1
|
Zhu D, Zhang J, Huang X, Wei N, Jiang J, Li J, Liu L, Liu Y, Zhou J, Jia J. Integrated network pharmacology and experimental validation to elucidate the mechanism of celastrol in mitigating sepsis-induced acute lung injury in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156678. [PMID: 40133025 DOI: 10.1016/j.phymed.2025.156678] [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: 09/23/2024] [Revised: 01/13/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Sepsis is an acute, life-threatening condition that precipitates multiple organ failure, including acute lung injury (ALI), characterized by a complex pathophysiological process and elevated mortality rates. Celastrol, a pentacyclic triterpenoid quinone derived from traditional Chinese medicine, exhibits diverse pharmacological properties, including immunomodulatory, anti-inflammatory, anticancer, and antifibrotic effects, and has demonstrated favorable safety profiles in vivo. However, the precise mechanism by which CSL contributes to sepsis-induced ALI remains to be elucidated. PURPOSE The study aimed to explore the mechanisms by which celastrol mitigates sepsis-induced ALI using network pharmacology, followed by experimental validation of its regulatory effects on sepsis-induced ALI. METHODS Utilizing a network pharmacology analysis, the potential targets and pathways of celastrol were identified. To explore celastrol's therapeutic effects on ALI, a rat model of sepsis was induced via cecal ligation and puncture, followed by assessment through hematoxylin-eosin staining, Real-time quantitative polymerase chain reaction (RT-qPCR), and Western blotting. Further investigation involved evaluating celastrol's influence on LPS-stimulated A549 and Raw264.7 cells, employing RT-qPCR, Western blotting, and immunofluorescence techniques. RESULTS Network pharmacological analysis identified 10 core targets and 31 pathways relevant to sepsis-induced ALI, with STAT3, TLR4, HIF-1α, and NF-κB1 emerging as central targets. Animal experiments demonstrated that celastrol treatment significantly reduced lung tissue inflammation, as evidenced by immunohistochemistry, Western blot, and RT-qPCR results, in comparison to the cecal ligation and puncture group. Notably, the levels of IL-1β, TNF-α, HIF-1α, STAT3, and NF-κB1 proteins and mRNA in the celastrol treatment group were significantly reduced compared to those in the cecal ligation and puncture (CLP) group and the LPS-treated group. Additionally, Western blot and immunofluorescence analyses confirmed the activation of the NF-κB pathway in vitro. CONCLUSION This study indicates that celastrol significantly suppresses the expression of inflammatory factors in sepsis-induced ALI by inhibiting the NF-κB/HIF-1α pathway in both in vivo and in vitro models, highlighting its therapeutic potential for modulating inflammation. These findings provide valuable evidence for future clinical research and drug development.
Collapse
Affiliation(s)
- Danli Zhu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinghan Zhang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaochun Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Na Wei
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinxiu Jiang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiayao Li
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yulin Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
2
|
Fu M, Lv M, Guo J, Mei A, Qian H, Yang H, Wu W, Liu Z, Zhong J, Wei Y, Min X, Wu H, Chen J. The clinical significance of T-cell regulation in hypertension treatment. Front Immunol 2025; 16:1550206. [PMID: 40079010 PMCID: PMC11897580 DOI: 10.3389/fimmu.2025.1550206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Hypertension, a globally prevalent condition, is closely associated with T cell-mediated inflammatory responses. Studies have shown that T cells, by secreting pro-inflammatory cytokines such as interferon-gamma (IFN-γ), Interleukin-17 (IL-17), and Tumor necrosis factor-alpha (TNF-α), directly lead to vascular dysfunction and elevated blood pressure. The activation of Th1 and Th17 cell subsets, along with the dysfunction of regulatory T cells (Tregs), is a critical mechanism in the onset and progression of hypertension. This review explores the role of T cells in the pathophysiology of hypertension and discusses potential therapeutic strategies targeting T cell regulation, such as immunotherapy and gene-editing technologies. These emerging treatments hold promise for providing personalized therapeutic options for hypertensive patients, reducing inflammatory complications, and improving treatment outcomes.
Collapse
Affiliation(s)
- Miaoxin Fu
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Mingzhu Lv
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jinyue Guo
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Aihua Mei
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hang Qian
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenwen Wu
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- School of Public Health, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhixin Liu
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Wei
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Haiyan Wu
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
3
|
Deveci G, Tek NA. Review on critical factor in monocyte adhesion: Nutrients. Cytokine 2025; 186:156845. [PMID: 39754794 DOI: 10.1016/j.cyto.2024.156845] [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: 10/14/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
Endogenous and exogenous factors play a role in endothelial dysfunction. Inflammation, leukocyte adhesion-aggregation, abnormal vascular proliferation, atherosclerosis, and hypertension are among the endogenous factors. Another factor that affects endothelial dysfunction is exogenous factors such as drug treatments, smoking, alcohol, and nutrition. According various studies on nutrition and endothelial function, it is supported that fatty acids, proteins, and phenolic compounds modulate this function. In vitro studies show that nutrients change the adhesion of monocytes to the endothelium. The pathways that play a role in the adhesion process of monocytes are also affected by nutrients. Particularly among these pathways, mTORC1, S6 plaques, monocyte chemotaxis protein, monocyte integrins, monocyte cytokines are transferred to the lesional area selectin protein. In this article, the effects of various nutrients on monocyte adhesion are examined. It explains the changes and possible mechanisms of nutrients such as fatty acids, protein, phenolic compounds, and other dietary components on monocyte adhesion, and examines the relationship between nutrients and monocyte adhesion in our country and allows us to look at our profession from a different perspective. Although not all nutritional elements are included, it is thought that our profession will play a role in taking the first step towards cell studies.
Collapse
Affiliation(s)
- Gülsüm Deveci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Çankırı Kartekin University, Çankırı 18000, Türkiye.
| | - Nilüfer Acar Tek
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Türkiye
| |
Collapse
|
4
|
Lilley T, Camera DM, Kwa FAA. Repairing muscle with broccoli-derived sulforaphane: A preclinical evaluation for the treatment of mitochondrial myopathies. Drug Discov Today 2025; 30:104283. [PMID: 39736463 DOI: 10.1016/j.drudis.2024.104283] [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: 10/18/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Skeletal muscle health relies on the production of adenosine triphosphate (ATP) in the mitochondria. ATP production is accompanied by oxidative phosphorylation, which generates reactive oxygen species (ROS). When there is an imbalance in ROS levels, oxidative stress and subsequent mitochondrial dysfunction, mitochondrial myopathies including sarcopenia, chronic progressive external ophthalmoplegia, and proximal myopathy can result. Such incurable myopathies are characterised by aberrant metabolism, limited ATP production, and muscle atrophy. Broccoli-derived sulforaphane has emerged as a novel treatment for mitochondrial myopathies because of its antioxidant and anti-inflammatory properties. This review discusses preclinical studies that reveal sulforaphane's potential therapeutic benefits and limitations in treating mitochondrial myopathies.
Collapse
Affiliation(s)
- Thomas Lilley
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Donny M Camera
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Faith A A Kwa
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia.
| |
Collapse
|
5
|
Narra F, Piragine E, Benedetti G, Ceccanti C, Florio M, Spezzini J, Troisi F, Giovannoni R, Martelli A, Guidi L. Impact of thermal processing on polyphenols, carotenoids, glucosinolates, and ascorbic acid in fruit and vegetables and their cardiovascular benefits. Compr Rev Food Sci Food Saf 2024; 23:e13426. [PMID: 39169551 PMCID: PMC11605278 DOI: 10.1111/1541-4337.13426] [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: 04/05/2024] [Revised: 06/19/2024] [Accepted: 07/28/2024] [Indexed: 08/23/2024]
Abstract
Bioactive compounds in fruit and vegetables have a positive impact on human health by reducing oxidative stress, inflammation, and the risk of chronic diseases such as cancer, cardiovascular (CV) diseases, and metabolic disorders. However, some fruit and vegetables must be heated before consumption and thermal processes can modify the amount of nutraceuticals, that is, polyphenols, carotenoids, glucosinolates, and ascorbic acid, that can increase or decrease in relation to different factors such as type of processing, temperature, and time but also the plant part (e.g., flower, leaf, tuber, and root) utilized as food. Another important aspect is related to the bioaccessibility and bioavailability of nutraceuticals. Indeed, the key stage of nutraceutical bioefficiency is oral bioavailability, which involves the release of nutraceuticals from fruit and vegetables in gastrointestinal fluids, the solubilization of nutraceuticals and their interaction with other components of gastrointestinal fluids, the absorption of nutraceuticals by the epithelial layer, and the chemical and biochemical transformations into epithelial cells. Several studies have shown that thermal processing can enhance the absorption of nutraceuticals from fruit and vegetable. Once absorbed, they reach the blood vessels and promote multiple biological effects (e.g., antioxidant, anti-inflammatory, antihypertensive, vasoprotective, and cardioprotective). In this review, we described the impact of different thermal processes (such as boiling, steaming and superheated steaming, blanching, and microwaving) on the retention/degradation of bioactive compounds and their health-promoting effects after the intake. We then summarized the impact of heating on the absorption of nutraceuticals and the biological effects promoted by natural compounds in the CV system to provide a comprehensive overview of the potential impact of thermal processing on the CV benefits of fruit and vegetables.
Collapse
Affiliation(s)
- Federica Narra
- Department of Agriculture, Food and EnvironmentUniversity of PisaPisaItaly
| | - Eugenia Piragine
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”University of PisaPisaItaly
- Department of PharmacyUniversity of PisaPisaItaly
| | | | - Costanza Ceccanti
- Department of Agriculture, Food and EnvironmentUniversity of PisaPisaItaly
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”University of PisaPisaItaly
| | - Marta Florio
- Department of Agriculture, Food and EnvironmentUniversity of PisaPisaItaly
| | | | | | - Roberto Giovannoni
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”University of PisaPisaItaly
- Department of BiologyUniversity of PisaPisaItaly
| | - Alma Martelli
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”University of PisaPisaItaly
- Department of PharmacyUniversity of PisaPisaItaly
| | - Lucia Guidi
- Department of Agriculture, Food and EnvironmentUniversity of PisaPisaItaly
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”University of PisaPisaItaly
| |
Collapse
|
6
|
Bontempo P, Capasso L, De Masi L, Nebbioso A, Rigano D. Therapeutic Potential of Natural Compounds Acting through Epigenetic Mechanisms in Cardiovascular Diseases: Current Findings and Future Directions. Nutrients 2024; 16:2399. [PMID: 39125279 PMCID: PMC11314203 DOI: 10.3390/nu16152399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain a leading global cause of morbidity and mortality. These diseases have a multifaceted nature being influenced by a multitude of biochemical, genetic, environmental, and behavioral factors. Epigenetic modifications have a crucial role in the onset and progression of CVD. Epigenetics, which regulates gene activity without altering the DNA's primary structure, can modulate cardiovascular homeostasis through DNA methylation, histone modification, and non-coding RNA regulation. The effects of environmental stimuli on CVD are mediated by epigenetic changes, which can be reversible and, hence, are susceptible to pharmacological interventions. This represents an opportunity to prevent diseases by targeting harmful epigenetic modifications. Factors such as high-fat diets or nutrient deficiencies can influence epigenetic enzymes, affecting fetal growth, metabolism, oxidative stress, inflammation, and atherosclerosis. Recent studies have shown that plant-derived bioactive compounds can modulate epigenetic regulators and inflammatory responses, contributing to the cardioprotective effects of diets. Understanding these nutriepigenetic effects and their reversibility is crucial for developing effective interventions to combat CVD. This review delves into the general mechanisms of epigenetics, its regulatory roles in CVD, and the potential of epigenetics as a CVD therapeutic strategy. It also examines the role of epigenetic natural compounds (ENCs) in CVD and their potential as intervention tools for prevention and therapy.
Collapse
Affiliation(s)
- Paola Bontempo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy; (L.C.); (A.N.)
| | - Lucia Capasso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy; (L.C.); (A.N.)
| | - Luigi De Masi
- National Research Council (CNR), Institute of Biosciences and BioResources (IBBR), Via Università 133, 80055 Portici, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy; (L.C.); (A.N.)
| | - Daniela Rigano
- Department of Pharmacy, University of Naples Federico II, Via Montesano 49, 80131 Naples, Italy;
| |
Collapse
|
7
|
Habtemariam S. Anti-Inflammatory Therapeutic Mechanisms of Isothiocyanates: Insights from Sulforaphane. Biomedicines 2024; 12:1169. [PMID: 38927376 PMCID: PMC11200786 DOI: 10.3390/biomedicines12061169] [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: 04/23/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Isothiocyanates (ITCs) belong to a group of natural products that possess a highly reactive electrophilic -N=C=S functional group. They are stored in plants as precursor molecules, glucosinolates, which are processed by the tyrosinase enzyme upon plant tissue damage to release ITCs, along with other products. Isolated from broccoli, sulforaphane is by far the most studied antioxidant ITC, acting primarily through the induction of a transcription factor, the nuclear factor erythroid 2-related factor 2 (Nrf2), which upregulates downstream antioxidant genes/proteins. Paradoxically, sulforaphane, as a pro-oxidant compound, can also increase the levels of reactive oxygen species, a mechanism which is attributed to its anticancer effect. Beyond highlighting the common pro-oxidant and antioxidant effects of sulforaphane, the present paper was designed to assess the diverse anti-inflammatory mechanisms reported to date using a variety of in vitro and in vivo experimental models. Sulforaphane downregulates the expression of pro-inflammatory cytokines, chemokines, adhesion molecules, cycloxyhenase-2, and inducible nitric oxide synthase. The signalling pathways of nuclear factor κB, activator protein 1, sirtuins 1, silent information regulator sirtuin 1 and 3, and microRNAs are among those affected by sulforaphane. These anti-inflammatory actions are sometimes due to direct action via interaction with the sulfhydryl structural moiety of cysteine residues in enzymes/proteins. The following are among the topics discussed in this paper: paradoxical signalling pathways such as the immunosuppressant or immunostimulant mechanisms; crosstalk between the oxidative and inflammatory pathways; and effects dependent on health and disease states.
Collapse
Affiliation(s)
- Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| |
Collapse
|
8
|
Lee J, Park J, Song KM, Lee YG, Choi HK. Actinidia arguta Extract Containing Myo-Inositol Suppresses TNF- α-Induced VCAM-1 Expression and Monocyte Adhesion to Endothelial Cells via Inhibition of the PTEN/Akt/GSK-3 β and NF- κB Signaling Pathways. J Med Food 2024; 27:419-427. [PMID: 38656897 DOI: 10.1089/jmf.2023.k.0326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
The primary inflammatory process in atherosclerosis, a major contributor to cardiovascular disease, begins with monocyte adhering to vascular endothelial cells. Actinidia arguta (kiwiberry) is an edible fruit that contains various bioactive components. While A. arguta extract (AAE) has been recognized for its anti-inflammatory characteristics, its specific inhibitory effect on early atherogenic events has not been clarified. We used tumor necrosis factor-α (TNF-α)-stimulated human umbilical vein endothelial cells (HUVECs) for an in vitro model. AAE effectively hindered the attachment of THP-1 monocytes and reduced the expression of vascular cell adhesion molecule-1 (VCAM-1) in HUVECs. Transcriptome analysis revealed that AAE treatment upregulated phosphatase and tensin homolog (PTEN), subsequently inhibiting phosphorylation of AKT and glycogen synthase kinase 3β (GSK3β) in HUVECs. AAE further hindered phosphorylation of AKT downstream of the nuclear factor kappa B (NF-κB) signaling pathway, leading to suppression of target gene expression. Oral administration of AAE suppressed TNF-α-stimulated VCAM-1 expression, monocyte-derived macrophage infiltration, and proinflammatory cytokine expression in C57BL/6 mouse aortas. Myo-inositol, identified as the major compound in AAE, played a key role in suppressing THP-1 monocyte adhesion in HUVECs. These findings suggest that AAE could serve as a nutraceutical for preventing atherosclerosis by inhibiting its initial pathogenesis.
Collapse
Affiliation(s)
- Jangho Lee
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Joon Park
- Divisions of Strategic Food Technology Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Kyung-Mo Song
- Divisions of Strategic Food Technology Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Yu Geon Lee
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Hyo-Kyoung Choi
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| |
Collapse
|
9
|
Luo J, Alkhalidy H, Jia Z, Liu D. Sulforaphane Ameliorates High-Fat-Diet-Induced Metabolic Abnormalities in Young and Middle-Aged Obese Male Mice. Foods 2024; 13:1055. [PMID: 38611359 PMCID: PMC11012181 DOI: 10.3390/foods13071055] [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: 03/05/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Type 2 diabetes (T2D) is still a fast-growing health problem globally. It is evident that chronic insulin resistance (IR) and progressive loss of β-cell mass and function are key features of T2D etiology. Obesity is a leading pathogenic factor for developing IR. The aim of the present study was to determine whether sulforaphane (SFN), a natural compound derived from cruciferous vegetables, can prevent (prevention approach) or treat (treatment approach) obesity and IR in mouse models. We show that dietary intake of SFN (0.5 g/kg of HFD) for 20 weeks suppressed high-fat diet (HFD)-induced fat accumulation by 6.04% and improved insulin sensitivity by 23.66% in young male mice. Similarly, dietary provision of SFN (0.25 g/kg) significantly improved blood lipid profile, glucose tolerance, and insulin sensitivity of the middle-aged male mice while it had little effects on body composition as compared with the HFD group. In the treatment study, oral administration of SFN (40 mg/kg) induced weight loss and improved insulin sensitivity and plasma lipid profile in the diet-induced-obesity (DIO) male mice. In all three studies, the metabolic effects of SFN administration were not associated with changes in food intake. In vitro, SFN increased glucose uptake in C2C12 myotubes and increased fatty acid and pyruvate oxidation in primary human skeletal muscle cells. Our results suggest that SFN may be a naturally occurring insulin-sensitizing agent that is capable of improving the metabolic processes in HFD-induced obesity and IR and thereby may be a promising compound for T2D prevention.
Collapse
Affiliation(s)
- Jing Luo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
- Department of Human Nutrition, Foods, and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA 24060, USA;
| | - Hana Alkhalidy
- Department of Human Nutrition, Foods, and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA 24060, USA;
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA;
| | - Dongmin Liu
- Department of Human Nutrition, Foods, and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA 24060, USA;
| |
Collapse
|
10
|
Ruhee RT, Suzuki K. The Immunomodulatory Effects of Sulforaphane in Exercise-Induced Inflammation and Oxidative Stress: A Prospective Nutraceutical. Int J Mol Sci 2024; 25:1790. [PMID: 38339067 PMCID: PMC10855658 DOI: 10.3390/ijms25031790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Sulforaphane (SFN) is a promising molecule for developing phytopharmaceuticals due to its potential antioxidative and anti-inflammatory effects. A plethora of research conducted in vivo and in vitro reported the beneficial effects of SFN intervention and the underlying cellular mechanisms. Since SFN is a newly identified nutraceutical in sports nutrition, only some human studies have been conducted to reflect the effects of SFN intervention in exercise-induced inflammation and oxidative stress. In this review, we briefly discussed the effects of SFN on exercise-induced inflammation and oxidative stress. We discussed human and animal studies that are related to exercise intervention and mentioned the underlying cellular signaling mechanisms. Since SFN could be used as a potential therapeutic agent, we mentioned briefly its synergistic attributes with other potential nutraceuticals that are associated with acute and chronic inflammatory conditions. Given its health-promoting effects, SFN could be a prospective nutraceutical at the forefront of sports nutrition.
Collapse
Affiliation(s)
- Ruheea Taskin Ruhee
- Research Fellow of Japan Society for the Promotion of Sciences, Tokyo 102-0083, Japan
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| |
Collapse
|
11
|
Mthembu SXH, Mazibuko-Mbeje SE, Moetlediwa MT, Muvhulawa N, Silvestri S, Orlando P, Nkambule BB, Muller CJF, Ndwandwe D, Basson AK, Tiano L, Dludla PV. Sulforaphane: A nutraceutical against diabetes-related complications. Pharmacol Res 2023; 196:106918. [PMID: 37703962 DOI: 10.1016/j.phrs.2023.106918] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
There is an increasing interest in the use of nutraceuticals and plant-derived bioactive compounds from foods for their potential health benefits. For example, as a major active ingredient found from cruciferous vegetables like broccoli, there has been growing interest in understanding the therapeutic effects of sulforaphane against diverse metabolic complications. The past decade has seen an extensive growth in literature reporting on the potential health benefits of sulforaphane to neutralize pathological consequences of oxidative stress and inflammation, which may be essential in protecting against diabetes-related complications. In fact, preclinical evidence summarized within this review supports an active role of sulforaphane in activating nuclear factor erythroid 2-related factor 2 or effectively modulating AMP-activated protein kinase to protect against diabetic complications, including diabetic cardiomyopathy, diabetic neuropathy, diabetic nephropathy, as well as other metabolic complications involving non-alcoholic fatty liver disease and skeletal muscle insulin resistance. With clinical evidence suggesting that foods rich in sulforaphane like broccoli can improve the metabolic status and lower cardiovascular disease risk by reducing biomarkers of oxidative stress and inflammation in patients with type 2 diabetes. This information remains essential in determining the therapeutic value of sulforaphane or its potential use as a nutraceutical to manage diabetes and its related complications. Finally, this review discusses essential information on the bioavailability profile of sulforaphane, while also covering information on the pathological consequences of oxidative stress and inflammation that drive the development and progression of diabetes.
Collapse
Affiliation(s)
- Sinenhlanhla X H Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | | | - Marakiya T Moetlediwa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | - Ndivhuwo Muvhulawa
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa; Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Duduzile Ndwandwe
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Albertus K Basson
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa.
| |
Collapse
|
12
|
Svolacchia F, Brongo S, Catalano A, Ceccarini A, Svolacchia L, Santarsiere A, Scieuzo C, Salvia R, Finelli F, Milella L, Saturnino C, Sinicropi MS, Fabrizio T, Giuzio F. Natural Products for the Prevention, Treatment and Progression of Breast Cancer. Cancers (Basel) 2023; 15:cancers15112981. [PMID: 37296944 DOI: 10.3390/cancers15112981] [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: 05/01/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
In this review, we summarize the most used natural products as useful adjuvants in BC by clarifying how these products may play a critical role in the prevention, treatment and progression of this disease. BC is the leading cancer, in terms of incidence, that affects women. The epidemiology and pathophysiology of BC were widely reported. Inflammation and cancer are known to influence each other in several tumors. In the case of BC, the inflammatory component precedes the development of the neoplasm through a slowly increasing and prolonged inflammation that also favors its growth. BC therapy involves a multidisciplinary approach comprising surgery, radiotherapy and chemotherapy. There are numerous observations that showed that the effects of some natural substances, which, in integration with the classic protocols, can be used not only for prevention or integration in order to prevent recurrences and induce a state of chemoquiescence but also as chemo- and radiosensitizers during classic therapy.
Collapse
Affiliation(s)
- Fabiano Svolacchia
- Department of Medical-Surgical Sciences and Biotechnologies, La Sapienza University, 00118 Rome, Italy
- Department of Medical Sciences, Policlinic Foundation Tor Vergata University, 00133 Rome, Italy
| | - Sergio Brongo
- Department of Plastic Surgery, University of Salerno, 84131 Campania, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126 Bari, Italy
| | - Agostino Ceccarini
- U.O.C. Primary Care and Territorial Health, Social and Health Department, State Hospital, 47893 San Marino, San Marino
| | - Lorenzo Svolacchia
- Department of Medical-Surgical Sciences and Biotechnologies, La Sapienza University, 00118 Rome, Italy
| | - Alessandro Santarsiere
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- CNRS, UMR 7042-LIMA, ECPM, Université de Strasbourg, Université de Haute-Alsace, 67000 Strasbourg, France
| | - Carmen Scieuzo
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- Spinoff XFlies s.r.l., University of Basilicata, 85100 Potenza, Italy
| | - Rosanna Salvia
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- Spinoff XFlies s.r.l., University of Basilicata, 85100 Potenza, Italy
| | | | - Luigi Milella
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Tommaso Fabrizio
- Department of Plastic Surgery, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Federica Giuzio
- U.O.C. Primary Care and Territorial Health, Social and Health Department, State Hospital, 47893 San Marino, San Marino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- Spinoff TNcKILLERS s.r.l., University of Basilicata, 85100 Potenza, Italy
| |
Collapse
|
13
|
Liu S, Zhang Y, Zheng X, Wang Z, Wang P, Zhang M, Shen M, Bao Y, Li D. Sulforaphane Inhibits Foam Cell Formation and Atherosclerosis via Mechanisms Involving the Modulation of Macrophage Cholesterol Transport and the Related Phenotype. Nutrients 2023; 15:2117. [PMID: 37432260 DOI: 10.3390/nu15092117] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 07/12/2023] Open
Abstract
Sulforaphane (SFN), an isothiocyanate, is one of the major dietary phytochemicals found in cruciferous vegetables. Many studies suggest that SFN can protect against cancer and cardiometabolic diseases. Despite the proposed systemic and local vascular protective mechanisms, SFN's potential to inhibit atherogenesis by targeting macrophages remains unknown. In this study, in high fat diet fed ApoE-deficient (ApoE-/-) mice, oral SFN treatment improved dyslipidemia and inhibited atherosclerotic plaque formation and the unstable phenotype, as demonstrated by reductions in the lesion areas in both the aortic sinus and whole aorta, percentages of necrotic cores, vascular macrophage infiltration and reactive oxygen species (ROS) generation. In THP-1-derived macrophages, preadministration SFN alleviated oxidized low-density lipoprotein (ox-LDL)-induced lipid accumulation, oxidative stress and mitochondrial injury. Moreover, a functional study revealed that peritoneal macrophages isolated from SFN-treated mice exhibited attenuated cholesterol influx and enhanced apolipoprotein A-I (apoA-I)- and high-density lipoprotein (HDL)-mediated cholesterol efflux. Mechanistic analysis revealed that SFN supplementation induced both intralesional and intraperitoneal macrophage phenotypic switching toward high expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) and ATP-binding cassette subfamily A/G member 1 (ABCA1/G1) and low expression of peroxisome proliferator-activated receptor γ (PPARγ) and cluster of differentiation 36 (CD36), which was further validated by the aortic protein expression. These results suggest that the regulation of macrophages' cholesterol transport and accumulation may be mainly responsible for SFN's potential atheroprotective properties, and the regulatory mechanisms might involve upregulating ABCA1/G1 and downregulating CD36 via the modulation of PPARγ and Nrf2.
Collapse
Affiliation(s)
- Shiyan Liu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Yuan Zhang
- Department of Geriatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xiangyu Zheng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Ziling Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Pan Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Mengdi Zhang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Mengfan Shen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, Norfolk, UK
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| |
Collapse
|
14
|
Therapeutic effect of the sulforaphane derivative JY4 on ulcerative colitis through the NF-κB-p65 pathway. Inflammopharmacology 2022; 30:1717-1728. [PMID: 35943671 DOI: 10.1007/s10787-022-01044-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/22/2022] [Indexed: 11/05/2022]
Abstract
The efficacy of the sulforaphane derivative JY4 was evaluated in acute and chronic mouse models of ulcerative colitis induced by dextran sodium sulfate. Oral administration of JY4 led to significant improvements in symptoms, with recovery of body weight and colorectal length, together with reduced diarrhoea, bloody stools, ulceration of colonic tissue and infiltration of inflammatory cells. The oral bioavailability of JY4, determined by comparing oral dosing with injection into the tail vein, was 5.67%, which was comply with the idea in the intestinal function. Using a dual-luciferase reporter assay, immunofluorescence studies, western blot analysis and immunohistochemical staining, JY4 was shown to significant interfere with the NF-κB-p65 signaling pathway. By preventing the activation of NF-κB-p65, JY4 inhibited the overexpression of downstream inflammatory factors, thereby exerting an anti-inflammatory effect on the intestinal tract. This study thus provides a promising candidate drug, and a new concept for the treatment of ulcerative colitis.
Collapse
|
15
|
Luo X, Zhao C, Wang S, Jia H, Yu B. TNF-α is a Novel Biomarker for Predicting Plaque Rupture in Patients with ST-Segment Elevation Myocardial Infarction. J Inflamm Res 2022; 15:1889-1898. [PMID: 35313673 PMCID: PMC8933622 DOI: 10.2147/jir.s352509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/14/2022] [Indexed: 01/09/2023] Open
Abstract
Background and Aims Patients with plaque rupture (PR) present with different cardiovascular risks, clinical strategies, and outcomes from those with plaque erosion (PE). However, there are lack of noninvasive biomarkers to distinguish PE from PR. Methods A prospective analysis of 382 patients with ST-segment elevation myocardial infarction (STEMI) was conducted. Of these patients, 262 and 120 presented with PR and PE, respectively. An additional 83 patients diagnosed with stable angina pectoris were enrolled as control group. Peripheral blood monocytes were collected pre-percutaneous coronary intervention and used to evaluate the mRNA expression of IL-4, IL-10, IL-1β, and TNF-α in all patients. Results STEMI patients had higher IL-4, IL-10, IL-1β, and TNF-α expression than the control patients. The mRNA levels of IL-4, IL-1β, and TNF-α were significantly higher in PR patients than PE; however, no significant difference was observed in IL-10 between PE and PR. The areas under the receiver-operating characteristic curves for IL-4, IL-1β, and TNF-α for PR versus PE were 0.685, 0.747, and 0.895, respectively. At the cut-off value of 2.52, TNF-α demonstrated a sensitivity of 70.61% and specificity of 93.33% for discriminating PR from PE patients. When added to the model of established clinical risk factors, TNF-α significantly improved the predictive accuracy of PR. Multivariable logistic regression analysis indicated that TNF-α mRNA level was independently associated with PR (odds ratio, 3.09; 95% confidence interval, 2.29–4.16; p < 0.001). Conclusion The inflammatory response of peripheral blood mononuclear cells in patients with PR was higher than that in patients with PE. TNF-α may be a potential biomarker for predicting PR that could facilitate risk stratification and management in STEMI patients.
Collapse
Affiliation(s)
- Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People’s Republic of China
| | - Chen Zhao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People’s Republic of China
| | - Shengfang Wang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People’s Republic of China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People’s Republic of China
- Correspondence: Haibo Jia; Bo Yu, Tel/Fax +86 0451 86297221; +86 0451 86297220, Email ;
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People’s Republic of China
| |
Collapse
|
16
|
Liang W, Greven J, Fragoulis A, Horst K, Bläsius F, Wruck C, Pufe T, Kobbe P, Hildebrand F, Lichte P. Sulforaphane-Dependent Up-Regulation of NRF2 Activity Alleviates Both Systemic Inflammatory Response and Lung Injury After Hemorrhagic Shock/Resuscitation in Mice. Shock 2022; 57:221-229. [PMID: 34559743 DOI: 10.1097/shk.0000000000001859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Hemorrhagic shock/resuscitation (HS/R) is closely associated with overwhelming oxidative stress and systemic inflammation. As an effective activator of the nuclear factor-erythroid factor 2 related factor 2 (Nrf2) pathway, sulforaphane (SFN) exerts antioxidant and anti-inflammatory effects. We explored SFN's effects on alveolar macrophages (AMs), systemic inflammation, and pulmonary damage in an isolated murine HS/R model. Male C57/BL6 wild type and transgenic antioxidant response element (ARE)-luciferase (luc) mice (both n = 6 per group) were exposed to either pressure-controlled HS/R (mean arterial pressure 35-45 mm Hg for 90 min) or sham procedure (surgery without HS/R) or were sacrificed without intervention (control group). Fluid resuscitation was performed via the reinfusion of withdrawn blood and 0.9% saline. Sulforaphane or 0.9% saline (vehicle) was administrated intraperitoneally. Mice were sacrificed 6, 24, or 72 h after resuscitation. Bioluminescence imaging of ARE-luc mice was conducted to measure pulmonary Nrf2 activity. Plasma was collected to determine systemic cytokine levels. Alveolar macrophages were isolated before measuring cytokines in the supernatant and performing immunofluorescence staining, as well as Western blot for intracellular Nrf2. Histological damage was assessed via the acute lung injury score and wet/dry ratio.Hemorrhagic shock/resuscitation was associated with pulmonary Nrf2 activation. Sulforaphane enhanced pulmonary Nrf2 activity and the Nrf2 activation of AM, while it decreased lung damage. Sulforaphane exerted down-regulatory effects on AM-generated and systemic pro-inflammatory mediators, while it did not have such effects on IL-10.In conclusion, SFN beneficially enhances pulmonary Nrf2 activity and promotes Nrf2 accumulation in AMs' nuclei. This may exert not only local protective effects but also systemic effects via the down-regulation of pro-inflammatory cytokines. The administration of Nrf2 activator post-HS/R may represent an innovative treatment strategy.
Collapse
Affiliation(s)
- Weiqiang Liang
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan City, Shandong Province, PR China
| | - Johannes Greven
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, Aachen, Germany
| | - Klemens Horst
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Felix Bläsius
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Wruck
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, Aachen, Germany
| | - Philipp Kobbe
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lichte
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
17
|
Kamal RM, Abdull Razis AF, Mohd Sukri NS, Perimal EK, Ahmad H, Patrick R, Djedaini-Pilard F, Mazzon E, Rigaud S. Beneficial Health Effects of Glucosinolates-Derived Isothiocyanates on Cardiovascular and Neurodegenerative Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030624. [PMID: 35163897 PMCID: PMC8838317 DOI: 10.3390/molecules27030624] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Neurodegenerative diseases (NDDs) and cardiovascular diseases (CVDs) are illnesses that affect the nervous system and heart, all of which are vital to the human body. To maintain health of the human body, vegetable diets serve as a preventive approach and particularly Brassica vegetables have been associated with lower risks of chronic diseases, especially NDDs and CVDs. Interestingly, glucosinolates (GLs) and isothiocyanates (ITCs) are phytochemicals that are mostly found in the Cruciferae family and they have been largely documented as antioxidants contributing to both cardio- and neuroprotective effects. The hydrolytic breakdown of GLs into ITCs such as sulforaphane (SFN), phenylethyl ITC (PEITC), moringin (MG), erucin (ER), and allyl ITC (AITC) has been recognized to exert significant effects with regards to cardio- and neuroprotection. From past in vivo and/or in vitro studies, those phytochemicals have displayed the ability to mitigate the adverse effects of reactive oxidation species (ROS), inflammation, and apoptosis, which are the primary causes of CVDs and NDDs. This review focuses on the protective effects of those GL-derived ITCs, featuring their beneficial effects and the mechanisms behind those effects in CVDs and NDDs.
Collapse
Affiliation(s)
- Ramla Muhammad Kamal
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Department of Pharmacology, Federal University Dutse, Dutse 720101, Jigawa State, Nigeria
| | - Ahmad Faizal Abdull Razis
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Correspondence:
| | - Nurul Syafuhah Mohd Sukri
- Faculty of Applied Science and Technology, Universiti Tun Hussein Onn Malaysia, Batu Pahat 86400, Johor, Malaysia;
| | - Enoch Kumar Perimal
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Hafandi Ahmad
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Rollin Patrick
- Université d’Orléans et CNRS, ICOA, UMR 7311, BP 6759, CEDEX 02, F-45067 Orléans, France;
| | - Florence Djedaini-Pilard
- LG2A UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu—UFR des Sciences, F-80000 Amiens, France; (F.D.-P.); (S.R.)
| | - Emanuela Mazzon
- Laboratorio di Neurologia Sperimentale, IRCCS Centro Neurolesi "Bonino Pulejo", 98124 Messina, Italy;
| | - Sébastien Rigaud
- LG2A UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu—UFR des Sciences, F-80000 Amiens, France; (F.D.-P.); (S.R.)
| |
Collapse
|
18
|
Natural Compound Resveratrol Attenuates TNF-Alpha-Induced Vascular Dysfunction in Mice and Human Endothelial Cells: The Involvement of the NF-κB Signaling Pathway. Int J Mol Sci 2021; 22:ijms222212486. [PMID: 34830366 PMCID: PMC8620472 DOI: 10.3390/ijms222212486] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/07/2021] [Indexed: 01/04/2023] Open
Abstract
Resveratrol, a natural compound in grapes and red wine, has drawn attention due to potential cardiovascular-related health benefits. However, its effect on vascular inflammation at physiologically achievable concentrations is largely unknown. In this study, resveratrol in concentrations as low as 1 μm suppressed TNF-α-induced monocyte adhesion to human EA.hy926 endothelial cells (ECs), a key event in the initiation and development of atherosclerosis. Low concentrations of resveratrol (0.25–2 μm) also significantly attenuated TNF-α-stimulated mRNA expressions of MCP-1/CCL2 and ICAM-1, which are vital mediators of EC-monocyte adhesion molecules and cytokines for cardiovascular plaque formation. Additionally, resveratrol diminished TNF-α-induced IκB-α degradation and subsequent nuclear translocation of NF-κB p65 in ECs. In the animal study, resveratrol supplementation in diet significantly diminished TNF-α-induced increases in circulating levels of adhesion molecules and cytokines, monocyte adhesion to mouse aortic ECs, F4/80-positive macrophages and VCAM-1 expression in mice aortas and restored the disruption in aortic elastin fiber caused by TNF-α treatment. The animal study also confirmed that resveratrol blocks the activation of NF-κB In Vivo. In conclusion, resveratrol at physiologically achievable concentrations displayed protective effects against TNF-α-induced vascular endothelial inflammation in vitro and In Vivo. The ability of resveratrol in reducing inflammation may be associated with its role as a down-regulator of the NF-κB pathway.
Collapse
|
19
|
Fowler J, Tsui MTK, Chavez J, Khan S, Ahmed H, Smith L, Jia Z. Methyl mercury triggers endothelial leukocyte adhesion and increases expression of cell adhesion molecules and chemokines. Exp Biol Med (Maywood) 2021; 246:2522-2532. [PMID: 34308659 DOI: 10.1177/15353702211033812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of morbidity, mortality, and health care costs in the USA, and around the world. Among the various risk factors of cardiovascular disease, environmental and dietary exposures to methyl mercury, a highly toxic metal traditionally labeled as a neurotoxin, have been epidemiologically linked to human cardiovascular disease development. However, its role in development and promotion of atherosclerosis, an initial step in more immediately life-threatening cardiovascular diseases, remains unclear. This study was conducted to examine the role that methyl mercury plays in the adhesion of monocytes to human microvascular endothelial cells (HMEC-1), and the underlying mechanisms. Methyl mercury treatment significantly induced the adhesion of monocyte to HMEC-1 endothelial cells, a critical step in atherosclerosis, while also upregulating the expression of proinflammatory cytokines interleukin-6, interleukin-8. Further, methyl mercury treatment also upregulated the chemotactic cytokine monocyte chemoattractant protein-1 and intercellular adhesion molecule-1. These molecules are imperative for the firm adhesion of leukocytes to endothelial cells. Additionally, our results further demonstrated that methyl mercury stimulated a significant increase in NF-κB activation. These findings suggest that NF-κB signaling pathway activation by methyl mercury is an important factor in the binding of monocytes to endothelial cells. Finally, by using flow cytometric analysis, methyl mercury treatment caused a significant increase in necrotic cell death only at higher concentrations without initiating apoptosis. This study provides new insights into the molecular actions of methyl mercury that can lead to endothelial dysfunction, inflammation, and subsequent atherosclerotic development.
Collapse
Affiliation(s)
- Joshua Fowler
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Martin Tsz-Ki Tsui
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA.,School of Life Sciences, Chinese University of Hong Kong, Hong Kong SAR 00000, China
| | - Jessica Chavez
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Safeera Khan
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Hassan Ahmed
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Lena Smith
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| |
Collapse
|
20
|
Belperain S, Kang ZY, Dunphy A, Priebe B, Chiu NHL, Jia Z. Anti-Inflammatory Effect and Cellular Uptake Mechanism of Carbon Nanodots in in Human Microvascular Endothelial Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1247. [PMID: 34068511 PMCID: PMC8151002 DOI: 10.3390/nano11051247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022]
Abstract
Cardiovascular disease (CVD) has become an increasingly important topic in the field of medical research due to the steadily increasing rates of mortality caused by this disease. With recent advancements in nanotechnology, a push for new, novel treatments for CVD utilizing these new materials has begun. Carbon Nanodots (CNDs), are a new form of nanoparticles that have been coveted due to the green synthesis method, biocompatibility, fluorescent capabilities and potential anti-antioxidant properties. With much research pouring into CNDs being used as bioimaging and drug delivery tools, few studies have been completed on their anti-inflammatory potential, especially in the cardiovascular system. CVD begins initially by endothelial cell inflammation. The cause of this inflammation can come from many sources; one being tumor necrosis factor (TNF-α), which can not only trigger inflammation but prolong its existence by causing a storm of pro-inflammatory cytokines. This study investigated the ability of CNDs to attenuate TNF-α induced inflammation in human microvascular endothelial cells (HMEC-1). Results show that CNDs at non-cytotoxic concentrations reduce the expression of pro-inflammatory genes, mainly Interleukin-8 (IL-8), and interleukin 1 beta (IL-1β). The uptake of CNDs by HMEC-1s was examined. Results from the studies involving channel blockers and endocytosis disruptors suggest that uptake takes place by endocytosis. These findings provide insights on the interaction CNDs and endothelial cells undergoing TNF-α induced cellular inflammation.
Collapse
Affiliation(s)
- Sarah Belperain
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Zi Yae Kang
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Andrew Dunphy
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Brandon Priebe
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Norman H. L. Chiu
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27412, USA;
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC 27401, USA
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| |
Collapse
|
21
|
Almushayti AY, Brandt K, Carroll MA, Scotter MJ. Current analytical methods for determination of glucosinolates in vegetables and human tissues. J Chromatogr A 2021; 1643:462060. [PMID: 33770631 DOI: 10.1016/j.chroma.2021.462060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/18/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
Numerous epidemiological studies have indicated the potential effects of glucosinolates and their metabolites against cancer as well as other non-communicable diseases, such as cardiovascular disease and neurodegenerative disorders. However, information on the presence and quantity of glucosinolates in commonly consumed vegetables and in human fluids is sparse, largely because well-standardised methods for glucosinolate determination are not available, resulting in published data being inconsistent and conflicting. Thus, studies published since 2002 on the most recent developments of glucosinolate extraction and identification have been collected and reviewed with emphasis on determination of the intact glucosinolates by LC-MS and LC-MS/MS. This overview highlights the glucosinolate extraction methods used, the stability of glucosinolates during extraction, the availability of stable isotope labelled internal standards and the use of NMR for purity analysis, as well as the current analytical techniques that have been applied for glucosinolate analysis, e.g. liquid chromatography with mass spectrometric detection (LC-MS). It aims to interpret the findings with a focus on the development of a validated method, which will help to determine the glucosinolate content of vegetative plants and human tissues, and the identification and determination of selected glucosinolate metabolites.
Collapse
Affiliation(s)
- Albatul Y Almushayti
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; College of Agriculture and Veterinary Medicine, Department of Food Science and Human Nutrition, Qassim University, Qassim, KSA.
| | - Kirsten Brandt
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Michael A Carroll
- School of Natural & Environmental Sciences-Chemistry, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| | | |
Collapse
|
22
|
Park CR, Lee JS, Son CG, Lee NH. A survey of herbal medicines as tumor microenvironment-modulating agents. Phytother Res 2021; 35:78-94. [PMID: 32658314 DOI: 10.1002/ptr.6784] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/24/2020] [Accepted: 06/06/2020] [Indexed: 12/18/2022]
Abstract
The tumor microenvironment (TME) is extremely complex, involving extensive interactions among stromal cells, immune cells, and signaling molecules. Therefore, an approach targeting the TME has emerged as a promising therapeutic strategy. Herbal medicines consist of multiple active compounds, which have multi-target effects. Therefore, they have been regarded as potential anticancer agents; multiple studies have explored their effects on the TME. In this review, we report the effects of 29 single herb medicines or herbal formulas on the TME, based on the findings of 64 published studies. Specifically, we describe the effects of these herbal medicines on cancer-associated fibroblasts/tumor-associated fibroblasts, tumor-associated endothelial cells, myeloid-derived suppressor cells, and tumor-associated macrophages. Among the reviewed herbal medicines, the most promising TME-modulating effects were exhibited by curcumin, DHA, EGCG, resveratrol, and silibinin; these medicines showed the ability to regulate two or more components of the TME. The findings of this review support the notion that the combination of herbal medicines with conventional anticancer therapies are likely to exhibit a clinical benefit, which should be further explored in clinical trials.
Collapse
Affiliation(s)
- Chan-Ran Park
- Dept. of Clinical Oncology, Cheonan Korean Medicine Hospital of Daejeon University, Cheonan-si, Republic of Korea
- Liver and Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon-si, Republic of Korea
- Dept. of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon-si, Republic of Korea
| | - Jin-Seok Lee
- Liver and Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon-si, Republic of Korea
- Dept. of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon-si, Republic of Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon-si, Republic of Korea
- Dept. of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon-si, Republic of Korea
| | - Nam-Hun Lee
- Dept. of Clinical Oncology, Cheonan Korean Medicine Hospital of Daejeon University, Cheonan-si, Republic of Korea
- Liver and Immunology Research Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon-si, Republic of Korea
- Dept. of Internal Medicine, Graduated School of Korean Medicine, University of Daejeon, Daejeon-si, Republic of Korea
| |
Collapse
|
23
|
Martelli A, Citi V, Calderone V. Recent efforts in drug discovery on vascular inflammation and consequent atherosclerosis. Expert Opin Drug Discov 2020; 16:411-427. [PMID: 33256484 DOI: 10.1080/17460441.2021.1850688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Preservation of vascular endothelium integrity and maintenance of its full functionality are fundamental aspects in order to avoid both cardiovascular and non-cardiovascular diseases.Areas covered: Although a massive endothelial disruption is a rare condition, caused by acute and uncontrolled inflammatory responses (e.g. the cytokine storm induced by SARS-CoV-2 infection), more frequently the vascular tree is the first target of slowly progressive inflammatory processes which affect the integrity of endothelium and its 'barrier' function, supporting the onset of atherosclerotic plaque and spreading inflammation. This endothelial dysfunction leads to decrease NO biosynthesis, impaired regulation of vascular tone, and increased platelet aggregation. Such chronic subclinic inflammation leads to macrophage infiltration in atherosclerotic lesions. Therefore, many efforts should be addressed to find useful approaches to preserve vascular endothelium from inflammation. In this review, the authors have evaluated the most recent strategies to counteract this pathological condition.Expert opinion: The therapeutic and nutraceutical approaches represent useful tools to treat or prevent different phases of vascular inflammation. In particular, the pharmacological approach should be used in advanced phases characterized by clinical signs of vascular disease, whilst the nutraceutical approach may represent a promising preventive strategy to preserve the integrity of the endothelial tissue.
Collapse
Affiliation(s)
- Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)", University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of Ageing, Biology and Pathology, University of Pisa, Pisa, Italy
| | | | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)", University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of Ageing, Biology and Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
24
|
Wang ZC, Chen Q, Wang J, Yu LS, Chen LW. Sulforaphane mitigates LPS-induced neuroinflammation through modulation of Cezanne/NF-κB signalling. Life Sci 2020; 262:118519. [PMID: 33010279 DOI: 10.1016/j.lfs.2020.118519] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 10/23/2022]
Abstract
AIM Neuroinflammation is a potent pathological process of various neurodegenerative diseases. Sulforaphane (SFN) is a natural product and acts as a neuroprotective agent to suppress inflammatory response in brain. The present study investigated the protective effect of Sulforaphane (SFN) on lipopolysaccharide (LPS)-induced neuroinflammation. MATERIALS AND METHODS Rats were divided into three groups: control group, LPS group and LPS + SFN group. Morris water maze test was carried out to evaluate the spatial memory and learning function of rats. The inflammatory cytokines levels in hippocampal tissues, plasma were measured by ELISA. The western blot was used to detect Cezanne/NF-κB signalling. For in vitro study, the Cezanne siRNA and scrambled control were transfected into BV2 cells, and then treated with or without 20 μM SFN before exposed to LPS. The inflammatory cytokines levels and Cezanne/NF-κB signalling were detected by ELISA and western blot, respectively. Co-IP assay were applied to investigate the regulation of Cezanne on ubiquitination of TRAF6 and RIP1. KEY FINDINGS SFN improved LPS-induced neurocognitive dysfunction in rats. It inhibited the neuroinflammation and activation of NF-κB pathway induced by LPS. The modulation of TRAF6 and RIP1 ubiquitination by Cezanne was playing a pivotal role in relation to the mechanism of SFN inhibiting NF-κB pathway. SIGNIFICANCE The results of our study demonstrated that SFN could attenuate LPS-induced neuroinflammation through the modulation of Cezanne/NF-κB signalling.
Collapse
Affiliation(s)
- Zeng-Chun Wang
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, China.
| | - Qiang Chen
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, China
| | - Jing Wang
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Ling-Shan Yu
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Liang-Wan Chen
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
25
|
Zhong L, Yang M, Zou X, Du T, Xu H, Sun J. Human umbilical cord multipotent mesenchymal stromal cells alleviate acute ischemia-reperfusion injury of spermatogenic cells via reducing inflammatory response and oxidative stress. Stem Cell Res Ther 2020; 11:294. [PMID: 32680554 PMCID: PMC7366899 DOI: 10.1186/s13287-020-01813-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/17/2020] [Accepted: 07/06/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND This study was designed to determine the effect of human umbilical cord multipotent mesenchymal stromal cells (hUC-MSC) on acute ischemia/reperfusion (I/R) injury of spermatogenic cells. METHOD The testicular I/R rat model was established through 720° torsion for 1 h. hUC-MSC were intravenously injected 10 min before detorsion. Injury severity of spermatogenic cells was estimated by Johnsen's score. The proliferating of recipient spermatogonia was measured by the immunostaining of antibodies against Ki67, and all germ cells were detected with DDX4 antibody. And recipient spermatogenesis was assessed by staining spermatozoa with lectin PNA. The levels of inflammatory factors were measured by real-time PCR. And the Selectin-E expression, neutrophil infiltration in the testes was detected by immunostaining. Germ cells apoptosis was tested by TUNEL assay and western blot. Furthermore, the oxidative stress was tested by reactive oxidative species (ROS) levels. In vitro, the condition medium (CM) of hUC-MSC was used to culture human umbilical vein endothelial cells (HUVECs), so as to assess the paracrine effect of hUC-MSC on HUVECs. The protein chip was used to measure the relative concentration of the secretory proteins in the CM of hUC-MSC. RESULT hUC-MSC greatly alleviated the testicular injury induced by testis I/R. The levels of proinflammatory factors were downregulated by hUC-MSC in vivo and in vitro. Neutrophil infiltration, ROS, and germ cell apoptosis in testicular tissues were greatly reduced in the group of hUC-MSC. Paracrine factors secreted by hUC-MSC including growth factors, cytokines, and anti-inflammatory cytokine were rich. CONCLUSION This study demonstrated that intravenously injected hUC-MSC could protect the spermatogenic cells against I/R injury by reducing the inflammatory response, apoptosis, and acute oxidative injury. Paracrine mechanism of hUC-MSC may contribute to the protection of spermatogenic cells against I/R injury. Therefore, the present study provides a method for clinical treatment of attenuate I/R injury of spermatogenic cells.
Collapse
Affiliation(s)
- Liang Zhong
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Mengbo Yang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiangyu Zou
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Tao Du
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou City, 450003, China
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
26
|
Lee J, Ha SJ, Park J, Kim YH, Lee NH, Kim YE, Hong YS, Song KM. Arctium lappa root extract containing L-arginine prevents TNF-α-induced early atherosclerosis in vitro and in vivo. Nutr Res 2020; 77:85-96. [PMID: 32388084 DOI: 10.1016/j.nutres.2020.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 11/28/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease affecting the aorta and is a major cause of cardiovascular disease. Arctium lappa root is a plant widely used in traditional Chinese medicine (TCM), and Arctium lappa root extract (ALE) has been reported to exhibit anti-inflammatory capacity and to ameliorate endothelial dysfunction. Thus, we hypothesized that ALE would inhibit the early atherosclerotic stage. In this study, we evaluated the inhibitory effect of ALE on early arteriosclerosis and its mechanisms of action. ALE suppressed TNF-α-induced monocyte adhesion to the vascular endothelium by suppressing NF-κB signaling in HUVECs. In an acute mouse model of atherosclerosis, ALE suppressed TNF-α-induced monocyte infiltration of the vascular endothelium and the expression of genes encoding inflammatory cytokines including IL-1β, IL-6, TNF-α, and MCP-1 in the mouse aorta. Moreover, inulin-type fructan and amino acids, especially L-aspartate and L-arginine (60.27 and 42.17 mg/g, respectively) were detected by NMR, MALDI-TOF MS, and HPLC analysis as the main components of ALE. Notably, L-arginine suppressed TNF-α-induced monocyte adhesion to HUVECs. Therefore, these results suggest that ALE may be a functional food for the suppression or prevention of early stages of atherosclerosis.
Collapse
Affiliation(s)
- Jangho Lee
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Su Jeong Ha
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea.
| | - Joon Park
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Young Ho Kim
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Nam Hyouck Lee
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Young Eon Kim
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Young-Shick Hong
- Division of Food and Nutrition, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju 500-757, Republic of Korea.
| | - Kyung-Mo Song
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| |
Collapse
|
27
|
Zhang M, Xu Y, Jiang L. Sulforaphane attenuates angiotensin II-induced human umbilical vein endothelial cell injury by modulating ROS-mediated mitochondrial signaling. Hum Exp Toxicol 2020; 39:734-747. [PMID: 31957488 DOI: 10.1177/0960327119893414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The study aimed to investigate whether sulforaphane (SFN) protects against angiotensin II (Ang II)-mediated human umbilical vein endothelial cell (HUVEC) injury. Ang II treatment decreased HUVEC viability, increased cell apoptosis, decreased mitochondria membrane potential (MMP), impaired cytochrome c release, activated caspase 3/9, and induced reactive oxygen species (ROS) production, and nicotinamide adenine dinucleotide phosphate oxidase activity. Moreover, SFN treatment blunted Ang II-stimulated oxidative stress and mitochondria-related apoptosis in HUVECs. The ROS scavenger N-acetyl-l-cysteine reduced Ang II-induced oxidative stress and apoptosis, indicating that ROS generation is involved in the Ang II-induced mitochondria-mediated apoptotic pathway. SFN induced nuclear factor erythroid 2 (Nrf2) activation and expression in Ang II-stimulated HUVECs. Downregulation of Nrf2 expression by a target-specific siRNA revealed an Nrf2-dependent effect on the SFN-mediated attenuation of Ang II-induced apoptosis in HUVECs. Pretreatment with brusatol, an Nrf2-specific inhibitor, reversed the protective effects of SFN on Ang II-induced HUVEC injury. SFN treatment protected HUVECs from Ang II-induced damage by decreasing oxidative stress and ameliorating mitochondrial injury.
Collapse
Affiliation(s)
- M Zhang
- Division of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Xu
- Division of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - L Jiang
- Division of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Martelli A, Citi V, Testai L, Brogi S, Calderone V. Organic Isothiocyanates as Hydrogen Sulfide Donors. Antioxid Redox Signal 2020; 32:110-144. [PMID: 31588780 DOI: 10.1089/ars.2019.7888] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Hydrogen sulfide (H2S), the "new entry" in the series of endogenous gasotransmitters, plays a fundamental role in regulating the biological functions of various organs and systems. Consequently, the lack of adequate levels of H2S may represent the etiopathogenetic factor of multiple pathological alterations. In these diseases, the use of H2S donors represents a precious and innovative opportunity. Recent Advances: Natural isothiocyanates (ITCs), sulfur compounds typical of some botanical species, have long been investigated because of their intriguing pharmacological profile. Recently, the ITC moiety has been proposed as a new H2S-donor chemotype (with a l-cysteine-mediated reaction). Based on this recent discovery, we can clearly observe that almost all the effects of natural ITCs can be explained by the H2S release. Consistently, the ITC function was also used as an original H2S-releasing moiety for the design of synthetic H2S donors and original "pharmacological hybrids." Very recently, the chemical mechanism of H2S release, resulting from the reaction between l-cysteine and some ITCs, has been elucidated. Critical Issues: Available literature gives convincing demonstration that H2S is the real player in ITC pharmacology. Further, countless studies have been carried out on natural ITCs, but this versatile moiety has been used only rarely for the design of synthetic H2S donors with optimal drug-like properties. Future Directions: The development of more ITC-based synthetic H2S donors with optimal drug-like properties and selectivity toward specific tissues/pathologies seem to represent a stimulating and indispensable prospect of future experimental activities.
Collapse
Affiliation(s)
- Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)," University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of "Ageing Biology and Pathology," University of Pisa, Pisa, Italy
| | | | - Lara Testai
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)," University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of "Ageing Biology and Pathology," University of Pisa, Pisa, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)," University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of "Ageing Biology and Pathology," University of Pisa, Pisa, Italy
| |
Collapse
|
29
|
Deng X, Chu X, Wang P, Ma X, Wei C, Sun C, Yang J, Li Y. MicroRNA-29a-3p Reduces TNFα-Induced Endothelial Dysfunction by Targeting Tumor Necrosis Factor Receptor 1. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:903-915. [PMID: 31760375 PMCID: PMC6883339 DOI: 10.1016/j.omtn.2019.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 09/18/2019] [Accepted: 10/12/2019] [Indexed: 12/25/2022]
Abstract
miR-29a-3p has been shown to be associated with cardiovascular diseases; however, the effect of miR-29a-3p on endothelial dysfunction is unclear. This study aimed to reveal the effects and mechanisms of miR-29a-3p on endothelial dysfunction. The levels of vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and E-selectin were determined by real-time PCR and immunofluorescence staining to reveal the degree of tumor necrosis factor alpha (TNFα)-induced endothelial dysfunction. A luciferase activity assay and cell transfection with a miR-29a-3p mimic or an inhibitor were used to reveal the underlying mechanisms of miR-29a-3p action. Furthermore, the effects of miR-29a-3p on endothelial dysfunction were assessed in C57BL/6 mice injected with TNFα and/or a miR-29a-3p agomir. The results showed that the expression of TNFα-induced adhesion molecules in vascular endothelial cells (EA.hy926 cells, human aortic endothelial cells [HAECs], and primary human umbilical vein endothelial cells [pHUVECs]) and smooth muscle cells (human umbilical vein smooth muscle cells [HUVSMCs]) was significantly decreased following transfection with miR-29a-3p. This effect was reversed by cotransfection with a miR-29a-3p inhibitor. As a key target of miR-29a-3p, tumor necrosis factor receptor 1 mediated the effect of miR-29a-3p. Moreover, miR-29a-3p decreased the plasma levels of TNFα-induced VCAM-1 (32.62%), ICAM-1 (38.22%), and E-selectin (39.32%) in vivo. These data indicate that miR-29a-3p plays a protective role in TNFα-induced endothelial dysfunction, suggesting that miR-29a-3p is a novel target for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xinrui Deng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Peng Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Xiaohui Ma
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Chunbo Wei
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Jianjun Yang
- Department of Nutrition and Food Hygiene, School of Public Health and Management, Ningxia Medical University, No. 1160 Shengli Road, Xingqing District, Yinchuan 750004, China.
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin 150081, China.
| |
Collapse
|
30
|
Tao S, Wang L, Zhu Z, Liu Y, Wu L, Yuan C, Zhang G, Wang Z. Adverse effects of bisphenol A on Sertoli cell blood-testis barrier in rare minnow Gobiocypris rarus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 171:475-483. [PMID: 30639874 DOI: 10.1016/j.ecoenv.2019.01.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 12/28/2018] [Accepted: 01/03/2019] [Indexed: 06/09/2023]
Abstract
Bisphenol A (BPA), an environmental contaminant, has been shown to disturb the dynamics of Sertoli cell blood-testis barrier (BTB) in mammal testis. However, the effects of BPA on Sertoli cell barrier (SC barrier) were little known in fish to date. To evaluate the potential mechanism of reproductive toxicity of BPA, we studied the damage of SC barrier using in vivo models. In this study, male adult rare minnow Gobiocypris rarus were exposed to 15 μg/L BPA for 7-35 days. Gonadal histology and the integrity of SC barrier were analyzed. Meanwhile, the expressions of SC barrier -associated proteins, tumor necrosis factor (TNFα) content, and the mRNA expressions of genes in the mitogen activated protein kinase (MAPK) pathway were detected. Histological analysis demonstrated 15 μg/L BPA promoted the infiltration of inflammatory cells in fish testes after 7-days exposure. The biotin tracer assay showed that 7-days BPA exposure increased permeability for spermatid cysts. In addition, the BPA treatment caused increased TNFα in testis, which was reportedly related to SC barrier impairment. The expressions of Occludin and β-Catenin protein were significantly decreased in the testes after 7- and 21-days exposure. BPA also altered the mRNA expressions of occludin, β-catenin, p38 MAPK and JNK. Therefore, the detrimental effects of BPA on reproduction of male fish may attribute to the disturbed expressions of SC junction proteins.
Collapse
Affiliation(s)
- Shiyu Tao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lihong Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zeliang Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Cong Yuan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Guo Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zaizhao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
31
|
Sulforaphane - role in aging and neurodegeneration. GeroScience 2019; 41:655-670. [PMID: 30941620 DOI: 10.1007/s11357-019-00061-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/14/2019] [Indexed: 12/16/2022] Open
Abstract
In the last several years, numerous molecules derived from plants and vegetables have been tested for their antioxidant, anti-inflammatory, and anti-aging properties. One of them is sulforaphane (SFN), an isothiocyanate present in cruciferous vegetables. SFN activates the antioxidant and anti-inflammatory responses by inducing Nrf2 pathway and inhibiting NF-κB. It also has an epigenetic effect by inhibiting HDAC and DNA methyltransferases and modifies mitochondrial dynamics. Moreover, SFN preserves proteome homeostasis (proteostasis) by activating the proteasome, which has been shown to lead to increased cellular lifespan and prevent neurodegeneration. In this review, we describe some of the molecular and physical characteristics of SFN, its mechanisms of action, and the effects that SFN treatment induces in order to discuss its relevance as a "miraculous" drug to prevent aging and neurodegeneration.
Collapse
|
32
|
Soundararajan P, Kim JS. Anti-Carcinogenic Glucosinolates in Cruciferous Vegetables and Their Antagonistic Effects on Prevention of Cancers. Molecules 2018; 23:E2983. [PMID: 30445746 PMCID: PMC6278308 DOI: 10.3390/molecules23112983] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022] Open
Abstract
Glucosinolates (GSL) are naturally occurring β-d-thioglucosides found across the cruciferous vegetables. Core structure formation and side-chain modifications lead to the synthesis of more than 200 types of GSLs in Brassicaceae. Isothiocyanates (ITCs) are chemoprotectives produced as the hydrolyzed product of GSLs by enzyme myrosinase. Benzyl isothiocyanate (BITC), phenethyl isothiocyanate (PEITC) and sulforaphane ([1-isothioyanato-4-(methyl-sulfinyl) butane], SFN) are potential ITCs with efficient therapeutic properties. Beneficial role of BITC, PEITC and SFN was widely studied against various cancers such as breast, brain, blood, bone, colon, gastric, liver, lung, oral, pancreatic, prostate and so forth. Nuclear factor-erythroid 2-related factor-2 (Nrf2) is a key transcription factor limits the tumor progression. Induction of ARE (antioxidant responsive element) and ROS (reactive oxygen species) mediated pathway by Nrf2 controls the activity of nuclear factor-kappaB (NF-κB). NF-κB has a double edged role in the immune system. NF-κB induced during inflammatory is essential for an acute immune process. Meanwhile, hyper activation of NF-κB transcription factors was witnessed in the tumor cells. Antagonistic activity of BITC, PEITC and SFN against cancer was related with the direct/indirect interaction with Nrf2 and NF-κB protein. All three ITCs able to disrupts Nrf2-Keap1 complex and translocate Nrf2 into the nucleus. BITC have the affinity to inhibit the NF-κB than SFN due to the presence of additional benzyl structure. This review will give the overview on chemo preventive of ITCs against several types of cancer cell lines. We have also discussed the molecular interaction(s) of the antagonistic effect of BITC, PEITC and SFN with Nrf2 and NF-κB to prevent cancer.
Collapse
Affiliation(s)
- Prabhakaran Soundararajan
- Genomics Division, Department of Agricultural Bio-Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wansan-gu, Jeonju 54874, Korea.
| | - Jung Sun Kim
- Genomics Division, Department of Agricultural Bio-Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wansan-gu, Jeonju 54874, Korea.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW We present a current perspective of epigenetic alterations that can lead to cardiovascular disease (CVD) and the potential of dietary factors to counteract their actions. In addition, we discuss the challenges and opportunities of dietary treatments as epigenetic modifiers for disease prevention and therapy. RECENT FINDINGS Recent epigenome-wide association studies along with candidate gene approaches and functional studies in cell culture and animal models have delineated mechanisms through which nutrients, food compounds and dietary patterns may affect the epigenome. Several risk factors for CVD, including adiposity, inflammation and oxidative stress, have been associated with changes in histone acetylation, lower global DNA methylation levels and shorter telomere length. A surplus of macronutrients such as in a high-fat diet or deficiencies of specific nutrients such as folate and other B-vitamins can affect the activity of DNA methyltransferases and histone-modifying enzymes, affecting foetal growth, glucose/lipid metabolism, oxidative stress, inflammation and atherosclerosis. Bioactive compounds such as polyphenols (resveratrol, curcumin) or epigallocatechin may activate deacetylases Sirtuins (SIRTs), histone deacetylases or acetyltransferases and in turn the response of inflammatory mediators. Adherence to cardioprotective dietary patterns, such as the Mediterranean diet (MedDiet), has been associated with altered methylation and expression of genes related to inflammation and immuno-competence. SUMMARY The mechanisms through which nutrients and dietary patterns may alter the cardiovascular epigenome remain elusive. The research challenge is to determine which of these nutriepigenetic effects are reversible, so that novel findings translate into effective dietary interventions to prevent CVD or its progression.
Collapse
Affiliation(s)
- Anastasia Z. Kalea
- Institute of Liver and Digestive Health, Division of Medicine, University College London (UCL), UK
- Institute of Cardiovascular Science, University College London (UCL), UK
- School of Human Sciences, London Metropolitan University, UK
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Temple University School of Medicine, Center for Translational Medicine, Department of Pharmacology, Philadelphia, PA, USA
| | - Jessica L. Buxton
- UCL Great Ormond Street Institute of Child Health, UK
- School of Life Sciences, Pharmacy and Chemistry, Kingston University, UK
| |
Collapse
|
34
|
Patel B, Mann GE, Chapple SJ. Concerted redox modulation by sulforaphane alleviates diabetes and cardiometabolic syndrome. Free Radic Biol Med 2018; 122:150-160. [PMID: 29427794 DOI: 10.1016/j.freeradbiomed.2018.02.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023]
Abstract
Diabetes and cardiometabolic disorders such as hypertension and obesity are major risk factors for the development of cardiovascular disease, with a wealth of evidence suggesting that oxidative stress is linked to the initiation and pathogenesis of these disease processes. With yearly increases in the global incidence of cardiovascular diseases (CVD) and diabetes, numerous studies have focused on characterizing whether upregulating antioxidant defenses through exogenous antioxidants (e.g. vitamin E, vitamin C) or activation of endogenous defenses (e.g. the Nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant defense pathway) may be of benefit. The dietary isothiocyanate sulforaphane (SFN) is currently the subject of several clinical trials for a variety of disease states, including the evaluation of its therapeutic potential to ameliorate diabetic and cardiometabolic complications. SFN is a well characterized and potent Nrf2 inducer, however recent studies suggest its protective actions may be in part mediated by its modulation of various pro-inflammatory (e.g. Nuclear factor-kappa B (NFκB)) and metabolic (e.g. Peroxisome Proliferator-Activator Receptor Gamma (PPARγ)) signaling pathways. The focus of this review is to provide a detailed analysis of the known mechanisms by which SFN modulates Nrf2, NFκB and PPARγ signaling and crosstalk and to provide a critical evaluation of the evidence linking these transcriptional pathways with diabetic and cardiometabolic complications and SFN mediated cytoprotection. To allow comparison between rodent and human studies, we discuss the published bioavailability of SFN metabolites achieved in rodents and man in the context of Nrf2, NFκB and PPARγ signaling. Furthermore, we provide an update on the functional outcomes and implicated signaling pathways reported in recent clinical trials with SFN in Type 2 diabetic patients.
Collapse
Affiliation(s)
- Bijal Patel
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Giovanni E Mann
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Sarah J Chapple
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
35
|
Tian H, Sun W, Zhang Q, Li X, Sang Y, Li J, Niu Y, Ding H. Procyanidin B2 mitigates behavioral impairment and protects myelin integrity in cuprizone-induced schizophrenia in mice. RSC Adv 2018; 8:23835-23846. [PMID: 35540280 PMCID: PMC9081829 DOI: 10.1039/c8ra03854f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 06/11/2018] [Indexed: 01/18/2023] Open
Abstract
Numerous studies have suggested that neuropathological changes in schizophrenia may be related to damage to white matter or demyelination. Procyanidin B2, which is a constituent of many fruits such as grapes and strawberries, has various biological activities such as anti-inflammatory and anti-tumor activity, as has been reported. This study aimed to estimate the effects of procyanidin B2 on behavioral impairment and the protection of myelin integrity in a cuprizone-induced schizophrenia model. Mice were exposed to cuprizone (0.2% w/w in chow) for five weeks to induce schizophrenia-like behavioral changes and demyelination. Procyanidin B2 (20 or 100 mg kg−1 day−1) or vehicle was administered orally to mice after withdrawal from cuprizone. Behavioral impairment was detected with an open-field test, a rotarod test and a Morris water maze. Myelin integrity was assessed using LFB staining and MBP expression, including immunofluorescence and western blotting. In addition, enhancements in the expression of HO-1 and NQO1 suggested that procyanidin B2 may regulate oxidative homeostasis via promoting the translation of Nrf2 to the nucleus. Data indicated that procyanidin B2 could mitigate behavioral impairment and protect myelin integrity in the cuprizone-induced model via regulating oxidative stress by activating Nrf2 signaling. Numerous studies have suggested that neuropathological changes in schizophrenia may be related to damage to white matter or demyelination.![]()
Collapse
Affiliation(s)
- Hui Tian
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Wanchun Sun
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Qianying Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Xiaofei Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Ying Sang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Jian Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Yunhui Niu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| |
Collapse
|
36
|
Circulating metabolites of strawberry mediate reductions in vascular inflammation and endothelial dysfunction in db/db mice. Int J Cardiol 2018; 263:111-117. [PMID: 29681407 DOI: 10.1016/j.ijcard.2018.04.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cardiovascular disease is 2-4-fold more prevalent in patients with diabetes. Human studies support the cardiovascular benefits of strawberry consumption but the effects of strawberry on diabetic vasculature are unknown. We tested the hypothesis that dietary strawberry supplementation attenuates vascular inflammation and dysfunction in diabetic mice. METHODS Seven-week-old diabetic db/db mice that consumed standard diet (db/db) or diet supplemented with 2.35% freeze-dried strawberry (db/db + SB) for ten weeks were compared to non-diabetic control mice (db/+). Indices of vascular inflammation and dysfunction were measured. Endothelial cells (ECs) were isolated from the vasculature to determine the influence of strawberry on them. The effect of metabolites of strawberry on endothelial inflammation was determined by incubating mouse aortic ECs (MAECs) with ±5% serum, obtained from strawberry fed mice (metabolites serum) or standard diet fed mice (control serum) ± 25 mM glucose and 100 μM palmitate. RESULTS db/db mice exhibited an increased monocyte binding to vessel, elevated blood pressure, and reduced endothelial-dependent vasorelaxation compared with db/+ mice but each defect was attenuated in db/db + SB mice. The elevation of inflammatory molecules, NOX2 and inhibitor-κB kinase observed in ECs from db/db vs. db/+ mice was suppressed in db/db + SB mice. Glucose and palmitate increased endothelial inflammation in MAECs but were normalized by co-incubation with metabolites serum. CONCLUSIONS Dietary supplementation of strawberry attenuates indices of vascular inflammation and dysfunction in diabetic db/db mice. The effect of strawberry on vasculature is endothelial-dependent and possibly mediated through their circulating metabolites. Strawberry might complement conventional therapies to improve vascular complications in diabetics.
Collapse
|
37
|
Bessler H, Djaldetti M. Broccoli and human health: immunomodulatory effect of sulforaphane in a model of colon cancer. Int J Food Sci Nutr 2018. [DOI: 10.1080/09637486.2018.1439901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Hanna Bessler
- Laboratory for Immunology and Hematology Research, Rabin Medical Center, Hasharon Hospital, Petah-Tiqva, Israel
- Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| | - Meir Djaldetti
- Laboratory for Immunology and Hematology Research, Rabin Medical Center, Hasharon Hospital, Petah-Tiqva, Israel
- Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| |
Collapse
|
38
|
Xu Y, Xu S, Liu P, Koroleva M, Zhang S, Si S, Jin ZG. Suberanilohydroxamic Acid as a Pharmacological Kruppel-Like Factor 2 Activator That Represses Vascular Inflammation and Atherosclerosis. J Am Heart Assoc 2017; 6:JAHA.117.007134. [PMID: 29191808 PMCID: PMC5779026 DOI: 10.1161/jaha.117.007134] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Kruppel-like factor 2 (KLF2) is an important zinc-finger transcription factor that maintains endothelial homeostasis by its anti-inflammatory, -thrombotic, -oxidative, and -proliferative effects in endothelial cells. In light of the potent vasoprotective effects of KLF2, modulating KLF2 expression or function could give rise to new therapeutic strategies to treat cardiovascular diseases. METHODS AND RESULTS High-throughput drug screening based on KLF2 promoter luciferase reporter assay was performed to screen KLF2 activators. Real-time PCR and western blot were used to detect gene and protein expression. Identified KLF2 activator was orally administered to ApoE-/- mice to evaluate anti-atherosclerotic efficacy. By screening 2400 compounds in the Spectrum library, we identified suberanilohydroxamic (SAHA) acid, also known as vorinostat as a pharmacological KLF2 activator through myocyte enhancer factor 2. We found that SAHA exhibited anti-inflammatory effects and attenuated monocyte adhesion to endothelial cells inflamed with tumor necrosis factor alpha. We further showed that the inhibitory effect of SAHA on endothelial inflammation and ensuing monocyte adhesion was KLF2 dependent using KLF2-deficient mouse lung endothelial cells or KLF2 small interfering RNA- depleted human endothelial cells. Importantly, we observed that oral administration of SAHA reduced diet-induced atherosclerotic lesion development in ApoE-/- mice without significant effect on serum lipid levels. CONCLUSIONS These results demonstrate that SAHA has KLF2-dependent anti-inflammatory effects in endothelial cells and provide the proof of concept that KLF2 activation could be a promising therapeutic strategy for treating atherosclerosis.
Collapse
Affiliation(s)
- Yanni Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY.,Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Suowen Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Peng Liu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY.,Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Marina Koroleva
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Shuya Zhang
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan, China
| | - Shuyi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Gen Jin
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
39
|
Kim HJ, Son JE, Kim JH, Lee CC, Yang H, Yaghmoor SS, Ahmed Y, Yousef JM, Abualnaja KO, Al-Malki AL, Kumosani TA, Kim JH, Yoon Park JH, Lee CY, Kim JE, Lee KW. Gingerenone A Attenuates Monocyte-Endothelial Adhesion via Suppression of I Kappa B Kinase Phosphorylation. J Cell Biochem 2017; 119:260-268. [PMID: 28513976 DOI: 10.1002/jcb.26138] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/16/2017] [Indexed: 12/19/2022]
Abstract
During the early stages of atherosclerosis, monocytes bind and migrate into the endothelial layer, promoting inflammation within the aorta. In order to prevent the development of atherosclerosis, it is critical to inhibit such inflammation. The therapeutic effects of ginger have been investigated in several models of cardiovascular disease. However, although a number of previous studies have focused on specific compounds, the mechanisms of action responsible remain unclear. Here, we investigated five major compounds present in ginger, and observed that gingerenone A exhibited the strongest inhibitory effects against tumor necrosis factor (TNF)-α and lipopolysaccharide (LPS) induced monocyte-endothelial adhesion. Furthermore, gingerenone A significantly suppressed the expression of TNF-α and LPS-induced vascular cell adhesion molecule-1 (VCAM-1) and chemokine (C-C motif) ligand 2 (CCL2), key mediators of the interaction between monocytes, and endothelial cells. Transactivation of nuclear factor-κB (NF-κB), which is a key transcription factor of VCAM-1 and CCL2, was induced by TNF-α and LPS, and inhibited by treatment of gingerenone A. Gingerenone A also inhibited the phosphorylation of NF-κB inhibitor (IκB) α and IκB Kinase. Taken together, these results demonstrate that gingerenone A attenuates TNF-α and LPS-induced monocyte adhesion and the expression of adhesion factors in endothelial cells via the suppression of NF-κB signaling. J. Cell. Biochem. 119: 260-268, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hee Joo Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Joe Eun Son
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea
| | - Jae Hwan Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Charles C Lee
- Department of Food Science, Cornell University, Ithaca, New York
| | - Hee Yang
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea
| | - Soonham Sami Yaghmoor
- Experimental Biochemistry Unit, King Fahd Medical Research Center and Production of Bioproducts for Industrial Applications Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Youssri Ahmed
- Faculty of Science and Production of Bioproducts for Industrial Applications Research Group, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jehad Mustafa Yousef
- Faculty of Science for Girl's, Department of Biochemistry, Experimental Biochemistry Unit, King Fahd Medical Research Center and Production of Bioproducts for Industrial Applications Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalid Omer Abualnaja
- Faculty of Science and Bioactive Natural Products Research Group, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman Labeed Al-Malki
- Faculty of Science, Department of Biochemistry, Experimental Biochemistry Unit, King Fahd Medical Research Center and Bioactive Natural Products Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Taha Abdullah Kumosani
- Faculty of Science, Department of Biochemistry, Experimental Biochemistry Unit, King Fahd Medical Research Center and Production of Bioproducts for Industrial Applications Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jong Hun Kim
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Han Yoon Park
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Chang Yong Lee
- Department of Food Science, Cornell University, Ithaca, New York.,Production of Bio-products for Industrial Applications Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jong-Eun Kim
- Research Institute of Biotechnology and Medical Converged Science, Dongguk University-Seoul, Goyang, South Korea
| | - Ki Won Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea.,Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
40
|
Park SA, Surh YJ. Modulation of tumor microenvironment by chemopreventive natural products. Ann N Y Acad Sci 2017. [DOI: 10.1111/nyas.13395] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Sin-Aye Park
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences; Seoul National University; Seoul South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences; Seoul National University; Seoul South Korea
- Cancer Research Institute; Seoul National University; Seoul South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, College of Pharmacy; Seoul National University; Seoul South Korea
| |
Collapse
|
41
|
da Costa JP. A current look at nutraceuticals – Key concepts and future prospects. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.02.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
42
|
Anti-nociceptive and anti-inflammatory actions of sulforaphane in chronic constriction injury-induced neuropathic pain mice. Inflammopharmacology 2017; 25:99-106. [PMID: 28054242 DOI: 10.1007/s10787-016-0307-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/21/2016] [Indexed: 01/25/2023]
Abstract
Neuropathic pain is still considered as incurable disease as current therapies are not ideal in terms of efficacy and tolerability. It is imperative to search for novel drugs to obtain better treatments. Sulforaphane (SFN), a derivative of glucoraphanin present in cruciferous vegetables, exhibits therapeutic effects on inflammation-related diseases. Since inflammation plays an important role in regulating chronic pain, in the present study, we investigated anti-nociceptive effects of SFN and its underlying mechanisms in a neuropathic pain mouse model, sciatic nerve chronic constriction injury (CCI). SFN (0.1-100 mg/kg) was injected intraperitoneally for 7 days when pain behaviors, including mechanical allodynia and thermal hyperalgesia, reached to the maximum in CCI mice. We observed that SFN dose-dependently attenuated CCI-induced pain behavioral hypersensitivity, accompanied by reduction in pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) and upregulation of an anti-inflammatory cytokine (IL-10). Moreover, SFN counteracted CCI enhancement of COX2 and iNOS in injured nerves, two key enzymes implicated in inflammation and neuropathic pain. Furthermore, pretreatment of naloxone, an antagonist of opioid receptors, significantly blocked SFN attenuation of behavioral hypersensitivity without affecting SFN modulation of inflammatory cytokines in CCI mice. Interestingly, CCI-induced increase in µ-opioid receptors in injured sciatic nerves was further increased by SFN treatment. Taken together, SFN has both anti-nociceptive and anti-inflammatory actions.
Collapse
|
43
|
Gwon WG, Joung EJ, Kwon MS, Lim SJ, Utsuki T, Kim HR. Sargachromenol protects against vascular inflammation by preventing TNF-α-induced monocyte adhesion to primary endothelial cells via inhibition of NF-κB activation. Int Immunopharmacol 2017; 42:81-89. [PMID: 27902962 DOI: 10.1016/j.intimp.2016.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/09/2016] [Accepted: 11/17/2016] [Indexed: 10/20/2022]
Abstract
Vascular inflammation is a key factor in the pathogenesis of atherosclerosis. The purpose of this study was to investigate the protective effects of sargachromenol (SCM) against tumor necrosis factor (TNF)-α-induced vascular inflammation. SCM decreased the expression of cell adhesion molecules, including intracellular adhesion molecule-1 and vascular cell adhesion molecule-1, in TNF-α-stimulated human umbilical vein endothelial cells (HUVECs), resulted in reduced adhesion of monocytes to HUVECs. SCM also decreased the production of monocyte chemoattractant protein-1 and matrix metalloproteinase-9 in TNF-α-induced HUVECs. Additionally, SCM inhibited activation of nuclear factor kappa B (NF-κB) induced by TNF-α through preventing the degradation of inhibitor kappa B. Moreover, SCM reduced the production of reactive oxygen species in TNF-α-treated HUVECs. Overall, SCM alleviated vascular inflammation through the regulation of NF-κB activation and through its intrinsic antioxidant activity in TNF-α-induced HUVECs. These results indicate that SCM may have potential application as a therapeutic agent against vascular inflammation.
Collapse
Affiliation(s)
- Wi-Gyeong Gwon
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Busan 48513, Republic of Korea
| | - Eun-Ji Joung
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Busan 48513, Republic of Korea
| | - Mi-Sung Kwon
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Busan 48513, Republic of Korea
| | - Su-Jin Lim
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Busan 48513, Republic of Korea
| | - Tadanobu Utsuki
- Department of Pathobiological Science, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Hyeung-Rak Kim
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Busan 48513, Republic of Korea.
| |
Collapse
|
44
|
Yamagishi SI, Matsui T. Protective role of sulphoraphane against vascular complications in diabetes. PHARMACEUTICAL BIOLOGY 2016; 54:2329-2339. [PMID: 26841240 DOI: 10.3109/13880209.2016.1138314] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Context Diabetes is a global health challenge. Although large prospective clinical trials have shown that intensive control of blood glucose or blood pressure reduces the risk for development and progression of vascular complications in diabetes, a substantial number of diabetic patients still experience renal failure and cardiovascular events, which could account for disabilities and high mortality rate in these subjects. Objective Sulphoraphane is a naturally occurring isothiocyanate found in widely consumed cruciferous vegetables, such as broccoli, cabbage and Brussels sprouts, and an inducer of phase II antioxidant and detoxification enzymes with anticancer properties. We reviewed here the protective role of sulphoraphane against diabetic vascular complications. Methods In this review, literature searches were undertaken in Medline and in CrossRef. Non-English language articles were excluded. Keywords [sulphoraphane and (diabetes, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, diabetic complications, vascular, cardiomyocytes, heart or glycation)] have been used to select the articles. Results There is accumulating evidence that sulphoraphane exerts beneficial effects on vascular damage in both cell culture and diabetic animal models via antioxidative properties. Furthermore, we have recently found that sulphoraphane inhibits in vitro formation of advanced glycation end products (AGEs), suppresses the AGE-induced inflammatory reactions in rat aorta by reducing receptor for AGEs (RAGE) expression and decreases serum levels of AGEs in humans. Conclusion These findings suggest that blockade of oxidative stress and/or the AGE-RAGE axis by sulphoraphane may be a novel therapeutic strategy for preventing vascular complications in diabetes.
Collapse
Affiliation(s)
- Sho-Ichi Yamagishi
- a Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications , Kurume University School of Medicine , Kurume , Japan
| | - Takanori Matsui
- a Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications , Kurume University School of Medicine , Kurume , Japan
| |
Collapse
|
45
|
Zhao X, Wen L, Dong M, Lu X. Sulforaphane activates the cerebral vascular Nrf2-ARE pathway and suppresses inflammation to attenuate cerebral vasospasm in rat with subarachnoid hemorrhage. Brain Res 2016; 1653:1-7. [PMID: 27693416 DOI: 10.1016/j.brainres.2016.09.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/25/2016] [Accepted: 09/26/2016] [Indexed: 10/20/2022]
Abstract
Nrf2-ARE pathway reportedly plays a protective role in several central nervous system diseases. No study has explored the role of the Nrf2-ARE pathway in cerebral vasospasm(CVS) after subarachnoid hemorrhage(SAH). The purpose of the present study was to investigate the activation of the cerebral vascular Nrf2-ARE pathway and to determine the potential role of this pathway in the development of CVS following SAH. We investigated whether the administration of sulforaphane (SFN, a specific Nrf2 activator) modulated vascular caliber, Nrf2-ARE pathway activity, proinflammatory cytokine expression, and clinical behavior in a rat model of SAH. A two-hemorrhage protocol was used to generate an animal model of SAH in male Sprague-Dawley rats. Administration of SFN to these rats following SAH enhanced the activity of the Nrf2-ARE pathway and suppressed the release of proinflammatory cytokines. Vasospasm was markedly attenuated in the basilar arteries after SFN therapy. Additionally, SFN administration significantly ameliorated two behavioral functions disrupted by SAH. These results suggest that SFN has a therapeutic benefit in post-SAH, and this may be due to elevated Nrf2-ARE pathway activity and inhibition of cerebral vascular proinflammatory cytokine expression.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Neurosurgery, Wuxi Second Hospital Affiliated Nanjing Medical University, 68 Zhong Shan Road, Wuxi, Jiangsu, China
| | - Liting Wen
- Department of Operating Room, Wuxi Second Hospital Affiliated Nanjing Medical University, 68 Zhong Shan Road, Wuxi, Jiangsu, China
| | - Min Dong
- Department of Neurosurgery, Wuxi Second Hospital Affiliated Nanjing Medical University, 68 Zhong Shan Road, Wuxi, Jiangsu, China
| | - Xiaojie Lu
- Department of Neurosurgery, Wuxi Second Hospital Affiliated Nanjing Medical University, 68 Zhong Shan Road, Wuxi, Jiangsu, China.
| |
Collapse
|
46
|
Matsui T, Nakamura N, Ojima A, Nishino Y, Yamagishi SI. Sulforaphane reduces advanced glycation end products (AGEs)-induced inflammation in endothelial cells and rat aorta. Nutr Metab Cardiovasc Dis 2016; 26:797-807. [PMID: 27212619 DOI: 10.1016/j.numecd.2016.04.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/04/2016] [Accepted: 04/12/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Advanced glycation end products (AGEs)-receptor RAGE interaction evokes oxidative stress and inflammatory reactions, thereby being involved in endothelial cell (EC) damage in diabetes. Sulforaphane is generated from glucoraphanin, a naturally occurring isothiocyanate found in widely consumed cruciferous vegetables, by myrosinase. Sulforaphane has been reported to protect against oxidative stress-mediated cell and tissue injury. However, effects of sulforaphane on AGEs-induced vascular damage remain unclear. METHODS AND RESULTS In this study, we investigated whether and how sulforaphane could inhibit inflammation in AGEs-exposed human umbilical vein ECs (HUVECs) and AGEs-injected rat aorta. Sulforaphane treatment for 4 or 24 h dose-dependently inhibited the AGEs-induced increase in RAGE, monocyte chemoattractant protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecular-1 (VCAM-1) gene expression in HUVECs. AGEs significantly stimulated MCP-1 production by, and THP-1 cell adhesion to, HUVECs, both of which were prevented by 1.6 μM sulforaphane. Sulforaphane significantly suppressed oxidative stress generation and NADPH oxidase activation evoked by AGEs in HUVECs. Furthermore, aortic RAGE, ICAM-1 and VCAM-1 expression in AGEs-injected rats were increased, which were suppressed by simultaneous infusion of sulforaphane. CONCLUSION The present study demonstrated for the first time that sulforaphane could inhibit inflammation in AGEs-exposed HUVECs and AGEs-infused rat aorta partly by suppressing RAGE expression through its anti-oxidative properties. Inhibition of the AGEs-RAGE axis by sulforaphane might be a novel therapeutic target for vascular injury in diabetes.
Collapse
Affiliation(s)
- T Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - N Nakamura
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - A Ojima
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Y Nishino
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - S-I Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan.
| |
Collapse
|
47
|
Holloway PM, Gillespie S, Becker F, Vital SA, Nguyen V, Alexander JS, Evans PC, Gavins FNE. Sulforaphane induces neurovascular protection against a systemic inflammatory challenge via both Nrf2-dependent and independent pathways. Vascul Pharmacol 2016; 85:29-38. [PMID: 27401964 DOI: 10.1016/j.vph.2016.07.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/01/2016] [Accepted: 07/03/2016] [Indexed: 12/25/2022]
Abstract
Sepsis is often characterized by an acute brain inflammation and dysfunction, which is associated with increased morbidity and mortality worldwide. Preventing cerebral leukocyte recruitment may provide the key to halt progression of systemic inflammation to the brain. Here we investigated the influence of the anti-inflammatory and anti-oxidant compound, sulforaphane (SFN) on lipopolysaccharide (LPS)-induced cellular interactions in the brain. The inflammatory response elicited by LPS was blunted by SFN administration (5 and 50mg/kg i.p.) 24h prior to LPS treatment in WT animals, as visualized and quantified using intravital microscopy. This protective effect of SFN was lost in Nrf2-KO mice at the lower dose tested, however 50mg/kg SFN revealed a partial effect, suggesting SFN works in part independently of Nrf2 activity. In vitro, SFN reduced neutrophil recruitment to human brain endothelial cells via a down regulation of E-selectin and vascular cell adhesion molecule 1 (VCAM-1). Our data confirm a fundamental dose-dependent role of SFN in limiting cerebral inflammation. Furthermore, our data demonstrate that not only is Nrf2 in part essential in mediating these neuroprotective effects, but they occur via down-regulation of E-selectin and VCAM-1. In conclusion, SFN may provide a useful therapeutic drug to reduce cerebral inflammation in sepsis.
Collapse
Affiliation(s)
- Paul M Holloway
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Scarlett Gillespie
- Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Felix Becker
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA; Department for General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Shantel A Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Victoria Nguyen
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Paul C Evans
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA; Division of Brain Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
48
|
Michl C, Vivarelli F, Weigl J, De Nicola GR, Canistro D, Paolini M, Iori R, Rascle A. The Chemopreventive Phytochemical Moringin Isolated from Moringa oleifera Seeds Inhibits JAK/STAT Signaling. PLoS One 2016; 11:e0157430. [PMID: 27304884 PMCID: PMC4909285 DOI: 10.1371/journal.pone.0157430] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 05/31/2016] [Indexed: 11/19/2022] Open
Abstract
Sulforaphane (SFN) and moringin (GMG-ITC) are edible isothiocyanates present as glucosinolate precursors in cruciferous vegetables and in the plant Moringa oleifera respectively, and recognized for their chemopreventive and medicinal properties. In contrast to the well-studied SFN, little is known about the molecular pathways targeted by GMG-ITC. We investigated the ability of GMG-ITC to inhibit essential signaling pathways that are frequently upregulated in cancer and immune disorders, such as JAK/STAT and NF-κB. We report for the first time that, similarly to SFN, GMG-ITC in the nanomolar range suppresses IL-3-induced expression of STAT5 target genes. GMG-ITC, like SFN, does not inhibit STAT5 phosphorylation, suggesting a downstream inhibitory event. Interestingly, treatment with GMG-ITC or SFN had a limited inhibitory effect on IFNα-induced STAT1 and STAT2 activity, indicating that both isothiocyanates differentially target JAK/STAT signaling pathways. Furthermore, we showed that GMG-ITC in the micromolar range is a more potent inhibitor of TNF-induced NF-κB activity than SFN. Finally, using a cellular system mimicking constitutive active STAT5-induced cell transformation, we demonstrated that SFN can reverse the survival and growth advantage mediated by oncogenic STAT5 and triggers cell death, therefore providing experimental evidence of a cancer chemopreventive activity of SFN. This work thus identified STAT5, and to a lesser extent STAT1/STAT2, as novel targets of moringin. It also contributes to a better understanding of the biological activities of the dietary isothiocyanates GMG-ITC and SFN and further supports their apparent beneficial role in the prevention of chronic illnesses such as cancer, inflammatory diseases and immune disorders.
Collapse
Affiliation(s)
- Carina Michl
- Stat5 Signaling Research Group, Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Fabio Vivarelli
- Stat5 Signaling Research Group, Institute of Immunology, University of Regensburg, Regensburg, Germany
- Molecular toxicology unit, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Julia Weigl
- Stat5 Signaling Research Group, Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Gina Rosalinda De Nicola
- Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria, Centro di ricerca per le colture industriali (CREA-CIN), Bologna, Italy
| | - Donatella Canistro
- Molecular toxicology unit, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Moreno Paolini
- Molecular toxicology unit, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Renato Iori
- Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria, Centro di ricerca per le colture industriali (CREA-CIN), Bologna, Italy
| | - Anne Rascle
- Stat5 Signaling Research Group, Institute of Immunology, University of Regensburg, Regensburg, Germany
- * E-mail:
| |
Collapse
|
49
|
Luang-In V, Albaser AA, Nueno-Palop C, Bennett MH, Narbad A, Rossiter JT. Glucosinolate and Desulfo-glucosinolate Metabolism by a Selection of Human Gut Bacteria. Curr Microbiol 2016; 73:442-451. [DOI: 10.1007/s00284-016-1079-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/09/2016] [Indexed: 11/24/2022]
|
50
|
Sulforaphane regulates phenotypic and functional switching of both induced and spontaneously differentiating human monocytes. Int Immunopharmacol 2016; 35:85-98. [DOI: 10.1016/j.intimp.2016.03.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/05/2016] [Accepted: 03/08/2016] [Indexed: 12/26/2022]
|