1
|
Ariyanto EF, Farahana AK, Sudirman GSJ, Widiarsih E, Qomarilla N, Rahayu NS, Wikayani TP, Heryaman H, Wira DW, Triatin RD, Bashari MH, Pamela Y, Pratiwi YS, Ghozali M. Oyster Mushroom ( Pleurotus ostreatus) Ethanolic Extract Inhibits Pparg Expression While Maintaining the Methylation of the Pparg Promoter During 3T3-L1 Adipocyte Differentiation. J Exp Pharmacol 2025; 17:27-36. [PMID: 39834594 PMCID: PMC11745172 DOI: 10.2147/jep.s494116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Purpose This study aims to provide new insights into the potential of oyster mushroom (Pleurotus ostreatus) ethanolic extract in preventing obesity through the inhibition of Pparg expression and modulation of methylation level on Pparg promoter during 3T3-L1 adipocyte differentiation. Methods This in vitro quantitative experimental study was conducted by treating the 3T3-L1 cell line differentiated using 0.5 mM methyl-isobutyl-xanthine, 1 μM dexamethasone, and 10 μg/mL insulin-containing medium with oyster mushroom ethanolic extract. The extract was obtained from 80 g of dried oyster mushroom powder extracted three times with 800 mL of ethanol, filtered, evaporated, and reconstituted in dimethyl sulfoxide (DMSO) to final concentrations of 0, 25, 50, and 100 µg/mL, with DMSO limited to 0.5% in all solutions. Pparg mRNA expression was quantified by qRT-PCR analysis and Pparg promoter methylation levels were measured quantitatively by pyrosequencing of bisulfite-treated DNA samples. Results The addition of 25 µg/mL oyster mushroom ethanolic extract significantly suppressed Pparg mRNA expression with no significant change in the Pparg promoter methylation levels. Conclusion Oyster mushroom ethanolic extract inhibited Pparg mRNA expression without altering Pparg promoter methylation, suggesting reduced adipocyte differentiation. This study emphasizes the potential of oyster mushroom in the prevention or treatment of obesity by inhibiting adipocyte differentiation.
Collapse
Affiliation(s)
- Eko Fuji Ariyanto
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Research Center for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Anastasya Kania Farahana
- Undergraduate Program of Medical Doctor, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | | | - Erlina Widiarsih
- Molecular Genetics Laboratory, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Nurul Qomarilla
- Cell Culture Laboratory, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Nurul Setia Rahayu
- Molecular Genetics Laboratory, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Tenny Putri Wikayani
- Cell Culture Laboratory, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Henhen Heryaman
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Dwi Wahyudha Wira
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Rima Destya Triatin
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Research Center for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Muhammad Hasan Bashari
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Yunisa Pamela
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Yuni Susanti Pratiwi
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Mohammad Ghozali
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| |
Collapse
|
2
|
Abdullah, Ahmad N, Xiao J, Tian W, Khan NU, Hussain M, Ahsan HM, Hamed YS, Zhong H, Guan R. Gingerols: Preparation, encapsulation, and bioactivities focusing gut microbiome modulation and attenuation of disease symptoms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156352. [PMID: 39740381 DOI: 10.1016/j.phymed.2024.156352] [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: 03/31/2024] [Revised: 10/17/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut dysbiosis, chronic diseases, and microbial recurrent infections concerns have driven the researchers to explore phytochemicals from medicinal and food homologous plants to modulate gut microbiota, mitigate diseases, and inhibit pathogens. Gingerols have attracted attention as therapeutic agents due to their diverse biological activities like gut microbiome regulation, gastro-protective, anti-inflammatory, anti-microbial, and anti-oxidative effects. PURPOSE This review aimed to summarize the gingerols health-promoting potential, specifically focusing on the regulation of gut microbiome, attenuation of disease symptoms, mechanisms of action, and signaling pathways involved. METHOD Research findings from experimental and clinical studies have been summarized regarding gingerols effects on the modulation of gut microbiome and its metabolites, and attenuation of disease symptoms. RESULTS Gingerols are phenolic compounds characterized by a common 3-methoxy-4-hydroxyphenyl moiety in their chemical structures, and further divided into different gingerol types, including gingerols (major), shogaols, paradols, gingerdiols, gingerdiones, and zingerones (minor). Advanced extraction techniques (e.g., ionic liquid-based-, enzyme-assisted-, microwave-assisted-, pressurized liquid-, ultrasound-assisted-, and supercritical fluid extractions) were reported as optimal alternatives to conventional methods for gingerols extraction. Research studies reported that gingerols positively modulated the composition of gut microbiome that helped to combat disease symptoms (e.g., obesity by decreasing weight gain- (Lactobacillus reuteri and Lachnospiraceae) and increasing weight loss associated-bacteria (Akkermansia, Muribaculaceae, and Alloprevotella). Gingerols intervention also ameliorated ulcerative colitis by increasing relative abundance of the beneficial bacteria (Akkermansia, Lachnospiraceae NK4A136, and Muribaculaceae_norank), and decreasing pathogenic microorganisms (Bacteroides, Parabacteroides, and Desulfovibrio). Emerging delivery systems (e.g., microcapsules, nanoparticles, nanostructured lipid carriers, nanoemulsions, and nanoliposomes) can enhance the bioavailability and therapeutic efficacy of gingerols by preserving their inherent properties and addressing challenges of stability, solubility, and absorption. CONCLUSION Gingerols are promising therapeutic agents to modulate gut microbiome (increase beneficial bacteria and inhibit pathogenic microbes), and attenuate chronic disease symptoms such as diabetes, colitis, obesity, oxidative stress, and cancer. Despite significant progress, challenges persist in transforming research findings into industrial applications, such as stability and solubility during processing and low bioavailability in the distal gut to impart desirable health benefits.
Collapse
Affiliation(s)
- Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Naveed Ahmad
- Multan College of Food & Nutrition Sciences, Multan Medical & Dental College, Multan, Pakistan
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenni Tian
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hafiz Muhammad Ahsan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yahya Saud Hamed
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
3
|
Tan L, Mei J, Tang R, Huang D, Qi K, Ossowski Z, Wang X. Can exercise as a complementary technique manage inflammatory markers in women with breast cancer who are overweight and obese? A systematic review and meta-analysis. Complement Ther Med 2024; 88:103119. [PMID: 39710346 DOI: 10.1016/j.ctim.2024.103119] [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: 09/24/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Inflammation can result in the development of breast cancer in women with overweight and obese, and also affects the outcome and prognosis of breast cancer patients, thereby decreasing the cure and survival rates of breast cancer patients. Exercise may benefit breast cancer patients as a supplement to conventional treatments. However, research on the effects of exercise on inflammatory markers in women with breast cancer who are overweight and obese remains incomplete. OBJECTIVE A systematic review and meta-analysis were used to study the effects of exercise on inflammatory markers in women with breast cancer who are overweight and obese. METHOD Literature up to May 2024 was searched from databases such as Cochrane, Embase, Pubmed, Web of Science, and EBSCO, and English-language randomized controlled trials (RCTs) that met the inclusion criteria were screened. The screening criteria were as follows (A) written in English; (B) RCT; (C) studied in women with overweight obese and breast cancer; (D) outcome measures: inflammatory markers; (E) the duration of the exercise intervention was unlimited. RESULTS A total of 14 articles and 1064 participants were included. Exercise significantly reduced C-reactive protein (CRP) (MD: -0.52, 95 % CI: -0.94 to -0.11; p = 0.01; heterogeneity p < 0.1), interleukin-6 (IL-6) (MD: -0.87, 95 % CI: -1.62 to -0.11; p = 0.02; heterogeneity p < 0.1), and leptin (MD: -0.92, 95 % CI: -1.71 to -0.13; p = 0.02; heterogeneity p < 0.1) levels and exercise significantly increased adiponectin levels (MD: 0.89, 95 % CI: 0.03-1.75, p = 0.04; heterogeneity p < 0.1) but had no effect on tumor necrosis factor-α (TNF-α) (MD: -0.26, 95 % CI: -0.82-0.29; p = 0.35; heterogeneity p < 0.1) and IL-10 (MD: 0.14, 95 % CI: -0.17-0.45; p = 0.37; heterogeneity p = 0.45) were not significant. In addition, subgroup analyses suggest that combination training (CE) may be the most recommended type of exercise to decrease pro-inflammatory markers, and increase anti-inflammatory markers in women with overweight obesity, and have breast cancer. CONCLUSION Exercise significantly reduced CRP, IL-6, and leptin levels and overall increased adiponectin levels in women with overweight obese, and breast cancer. However, the effects on TNF-α and IL-10 levels were not significant. CE may be the most recommended type of exercise for reducing pro-inflammatory factors and increasing anti-inflammatory factors. Therefore, this study considers exercise as an effective complementary approach to managing inflammatory markers in women with breast cancer who are overweight and obese. Future researchers may consider exploring the combined effects of exercise and dietary control, weight loss, and other factors, and formulate a comprehensive treatment plan accordingly.
Collapse
Affiliation(s)
- Liang Tan
- Gdansk University of Physical Education and Sport, Gdansk 80-336, Poland.
| | - Jinyu Mei
- Department of Physical Education, Harbin Institute of Technology (Weihai), Weihai 264209, China.
| | - Ruihong Tang
- Education University of Hong Kong (EdUHK), Hongkong, 999077, China; Hunan First Normal University, Changsha 410002, China.
| | - Duo Huang
- Shangrao Normal University, Shangrao 334001, China.
| | - Kai Qi
- Gdansk University of Physical Education and Sport, Gdansk 80-336, Poland.
| | - Zbigniew Ossowski
- Gdansk University of Physical Education and Sport, Gdansk 80-336, Poland.
| | - Xiaoning Wang
- School of Physical Education, Shandong University, Jinan 250061, China.
| |
Collapse
|
4
|
Tian X, Liu P, Wang R, Hou Y, Zhou Y, Wang C, Zhang G. A review on the treatment of hyperlipidemia with Erchen Decoction. Front Pharmacol 2024; 15:1445950. [PMID: 39605912 PMCID: PMC11598526 DOI: 10.3389/fphar.2024.1445950] [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: 06/08/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Hyperlipidemia, commonly referred to as dyslipidemia, is characterized by elevated serum cholesterol and/or triglyceride levels. This condition contributes significantly to the high mortality rates associated with cardiovascular diseases, posing a serious threat to global health. Although statins remain the predominant pharmacological treatment for hyperlipidemia, their associated side effects have led to a growing interest in alternative therapeutic approaches. Traditional Chinese Medicine (TCM) is exploring these alternatives, with the Erchen Decoction (ECD) emerging as a promising candidate. This review aims to summarize current clinical research, elucidate the mechanisms of action, and assess the compatibility of ECD in the management of hyperlipidemia. By doing so, we hope to provide valuable insights and references for clinical practice and future research.
Collapse
Affiliation(s)
- Xinyang Tian
- Institute of Traditional Chinese Medicine Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peiyu Liu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ruolin Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yawei Hou
- Institute of Traditional Chinese Medicine Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ying Zhou
- Institute of Traditional Chinese Medicine Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Chunyan Wang
- Institute of Traditional Chinese Medicine Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Guiju Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Li P, Wang T, Qiu H, Zhang R, Yu C, Wang J. 6-Gingerol Inhibits De Novo Lipogenesis by Targeting Stearoyl-CoA Desaturase to Alleviate Fructose-Induced Hepatic Steatosis. Int J Mol Sci 2024; 25:11289. [PMID: 39457074 PMCID: PMC11508832 DOI: 10.3390/ijms252011289] [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: 09/26/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), also known as non-alcoholic fatty liver disease (NAFLD), is a worldwide liver disease without definitive or widely used therapeutic drugs in clinical practice. In this study, we confirm that 6-gingerol (6-G), an active ingredient of ginger (Zingiber officinale Roscoe) in traditional Chinese medicine (TCM), can alleviate fructose-induced hepatic steatosis. It was found that 6-G significantly decreased hyperlipidemia caused by high-fructose diets (HFD) in rats, and reversed the increase in hepatic de novo lipogenesis (DNL) and triglyceride (TG) levels induced by HFD, both in vivo and in vitro. Mechanistically, chemical proteomics and cellular thermal shift assay (CETSA)-proteomics approaches revealed that stearoyl-CoA desaturase (SCD) is a direct binding target of 6-G, which was confirmed by further CETSA assay and molecular docking. Meanwhile, it was found that 6-G could not alter SCD expression (in either mRNA or protein levels), but inhibited SCD activity (decreasing the desaturation levels of fatty acids) in HFD-fed rats. Furthermore, SCD deficiency mimicked the ability of 6-G to reduce lipid accumulation in HF-induced HepG2 cells, and impaired the improvement in hepatic steatosis brought about by 6-G treatment in HFD supplemented with oleic acid diet-induced SCD1 knockout mice. Taken together, our present study demonstrated that 6-G inhibits DNL by targeting SCD to alleviate fructose diet-induced hepatic steatosis.
Collapse
Affiliation(s)
- Pan Li
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Tingting Wang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Hongmei Qiu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Ruoyu Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China;
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Chao Yu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China;
| |
Collapse
|
6
|
Moon BR, Park JE, Han JS. HM-chromanone attenuates obesity and adipose tissue inflammation by downregulating SREBP-1c and NF-κb pathway in high-fat diet-fed mice. Arch Physiol Biochem 2024:1-9. [PMID: 39359053 DOI: 10.1080/13813455.2024.2399554] [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: 12/08/2023] [Revised: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024]
Abstract
Background: Obese adipose tissue produces various pro-inflammatory cytokines that are major contributors to adipose tissue inflammation. Objective: The present study aimed to determine the effects of HM-chromanone (HMC) against obesity and adipose tissue inflammation in high-fat diet-fed mice. Materials and methods: Twenty-four C57BL/6J male mice were divided into three groups: ND (normal diet), HFD (high-fat diet), and HFD + HMC. The ND group was fed a normal diet, whereas the HFD and HFD + HMC groups were fed a high-fat diet. After 10 weeks of feeding, the animals were orally administered the treatments daily for 9 weeks. The ND and HFD group received distilled water as treatment. The HFD+HMC group was treated with HM-chromaone (50 mg/kg). Results: HM-chromanone administration decreased body weight, fat mass, and adipocyte diameter. HM-chromanone also improved plasma lipid profiles, decreased leptin levels, and increased adiponectin levels. The inhibiting effect of HM-chromanone on SREBP-1c, PPARγ, C/EBPα, and FAS decreased adipogenesis, thereby alleviating lipid accumulation. Furthermore, HM-chromanone administration exhibited a reduction in macrophage infiltration and the expression of pro-inflammatory cytokines. HM-chromanone suppressed the phosphorylation of IκBα and NF-κB, leading to the inhibition of iNOS and COX2 expressions, resulting in decreased inflammation in adipose tissue. Discussion and conclusion: These results highlight the anti-obesity and anti-inflammatory properties of HM-chromanone, achieved through the downregulation of the SREBP-1c and NF-κB pathway in high-fat diet-fed mice.
Collapse
Affiliation(s)
- Bo Ra Moon
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan, Republic of Korea
| | - Jae Eun Park
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan, Republic of Korea
| | - Ji Sook Han
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
7
|
Qian J, Li Z, Wang J, Lin Y, Yu Y. 6-gingerol and its derivatives inhibit Helicobacter pylori-induced gastric mucosal inflammation and improve gastrin and somatostatin secretion. Front Microbiol 2024; 15:1451563. [PMID: 39234535 PMCID: PMC11371576 DOI: 10.3389/fmicb.2024.1451563] [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: 06/19/2024] [Accepted: 07/17/2024] [Indexed: 09/06/2024] Open
Abstract
The resistance of Helicobacter pylori (H. pylori) has increased in recent years, prompting a trend in the research and development of new drugs. In our study, three derivatives (JF-1, JF-2, and JF-3) were synthesized using 6-gingerol as the main component, while JF-4, containing both 6-gingerol and 6-shogaol as the main components, was extracted from dried ginger. The minimum inhibitory concentrations (MICs), determined using the ratio dilution method, were 80 μg/mL for JF-1, 40 μg/mL for JF-2, 30 μg/mL for JF-3, 40 μg/mL for JF-4, 60 μg/mL for 6-gingerol standard (SS), and 0.03 μg/mL for amoxicillin (AMX). After treating H. pylori-infected mice, the inflammation of the gastric mucosa was suppressed. The eradication rate of H. pylori was 16.7% of JF-3 low-dose treatment (LDT), 25.0% of JF-3 high-dose treatment (HDT), 16.7% of JF-4 LDT, 16.7% of JF-4 HDT, 30% of SS LDT, 50% of SS HDT, and 36.4% of the positive control group (PCG). The levels of gastrin, somatostatin (SST), IFN-γ, IL-4, and IL-8 were significantly recovered in the JF-3 and JF-4 administration groups, but the effect was stronger in the high-dose group. These results demonstrate that 6-gingerol and its derivatives have significant anti-Helicobacter pylori effects and are promising potential treatments for H. pylori infection.
Collapse
Affiliation(s)
- Jiali Qian
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhennan Li
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Jinhui Wang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuxian Lin
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China
| | - Yingcong Yu
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- School of Medicine, Shanghai University, Shanghai, China
- The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Zhang C, Rao A, Chen C, Li Y, Tan X, Long J, Wang X, Cai J, Huang J, Luo H, Li C, Dang Y. Pharmacological activity and clinical application analysis of traditional Chinese medicine ginger from the perspective of one source and multiple substances. Chin Med 2024; 19:97. [PMID: 38997763 PMCID: PMC11241951 DOI: 10.1186/s13020-024-00969-z] [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: 03/08/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
All types of ginger have common fundamental components, although they possess distinct strengths and inclinations when it comes to effectiveness and medicinal applications. Fresh ginger possesses the ability to effectively stimulate movement within the body, alleviate the act of vomiting, induce sweating, and provide relief for external syndromes. Dried ginger possesses both defensive and stimulant characteristics, which effectively raise the internal temperature and enhance the Yang energy. Fresh ginger is more hydrating than dried ginger, highly skilled at heating the Middle-jiao, alleviating pain, halting bleeding, and managing diarrhea. Dried ginger possesses the ability to alleviate coldness when consumed in a heated form, as well as to alleviate diarrhea when consumed in a heated form. It thrives in warm conditions and has a tendency to revert back to its warm nature. The moisture content of baked ginger is inferior to that of dried ginger, but it is highly effective in alleviating pain, bleeding, and diarrhea by warming the Middle-jiao. Ginger charcoal and stir-fried charcoal, produced through carbonization, have excellent heat retention properties and are effective in warming meridians and stopping bleeding. The potency and ability to spread of roasted ginger is less intense compared to fresh ginger, and its moisture content is not as low as that of dried ginger. The medicinal characteristics of this substance are gentle, making it beneficial for alleviating vomiting in patients who are physically frail. Its primary mode of action is on the Middle-jiao. Nevertheless, the main chemical compositions of various traditional Chinese medicines are nearly identical due to their shared base element. Ginger, in particular, possesses a range of pharmacological activities including antioxidant, anti-inflammatory, anti-tumor, anti-bacterial, and anticoagulant properties. However, modern pharmacological research has not fully acknowledged the clinical medicinal value of ginger and consequently, fails to provide accurate guidance for clinical medication. This situation has a negative impact on the contemporary advancement of traditional Chinese medicine (TCM). The research on modernizing ginger is conducted by analyzing and considering the prospects. It is based on Traditional Chinese Medicine (TCM) theory and incorporates the comprehensive perspective of TCM philosophy. In order to modernize ginger, it is essential to have a proper knowledge of the concepts of "recognizing nature by efficacy, homology, and mutual expression of nature and efficacy" and "rationally utilizing modern drug research technology". By applying these principles, we can construct a bridge towards the advancement of ginger.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Pharmacy, Chengdu University of Traditional Chinese, Medicine, Chengdu, 611137, China
| | - Anyang Rao
- Hongxing Town Government of Ruoergai County, Aba Tibetan and Qiang Autonomous Prefecture, Sichuan, 624504, China
| | - Cui Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology & NMPA & State Key Laboratory, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuqing Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology & NMPA & State Key Laboratory, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiuchi Tan
- Department of Pharmacy, Chengdu University of Traditional Chinese, Medicine, Chengdu, 611137, China
| | - Jiaxin Long
- Department of Pharmacy, Chengdu University of Traditional Chinese, Medicine, Chengdu, 611137, China
| | - Xinyue Wang
- Department of Pharmacy, Chengdu University of Traditional Chinese, Medicine, Chengdu, 611137, China
| | - Junjie Cai
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology & NMPA & State Key Laboratory, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiquan Huang
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology & NMPA & State Key Laboratory, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hua Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China.
| | - Chuwen Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology & NMPA & State Key Laboratory, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Yuanye Dang
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology & NMPA & State Key Laboratory, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
9
|
Zhao XY, Wang JQ, Neely GG, Shi YC, Wang QP. Natural compounds as obesity pharmacotherapies. Phytother Res 2024; 38:797-838. [PMID: 38083970 DOI: 10.1002/ptr.8083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 02/15/2024]
Abstract
Obesity has become a serious global public health problem, affecting over 988 million people worldwide. Nevertheless, current pharmacotherapies have proven inadequate. Natural compounds have garnered significant attention due to their potential antiobesity effects. Over the past three decades, ca. 50 natural compounds have been evaluated for the preventive and/or therapeutic effects on obesity in animals and humans. However, variations in the antiobesity efficacies among these natural compounds have been substantial, owing to differences in experimental designs, including variations in animal models, dosages, treatment durations, and administration methods. The feasibility of employing these natural compounds as pharmacotherapies for obesity remained uncertain. In this review, we systematically summarized the antiobesity efficacy and mechanisms of action of each natural compound in animal models. This comprehensive review furnishes valuable insights for the development of antiobesity medications based on natural compounds.
Collapse
Affiliation(s)
- Xin-Yuan Zhao
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ji-Qiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - G Gregory Neely
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Yan-Chuan Shi
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Qiao-Ping Wang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
10
|
Omidkhoda N, Mahdiani S, Hayes AW, Karimi G. Natural compounds against nonalcoholic fatty liver disease: A review on the involvement of the LKB1/AMPK signaling pathway. Phytother Res 2023; 37:5769-5786. [PMID: 37748097 DOI: 10.1002/ptr.8020] [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: 04/18/2023] [Revised: 08/18/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023]
Abstract
Although various therapeutic approaches are used to manage nonalcoholic fatty liver disease (NAFLD), the best approach to NAFLD management is unclear. NAFLD is a liver disorder associated with obesity, metabolic syndrome, and diabetes mellitus. NAFLD progression can lead to cirrhosis and end-stage liver disease. Hepatic kinase B1 (LKB1) is an upstream kinase of 5'-adenosine monophosphate-activated protein kinase (AMPK), a crucial regulator in hepatic lipid metabolism. Activation of LKB1/AMPK inhibits fatty acid synthesis, increases mitochondrial β-oxidation, decreases the expression of genes encoding lipogenic enzymes, improves nonalcoholic steatohepatitis, and suppresses NAFLD progression. One potential opening for new and safe chemicals that can tackle the NAFLD pathogenesis through the LKB1-AMPK pathway includes natural bioactive compounds. Accordingly, we summarized in vitro and in vivo studies regarding the effect of natural bioactive compounds such as a few members of the polyphenols, terpenoids, alkaloids, and some natural extracts on NAFLD through the LKB1/AMPK signaling pathway. This manuscript may shed light on the way to finding a new therapeutic agent for NAFLD management.
Collapse
Affiliation(s)
- Navid Omidkhoda
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Mahdiani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- College of Public Health, University of South Florida, Tampa, Florida, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Preciado-Ortiz ME, Martinez-Lopez E, Rodriguez-Echevarría R, Perez-Robles M, Gembe-Olivarez G, Rivera-Valdés JJ. 10‑Gingerol, a novel ginger compound, exhibits antiadipogenic effects without compromising cell viability in 3T3‑L1 cells. Biomed Rep 2023; 19:105. [PMID: 38025831 PMCID: PMC10646760 DOI: 10.3892/br.2023.1687] [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: 05/29/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is defined as excessive fat accumulation that can be detrimental to health and currently affects a large part of the global population. Obesity arises from excessive energy intake along with a sedentary lifestyle and leads to adipocytes with aggravated hypertrophy. Strategies have been designed to prevent and treat obesity. Nutrigenomics may serve a role in prevention of obesity using bioactive compounds present in certain foods with anti-obesogenic effects. Ginger (Zingiber officinale Roscoe) contains gingerols, key bioactive compounds that inhibit hypertrophy and hyperplasia of adipocytes. The present study aimed to evaluate the antiadipogenic activity of 10-gingerol (10-G) in the 3T3-L1 cell line. Three study groups were formed: Negative (3T3-L1 preadipocytes) and positive control (mature 3T3-L1 adipocytes) and 10-G (3T3-L1 preadipocytes stimulated with 10-G during adipogenic differentiation). Cell viability and lipid content were evaluated by MTT assay and Oil Red O staining, respectively. mRNA expression of CCAAT enhancer-binding protein α (C/ebpα), peroxisome proliferator-activated receptor γ (Pparγ), mechanistic target of rapamycin complex (Mtor), sterol regulatory element binding transcription factor 1 (Srebf1), acetyl-coenzyme A carboxylase (Acaca), fatty acid binding protein 4 (Fabp4), and 18S rRNA (Rn18s), was quantified by quantitative PCR. The protein expression of C/EPBα was analyzed by western blot. In the 10-G group, lipid content was decreased by 28.83% (P<0.0001) compared with the positive control; notably, cell viability was not affected (P=0.336). The mRNA expression in the 10-G group was higher for C/ebpα (P<0.001) and lower for Acaca (P<0.001), Fabp4 (P<0.001), Mtor (P<0.0001) and Srebf1 (P<0.0001) compared with the positive control group, while gene expression of Pparγ did not present significant changes. The presence of 10-G notably decreased C/EBPα protein levels in 3T3-L1 adipocytes. In summary, the antiadipogenic effect of 10-G during the differentiation of 3T3-L1 cells into adipocytes may be explained by mRNA downregulation of adipogenic transcriptional factors and lipid metabolism-associated genes.
Collapse
Affiliation(s)
- María Elizabeth Preciado-Ortiz
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
- PhD Program in Translational Nutrition Sciences, Department of Human Reproduction and Child Growth and Development, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Erika Martinez-Lopez
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Roberto Rodriguez-Echevarría
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Mariana Perez-Robles
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Gildardo Gembe-Olivarez
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
- Bachelor's Nutrition Program, Department of Human Reproduction and Child Growth and Development, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Juan José Rivera-Valdés
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| |
Collapse
|
12
|
Chuljerm H, Paradee N, Katekaew D, Nantachai P, Settakorn K, Srichairatanakool S, Koonyosying P. Iron Chelation Property, Antioxidant Activity, and Hepatoprotective Effect of 6-Gingerol-Rich Ginger ( Zingiber officinale) Extract in Iron-Loaded Huh7 Cells. PLANTS (BASEL, SWITZERLAND) 2023; 12:2936. [PMID: 37631148 PMCID: PMC10459954 DOI: 10.3390/plants12162936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023]
Abstract
Iron is essential for numerous biological processes; however, an iron imbalance can contribute to a number of diseases. An excess of iron can accumulate in the body and subsequently induce the production of reactive oxygen species (ROS), leading to oxidative tissue damage and organ dysfunction. The liver, a major iron storage site, is vulnerable to this iron-induced oxidative damage; however, this issue can be overcome by the chelation of excess iron. This study aimed to investigate the effect of 6-gingerol-rich ginger (Zingiber officinale) extract on iron chelation, antioxidation, and hepatoprotective function in protecting against iron-induced oxidative liver cell injury. In experiments, 6-gingerol was confirmed to be a main bioactive component of the ginger extract and possessed free radical scavenging activity, decreasing ABTS•+ and DPPH• radical levels, and inhibiting AAPH-induced red blood cell hemolysis. Interestingly, the extract significantly reduced the levels of labile cellular iron (LCI), intracellular ROS, and lipid peroxidation products (TBARS) in iron-loaded human hepatoma (Huh7) cells. In conclusion, this work highlights the iron chelation property of 6-gingerol-rich ginger extract and its antioxidant activity, which could potentially protect the liver from iron-induced oxidative tissue damage.
Collapse
Affiliation(s)
- Hataichanok Chuljerm
- School of Health Sciences Research, Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
- Environmental-Occupational Health Sciences and Non Communicable Diseases Research Center, Research Institute for Health Sciences Chiang Mai University, Chiang Mai 50200, Thailand
| | - Narisara Paradee
- Department of Biochemistry, Faculty of Medicine Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (K.S.); (S.S.)
| | - Dabudsawin Katekaew
- Science Classroom Affiliated School Project, Chiang Mai University Demonstration School, Chiang Mai University, Chiang Mai 50200, Thailand; (D.K.); (P.N.)
| | - Panaphat Nantachai
- Science Classroom Affiliated School Project, Chiang Mai University Demonstration School, Chiang Mai University, Chiang Mai 50200, Thailand; (D.K.); (P.N.)
| | - Kornvipa Settakorn
- Department of Biochemistry, Faculty of Medicine Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (K.S.); (S.S.)
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (K.S.); (S.S.)
| | - Pimpisid Koonyosying
- Department of Biochemistry, Faculty of Medicine Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (K.S.); (S.S.)
| |
Collapse
|
13
|
Hong KH, Um MY, Ahn J, Ha TY. 6-Gingerol Ameliorates Adiposity and Inflammation in Adipose Tissue in High Fat Diet-Induced Obese Mice: Association with Regulating of Adipokines. Nutrients 2023; 15:3457. [PMID: 37571394 PMCID: PMC10421254 DOI: 10.3390/nu15153457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
We investigated the effects of 6-gingerol on adiposity and obesity-induced inflammation by focusing on the regulation of adipogenesis and adipokines in white adipose tissue (WAT) of diet-induced obese mice. C57BL/6 mice were fed a high-fat diet (HFD) containing 0.05% 6-gingerol for 8 weeks. 6-Gingerol supplementation significantly reduced body weight, WAT mass, serum triglyceride, leptin and insulin levels, and HOMA-IR in HFD-fed mice. Additionally, the size of adipocytes in epididymal fat pads was reduced in HFD-fed mice by 6-gingerol supplementation. 6-Gingerol reduced the mRNA and protein levels of adipogenesis-related transcription factors, such as SREBP-1, PPARγ, and C/EBPα in WAT. Furthermore, 6-gingerol suppressed the expression of lipogenesis-related genes, such as fatty acid synthase and CD36 in WAT. Adiponectin expression was significantly increased, whereas inflammatory adipokines (leptin, resistin, TNF-α, MCP-1, and PAI-1) and the macrophage marker F4/80 were significantly reduced in the WAT of HFD-fed mice by 6-gingerol supplementation. In conclusion, 6-gingerol effectively contributed to the alleviation of adiposity and inflammation in WAT, which is associated with the regulation of adipokines in diet-induced obese mice.
Collapse
Affiliation(s)
- Kyung Hee Hong
- Department of Food Science and Nutrition, Dongseo University, Busan 47011, Republic of Korea;
| | - Min Young Um
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (M.Y.U.); (J.A.)
- Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Jiyun Ahn
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (M.Y.U.); (J.A.)
- Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Tae Youl Ha
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (M.Y.U.); (J.A.)
- Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
14
|
Gembe-Olivarez G, Preciado-Ortiz ME, Campos-Perez W, Rodríguez-Reyes SC, Martínez-López E, Rivera-Valdés JJ. A mix of ginger phenols exhibits anti‑adipogenic and lipolytic effects in mature adipocytes derived from 3T3‑L1 cells. Exp Ther Med 2023; 26:336. [PMID: 37383373 PMCID: PMC10294600 DOI: 10.3892/etm.2023.12035] [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: 02/01/2023] [Accepted: 04/28/2023] [Indexed: 06/30/2023] Open
Abstract
The prevalence of obesity has increased rapidly worldwide. Obesity is characterized by excessive adipose tissue in the body, which is related to hyperplasia and hypertrophy in adipocytes. Ginger (Zingiber officinale Roscoe) is a medicinal plant that possesses an anti-obesogenic effect mostly attributed to gingerols, the most abundant bioactive compounds in ginger. The anti-adipogenic and lipolytic effects of these phenols have been shown when investigated individually. Therefore, the present study aimed to evaluate the lipolytic and anti-adipogenic activity of a mix of the main ginger phenols 6-gingerol, 8-gingerol, 10-gingerol, 6-shogaol, 8-shogaol and 10-shogaol on the 3T3-L1 cell line. A total of four study groups were designed: Negative control (3T3-L1 preadipocytes); positive control (mature 3T3-L1 adipocytes); phenols-pre (3T3-L1 cells stimulated with the phenols mix during adipogenic differentiation); and phenols-post (mature 3T3-L1 adipocytes stimulated with the phenols mix). MTT viability cell assay and Oil Red O staining were performed. Glycerol concentration supernatants were determined using the VITROS 350 Chemistry System. Expression of mRNA was measured using qPCR. The treatment with a 2 µg/ml ginger phenol dose reduced the lipid content by 45.52±7.8 and 35.95±0.76% in the phenols-pre and -post group, respectively, compared with that in the positive control group. The phenols-post group presented a higher glycerol concentration in the supernatant compared with that in the positive control and the phenols-pre groups. The mRNA expression levels of CCAAT/enhancer-binding protein alpha, peroxisome proliferator activated receptor-γ, fatty acid-binding protein 4 and fatty acid synthase were higher in the phenols-pre and lower in the phenols-post groups, compared with those in the positive control group. To the best of our knowledge, the current study demonstrated for the first time the anti-adipogenic and lipolytic effects of a mix of the main bioactive compounds found in ginger, and it also established the basis to use this mix of phenols in in vivo studies and clinical trials.
Collapse
Affiliation(s)
- Gildardo Gembe-Olivarez
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
- Bachelor's Nutrition Program, Department of Human Reproduction and Child Growth and Development, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - María Elizabeth Preciado-Ortiz
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
- PhD Program in Translational Nutrition Sciences, Department of Human Reproduction and Child Growth and Development, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Wendy Campos-Perez
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Sarai Citlalic Rodríguez-Reyes
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Erika Martínez-López
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Juan José Rivera-Valdés
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| |
Collapse
|
15
|
Shaik Mohamed Sayed UF, Moshawih S, Goh HP, Kifli N, Gupta G, Singh SK, Chellappan DK, Dua K, Hermansyah A, Ser HL, Ming LC, Goh BH. Natural products as novel anti-obesity agents: insights into mechanisms of action and potential for therapeutic management. Front Pharmacol 2023; 14:1182937. [PMID: 37408757 PMCID: PMC10318930 DOI: 10.3389/fphar.2023.1182937] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 07/07/2023] Open
Abstract
Obesity affects more than 10% of the adult population globally. Despite the introduction of diverse medications aimed at combating fat accumulation and obesity, a significant number of these pharmaceutical interventions are linked to substantial occurrences of severe adverse events, occasionally leading to their withdrawal from the market. Natural products serve as attractive sources for anti-obesity agents as many of them can alter the host metabolic processes and maintain glucose homeostasis via metabolic and thermogenic stimulation, appetite regulation, pancreatic lipase and amylase inhibition, insulin sensitivity enhancing, adipogenesis inhibition and adipocyte apoptosis induction. In this review, we shed light on the biological processes that control energy balance and thermogenesis as well as metabolic pathways in white adipose tissue browning, we also highlight the anti-obesity potential of natural products with their mechanism of action. Based on previous findings, the crucial proteins and molecular pathways involved in adipose tissue browning and lipolysis induction are uncoupling protein-1, PR domain containing 16, and peroxisome proliferator-activated receptor-γ in addition to Sirtuin-1 and AMP-activated protein kinase pathway. Given that some phytochemicals can also lower proinflammatory substances like TNF-α, IL-6, and IL-1 secreted from adipose tissue and change the production of adipokines like leptin and adiponectin, which are important regulators of body weight, natural products represent a treasure trove for anti-obesity agents. In conclusion, conducting comprehensive research on natural products holds the potential to accelerate the development of an improved obesity management strategy characterized by heightened efficacy and reduced incidence of side effects.
Collapse
Affiliation(s)
| | - Said Moshawih
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Hui Poh Goh
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Nurolaini Kifli
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Andi Hermansyah
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
| | - Hooi Leng Ser
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Liu S, Jiang W, Liu C, Guo S, Wang H, Chang X. Chinese chestnut shell polyphenol extract regulates the JAK2/STAT3 pathway to alleviate high-fat diet-induced, leptin-resistant obesity in mice. Food Funct 2023; 14:4807-4823. [PMID: 37128963 DOI: 10.1039/d3fo00604b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Chinese chestnut shell is a by-product of chestnut food processing and is rich in polyphenols. This study sought to investigate the effect of chestnut shell polyphenol extract (CSP) on weight loss and lipid reduction in a 12-week high-fat diet (HFD)-induced murine obesity model. CSP (300 mg per kg body weight) was administered intragastrically daily. AG490, a JAK2 protein tyrosine kinase inhibitor, was also intraperitoneally injected. The results showed that an HFD induced leptin resistance (LR). Compared to corresponding values in the HFD group, CSP treatment improved blood lipid levels, weight, and leptin levels in obese mice (p < 0.01). Additionally, CSP treatment enhanced enzyme activity by improving total antioxidant capacity, attenuating oxidative stress, and reducing fat droplet accumulation and inflammation in the liver, epididymal, and retroperitoneal adipose tissue. CSP also activated the LEPR-JAK2/STAT3-PTP1B-SOCS-3 signal transduction pathway in hypothalamus tissue and improved LR while regulating the expression of proteins related to lipid metabolism (PPARγ, FAS, and LPL) in white adipose tissue in the retroperitoneal cavity. However, the amelioration of lipid metabolism by CSP was dependent on JAK2. Molecular docking simulation further demonstrated the strong binding affinity of procyanidin C1 (-10.3983297 kcal mol-1) and procyanidin B1 (-9.12686729 kcal mol-1) to the crystal structure of JAK2. These results suggest that CSP may be used to reduce HFD-induced obesity with potential application as a functional food additive.
Collapse
Affiliation(s)
- Suwen Liu
- Engineering Research Center of Chestnut Industry Technology of Ministry of Education, Hebei Key Laboratory of Active Components and Functions in Natural Products, College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China.
- Hebei Yanshan Special Industrial Technology Research Institute, Qinhuangdao, Hebei 066004, China
| | - Wenhong Jiang
- Hebei Yanshan Special Industrial Technology Research Institute, Qinhuangdao, Hebei 066004, China
- Cofco Great Wall Wine (Ningxia) Co., Ltd, Yinchuan, Ningxia, 750100, China
| | - Chang Liu
- Engineering Research Center of Chestnut Industry Technology of Ministry of Education, Hebei Key Laboratory of Active Components and Functions in Natural Products, College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China.
| | - Shuo Guo
- Engineering Research Center of Chestnut Industry Technology of Ministry of Education, Hebei Key Laboratory of Active Components and Functions in Natural Products, College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China.
| | - Hao Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin 300457, China
| | - Xuedong Chang
- Engineering Research Center of Chestnut Industry Technology of Ministry of Education, Hebei Key Laboratory of Active Components and Functions in Natural Products, College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China.
- Hebei Yanshan Special Industrial Technology Research Institute, Qinhuangdao, Hebei 066004, China
| |
Collapse
|
17
|
Liu Y, Li D, Wang S, Peng Z, Tan Q, He Q, Wang J. 6-Gingerol Ameliorates Hepatic Steatosis, Inflammation and Oxidative Stress in High-Fat Diet-Fed Mice through Activating LKB1/AMPK Signaling. Int J Mol Sci 2023; 24:ijms24076285. [PMID: 37047258 PMCID: PMC10094681 DOI: 10.3390/ijms24076285] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/18/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
6-Gingerol, one of the major pharmacologically active ingredients extracted from ginger, has been reported experimentally to exert hepatic protection in non-alcoholic fatty liver disease (NAFLD). However, the molecular mechanism remains largely elusive. RNA sequencing indicated the significant involvement of the AMPK signaling pathway in 6-gingerol-induced alleviation of NAFLD in vivo. Given the significance of the LKB1/AMPK pathway in metabolic homeostasis, this study aims to investigate its role in 6-gingerol-induced mitigation on NAFLD. Our study showed that 6-gingerol ameliorated hepatic steatosis, inflammation and oxidative stress in vivo and in vitro. Further experiment validation suggested that 6-gingerol activated an LKB1/AMPK pathway cascade in vivo and in vitro. Co-immunoprecipitation analysis demonstrated that the 6-gingerol-elicited activation of an LKB1/AMPK pathway cascade was related to the enhanced stability of the LKB1/STRAD/MO25 complex. Furthermore, radicicol, an LKB1 destabilizer, inhibited the activating effect of 6-gingerol on an LKB1/AMPK pathway cascade via destabilizing LKB1/STRAD/MO25 complex stability in vitro, thus reversing the 6-gingerol-elicited ameliorative effect. In addition, molecular docking analysis further predicated the binding pockets of LKB1 necessary for binding with 6-gingerol. In conclusion, our results indicate that 6-gingerol plays an important role in regulating the stability of the LKB1/STRAD/MO25 complex and the activation of LKB1, which might weigh heavily in the 6-gingerol alleviation of NAFLD.
Collapse
Affiliation(s)
- Yuzhe Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Dong Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ze Peng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qi Tan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qifeng He
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
- Correspondence:
| |
Collapse
|
18
|
Wang H, Li Y, Dai Y, Ma L, Di D, Liu J. Screening, structural characterization and anti-adipogenesis effect of a water-soluble polysaccharide from Lycium barbarum L. by an activity-oriented approach. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
19
|
Wu S, Zhu J, Wu G, Hu Z, Ying P, Bao Z, Ding Z, Tan X. 6-Gingerol Alleviates Ferroptosis and Inflammation of Diabetic Cardiomyopathy via the Nrf2/HO-1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3027514. [PMID: 36624878 PMCID: PMC9825225 DOI: 10.1155/2022/3027514] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Diabetes mellitus (DM) can induce cardiomyocyte injury and lead to diabetic cardiomyopathy (DCM) which presently has no specific treatments and consequently increase risk of mortality. OBJECTIVE To characterize the therapeutic effect of 6-gingerol (6-G) on DCM and identify its potential mechanism. METHODS In vivo streptozotocin- (STZ-) induced DM model was established by using a high-fat diet and STZ, followed by low-dose (25 mg/kg) and high-dose (75 mg/kg) 6-G intervention. For an in vitro DCM model, H9c2 rat cardiomyoblast cells were stimulated with high glucose (glucose = 33 mM) and palmitic acid (100 μM) and then treated with 6-G (100 μM). Histological and echocardiographic analyses were used to assess the effect of 6-G on cardiac structure and function in DCM. Western blotting, ELISA, and real-time qPCR were used to assess the expression of ferroptosis, inflammation, and the Nrf2/HO-1 pathway-related proteins and RNAs. Protein expression of collagen I and collagen III was assessed by immunohistochemistry, and kits were used to assay SOD, MDA, and iron levels. RESULTS The results showed that 6-G decreased cardiac injury in both mouse and cell models of DCM. The cardiomyocyte hypertrophy and interstitial fibrosis were attenuated by 6-G treatment in vivo and resulted in an improved heart function. 6-G inhibited the expression of ferroptosis-related protein FACL4 and the content of iron and enhanced the expression of anti-ferroptosis-related protein GPX4. In addition, 6-G also diminished the secretion of inflammatory cytokines, including IL-1β, IL-6, and TNF-α. 6-G treatment activated the Nrf2/HO-1 pathway, enhanced antioxidative stress capacity proved by increased activity of SOD, and decreased MDA production. Compared with in vivo, 6-G treatment of H9c2 cells treated with high glucose and palmitic acid could produce a similar effect. CONCLUSION These findings suggest that 6-G could protect against DCM by the mechanism of ferroptosis inhibition and inflammation reduction via enhancing the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Shenglin Wu
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Jinxiu Zhu
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
- Longgang Maternity and Child Institute of Shantou University Medica College, Shenzhen 518100, Guangdong, China
| | - Guihai Wu
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Zuoqi Hu
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Pengxiang Ying
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Zhijun Bao
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Zipeng Ding
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Xuerui Tan
- Institute of Clinical Electrocardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| |
Collapse
|
20
|
Health benefits of bioactive components in pungent spices mediated via the involvement of TRPV1 channel. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|