellular integrity, suppresses the expression of nuclear factor erythroid 2-related factor 2 (Nrf2). In addition, allicin attenuates oxidative damage to bovine hepatic cells caused by AFB1 by promoting the expression of the Nrf2 pathway and reducing cell apoptosis. In conclusion, the results of this study will help advance clinical research and applications, providing new options and directions for the prevention and treatment of liver diseases.
Collapse
Affiliation(s)
- Xin Jin
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Chenlong Wang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Chuangjiang Chen
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Sirao Hai
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Sajid Ur Rahman
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China; Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chang Zhao
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Wanyue Huang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, Hefei 230036, China.
| |
Collapse
|
11
|
Khoshvaghti A, Rahbari R. The effect of ellagic acid on sex hormones and miRNA-21 expression in rats with polycystic ovary syndrome. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4263-4273. [PMID: 38078918 DOI: 10.1007/s00210-023-02895-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/05/2023] [Indexed: 05/23/2024]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age. It is characterized by irregular menstrual cycles, hyperandrogenism, and polycystic ovaries. The syndrome's etiology is multifactorial, involving genetic, hormonal, metabolic, and environmental factors. Given its diverse effects, managing PCOS requires a comprehensive approach. METHODS This study employed a Sprague-Dawley rat model to investigate the effects of ellagic acid on polycystic ovary syndrome (PCOS). Forty adult female rats were randomly divided into four groups: a control group, a healthy group receiving ellagic acid (200 mg/kg), a PCOS group, and an ellagic acid + PCOS group. PCOS was induced in the relevant groups through subcutaneous injection of estradiol valerate (2 mg/kg), and ellagic acid was administered via subcutaneous injections for 14 days. Blood samples were collected for hormone analysis using the ELISA method, and ovarian tissues were processed for histological examination. RESULT Ellagic acid treatment showed reduced LH levels and restoration of follicular development, particularly primordial and graafian follicles, along with modulation of miRNA-21 expression. Moreover, ellagic acid exhibited positive effects on ovarian morphology, including decreased theca layer thickness, increased oocyte diameter, and improvements in antral and preovulatory follicles. This suggests ellagic acid's potential in addressing follicular development and oocyte quality in PCOS. CONCLUSIONS These findings suggest ellagic acid as a potential complementary approach in PCOS management. While the study is promising, further research, including clinical trials, is required to elucidate ellagic acid's mechanisms and clinical efficacy in human PCOS subjects.
Collapse
Affiliation(s)
- Ameneh Khoshvaghti
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran.
| | - Raha Rahbari
- Graduated of Faculty of Veterinary Medicine Kazerun Branch, Islamic Azad University, Kazerun, Iran
| |
Collapse
|
12
|
Lu Z, Wang Y, Liu C, Fan H. Efficacy and Safety of Asparagusic Acid against Echinococcus multilocularis In Vitro and in a Murine Infection Model. Trop Med Infect Dis 2024; 9:110. [PMID: 38787043 PMCID: PMC11126102 DOI: 10.3390/tropicalmed9050110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Alveolar echinococcosis (AE) stands as a perilous zoonotic affliction caused by the larvae of Echinococcus multilocularis. There is an imperative need to explore novel therapeutic agents or lead compounds for the treatment of AE. Asparagusic acid, characterized by its low toxicity and possessing antimicrobial, antioxidant, and anti-parasitic attributes, emerges as a promising candidate. The aim of this study was to investigate the in vivo and in vitro efficacy of asparagusic acid against E. multilocularis. Morphological observations, scanning electron microscopy, ROS assays, mitochondrial membrane potential assays, and Western blot were used to evaluate the in vitro effects of asparagusic acid on protoscoleces. The effects of asparagusic acid on vesicles were assessed via PGI release, γ-GGT release, and transmission electron microscopy observations. CellTiter-Glo assays, Caspase3 activity assays, flow cytometry, and Western blot were used for an evaluation of the effect of asparaginic acid on the proliferation and apoptosis of germinal cells. The in vivo efficacy of asparagusic acid was evaluated in a murine AE model. Asparagusic acid exhibited a pronounced killing effect on the protoscoleces post-treatment. Following an intervention with asparagusic acid, there was an increase in ROS levels and a decline in mitochondrial membrane potential in the protoscolex. Moreover, asparagusic acid treatment resulted in the upregulation of PGI and γ-GGT release in metacestode vesicles, concomitant with the inhibition of germinal cell viability. Furthermore, asparagusic acid led to an enhanced relative expression of Caspase3 in the culture supernatant of both the protoscoleces and germinal cells, accompanied by an increase in the proportion of apoptotic germinal cells. Notably, asparagusic acid induced an augmentation in Bax and Caspase3 protein expression while reducing Bcl2 protein expression in both the protoscoleces and germinal cells. In vitro cytotoxicity assessments demonstrated the low toxicity of asparagusic acid towards normal human hepatocytes and HFF cells. Additionally, in vivo experiments revealed that asparagusic acid administration at doses of 10 mg/kg and 40 mg/kg significantly reduced metacestode wet weight. A histopathological analysis displayed the disruption of the germinal layer structure within lesions post-asparagusic acid treatment, alongside the preservation of laminated layer structures. Transmission electron microscopy further revealed mitochondrial swelling and heightened cell necrosis subsequent to the asparagusic acid treatment. Furthermore, asparagusic acid promoted Caspase3 and Bax protein expression while decreasing Bcl2 protein expression in perilesional tissues. Subsequently, it inhibited the expression of Ki67, MMP2, and MMP9 proteins in the perilesional tissues and curbed the activation of the PI3K/Akt signaling pathway within the lesion-host microenvironmental tissues. Asparagusic acid demonstrated a pronounced killing effect on E. multilocularis, suggesting its potential as a promising therapeutic agent for the management of AE.
Collapse
Affiliation(s)
- Zhuanhong Lu
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
| | - Yating Wang
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
| | - Chuanchuan Liu
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
- Qinghai Key Laboratory of Echinococcosis Research, Xining 810001, China
| | - Haining Fan
- School of Clinical Medicine, Qinghai University Affiliated Hospital, Xining 810001, China; (Z.L.); (Y.W.)
- Qinghai Key Laboratory of Echinococcosis Research, Xining 810001, China
| |
Collapse
|
13
|
Singh T, Sharma D, Sharma R, Tuli HS, Haque S, Ramniwas S, Mathkor DM, Yadav V. The Role of Phytonutrient Kaempferol in the Prevention of Gastrointestinal Cancers: Recent Trends and Future Perspectives. Cancers (Basel) 2024; 16:1711. [PMID: 38730663 PMCID: PMC11083332 DOI: 10.3390/cancers16091711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
In recent years, kaempferol, a natural flavonoid present in various fruits and vegetables, has received significant attention in gastrointestinal cancer research due to its varied therapeutic effects. Kaempferol has been proven to alter several molecular mechanisms and pathways, such as the PI3/Akt, mTOR, and Erk/MAPK pathway involved in cancer progression, showing its inhibitory effects on cell proliferation, survival, angiogenesis, metastasis, and migration. Kaempferol is processed in the liver and small intestine, but limited bioavailability has been a major concern in the clinical implications of kaempferol. Nano formulations have been proven to enhance kaempferol's efficacy in cancer prevention. The synergy of nanotechnology and kaempferol has shown promising results in in vitro studies, highlighting the importance for more in vivo research and clinical trials to determine safety and efficacy. This review aims to focus on the role of kaempferol in various types of gastrointestinal cancer and how the combination of kaempferol with nanotechnology helps in improving therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi 110007, India; (D.S.); (R.S.)
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences-Defence Research and Development Organization, (INMAS-DRDO) New Delhi, Delhi 110054, India
| | - Deepika Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi 110007, India; (D.S.); (R.S.)
| | - Rishabh Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi 110007, India; (D.S.); (R.S.)
- Amity Stem Cell Institute, Amity Medical School, Amity University, Gurugram 122412, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India;
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (S.H.); (D.M.M.)
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 11022801, Lebanon
| | - Seema Ramniwas
- University Centre for Research & Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali 140413, India;
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (S.H.); (D.M.M.)
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, SE-20213 Malmö, Sweden
| |
Collapse
|
14
|
Tanabe S. Advances in Molecular Mechanisms of Gastrointestinal Tumors. Cancers (Basel) 2024; 16:1603. [PMID: 38672684 PMCID: PMC11049183 DOI: 10.3390/cancers16081603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Gastrointestinal cancer is one of the most common malignancies worldwide [...].
Collapse
Affiliation(s)
- Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, Kawasaki 210-9501, Japan
| |
Collapse
|
15
|
Yan S, He Y, Zhu Y, Ye W, Chen Y, Zhu C, Zhan F, Ma Z. Human patient derived organoids: an emerging precision medicine model for gastrointestinal cancer research. Front Cell Dev Biol 2024; 12:1384450. [PMID: 38638528 PMCID: PMC11024315 DOI: 10.3389/fcell.2024.1384450] [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: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Gastrointestinal cancers account for approximately one-third of the total global cancer incidence and mortality with a poor prognosis. It is one of the leading causes of cancer-related deaths worldwide. Most of these diseases lack effective treatment, occurring as a result of inappropriate models to develop safe and potent therapies. As a novel preclinical model, tumor patient-derived organoids (PDOs), can be established from patients' tumor tissue and cultured in the laboratory in 3D architectures. This 3D model can not only highly simulate and preserve key biological characteristics of the source tumor tissue in vitro but also reproduce the in vivo tumor microenvironment through co-culture. Our review provided an overview of the different in vitro models in current tumor research, the derivation of cells in PDO models, and the application of PDO model technology in gastrointestinal cancers, particularly the applications in combination with CRISPR/Cas9 gene editing technology, tumor microenvironment simulation, drug screening, drug development, and personalized medicine. It also elucidates the ethical status quo of organoid research and the current challenges encountered in clinical research, and offers a forward-looking assessment of the potential paths for clinical organoid research advancement.
Collapse
Affiliation(s)
- Sicheng Yan
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuxuan He
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuehong Zhu
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wangfang Ye
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Chen
- Department of Colorectal Surgery, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Cong Zhu
- Department of Colorectal Surgery, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Fuyuan Zhan
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhihong Ma
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
16
|
Zhang H, Wang H, Qin L, Lin S. Garlic-derived compounds: Epigenetic modulators and their antitumor effects. Phytother Res 2024; 38:1329-1344. [PMID: 38194996 DOI: 10.1002/ptr.8108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/26/2023] [Accepted: 12/09/2023] [Indexed: 01/11/2024]
Abstract
Cancer is a highly heterogeneous disease that poses a serious threat to human health worldwide. Despite significant advances in the diagnosis and treatment of cancer, the prognosis and survival rate of cancer remain poor due to late diagnosis, drug resistance, and adverse reactions. Therefore, it is very necessary to study the development mechanism of cancer and formulate effective therapeutic interventions. As widely available bioactive substances, natural products have shown obvious anticancer potential, especially by targeting abnormal epigenetic changes. The main active part of garlic is organic sulfur compounds, of which diallyl trisulfide (DATS) content is the highest, accounting for more than 40% of the total composition. The garlic-derived compounds have been recognized as an antioxidant for cancer prevention and treatment. However, the molecular mechanism of the antitumor effect of garlic-derived compounds remains unclear. Recent studies have identified garlic-derived compound DATS that plays critical roles in enhancing CpG demethylation or promoting histone acetylation as an epigenetic inhibitor. Here, we review the therapeutic progress of garlic-derived compounds against cancer through epigenetic pathways.
Collapse
Affiliation(s)
- Huan Zhang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Haichao Wang
- Institute of Resources and Environment, Beijing Academy of Science and Technology, Beijing, China
| | - Lin Qin
- Department of Endoscopic Diagnosis and Treatment, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Shuye Lin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
17
|
Hakami ZH. Biomarker discovery and validation for gastrointestinal tumors: A comprehensive review of colorectal, gastric, and liver cancers. Pathol Res Pract 2024; 255:155216. [PMID: 38401376 DOI: 10.1016/j.prp.2024.155216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 02/26/2024]
Abstract
Gastrointestinal (GI) malignancies, encompassing gastric, hepatic, colonic, and rectal cancers, are prevalent forms of cancer globally and contribute substantially to cancer-related mortality. Although there have been improvements in methods for diagnosing and treating GI cancers, the chances of survival for these types of cancers are still extremely low. According to the World Cancer Research International Fund's most recent figures, stomach cancer was responsible for roughly one million deaths worldwide in 2020. This emphasizes the importance of developing more effective tools for detecting, diagnosing, and predicting the outcome of these cancers at an early stage. Biomarkers, quantitative indications of biological processes or disease states, have emerged as promising techniques for enhancing the diagnosis and prognosis of GI malignancies. Recently, there has been a considerable endeavor to discover and authenticate biomarkers for various GI cancers by the utilization of diverse methodologies, including genomics, proteomics, and metabolomics. This review provides a thorough examination of the current state of biomarker research in the field of gastrointestinal malignancies, with a specific emphasis on colorectal, stomach, and liver cancers. A thorough literature search was performed on prominent databases such as PubMed, Scopus, and Web of Science to find pertinent papers published until November, 2023 for the purpose of compiling this review. The diverse categories of biomarkers, encompassing genetic, epigenetic, and protein-based biomarkers, and their potential utility in the fields of diagnosis, prognosis, and treatment selection, are explored. Recent progress in identifying and confirming biomarkers, as well as the obstacles that persist in employing biomarkers in clinical settings are emphasized. The utilization of biomarkers in GI cancers has significant potential in enhancing patient outcomes. Ongoing research is expected to uncover more efficient biomarkers for the diagnosis and prognosis of these cancers.
Collapse
Affiliation(s)
- Zaki H Hakami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Science, Jazan University, Jazan 45142, Saudi Arabia.
| |
Collapse
|
18
|
Lee HJ, Kwon YS, Lee JH, Moon YG, Choi J, Hyun M, Tak TK, Kim JH, Heo JD. Pectolinarigenin regulates the tumor-associated proteins in AGS-xenograft BALB/c nude mice. Mol Biol Rep 2024; 51:305. [PMID: 38361124 PMCID: PMC10869406 DOI: 10.1007/s11033-023-09046-4] [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: 03/07/2023] [Accepted: 10/30/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Pectolinarigenin (PEC) is a flavone extracted from Cirsium, and because it has anti-inflammatory properties, anti-cancer research is also being conducted. The objective of this work was to find out if PEC is involved in tumor control and which pathways it regulates in vivo and in vitro. METHODS AGS cell lines were xenografted into BALB/c nude mice to create tumors, and PEC was administered intraperitoneally to see if it was involved in tumor control. Once animal testing was completed, tumor proteins were isolated and identified using LC-MS analysis, and gene ontology of the found proteins was performed. RESULTS Body weight and hematological measurements on the xenograft mice model demonstrated that PEC was not harmful to non-cancerous cells. We found 582 proteins in tumor tissue linked to biological reactions such as carcinogenesis and cell death signaling. PEC regulated 6 out of 582 proteins in vivo and in vitro in the same way. CONCLUSION Our findings suggested that PEC therapy may inhibit tumor development in gastric cancer (GC), and proteomic research gives fundamental information about proteins that may have great promise as new therapeutic targets in GC.
Collapse
Affiliation(s)
- Ho Jeong Lee
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Young Sang Kwon
- Environmental Safety Assessment Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Ju Hong Lee
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Yeon Gyu Moon
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Jungil Choi
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Moonjung Hyun
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Tae Kil Tak
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Je-Hein Kim
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Jeong Doo Heo
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea.
| |
Collapse
|
19
|
Zhang Q, Yang C, Gao X, Dong J, Zhong C. Phytochemicals in regulating PD-1/PD-L1 and immune checkpoint blockade therapy. Phytother Res 2024; 38:776-796. [PMID: 38050789 DOI: 10.1002/ptr.8082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/27/2023] [Accepted: 11/12/2023] [Indexed: 12/06/2023]
Abstract
Clinical treatment and preclinical studies have highlighted the role of immune checkpoint blockade in cancer treatment. Research has been devoted to developing immune checkpoint inhibitors in combination with other drugs to achieve better efficacy or reduce adverse effects. Phytochemicals sourced from vegetables and fruits have demonstrated antiproliferative, proapoptotic, anti-migratory, and antiangiogenic effects against several cancers. Phytochemicals also modulate the tumor microenvironment such as T cells, regulatory T cells, and cytokines. Recently, several phytochemicals have been reported to modulate immune checkpoint proteins in in vivo or in vitro models. Phytochemicals decreased programmed cell death ligand-1 expression and synergized programmed cell death receptor 1 (PD-1) monoclonal antibody to suppress tumor growth. Combined administration of phytochemicals and PD-1 monoclonal antibody enhanced the tumor growth inhibition as well as CD4+ /CD8+ T-cell infiltration. In this review, we discuss immune checkpoint molecules as potential therapeutic targets of cancers. We further assess the impact of phytochemicals including carotenoids, polyphenols, saponins, and organosulfur compounds on cancer PD-1/programmed cell death ligand-1 immune checkpoint molecules and document their combination effects with immune checkpoint inhibitors on various malignancies.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenying Yang
- Yinzhou Center for Disease Control and Prevention, Ningbo, China
| | - Xingsu Gao
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ju Dong
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Jiang W, Zhang T, Zhang H, Han T, Ji P, Ou Z. Metabolic Patterns of High-Invasive and Low-Invasive Oral Squamous Cell Carcinoma Cells Using Quantitative Metabolomics and 13C-Glucose Tracing. Biomolecules 2023; 13:1806. [PMID: 38136676 PMCID: PMC10742159 DOI: 10.3390/biom13121806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/24/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Most current metabolomics studies of oral squamous cell carcinoma (OSCC) are mainly focused on identifying potential biomarkers for early screening and diagnosis, while few studies have investigated the metabolic profiles promoting metastasis. In this study, we aimed to explore the altered metabolic pathways associated with metastasis of OSCC. Here, we identified four OSCC cell models (CAL27, HN6, HSC-3, SAS) that possess different invasive heterogeneity via the transwell invasion assay and divided them into high-invasive (HN6, SAS) and low-invasive (CAL27, HSC-3) cells. Quantitative analysis and stable isotope tracing using [U-13C6] glucose were performed to detect the altered metabolites in high-invasive OSCC cells, low-invasive OSCC cells and normal human oral keratinocytes (HOK). The metabolic changes in the high-invasive and low-invasive cells included elevated glycolysis, increased fatty acid metabolism and an impaired TCA cycle compared with HOK. Moreover, pathway analysis demonstrated significant differences in fatty acid biosynthesis; arachidonic acid (AA) metabolism; and glycine, serine and threonine metabolism between the high-invasive and low-invasive cells. Furthermore, the high-invasive cells displayed a significant increase in the percentages of 13C-glycine, 13C-palmitate, 13C-stearic acid, 13C-oleic acid, 13C-AA and estimated FADS1/2 activities compared with the low-invasive cells. Overall, this exploratory study suggested that the metabolic differences related to the metastatic phenotypes of OSCC cells were concentrated in glycine metabolism, de novo fatty acid synthesis and polyunsaturated fatty acid (PUFA) metabolism, providing a comprehensive understanding of the metabolic alterations and a basis for studying related molecular mechanisms in metastatic OSCC cells.
Collapse
Affiliation(s)
- Wenrong Jiang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; (W.J.); (T.Z.)
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Ting Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; (W.J.); (T.Z.)
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Hua Zhang
- Ministry of Education of China International Collaborative Joint Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China; (H.Z.); (T.H.)
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tingli Han
- Ministry of Education of China International Collaborative Joint Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China; (H.Z.); (T.H.)
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; (W.J.); (T.Z.)
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Zhanpeng Ou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; (W.J.); (T.Z.)
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
21
|
Ma S, Liu H, Sun C, Meng M, Qu G, Jiang Y, Wu B, Gao J, Feng L, Xie P, Xia W, Sun Y. Effect of physical activity on incidence and mortality in patients with gastric cancer: evidence from real-world studies. Cancer Causes Control 2023; 34:1095-1111. [PMID: 37491662 DOI: 10.1007/s10552-023-01763-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023]
Abstract
PURPOSE Physical activity (PA) has been suggested to reduce the risk of cancer. However, previous studies have been inconsistent regarding the relationship between PA and the risk of developing gastric cancer (GC). The purpose of this study was to evaluate the impact of PA on the incidence and mortality risk of GC through a meta-analysis, as well as investigate potential dose-response relationships. METHODS A systematic literature search was conducted in 10 electronic databases and 4 registries. The combined relative risks (RRs) were calculated using a random-effects model with 95% confidence interval (CIs) to assess the effect of PA on the risk of GC. Relevant subgroup analyses and sensitivity analyses were performed. RESULTS The results showed that PA correlated with lower incidence of GC (RR: 0.83, 95% CI: 0.77-0.90), decreased risk of GC mortality (RR: 0.76, 95% CI: 0.66-0.89). The results of the subgroup analysis showed that PA was associated with reduced incidence of GC across gender, different regions, study designs, different sites of GC and different types of PA. A linear relationship was found for frequency of PA. CONCLUSIONS This meta-analysis found that PA was associated with a reduced risk of GC incidence and mortality. The correlation between PA and GC occurrence was in a dose-response relationship.
Collapse
Affiliation(s)
- Shaodi Ma
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Haixia Liu
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Chenyu Sun
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230032, Anhui, P.R. China
| | - Muzi Meng
- UK Program Site, American University of the Caribbean School of Medicine, Vernon Building Room 64, Sizer St, Preston, PR1 1JQ, UK
- Bronxcare Health System, 1650 Grand Concourse, The Bronx, NY, 10457, USA
| | - Guangbo Qu
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Yuemeng Jiang
- Department of Infectious Diseases, the First Affiliated Hospital of Anhui Medical University North District, No. 100 Huaihai Dadao, Hefei, 230032, Anhui, P.R. China
- Anhui Public Health Clinical Center, No. 100 Huaihai Dadao, Hefei, 230032, Anhui, P.R. China
| | - Birong Wu
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Juan Gao
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Linya Feng
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Peng Xie
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Weihang Xia
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China
| | - Yehuan Sun
- Department of Epidemiology and Health Statistics, School of Public Health Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, P.R. China.
- Chaohu Hospital, Anhui Medical University, No. 64 Chaohubei Road, Hefei, 238000, Anhui, China.
- Center for Evidence-Based Practice, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
22
|
Sa P, Mohapatra P, Swain SS, Khuntia A, Sahoo SK. Phytochemical-Based Nanomedicine for Targeting Tumor Microenvironment and Inhibiting Cancer Chemoresistance: Recent Advances and Pharmacological Insights. Mol Pharm 2023; 20:5254-5277. [PMID: 37596986 DOI: 10.1021/acs.molpharmaceut.3c00286] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2023]
Abstract
Cancer remains the leading cause of death and rapidly evolving disease worldwide. The understanding of disease pathophysiology has improved through advanced research investigation, and several therapeutic strategies are being used for better cancer treatment. However, the increase in cancer relapse and metastatic-related deaths indicate that available therapies and clinically approved chemotherapy drugs are not sufficient to combat cancer. Further, the constant crosstalk between tumor cells and the tumor microenvironment (TME) is crucial for the development, progression, metastasis, and therapeutic response to tumors. In this regard, phytochemicals with multimodal targeting abilities can be used as an alternative to current cancer therapy by inhibiting cancer survival pathways or modulating TME. However, due to their poor pharmacokinetics and low bioavailability, the success of phytochemicals in clinical trials is limited. Therefore, developing phytochemical-based nanomedicine or phytonanomedicine can improve the pharmacokinetic profile of these phytochemicals. Herein, the molecular characteristics and pharmacological insights of the proposed phytonanomedicine in cancer therapy targeting tumor tissue and altering the characteristics of cancer stem cells, chemoresistance, TME, and cancer immunity are well discussed. Further, we have highlighted the clinical perspective and challenges of phytonanomedicine in filling the gap in potential cancer therapeutics using various nanoplatforms. Overall, we have discussed how clinical success and pharmacological insights could make it more beneficial to boost the concept of nanomedicine in the academic and pharmaceutical fields to counter cancer metastases and drug resistance.
Collapse
Affiliation(s)
- Pratikshya Sa
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, NCR Delhi, India
| | - Priyanka Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, NCR Delhi, India
| | | | - Auromira Khuntia
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, NCR Delhi, India
| | | |
Collapse
|
23
|
Tandoro Y, Chen BK, Ali A, Wang CK. Review of Phytochemical Potency as a Natural Anti- Helicobacter pylori and Neuroprotective Agent. Molecules 2023; 28:7150. [PMID: 37894629 PMCID: PMC10609179 DOI: 10.3390/molecules28207150] [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: 09/01/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Phytochemicals are plant secondary metabolites that show health benefits for humans due to their bioactivity. There is a huge variety of phytochemicals that have already been identified, and these compounds can act as antimicrobial and neuroprotection agents. Due to their anti-microbial activity and neuroprotection, several phytochemicals might have the potency to be used as natural therapeutic agents, especially for Helicobacter pylori infection and neurodegenerative disease, which have become a global health concern nowadays. According to previous research, there are some connections between H. pylori infection and neurodegenerative diseases, especially Alzheimer's disease. Hence, this comprehensive review examines different kinds of phytochemicals from natural sources as potential therapeutic agents to reduce H. pylori infection and improve neurodegenerative disease. An additional large-scale study is needed to establish the connection between H. pylori infection and neurodegenerative disease and how phytochemicals could improve this condition.
Collapse
Affiliation(s)
- Yohanes Tandoro
- Department of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung 40201, Taiwan; (Y.T.); (B.-K.C.); (A.A.)
- Faculty of Agricultural Technology, Widya Mandala Catholic University Surabaya, Surabaya 60265, Indonesia
| | - Bo-Kai Chen
- Department of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung 40201, Taiwan; (Y.T.); (B.-K.C.); (A.A.)
| | - Asif Ali
- Department of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung 40201, Taiwan; (Y.T.); (B.-K.C.); (A.A.)
| | - Chin-Kun Wang
- Department of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung 40201, Taiwan; (Y.T.); (B.-K.C.); (A.A.)
| |
Collapse
|
24
|
Akash S, Bayıl I, Mahmood S, Mukerjee N, Mili TA, Dhama K, Rahman MA, Maitra S, Mohany M, Al-Rejaie SS, Ali N, Semwal P, Sharma R. Mechanistic inhibition of gastric cancer-associated bacteria Helicobacter pylori by selected phytocompounds: A new cutting-edge computational approach. Heliyon 2023; 9:e20670. [PMID: 37876433 PMCID: PMC10590806 DOI: 10.1016/j.heliyon.2023.e20670] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/09/2023] [Accepted: 10/04/2023] [Indexed: 10/26/2023] Open
Abstract
Background Helicobacter pylori (H. pylori) is a persistent bacterial inhabitant in the stomachs of approximately half the global populace. This bacterium is directly linked to chronic gastritis, leading to a heightened risk of duodenal and gastric ulcer diseases, and is the predominant risk factor for gastric cancer - the second most common cause of cancer-related deaths globally. The increasing prevalence of antibiotic resistance necessitates the exploration of innovative treatment alternatives to mitigate the H. pylori menace. Methods Initiating our study, we curated a list of thirty phytochemicals based on previous literature and subjected them to molecular docking studies. Subsequently, eight phytocompounds-Glabridin, Isoliquiritin, Sanguinarine, Liquiritin, Glycyrrhetic acid, Beta-carotin, Diosgenin, and Sarsasapogenin-were meticulously chosen based on superior binding scores. These were further subjected to an extensive computational analysis encompassing ADMET profiling, drug-likeness evaluation, principal component analysis (PCA), and molecular dynamic simulations (MDs) in comparison with the conventional drug, Mitomycin. Results The natural compounds investigated demonstrated superior docking affinities to H. pylori targets compared to the standard Mitomycin. Notably, the phytocompounds Diosgenin and Sarsasapogenin stood out due to their exceptional binding affinities and pharmacokinetic properties, including favorable ADMET profiles. Conclusion Our comprehensive and technologically-advanced approach showcases the potential of identified phytocompounds as pioneering therapeutic agents against H. pylori-induced gastric malignancies. In light of our promising in silico results, we recommend these natural compounds as potential candidates for advancing H. pylori-targeted drug development. Given their potential, we strongly advocate for subsequent in vitro and in vivo studies to validate their therapeutic efficacy against this formidable gastrointestinal bacterium.
Collapse
Affiliation(s)
- Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Birulia, 1216, Ashulia, Dhaka, Bangladesh
| | - Imren Bayıl
- Department of Bioinformatics and Computational Biology, Gaziantep University, Turkey
| | - Sajjat Mahmood
- Department of Microbiology, Jagannath University, Chittaranjan Avenue in Sadarghat, Dhaka, 1100, Bangladesh
| | - Nobendu Mukerjee
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute Of Medical and Technical Sciences, Chennai, India
- Department of Microbiology, West Bengal State University, West Bengal, Kolkata, 700126, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Tamanna Akter Mili
- Department of Pharmacy, University of Asia Pacific, 74/A Green Rd, Dhaka, 1205, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | | | - Swastika Maitra
- Department of Microbiology, Adamas University, West Bengal, Kolkata, 700126, India
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh, 1145, Saudi Arabia
| | - Salim S. Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh, 1145, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh, 1145, Saudi Arabia
| | - Prabhakar Semwal
- Department of Biotechnology, Graphic Era University, Dehradun, Uttarakhand, 248002, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
25
|
AL-Ishaq RK, Kubatka P, Büsselberg D. Sweeteners and the Gut Microbiome: Effects on Gastrointestinal Cancers. Nutrients 2023; 15:3675. [PMID: 37686707 PMCID: PMC10489909 DOI: 10.3390/nu15173675] [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: 07/19/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Worldwide, the demand for natural and synthetic sweeteners in the food industry as an alternative to refined sugar is increasing. This has prompted more research to be conducted to estimate its safety and effects on health. The gut microbiome is critical in metabolizing selected sweeteners which might affect overall health. Recently, more studies have evaluated the relationship between sweeteners and the gut microbiome. This review summarizes the current knowledge regarding the role played by the gut microbiome in metabolizing selected sweeteners. It also addresses the influence of the five selected sweeteners and their metabolites on GI cancer-related pathways. Overall, the observed positive effects of sweetener consumption on GI cancer pathways, such as apoptosis and cell cycle arrest, require further investigation in order to understand the underlying mechanism.
Collapse
Affiliation(s)
- Raghad Khalid AL-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
26
|
Li S, Hao L, Hu X. Natural products target glycolysis in liver disease. Front Pharmacol 2023; 14:1242955. [PMID: 37663261 PMCID: PMC10469892 DOI: 10.3389/fphar.2023.1242955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
Mitochondrial dysfunction plays an important role in the occurrence and development of different liver diseases. Oxidative phosphorylation (OXPHOS) dysfunction and production of reactive oxygen species are closely related to mitochondrial dysfunction, forcing glycolysis to become the main source of energy metabolism of liver cells. Moreover, glycolysis is also enhanced to varying degrees in different liver diseases, especially in liver cancer. Therefore, targeting the glycolytic signaling pathway provides a new strategy for the treatment of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis associated with liver cancer. Natural products regulate many steps of glycolysis, and targeting glycolysis with natural products is a promising cancer treatment. In this review, we have mainly illustrated the relationship between glycolysis and liver disease, natural products can work by targeting key enzymes in glycolysis and their associated proteins, so understanding how natural products regulate glycolysis can help clarify the therapeutic mechanisms these drugs use to inhibit liver disease.
Collapse
Affiliation(s)
- Shenghao Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
27
|
Delkhah AMD, Karimi E, Farivar S. Herniarin-loaded solid lipid nanoparticles: promising molecular mechanism and therapeutic potential against pancreatic cancer line. Mol Biol Rep 2023; 50:6469-6479. [PMID: 37326747 DOI: 10.1007/s11033-023-08560-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND The notion of cancer therapy is intrinsically subjected to multiple challenges due to the drug resistance and drug toxicity for normal tissues. Herniarin (7-methoxycoumarin) belongs to the naturally occurring aromatic phytochemicals and coumarins. Considering the boosting effect of nanocarriers in drug delivery, we investigated the proapoptotic, anti-metastatic properties, and molecular mechanism of herniarin-loaded solid lipid nanoparticles on human gastric adenocarcinoma (AGS), human colon adenocarcinoma (HT-29), human pancreatic carcinoma (Panc-1), and normal human skin fibroblast (HFF) cell lines. METHODS AND RESULTS The cytotoxicity of synthesized nanoparticle have been tested using MTT assay. The obtained results manifested that concentration of herniarin that exerts 50% cell growth inhibition (IC50) against HT-29, AGS, and Panc-1 was calculated 138.34, 123.46, and 83.744 µL, respectively. Given that nanoparticles showed lowest IC50 values on Panc-1 cell line, these cells were selected for further analysis. The apoptosis induction and cell cycle arrest were examined performing real-time PCR, flow cytometry, and DAPI/acridine orange-propidium iodide staining. The expression of apoptosis-related genes, including BCL-2, was decreased, while the expression of CASP9, CASP8, and CASP3 was increased in response to the treatment. Moreover, the expression of metastasis-related gene (MMP2) was significantly suppressed under Her-SLN-NPs treatment. According to the flow cytometry findings, we observed no cell cycle arrest at any stage. CONCLUSION Our funding manifested herniarin encapsulated solid lipid nanoparticles has potent therapeutic target against Panc-1 cell line.
Collapse
Affiliation(s)
- Arman Mokaram Doust Delkhah
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Ehsan Karimi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Shirin Farivar
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
28
|
Melo LFMD, Aquino-Martins VGDQ, Silva APD, Oliveira Rocha HA, Scortecci KC. Biological and pharmacological aspects of tannins and potential biotechnological applications. Food Chem 2023; 414:135645. [PMID: 36821920 DOI: 10.1016/j.foodchem.2023.135645] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/29/2023] [Accepted: 02/04/2023] [Indexed: 02/09/2023]
Abstract
Secondary metabolites are divided into three classes: phenolic, terpenoid, and nitrogenous compounds. Phenolic compounds are also known as polyphenols and include tannins, classified as hydrolysable or condensed. Herein, we explored tannins for their ROS reduction characteristics and role in homeostasis. These activities are associated with the numbers and degree of polymerisation of reactive hydroxyl groups present in the phenolic rings of tannins. These characteristics are associated with anti-inflammatory, anti-aging, and anti-proliferative health benefits. Tannins can reduce the risk of cancer and neurodegenerative diseases, such as cardiovascular diseases and Alzheimer's, respectively. These biomolecules may be used as nutraceuticals to maintain good gut microbiota. Industrial applications include providing durability to leather, anti-corrosive properties to metals, and substrates for 3D printing and in bio-based foam manufacture. This review updates regarding tannin-based research and highlights its biological and pharmacological relevance and potential applications.
Collapse
Affiliation(s)
- Luciana Fentanes Moura de Melo
- Departamento de Biologia Celular e Genética - Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59072-970, Bairro Lagoa Nova, Natal, RN, Brazil; Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59078-970, Bairro Lagoa Nova, Natal, RN, Brazil
| | - Verônica Giuliani de Queiroz Aquino-Martins
- Departamento de Biologia Celular e Genética - Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59072-970, Bairro Lagoa Nova, Natal, RN, Brazil; Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59078-970, Bairro Lagoa Nova, Natal, RN, Brazil
| | - Ariana Pereira da Silva
- Departamento de Biologia Celular e Genética - Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59072-970, Bairro Lagoa Nova, Natal, RN, Brazil; Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59078-970, Bairro Lagoa Nova, Natal, RN, Brazil
| | - Hugo Alexandre Oliveira Rocha
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59078-970, Bairro Lagoa Nova, Natal, RN, Brazil; Departamento de Bioquímica - Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59078-970, Bairro Lagoa Nova, Natal, RN, Brazil
| | - Katia Castanho Scortecci
- Departamento de Biologia Celular e Genética - Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59072-970, Bairro Lagoa Nova, Natal, RN, Brazil; Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Campus Universitário UFRN, 59078-970, Bairro Lagoa Nova, Natal, RN, Brazil.
| |
Collapse
|
29
|
Esquivel-Chirino C, Bolaños-Carrillo MA, Carmona-Ruiz D, Lopéz-Macay A, Hernández-Sánchez F, Montés-Sánchez D, Escuadra-Landeros M, Gaitán-Cepeda LA, Maldonado-Frías S, Yáñez-Ocampo BR, Ventura-Gallegos JL, Laparra-Escareño H, Mejía-Velázquez CP, Zentella-Dehesa A. The Protective Role of Cranberries and Blueberries in Oral Cancer. PLANTS (BASEL, SWITZERLAND) 2023; 12:2330. [PMID: 37375955 DOI: 10.3390/plants12122330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Oral cancer has a high prevalence worldwide, and this disease is caused by genetic, immunological, and environmental factors. The main risk factors associated with oral cancer are smoking and alcohol. RESULTS There are various strategies to reduce risk factors, including prevention programs as well as the consumption of an adequate diet that includes phytochemical compounds derived from cranberries (Vaccinium macrocarpon A.) and blueberries (Vaccinium corymbosum L.); these compounds exhibit antitumor properties. RESULTS The main outcome of this review is as follows: the properties of phytochemicals derived from cranberries were evaluated for protection against risk factors associated with oral cancer. CONCLUSIONS The secondary metabolites of cranberries promote biological effects that provide protection against smoking and alcoholism. An alternative for the prevention of oral cancer can be the consumption of these cranberries and blueberries.
Collapse
Affiliation(s)
- César Esquivel-Chirino
- Área de Básicas Médicas, División de Estudios Profesionales, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Mario Augusto Bolaños-Carrillo
- Área de Ciencias Naturales, Departamento de Bachillerato, Universidad del Valle de México, Campus Guadalajara Sur, Guadalajara 045601, Mexico
| | - Daniela Carmona-Ruiz
- Área de Ortodoncia, División de Estudios Profesionales, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Ambar Lopéz-Macay
- Laboratorio de Liquído Sinovial, Instituto Nacional de Rehabilitación LGII, Ciudad de México 14389, Mexico
| | - Fernando Hernández-Sánchez
- Departamento de Virología y Micología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Ciudad de México 04502, Mexico
| | - Delina Montés-Sánchez
- Investigación Biomédica Básica, Licenciatura en Estomatología, Benemérita Universidad Autónoma de Puebla, Puebla 75770, Mexico
| | | | - Luis Alberto Gaitán-Cepeda
- Departamento de Medicina y Patología Oral Clínica, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Silvia Maldonado-Frías
- Laboratorio de Bioingeniería de Tejidos, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04360, Mexico
| | - Beatriz Raquel Yáñez-Ocampo
- Especialidad en Periodoncia e Implantología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - José Luis Ventura-Gallegos
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México 04510, Mexico
| | - Hugo Laparra-Escareño
- Departamento de Cirugía, Sección de Cirugía Vascular y Terapia, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Claudia Patricia Mejía-Velázquez
- Departamento de Patología, Medicina Bucal y Maxilofacial, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Alejandro Zentella-Dehesa
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México 04510, Mexico
- Unidad de Bioquímica, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| |
Collapse
|
30
|
Cid-Gallegos MS, Jiménez-Martínez C, Sánchez-Chino XM, Madrigal-Bujaidar E, Vásquez-Garzón VR, Baltiérrez-Hoyos R, Álvarez-González I. Chemopreventive Effect of Cooked Chickpea on Colon Carcinogenesis Evolution in AOM/DSS-Induced Balb/c Mice. PLANTS (BASEL, SWITZERLAND) 2023; 12:2317. [PMID: 37375942 PMCID: PMC10304688 DOI: 10.3390/plants12122317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023]
Abstract
Chickpeas are one of the most widely consumed legumes worldwide and they might prevent diseases such as cancer. Therefore, this study evaluates the chemopreventive effect of chickpea (Cicer arietinum L.) on the evolution of colon carcinogenesis induced with azoxymethane (AOM) and dextran sodium sulfate (DSS) in a mice model at 1, 7, and 14 weeks after induction. Accordingly, the expression of biomarkers-such as argyrophilic nucleolar organizing regions (AgNOR), cell proliferation nuclear antigen (PCNA), β-catenin, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2)-was assessed in the colon of BALB/c mice fed diets supplemented with 10 and 20% cooked chickpea (CC). The results showed that a 20% CC diet significantly reduced tumors and biomarkers of proliferation and inflammation in AOM/DSS-induced colon cancer mice. Moreover, body weight loss decreased and the disease activity index (DAI) was lower than the positive control. Lastly, tumor reduction was more evident at week 7 in the groups fed a 20% CC diet. In conclusion, both diets (10% and 20% CC) exert a chemopreventive effect.
Collapse
Affiliation(s)
- María Stephanie Cid-Gallegos
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City 07738, Mexico;
| | - Cristian Jiménez-Martínez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City 07738, Mexico;
| | - Xariss M. Sánchez-Chino
- Catedra-CONAHCYT, Departamento de Salud, El Colegio de la Frontera Sur-Villahermosa, Tabasco 86280, Mexico;
| | - Eduardo Madrigal-Bujaidar
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City 07738, Mexico;
| | - Verónica R. Vásquez-Garzón
- Catedra-CONAHCYT, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (V.R.V.-G.); (R.B.-H.)
| | - Rafael Baltiérrez-Hoyos
- Catedra-CONAHCYT, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (V.R.V.-G.); (R.B.-H.)
| | - Isela Álvarez-González
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City 07738, Mexico;
| |
Collapse
|
31
|
Yuan Z, Lu X, Lei F, Sun H, Jiang J, Xing D, Du L. Novel Effect of p-Coumaric Acid on Hepatic Lipolysis: Inhibition of Hepatic Lipid-Droplets. Molecules 2023; 28:4641. [PMID: 37375195 DOI: 10.3390/molecules28124641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
p-coumaric acid (p-CA), a common plant phenolic acid with multiple bioactivities, has a lipid-lowering effect. As a dietary polyphenol, its low toxicity, with the advantages of prophylactic and long-term administration, makes it a potential drug for prophylaxis and the treatment of nonalcoholic fatty liver disease (NAFLD). However, the mechanism by which it regulates lipid metabolism is still unclear. In this study, we studied the effect of p-CA on the down-regulation of accumulated lipids in vivo and in vitro. p-CA increased a number of lipase expressions, including hormone-sensitive lipase (HSL), monoacylglycerol lipase (MGL) and hepatic triglyceride lipase (HTGL), as well as the expression of genes related to fatty acid oxidation, including long-chain fatty acyl-CoA synthetase 1 (ACSL1), carnitine palmitoyltransferase-1 (CPT1), by activating peroxisome proliferator-activated receptor α, and γ (PPARα and γ). Furthermore, p-CA promoted adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation and enhanced the expression of the mammalian suppressor of Sec4 (MSS4), a critical protein that can inhibit lipid droplet growth. Thus, p-CA can decrease lipid accumulation and inhibit lipid droplet fusion, which are correlated with the enhancement of liver lipases and genes related to fatty acid oxidation as an activator of PPARs. Therefore, p-CA is capable of regulating lipid metabolism and is a potential therapeutic drug or health care product for hyperlipidemia and fatty liver.
Collapse
Affiliation(s)
- Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Xi Lu
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Fan Lei
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hong Sun
- Institute of Medicinal Plant and Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100094, China
| | - Jingfei Jiang
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongming Xing
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lijun Du
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
32
|
Alzate-Yepes T, Pérez-Palacio L, Martínez E, Osorio M. Mechanisms of Action of Fruit and Vegetable Phytochemicals in Colorectal Cancer Prevention. Molecules 2023; 28:molecules28114322. [PMID: 37298797 DOI: 10.3390/molecules28114322] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and its incidence is expected to increase by almost 80% by 2030. CRC apparition is related to poor diet, mainly due to low consumption of phytochemicals present in fruits and vegetables. Hence, this paper reviews the most promising phytochemicals in the literature, presenting scientific evidence regarding potential CRC chemopreventive effects. Moreover, this paper reveals the structure and action of CRC mechanisms that these phytochemicals are involved in. The review reveals that vegetables rich in phytochemicals such as carrots and green leafy vegetables, as well as some fruits such as pineapple, citrus fruits, papaya, mango, and Cape gooseberry, that have antioxidant, anti-inflammatory, and chemopreventive properties can promote a healthy colonic environment. Fruits and vegetables in the daily diet promote antitumor mechanisms by regulating cell signaling and/or proliferation pathways. Hence, daily consumption of these plant products is recommended to reduce the risk of CRC.
Collapse
Affiliation(s)
- Teresita Alzate-Yepes
- School of Nutrition and Dietetics, University of Antioquia, Carrera 75 # 65-87, Medellín 050010, Antioquia, Colombia
| | - Lorena Pérez-Palacio
- School of Nutrition and Dietetics, University of Antioquia, Carrera 75 # 65-87, Medellín 050010, Antioquia, Colombia
| | - Estefanía Martínez
- School of Engineering, Pontifical Bolivarian University, Circular 1 No. 70-01, Medellín 050031, Antioquia, Colombia
| | - Marlon Osorio
- School of Engineering, Pontifical Bolivarian University, Circular 1 No. 70-01, Medellín 050031, Antioquia, Colombia
- Systems Biology Group, School of Health Sciences, Pontifical Bolivarian University, Calle 78 B # 72 A 10, Medellín 050034, Antioquia, Colombia
| |
Collapse
|
33
|
Al-Ishaq RK, Samuel SM, Büsselberg D. The Influence of Gut Microbial Species on Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24098118. [PMID: 37175825 PMCID: PMC10179351 DOI: 10.3390/ijms24098118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with an alarming incidence rate and a considerable burden on the patient's life and health care providers. An increase in blood glucose level and insulin resistance characterizes it. Internal and external factors such as urbanization, obesity, and genetic mutations could increase the risk of DM. Microbes in the gut influence overall health through immunity and nutrition. Recently, more studies have been conducted to evaluate and estimate the role of the gut microbiome in diabetes development, progression, and management. This review summarizes the current knowledge addressing three main bacterial species: Bifidobacterium adolescentis, Bifidobacterium bifidum, and Lactobacillus rhamnosus and their influence on diabetes and its underlying molecular mechanisms. Most studies illustrate that using those bacterial species positively reduces blood glucose levels and activates inflammatory markers. Additionally, we reported the relationship between those bacterial species and metformin, one of the commonly used antidiabetic drugs. Overall, more research is needed to understand the influence of the gut microbiome on the development of diabetes. Furthermore, more efforts are required to standardize the model used, concentration ranges, and interpretation tools to advance the field further.
Collapse
Affiliation(s)
- Raghad Khalid Al-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| |
Collapse
|
34
|
Nie F, Liu L, Cui J, Zhao Y, Zhang D, Zhou D, Wu J, Li B, Wang T, Li M, Yan M. Oligomeric Proanthocyanidins: An Updated Review of Their Natural Sources, Synthesis, and Potentials. Antioxidants (Basel) 2023; 12:antiox12051004. [PMID: 37237870 DOI: 10.3390/antiox12051004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Oligomeric Proanthocyanidins (OPCs), as a class of compounds widely found in plants, are particularly abundant in grapes and blueberries. It is a polymer comprising many different monomers, such as catechins and epicatechins. The monomers are usually linked to each other by two types of links, A-linkages (C-O-C) and B-linkages (C-C), to form the polymers. Numerous studies have shown that compared to high polymeric procyanidins, OPCs exhibit antioxidant properties due to the presence of multiple hydroxyl groups. This review describes the molecular structure and natural source of OPCs, their general synthesis pathway in plants, their antioxidant capacity, and potential applications, especially the anti-inflammatory, anti-aging, cardiovascular disease prevention, and antineoplastic functions. Currently, OPCs have attracted much attention, being non-toxic and natural antioxidants of plant origin that scavenge free radicals from the human body. This review would provide some references for further research on the biological functions of OPCs and their application in various fields.
Collapse
Affiliation(s)
- Fanxuan Nie
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Lili Liu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Jiamin Cui
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yuquan Zhao
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Dawei Zhang
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Dinggang Zhou
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Jinfeng Wu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Bao Li
- Crop Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
- Hunan Engineering and Technology Research Center of Hybrid Rapeseed, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Tonghua Wang
- Crop Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
- Hunan Engineering and Technology Research Center of Hybrid Rapeseed, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Mei Li
- Crop Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
- Hunan Engineering and Technology Research Center of Hybrid Rapeseed, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Mingli Yan
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, Hunan University of Science and Technology, Xiangtan 411201, China
- Crop Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
- Hunan Engineering and Technology Research Center of Hybrid Rapeseed, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| |
Collapse
|
35
|
Xing P, Zhong Y, Cui X, Liu Z, Wu X. Natural products in digestive tract tumors metabolism: Functional and application prospects. Pharmacol Res 2023; 191:106766. [PMID: 37061144 DOI: 10.1016/j.phrs.2023.106766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
Digestive tract diseases are presently the hotspot of clinical diagnosis and treatment, and the incidence of digestive tract tumor is increasing annually. Surgery remains the main therapeutic schedule for digestive tract tumor. Though benefits were brought by neoadjuvant chemotherapy, a part of patients lose the chance of surgery because of late detection or inappropriate intervention. Therefore, the treatment of inoperable patients has become an urgent need. At the same time, tumor metabolism is an extremely complex and diverse process. Natural products are confirmed effective to inhibit the development of tumors in vitro and in vitro. There are many kinds of natural products and their functions remain not clear. However, some natural products such as polyphenols have been proven to have definite anti-cancer effects, and some terpenoids have definite anti-inflammatory, anti-ulcer, anti-tumor, and other effects. Therefore, the anti-tumor characteristics of natural products should arouse our high attention. Although there are many obstacles to study the activities of natural products in tumor, including the difficulty in detection or distinguishing each component due to their low levels in tumor tissue, etc., the emergence of highly sensitive and locatable spatial metabolomics make the research and application of natural products a big step forward. In this review, natural products such as phenols, terpenoids and biotinoids were summarized to further discuss the development and therapeutic properties of natural metabolites on digestive tract tumors.
Collapse
Affiliation(s)
- Peng Xing
- Department of Surgical Oncology, Breast Surgery, General Surgery, First Hospital of China Medical University, Shenyang, China
| | - Yifan Zhong
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Xiao Cui
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China.
| | - Xingda Wu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
36
|
Pimenta BV, Madrid RRM, Mathews PD, Riske KA, Loh W, Angelov B, Angelova A, Mertins O. Interaction of polyelectrolyte-shell cubosomes with serum albumin for triggering drug release in gastrointestinal cancer. J Mater Chem B 2023; 11:2490-2503. [PMID: 36852541 DOI: 10.1039/d2tb02670h] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nano-structured and functionalized materials for encapsulation, transport, targeting and controlled release of drugs are of high interest to overcome low bioavailability in oral administration. We develop lipid-based cubosomes, which are surface-functionalized with biocompatible chitosan-N-arginine and alginate, displaying internal liquid crystalline structures. Polyelectrolyte-shell (PS) cubosomes have pH-responsive characteristics profitable for oral delivery. The obtained PScubosomes can strongly interact with serum albumin, a protein which is released in the stomach under gastric cancer conditions. An effective thermodynamic PScubosome-protein interaction was characterized at pH 2.0 and 7.4 by isothermal titration calorimetry at 37 °C. A high increment of the albumin conformation transition temperature was evidenced by differential scanning calorimetry upon incubation with PScubosomes. The performed structural studies by synchrotron small-angle X-ray scattering (SAXS) revealed essential alterations in the internal liquid crystalline topology of the nanocarriers including an Im3m to Pn3m transition and a reduction of the cubic lattice parameters. The PScubosome nanoparticle interaction with serum albumin, leading to inner structural changes in a range of temperatures, promoted the release of water from the cubosomal nanochannels. Altogether, the results revealed effective interactions of the PScubosomes with albumin under simulated gastrointestinal pH conditions and suggested promising nanocarrier characteristics for triggered oral drug release.
Collapse
Affiliation(s)
- Barbara V Pimenta
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Rafael R M Madrid
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Patrick D Mathews
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Karin A Riske
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Watson Loh
- Institute of Chemistry, State University of Campinas (UNICAMP), 13083-970 Campinas, Brazil
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, CZ-18221 Prague, Czech Republic
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | - Omar Mertins
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil. .,Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
37
|
Guillamón E, Mut-Salud N, Rodríguez-Sojo MJ, Ruiz-Malagón AJ, Cuberos-Escobar A, Martínez-Férez A, Rodríguez-Nogales A, Gálvez J, Baños A. In Vitro Antitumor and Anti-Inflammatory Activities of Allium-Derived Compounds Propyl Propane Thiosulfonate (PTSO) and Propyl Propane Thiosulfinate (PTS). Nutrients 2023; 15:nu15061363. [PMID: 36986093 PMCID: PMC10058678 DOI: 10.3390/nu15061363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Increasing rates of cancer incidence and the side-effects of current chemotherapeutic treatments have led to the research on novel anticancer products based on dietary compounds. The use of Allium metabolites and extracts has been proposed to reduce the proliferation of tumor cells by several mechanisms. In this study, we have shown the in vitro anti-proliferative and anti-inflammatory effect of two onion-derived metabolites propyl propane thiosulfinate (PTS) and propyl propane thiosulfonate (PTSO) on several human tumor lines (MCF-7, T-84, A-549, HT-29, Panc-1, Jurkat, PC-3, SW-837, and T1-73). We observed that this effect was related to their ability to induce apoptosis regulated by oxidative stress. In addition, both compounds were also able to reduce the levels of some pro-inflammatory cytokines, such as IL-8, IL-6, and IL-17. Therefore, PTS and PTSO may have a promising role in cancer prevention and/or treatment.
Collapse
Affiliation(s)
| | | | - María Jesús Rodríguez-Sojo
- Department of Pharmacology, Center for Biomedical Research (CIBM), Instituto de Investigacion Biosanitaria de Granada (ibs.GRANADA), University of Granada, 18071 Granada, Spain
| | - Antonio Jesús Ruiz-Malagón
- Department of Pharmacology, Center for Biomedical Research (CIBM), Instituto de Investigacion Biosanitaria de Granada (ibs.GRANADA), University of Granada, 18071 Granada, Spain
| | | | - Antonio Martínez-Férez
- Chemical Engineering Department, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM), Instituto de Investigacion Biosanitaria de Granada (ibs.GRANADA), University of Granada, 18071 Granada, Spain
- Correspondence: (A.R.-N.); (J.G.)
| | - Julio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM), Instituto de Investigacion Biosanitaria de Granada (ibs.GRANADA), University of Granada, 18071 Granada, Spain
- CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (A.R.-N.); (J.G.)
| | | |
Collapse
|
38
|
Hashemy SI, Amiri H, Hosseini H, Sadeghzadeh F, Jaseem MMM, Tabrizi MH. PEGylated Lecithin-Chitosan-Folic Acid Nanoparticles as Nanocarriers of Allicin for In Vitro Controlled Release and Anticancer Effects. Appl Biochem Biotechnol 2023:10.1007/s12010-022-04310-y. [PMID: 36652093 DOI: 10.1007/s12010-022-04310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2022] [Indexed: 01/19/2023]
Abstract
In this study, chitosan-lecithin nanoparticles modified with polyethylene glycol (PEG) and folic acid (FA) were used to deliver allicin (AC) to colon cancer cells. AC-loaded polyethylene glycol (PEG) and folic acid (FA)-modified chitosan-lecithin nanoparticles (AC-PLCF-NPs) were fabricated via self-assembling procedure. HPLC for AC encapsulation and FA binding, MTT for viability assay, ABTS and DPPH for antioxidant capacity, disc diffusion, MIC and MBC for antibacterial assay, qPCR and AO/PI staining for apoptotic, and CAM assay for angiogenesis effects of AC-PLCF-NPs were used. AC-PLCF-NPs (113.55 nm) were synthesized as single dispersed (PDI: 0.28) and stable (ZP: + 33.18 mV) with 81% AC encapsulation and 48% FA binding. The antioxidant power of AC-PLCF-NPs was confirmed by inhibiting free radicals ABTS (74.25 µg/mL) and DPPH (366.214 µg/mL) and its antibacterial capacity with very high inhibitory effects against gram-negative bacterial strains. MTT results showed higher toxicity of AC-PLCF-NPs (68.06 µg/mL) compared to AC (171.45 µg/mL). Increased expression of caspase 3 and 9 genes showed activation of the intrinsic apoptosis pathway in treated cells, and on the other hand, reduction of vascular and embryonic growth factors in CAM model confirmed the anti-angiogenesis effects of AC-PLCF-NPs. AC-PLCF-NPs can be suggested as a promising therapeutic agent for studies in the field of colon cancer treatment.
Collapse
Affiliation(s)
- Seyed Isaac Hashemy
- Faculty of Medicine, Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Amiri
- Faculty of Medicine, Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseini
- Faculty of Medicine, Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzaneh Sadeghzadeh
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | | | | |
Collapse
|
39
|
de Araujo IG, Pattaro-Júnior JR, Barbosa CG, Philippsen GS, Silva AR, Ioshino RS, Moraes CB, Freitas-Junior LH, Barros L, Peralta RM, Fernandez MA, Seixas FAV. Potential of plant extracts in targeting SARS-CoV-2 main protease: an in vitro and in silico study. J Biomol Struct Dyn 2023; 41:12204-12213. [PMID: 36651196 DOI: 10.1080/07391102.2023.2166589] [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: 08/29/2022] [Accepted: 01/01/2023] [Indexed: 01/19/2023]
Abstract
The deaths caused by the covid-19 pandemic have recently decreased due to a worldwide effort in vaccination campaigns. However, even vaccinated people can develop a severe form of the disease that requires ICU admission. As a result, the search for antiviral drugs to treat these severe cases has become a necessity. In this context, natural products are an interesting alternative to synthetic medicines used in drug repositioning, as they have been consumed for a long time through traditional medicine. Many natural compounds found in plant extracts have already been shown to be effective in treating viral and bacterial diseases, making them possible hits to exploit against covid-19. The objective of this work was to evaluate the antiviral activity of different plant extracts available in the library of natural products of the Universidade Estadual de Maringá, by inhibiting the SARS-CoV-2 main protease (Mpro), and by preventing viral infection in a cellular model. As a result, the extract of Cytinus hypocistis, obtained by ultrasound, showed a Mpro inhibition capacity greater than 90%. In the infection model assays using Vero cells, an inhibition of 99.6% was observed, with a selectivity index of 42.7. The in silico molecular docking simulations using the extract compounds against Mpro, suggested Tellimagrandin II as the component of C. hypocistis extract most likely to inhibit the viral enzyme. These results demonstrate the potential of C. hypocistis extract as a promising source of natural compounds with antiviral activity against covid-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Cecilia Gomes Barbosa
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Universidade Municipal de São Caetano do Sul (USCS), São Caetano, Brazil
- Bela Vista, São Paulo, Brazil
| | | | - Ana Rita Silva
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha, (SusTEC), Instituto Politécnico de Bragança, Bragança, Portugal
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS-IBSAL, Universidad de Salamanca, Salamanca, España
| | | | | | | | - Lillian Barros
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha, (SusTEC), Instituto Politécnico de Bragança, Bragança, Portugal
| | | | - Maria Aparecida Fernandez
- Department of Biotechnology, Genetics and Cell Biology, Universidade Estadual de Maringá, Maringá, P.R. Brazil
| | | |
Collapse
|
40
|
Liang Z, Xu Y, Zhang Y, Zhang X, Song J, Qian H, Jin J. Anticancer applications of phytochemicals in gastric cancer: Effects and molecular mechanism. Front Pharmacol 2023; 13:1078090. [PMID: 36712679 PMCID: PMC9877357 DOI: 10.3389/fphar.2022.1078090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common malignant cancer and is a life-threatening disease worldwide. Phytochemicals have been shown to be a rational, safe, non-toxic, and very promising approach to the prevention and treatment of cancer. It has been found that phytochemicals have protective effects against GC through inhibiting cell proliferation, inducing apoptosis and autophagy, suppressing cell invasion and migration, anti-angiogenesis, inhibit Helicobacter pylori infection, regulating the microenvironment. In recent years, the role of phytochemicals in the occurrence, development, drug resistance and prognosis of GC has attracted more and more attention. In order to better understand the relationship between phytochemicals and gastric cancer, we briefly summarize the roles and functions of phytochemicals in GC tumorigenesis, development and prognosis. This review will probably help guide the public to prevent the occurrence and development of GC through phytochemicals, and develop functional foods or drugs for the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Zhaofeng Liang
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Chang Zhou, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yumeng Xu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yue Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xinyi Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiajia Song
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hui Qian
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Chang Zhou, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jianhua Jin
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Chang Zhou, China
| |
Collapse
|
41
|
Gong YQ, Hou FT, Xiang CL, Li CL, Hu GH, Chen CW. The mechanisms and roles of melatonin in gastrointestinal cancer. Front Oncol 2022; 12:1066698. [PMID: 36591447 PMCID: PMC9798083 DOI: 10.3389/fonc.2022.1066698] [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: 10/11/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Gastrointestinal (GI) cancer is a global health problem with wide lesions and numerous cases. The increased morbidity and mortality of GI cancer is a socio-economic challenge for decades to come. Melatonin, a nature indolamine, exerts a crucial role in molecular interactions involved in multiple functional and physiological processes. Increasing evidence indicates that melatonin can modulate GI tract, decrease the occurrence of GI cancer, and enhance the sensitivity to chemoradiotherapy. However, little is known about the exact role of melatonin in anti-carcinogenesis. In this review, we discuss the action of the beneficial effects of melatonin in GI carcinogenesis. Furthermore, we compile the understanding of the role of melatonin in GI cancer, including esophageal cancer (EC), gastric cancer (GC), hepatocellular carcinoma (HCC), colorectal cancer (CRC), and pancreatic cancer (PC). In addition, the potential therapeutic application and clinical evaluation of melatonin in GI cancer are also discussed.
Collapse
Affiliation(s)
- Yong-Qiang Gong
- Department of Gastrointestinal Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Fu-Tao Hou
- Department of Gastrointestinal Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Cai-Ling Xiang
- Department of Gastrointestinal Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Cheng-Long Li
- Department of Gastrointestinal Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Guo-Huang Hu
- Department of General Surgery, Institute of Digestive Surgery of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China,*Correspondence: Guo-Huang Hu, ; Chao-Wu Chen,
| | - Chao-Wu Chen
- Department of Gastrointestinal Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China,*Correspondence: Guo-Huang Hu, ; Chao-Wu Chen,
| |
Collapse
|
42
|
Flavonoids' Dual Benefits in Gastrointestinal Cancer and Diabetes: A Potential Treatment on the Horizon? Cancers (Basel) 2022; 14:cancers14246073. [PMID: 36551558 PMCID: PMC9776408 DOI: 10.3390/cancers14246073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/08/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Diabetes and gastrointestinal cancers (GI) are global health conditions with a massive burden on patients' lives worldwide. The development of both conditions is influenced by several factors, such as diet, genetics, environment, and infection, which shows a potential link between them. Flavonoids are naturally occurring phenolic compounds present in fruits and vegetables. Once ingested, unabsorbed flavonoids reaching the colon undergo enzymatic modification by the gut microbiome to facilitate absorption and produce ring fission products. The metabolized flavonoids exert antidiabetic and anti-GI cancer properties, targeting major impaired pathways such as apoptosis and cellular proliferation in both conditions, suggesting the potentially dual effects of flavonoids on diabetes and GI cancers. This review summarizes the current knowledge on the impact of flavonoids on diabetes and GI cancers in four significant pathways. It also addresses the synergistic effects of selected flavonoids on both conditions. While this is an intriguing approach, more studies are required to better understand the mechanism of how flavonoids can influence the same impaired pathways with different outcomes depending on the disease.
Collapse
|
43
|
Deepika, Maurya PK. Ellagic acid: insight into its protective effects in age-associated disorders. 3 Biotech 2022; 12:340. [PMID: 36340805 PMCID: PMC9633905 DOI: 10.1007/s13205-022-03409-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
The disparity in the free radical generation and the production of antioxidants to counteract its effect is known as oxidative stress. Oxidative stress causes damage to the macromolecules such as lipids, carbohydrates, proteins, and DNA and RNA. The oxidative damage to the cellular components leads to a process of aging and various age-associated disorders. The literature survey for this review was done using PubMed, Google Scholar, and Science Direct. The papers showing the studies related to aging and age-associated disorders have been selected for reviewing this paper. Ellagic acid has been used as the keyword, and more emphasis has been put on papers from the last 10 years. However, some papers with significant studies prior to 10 years have also been considered. Almost 250 papers have been studied for reviewing this paper, and about 135 papers have been cited. Ellagic acid (EA) is present in high quantities in pomegranate and various types of berries. It is known to possess the antioxidant potential and protects from the harmful effects of free radicals. Various studies have shown its effect to protect cardiovascular, neurodegenerative, cancer, and diabetes. The present review focuses on the protective effect of ellagic acid in age-associated disorders. The effect of EA has been studied in various chronic disorders but the scope of this review is limited to cancer, diabetes, cardiovascular and neurodegenerative disorders. All the disease aspects have not been addressed in this particular review.
Collapse
Affiliation(s)
- Deepika
- Department of Biochemistry, Central University of Haryana, Mahendragarh, 123031 India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendragarh, 123031 India
| |
Collapse
|
44
|
Chen PN, Lin CW, Yang SF, Chang YC. CLEFMA Induces the Apoptosis of Oral Squamous Carcinoma Cells through the Regulation of the P38/HO-1 Signalling Pathway. Cancers (Basel) 2022; 14:cancers14225519. [PMID: 36428612 PMCID: PMC9688613 DOI: 10.3390/cancers14225519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
The purpose of this research was to evaluate the impact and the underlying molecular mechanism of CLEFMA-induced cell death in human OSCC. The anti-tumour properties of CLEFMA in oral cancer were explored using colony formation, flow cytometry, human apoptosis array, Western blot, and immunohistochemistry assays. The in vivo anti-tumour effect of CLEFMA administered by oral gavage was evaluated using SCC-9-derived xenograft-bearing nude mouse models. CLEFMA significantly suppressed colony formation and elicited cellular apoptosis in oral cancer cells. CLEFMA treatment remarkably increased phosphorylated p38 and HO-1 along with cleavage of poly ADP-ribose polymerase and activation of caspase-8, -9, and -3 in HSC-3 and SCC-9 cells. Administration of HO-1 small interfering RNA significantly protected the cells from CLEFMA-induced caspase-3, -8, and -9 activation. Attenuation of p38 activity by the pharmacologic inhibitor SB203580 dramatically reduced CLEFMA-induced caspase-3, -8, and -9 activation and HO-1 expression in OSCC. The subcutaneous murine xenograft models showed that CLEFMA in vivo suppressed tumour growth in implanted SCC-9 cells. All of these findings indicated that CLEFMA induced apoptosis through the p38-dependent rise in HO-1 signal transduction cascades in OSCC.
Collapse
Affiliation(s)
- Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Yu-Chao Chang
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung 402, Taiwan
- Correspondence:
| |
Collapse
|
45
|
Geng Y, Wang J, Chen K, Li Q, Ping Z, Xue R, Zhang S. Effects of sea buckthorn (Hippophae rhamnoides L.) on factors related to metabolic syndrome: A systematic review and meta-analysis of randomized controlled trial. Phytother Res 2022; 36:4101-4114. [PMID: 36043374 DOI: 10.1002/ptr.7596] [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: 04/04/2022] [Revised: 06/24/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022]
Abstract
The purpose of this meta-analysis is to explore whether the supplement of sea buckthorn affects the factors related to metabolic syndrome. The related RCTs from five databases were systematically searched and comprehensively random effects model was used to calculate SMD and 95% CI. The Cochrane deviation risk tool was used to evaluate the deviation risk. Fifteen studies were involved in the meta-analysis. First, sea buckthorn supplementation reduced triglycerides [-0.722 (-1.129, -0.316); p < .001], total cholesterol [-0.345 (-0.639, -0.051); p = .021], low density lipoprotein cholesterol [-0.396 (-0.755, -0.037); p = .031], and increased high density lipoprotein cholesterol [0.370 (0.056, 0.684); p = .021] in overall subjects. Second, subgroup analysis showed that sea buckthorn supplementation reduced lipids only in people with abnormal lipid metabolism. Third, sea buckthorn had no effect on blood sugar, blood pressure, and BMI of the overall subjects. Sea buckthorn may affect the lipid metabolism in circulation, but it cannot affect blood glucose, blood pressure, and BMI. These indicators are closely associated with metabolic syndrome. This study may contribute to the development and utilization of sea buckthorn, and may provide a new and safer way for the prevention and treatment of metabolic syndrome. The limitation of this study is high heterogeneity, even if subgroup analysis is used. However, more clinical studies are needed to determine the real effect of sea buckthorn on metabolic syndrome.
Collapse
Affiliation(s)
- Yaping Geng
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jing Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ke Chen
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Qianwen Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Zhiguang Ping
- Department of Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ran Xue
- Institution of tuberculosis control, Jinan Municipal Center for Disease Control and Prevention, Jinan, China
| | - Shenshen Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
46
|
Mitra S, Emran TB, Chandran D, Zidan BMRM, Das R, Mamada SS, Masyita A, Salampe M, Nainu F, Khandaker MU, Idris AM, Simal-Gandara J. Cruciferous vegetables as a treasure of functional foods bioactive compounds: Targeting p53 family in gastrointestinal tract and associated cancers. Front Nutr 2022; 9:951935. [PMID: 35990357 PMCID: PMC9386315 DOI: 10.3389/fnut.2022.951935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022] Open
Abstract
In the past few years, phytochemicals from natural products have gotten the boundless praise in treating cancer. The promising role of cruciferous vegetables and active components contained in these vegetables, such as isothiocyanates, indole-3-carbinol, and isothiocyanates, has been widely researched in experimental in vitro and in vivo carcinogenesis models. The chemopreventive agents produced from the cruciferous vegetables were recurrently proven to affect carcinogenesis throughout the onset and developmental phases of cancer formation. Likewise, findings from clinical investigations and epidemiological research supported this statement. The anticancer activities of these functional foods bioactive compounds are closely related to their ability to upregulate p53 and its related target genes, e.g., p21. As the “guardian of the genome,” the p53 family (p53, p63, and p73) plays a pivotal role in preventing the cancer progression associated with DNA damage. This review discusses the functional foods bioactive compounds derived from several cruciferous vegetables and their use in altering the tumor-suppressive effect of p53 proteins. The association between the mutation of p53 and the incidence of gastrointestinal malignancies (gastric, small intestine, colon, liver, and pancreatic cancers) is also discussed. This review contains crucial information about the use of cruciferous vegetables in the treatment of gastrointestinal tract malignancies.
Collapse
Affiliation(s)
- Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh.,Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, Tamil Nadu, India
| | | | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | | | - Ayu Masyita
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | | | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Subang Jaya, Selangor, Malaysia
| | - Abubakr M Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia.,Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade de Vigo, Ourense, Spain
| |
Collapse
|
47
|
Carotenoids in Palliative Care—Is There Any Benefit from Carotenoid Supplementation in the Adjuvant Treatment of Cancer-Related Symptoms? Nutrients 2022; 14:nu14153183. [PMID: 35956359 PMCID: PMC9370407 DOI: 10.3390/nu14153183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/22/2022] Open
Abstract
Carotenoids are organic, liposoluble pigments found in nature, which are responsible for the characteristic colors of ripe tomatoes, carrots, peppers, and crustaceans, among others. Palliative care provided to patients with an incurable disease is aimed at improving the patient’s quality of life through appropriate treatment of symptoms accompanying the disease. Palliative care patients with burdensome symptoms related to advanced-stage cancers are especially interested in the use of natural dietary supplements and herbal remedies to reduce symptoms’ intensity and ameliorate the quality of life. Carotenoids seem to be a group of natural compounds with particularly promising properties in relieving symptoms, mainly due to their strong antioxidant, anti-inflammatory, and neuroprotective properties. Moreover, carotenoids have been used in folk medicine to treat various diseases and alleviate the accompanying symptoms. In this narrative review, the authors decided to determine whether there is any scientific evidence supporting the rationale for carotenoid supplementation in advanced-stage cancer patients, with particular emphasis on the adjuvant treatment of cancer-related symptoms, such as neuropathic pain and cancer-related cachexia.
Collapse
|
48
|
Effect of aqueous extract of seed of broccoli on inflammatory cytokines and Helicobacter pylori infection: a randomized, double-blind, controlled trial in patients without atrophic gastritis. Inflammopharmacology 2022; 30:1659-1668. [PMID: 35831736 DOI: 10.1007/s10787-022-01030-x] [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: 04/11/2022] [Accepted: 06/25/2022] [Indexed: 11/09/2022]
Abstract
The purpose of this study was to investigate the anti-inflammatory effect of an aqueous extract of seed of broccoli (AESB) in Helicobacter pylori (HP)-infected patients without atrophic gastritis. This was a double-centre, randomized, double-blind, controlled study. A total of 110 HP-infected subjects were randomized to receive either AESB or placebo for 2 months. Inflammatory cytokine (IL-8, IFN-γ, TNF-α, CRP, IL-17A, IL-1β, IL-18), pepsinogen I, II (PG I, PG II), and gastrin-17 (G-17) measurements and 13C-urea breath tests were performed at baseline and at 60 days. At 60 days, there was no significant difference in any of the inflammatory cytokines, pepsinogen or gastrin between the two groups. However, IL-8, IFN-γ, PG I, PG I/PG II ratio (PGR), and G-17 were reduced by 9.02 pg/mL, 5.08 pg/mL, 24.56 ng/mL, 1.75 and 0.3 pmol/L, respectively, in the AESB group compared with baseline (all P < 0.05). The HP eradication rates in the AESB group and placebo group were 11.11 and 3.70% at 60 days, respectively (P > 0.05). No treatment-related adverse events were reported. Thus, AESB may reduce the risk of gastric mucosal lesions and decrease the risk of gastric cancer by relieving inflammatory cytokines. The safety profile of AESB was satisfactory. This study is registered with the Chinese Clinical Trials Registry (Registration No. ChiCTR2100054249).
Collapse
|
49
|
Chuang CW, Chang KP, Cho HY, Chuang TH, Yu MC, Wu CL, Wu SN. Characterization of Inhibitory Capability on Hyperpolarization-Activated Cation Current Caused by Lutein (β,ε-Carotene-3,3'-Diol), a Dietary Xanthophyll Carotenoid. Int J Mol Sci 2022; 23:7186. [PMID: 35806190 PMCID: PMC9266545 DOI: 10.3390/ijms23137186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023] Open
Abstract
Lutein (β,ε-carotene-3,3'-diol), a xanthophyll carotenoid, is found in high concentrations in the macula of the human retina. It has been recognized to exert potential effectiveness in antioxidative and anti-inflammatory properties. However, whether and how its modifications on varying types of plasmalemmal ionic currents occur in electrically excitable cells remain incompletely answered. The current hypothesis is that lutein produces any direct adjustments on ionic currents (e.g., hyperpolarization-activated cation current, Ih [or funny current, If]). In the present study, GH3-cell exposure to lutein resulted in a time-, state- and concentration-dependent reduction in Ih amplitude with an IC50 value of 4.1 μM. There was a hyperpolarizing shift along the voltage axis in the steady-state activation curve of Ih in the presence of this compound, despite being void of changes in the gating charge of the curve. Under continued exposure to lutein (3 μM), further addition of oxaliplatin (10 μM) or ivabradine (3 μM) could be effective at either reversing or further decreasing lutein-induced suppression of hyperpolarization-evoked Ih, respectively. The voltage-dependent anti-clockwise hysteresis of Ih responding to long-lasting inverted isosceles-triangular ramp concentration-dependently became diminished by adding this compound. However, the addition of 10 μM lutein caused a mild but significant suppression in the amplitude of erg-mediated or A-type K+ currents. Under current-clamp potential recordings, the sag potential evoked by long-lasting hyperpolarizing current stimulus was reduced under cell exposure to lutein. Altogether, findings from the current observations enabled us to reflect that during cell exposure to lutein used at pharmacologically achievable concentrations, lutein-perturbed inhibition of Ih would be an ionic mechanism underlying its changes in membrane excitability.
Collapse
Affiliation(s)
- Chao-Wei Chuang
- Department of Ophthalmology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan; (C.-W.C.); (K.-P.C.)
| | - Kuo-Pin Chang
- Department of Ophthalmology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan; (C.-W.C.); (K.-P.C.)
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.); (M.-C.Y.)
| | - Tzu-Hsien Chuang
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.); (M.-C.Y.)
| | - Meng-Cheng Yu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.); (M.-C.Y.)
| | - Chao-Liang Wu
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan;
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.); (M.-C.Y.)
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| |
Collapse
|
50
|
Muema FW, Liu Y, Zhang Y, Chen G, Guo M. Flavonoids from Selaginella doederleinii Hieron and Their Antioxidant and Antiproliferative Activities. Antioxidants (Basel) 2022; 11:antiox11061189. [PMID: 35740086 PMCID: PMC9229023 DOI: 10.3390/antiox11061189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/02/2022] [Accepted: 06/09/2022] [Indexed: 01/13/2023] Open
Abstract
Selaginella doederleinii Hieron. (S. doederleinii) is a traditional herb that is widely used in China to treat several ailments, but mainly cancer. Studies have been carried out to determine the phytochemicals ascribed to its pharmacological activity. However, both phytochemical and pharmacological profiles have not been fully explored as few compounds have been reported. This study evaluated the flavonoid content of the ethanol extract and its four fractions (petroleum ether, dichloromethane, ethyl acetate, and n-butanol) together with their antioxidant activity (DPPH and FRAP assays). Further, the antiproliferative activity was evaluated. Two new secondary metabolites (1 and 3) were isolated from S. doederleinii, which comprised of an apigenin skeleton with a phenyl attached at C-8 of ring A and an acetyl group. Additionally, other known metabolites 2 and 4–16 were isolated, whereby compounds 2, 4, 5, 8, 12, 15, and 16 were reported for the first time in this species. These compounds were evaluated for their antioxidative potentials by both DPPH and FRAP assays, and for their antiproliferative activities by the MTT assay on three human cancer cell lines: colon cancer (HT-29), cervical cancer (HeLa), and lung cancer (A549). Compound 7 exhibited the best activity on the three cancer cell lines (HT-29, HeLa, A549) by inhibiting the rate of growth of the cancer cells in a dose-dependent manner with IC50 values of 27.97, 35.47, and 20.71 µM, respectively. The structure–activity relationship of the pure compounds was highlighted in this study. Hence, the study enriched both the phytochemical and pharmacological profiles of S. doederleinii.
Collapse
Affiliation(s)
- Felix Wambua Muema
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China; (F.W.M.); (Y.L.); (Y.Z.); (G.C.)
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 430074, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ye Liu
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China; (F.W.M.); (Y.L.); (Y.Z.); (G.C.)
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 430074, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongli Zhang
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China; (F.W.M.); (Y.L.); (Y.Z.); (G.C.)
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 430074, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guilin Chen
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China; (F.W.M.); (Y.L.); (Y.Z.); (G.C.)
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 430074, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mingquan Guo
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China; (F.W.M.); (Y.L.); (Y.Z.); (G.C.)
- Sino-Africa Joint Research Center, Chinese Academy of Sciences, Wuhan 430074, China
- Innovation Academy for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: ; Tel.: +86-027-8770-0850
| |
Collapse
|