1
|
Xavier LEMDS, Reis TCG, Martins ASDP, Santos JCDF, Bueno NB, Goulart MOF, Moura FA. Antioxidant Therapy in Inflammatory Bowel Diseases: How Far Have We Come and How Close Are We? Antioxidants (Basel) 2024; 13:1369. [PMID: 39594511 PMCID: PMC11590966 DOI: 10.3390/antiox13111369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Inflammatory bowel diseases (IBD) pose a growing public health challenge with unclear etiology and limited efficacy of traditional pharmacological treatments. Alternative therapies, particularly antioxidants, have gained scientific interest. This systematic review analyzed studies from MEDLINE, Cochrane, Web of Science, EMBASE, and Scopus using keywords like "Inflammatory Bowel Diseases" and "Antioxidants." Initially, 925 publications were identified, and after applying inclusion/exclusion criteria-covering studies from July 2015 to June 2024 using murine models or clinical trials in humans and evaluating natural or synthetic substances affecting oxidative stress markers-368 articles were included. This comprised 344 animal studies and 24 human studies. The most investigated antioxidants were polyphenols and active compounds from medicinal plants (n = 242; 70.3%). The review found a strong link between oxidative stress and inflammation in IBD, especially in studies on nuclear factor kappa B and nuclear factor erythroid 2-related factor 2 pathways. However, it remains unclear whether inflammation or oxidative stress occurs first in IBD. Lipid peroxidation was the most studied oxidative damage, followed by DNA damage. Protein damage was rarely investigated. The relationship between antioxidants and the gut microbiota was examined in 103 animal studies. Human studies evaluating oxidative stress markers were scarce, reflecting a major research gap in IBD treatment. PROSPERO registration: CDR42022335357 and CRD42022304540.
Collapse
Affiliation(s)
| | | | - Amylly Sanuelly da Paz Martins
- Postgraduate Studies at the Northeast Biotechnology Network (RENORBIO), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| | - Juliana Célia de Farias Santos
- Postgraduate Degree in Medical Sciences (PPGCM/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| | - Nassib Bezerra Bueno
- Postgraduate Degree in Nutrition (PPGNUT), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil; (L.E.M.d.S.X.); (N.B.B.)
| | - Marília Oliveira Fonseca Goulart
- Postgraduate Studies at the Northeast Biotechnology Network (RENORBIO), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
- Institute of Chemistry and Biotechnology (IQB/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil
| | - Fabiana Andréa Moura
- Postgraduate Degree in Nutrition (PPGNUT), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil; (L.E.M.d.S.X.); (N.B.B.)
- Postgraduate Degree in Medical Sciences (PPGCM/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| |
Collapse
|
2
|
Posta E, Fekete I, Varkonyi I, Zold E, Barta Z. The Versatile Role of Peroxisome Proliferator-Activated Receptors in Immune-Mediated Intestinal Diseases. Cells 2024; 13:1688. [PMID: 39451206 PMCID: PMC11505700 DOI: 10.3390/cells13201688] [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: 07/30/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that sense lipophilic molecules and act as transcription factors to regulate target genes. PPARs have been implicated in the regulation of innate immunity, glucose and lipid metabolism, cell proliferation, wound healing, and fibrotic processes. Some synthetic PPAR ligands are promising molecules for the treatment of inflammatory and fibrotic processes in immune-mediated intestinal diseases. Some of these are currently undergoing or have previously undergone clinical trials. Dietary PPAR ligands and changes in microbiota composition could modulate PPARs' activation to reduce inflammatory responses in these immune-mediated diseases, based on animal models and clinical trials. This narrative review aims to summarize the role of PPARs in immune-mediated bowel diseases and their potential therapeutic use.
Collapse
Affiliation(s)
- Edit Posta
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (I.V.); (Z.B.)
| | - Istvan Fekete
- Institute of Food Technology, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi út 138, 4032 Debrecen, Hungary;
| | - Istvan Varkonyi
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (I.V.); (Z.B.)
| | - Eva Zold
- Department of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, Móricz Zsigmond str. 22, 4032 Debrecen, Hungary;
| | - Zsolt Barta
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (I.V.); (Z.B.)
| |
Collapse
|
3
|
Jia CL, Li BL, Zhao ZH, Zhang Z, Qi-Chen, Song JX, Gou Y, Gao SY, Sun CS, He Y, Ji ES, Zhao Y. Rosmarinic Acid Liposomes Downregulate Hepcidin Expression via BMP6-SMAD1/5/8 Pathway in Mice with Iron Overload. Appl Biochem Biotechnol 2024; 196:6028-6044. [PMID: 38175414 DOI: 10.1007/s12010-023-04828-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
The objective of this study is to examine the potential protective effect of rosmarinic acid (RosA) encapsulated within nanoliposomes (RosA-LIP) on hepatic damage induced by iron overload. The characteristics, stability, and release of RosA-LIP in vitro were identified. The mice were randomly assigned to five groups: Control, Model, Model+DFO (DFO), Model+RosA (RosA), and Model+RosA-LIP (RosA-LIP). The iron overload model was induced by administering iron dextran (i.p.). The DFO, RosA, and RosA-LIP groups received iron dextran and were subsequently treated with DFO, RosA, and RosA-LIP for 14 days. We developed a novel formulation of RosA-LIP that exhibited stability and controlled release properties. Firstly, RosA-LIP improved liver function and ameliorated pathological changes in a mouse model of iron overload. Secondly, RosA-LIP demonstrated the ability to enhance the activities of T-SOD, GSH-Px, and CAT, while reducing the levels of MDA and 4-HNE, thereby effectively mitigating oxidative stress damage induced by iron overload. Thirdly, RosA-LIP reduced hepatic iron levels by downregulating FTL, FTH, and TfR1 levels. Additionally, RosA-LIP exerted a suppressive effect on hepcidin expression through the BMP6-SMAD1/5/8 signaling pathway. Furthermore, RosA-LIP upregulated FPN1 expression in both the liver and duodenum, thereby alleviating iron accumulation in these organs in mice with iron overload. Notably, RosA exhibited a comparable iron chelation effect, and RosA-LIP demonstrated superior efficacy in mitigating liver damage induced by excessive iron overload. RosA-LIP exhibited favorable sustained release properties, targeted delivery, and efficient protection against iron overload-induced liver damage.
Collapse
Affiliation(s)
- Cui-Ling Jia
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Bo-Liang Li
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Zi-Hao Zhao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang City, 050200, Hebei Province, China
| | - Zhi Zhang
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Qi-Chen
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Ji-Xian Song
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Yujing Gou
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Si-Yu Gao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Chen-Sha Sun
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China
| | - Yingna He
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang City, 050200, Hebei Province, China.
- Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, 050200, Hebei, China.
| | - En-Sheng Ji
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China.
| | - Yashuo Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Luquan Xingyuan Road 3, Shijiazhuang, 050200, China.
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, 050013, China.
| |
Collapse
|
4
|
Jia CL, Gou Y, Gao Y, Pei X, Jin X, Li BL, Zhang Z, He Y, Ji ES, Zhao Y. Rosmarinic acid liposomes suppress ferroptosis in ischemic brain via inhibition of TfR1 in BMECs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155835. [PMID: 38968791 DOI: 10.1016/j.phymed.2024.155835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Iron deposition and ferroptosis are involved in ischemic stroke injury, but the choice of drugs for treatment is limited. PURPOSE To investigate the potential neuroprotective effects of Rosmarinic acid (RosA) encapsulated within nanoliposomes (RosA-LIP) on ischemic stroke. METHODS Wild-type (WT) and TfR1EC cKO (specific knockout of the TfR1 gene in BMECs) mice used to establish a dMCAO model, with simultaneous administration of RosA-LIP (20 mg/kg/d, i.p.) or RosA (20 mg/kg/d, i.p.). RESULTS The successful synthesis of RosA-LIP resulted in enhanced stability and precise delivery in both the serum and brain. The administration of RosA-LIP effectively mitigated ischemia-induced behavioral abnormalities and pathological damage. RosA-LIP inhibited ferroptosis by ameliorating mitochondrial abnormalities, increasing GPX4 levels, and decreasing ACSL4/LPCAT3/Lox-dependent lipid peroxidation. RosA-LIP effectively improved blood‒brain barrier (BBB) permeability, increased tight junctions (TJs) protein expression and reduced iron levels in ischemic tissue and brain microvascular endothelial cells (BMECs) by modulating FPN1 and TfR1 levels. Furthermore, RosA-LIP suppressed TfR1 to attenuate ACSL4/LPCAT3/Lox-mediated ferroptosis in TfR1EC cKO mice subjected to dMCAO. CONCLUSION RosA-LIP effectively increased the brain level of RosA and protected against ferroptosis through the regulation of TfR1 in BMECs.
Collapse
Affiliation(s)
- Cui-Ling Jia
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yujing Gou
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuhui Gao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaocui Pei
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China
| | - Xiaofei Jin
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Bo-Liang Li
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhi Zhang
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yingna He
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China.
| | - En-Sheng Ji
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| | - Yashuo Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China.
| |
Collapse
|
5
|
Zhang D, Wan H, Zhao R, Zhang Y, Chen H. Eudragit S100 coated iron oxide-chitosan nanocomposites for colon targeting of 5-aminosalicylic acid ameliorate ulcerative colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome. Int Immunopharmacol 2024; 139:112661. [PMID: 39008936 DOI: 10.1016/j.intimp.2024.112661] [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: 04/15/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
The therapeutic effect of 5-amino salicylic acid (5-ASA), a first-line therapeutic agent for the treatment of ulcerative colitis (UC), is limited by the modest bioavailability afforded by its oral administration. In this study, a 5-ASA oral delivery system was developed using Eudragit S100-coated iron oxide-chitosan nanocomposites (ES-IOCS/5-ASA) to address this issue. According to drug release studies in vitro, ES-IOCS/5-ASA only released a small amount of drug in simulated gastric fluid with a pH of 1.2. However, in a medium with a pH of 7.5, a relatively rapid and complete release was noted. 5-ASA-loaded iron oxide-chitosan nanocomposites (IOCS/5-ASA) could be effectively taken up by NCM460 cells and performed better anti-inflammatory effects than free 5-ASA. At the same time, IOCS/5-ASA improved barrier damage in DSS-induced NCM460 cells. In vivo models of dextran sulphate sodium (DSS)-induced colitis were used to assess the therapeutic efficacy of oral administration of ES-IOCS/5-ASA. ES-IOCS/5-ASA significantly relieved DSS-induced colitis and enhanced the integrity of the intestinal epithelial barrier. ES-IOCS/5-ASA also reduced the expression of NLRP3, ASC and IL-1β. Additionally, iron oxide nanoparticles used as nanozymes could alleviate inflammation. In summary, this study indicates that ES-IOCS/5-ASA exert anti-inflammatory effects on DSS-induced colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome expression, presenting a viable therapeutic choice for the treatment of UC.
Collapse
Affiliation(s)
- Dandan Zhang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Hao Wan
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China
| | - Ran Zhao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yu Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China.
| | - Hong Chen
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
You QY, Hu MD, Qian H. Advanced Nanoarchitectonics of Drug Delivery Systems with Pyroptosis Inhibition for Noncancerous Disease Treatment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202315199] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 01/06/2025]
Abstract
AbstractProgrammed cell death (PCD) is a controlled and organized form of death regulated by genes, allowing cells to adapt to their environment. Pyroptosis, a recently discovered type of programmed cell death, differs from apoptosis and necrosis. It is characterized by the activation of caspase and the cleavage of gasdermin. Many studies have focused on understanding the mechanisms and roles of pyroptosis, particularly in cancer research. While inducing pyroptosis in tumor cells for cancer treatment is a major research focus, it is equally important to explore methods of reducing pyroptosis in noncancerous diseases. Recent advancements in drug delivery systems, specifically nanoarchitectonics, offer site‐specific targeting, prolonged drug circulation, enhanced efficacy, improved solubility, and better absorption. Although several reviews have described how nanoarchitectonics can trigger pyroptosis in tumor cells, little attention is given to their potential to inhibit pyroptosis in noncancerous diseases. Therefore, it is crucial to bridge this gap and explore the future directions for utilizing nanoarchitectonics as a powerful tool against noncancerous diseases. This review aims to delve into the recent progress made in nanoarchitectonics‐based advanced drug delivery systems for the treatment of noncancerous diseases by reducing pyroptosis, while also highlighting potential future perspectives in this emerging field.
Collapse
Affiliation(s)
- Qian Yi You
- Department of Geriatrics and Special Services Medicine Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
- Institute of Respiratory Diseases Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
| | - Ming Dong Hu
- Department of Geriatrics and Special Services Medicine Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
| | - Hang Qian
- Institute of Respiratory Diseases Xinqiao Hospital of Army Medical University (Third Military Medical University) 183 Xinqiao Street Chongqing 400037 P. R. China
| |
Collapse
|
7
|
Kolodziejczyk-Czepas J. Clovamide and Its Derivatives-Bioactive Components of Theobroma cacao and Other Plants in the Context of Human Health. Foods 2024; 13:1118. [PMID: 38611422 PMCID: PMC11011365 DOI: 10.3390/foods13071118] [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: 03/15/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Clovamide (N-caffeoyl-L-3,4-dihydroxyphenylalanine, N-caffeoyldopamine, N-caffeoyl-L-DOPA) is a derivative of caffeic acid, belonging to phenolamides (hydroxycinnamic acid amides). Despite a growing interest in the biological activity of natural polyphenolic substances, studies on the properties of clovamide and related compounds, their significance as bioactive components of the diet, as well as their effects on human health are a relatively new research trend. On the other hand, in vitro and in vivo evidence indicates the considerable potential of these substances in the context of maintaining human health or using them as pharmacophores. The name "clovamide" directly derives from red clover (Trifolium pratense L.), being the first identified source of this compound. In the human diet, clovamides are mainly present in chocolate and other cocoa-containing products. Furthermore, their occurrence in some medicinal plants has also been confirmed. The literature reports deal with the antioxidant, anti-inflammatory, neuroprotective, antiplatelet/antithrombotic and anticancer properties of clovamide-type compounds. This narrative review summarizes the available data on the biological activity of clovamides and their potential health-supporting properties, including prospects for the use of these compounds for therapeutic purposes.
Collapse
Affiliation(s)
- Joanna Kolodziejczyk-Czepas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
8
|
Fadaei MS, Fadaei MR, Kheirieh AE, Rahmanian-Devin P, Dabbaghi MM, Nazari Tavallaei K, Shafaghi A, Hatami H, Baradaran Rahimi V, Nokhodchi A, Askari VR. Niosome as a promising tool for increasing the effectiveness of anti-inflammatory compounds. EXCLI JOURNAL 2024; 23:212-263. [PMID: 38487088 PMCID: PMC10938253 DOI: 10.17179/excli2023-6868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/16/2024] [Indexed: 03/17/2024]
Abstract
Niosomes are drug delivery systems with widespread applications in pharmaceutical research and the cosmetic industry. Niosomes are vesicles of one or more bilayers made of non-ionic surfactants, cholesterol, and charge inducers. Because of their bilayer characteristics, similar to liposomes, niosomes can be loaded with lipophilic and hydrophilic cargos. Therefore, they are more stable and cheaper in preparation than liposomes. They can be classified into four categories according to their sizes and structures, namely small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs,), multilamellar vesicles (MLVs), and multivesicular vesicles (MVVs). There are many methods for niosome preparation, such as thin-film hydration, solvent injection, and heating method. The current study focuses on the preparation methods and pharmacological effects of niosomes loaded with natural and chemical anti-inflammatory compounds in kinds of literature during the past decade. We found that most research was carried out to load anti-inflammatory agents like non-steroidal anti-inflammatory drugs (NSAIDs) into niosome vesicles. The studies revealed that niosomes could improve anti-inflammatory agents' physicochemical properties, including solubility, cellular uptake, stability, encapsulation, drug release and liberation, efficiency, and oral bioavailability or topical absorption. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Mohammad Saleh Fadaei
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Emad Kheirieh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Rahmanian-Devin
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Abouzar Shafaghi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hooman Hatami
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Nokhodchi
- Lupin Pharmaceutical Research Center, 4006 NW 124th Ave., Coral Springs, Florida, FL 33065, USA
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK
| | - Vahid Reza Askari
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Ramos-Tovar E, Muriel P. NLRP3 inflammasome in hepatic diseases: A pharmacological target. Biochem Pharmacol 2023; 217:115861. [PMID: 37863329 DOI: 10.1016/j.bcp.2023.115861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome pathway is mainly responsible for the activation and release of a cascade of proinflammatory mediators that contribute to the development of hepatic diseases. During alcoholic liver disease development, the NLRP3 inflammasome pathway contributes to the maturation of caspase-1, interleukin (IL)-1β, and IL-18, which induce a robust inflammatory response, leading to fibrosis by inducing profibrogenic hepatic stellate cell (HSC) activation. Substantial evidence demonstrates that nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis (NASH) via NLRP3 inflammasome activation, ultimately leading to fibrosis and hepatocellular carcinoma (HCC). Activation of the NLRP3 inflammasome in NASH can be attributed to several factors, such as reactive oxygen species (ROS), gut dysbiosis, leaky gut, which allow triggers such as cardiolipin, cholesterol crystals, endoplasmic reticulum stress, and uric acid to reach the liver. Because inflammation triggers HSC activation, the NLRP3 inflammasome pathway performs a central function in fibrogenesis regardless of the etiology. Chronic hepatic activation of the NLRP3 inflammasome can ultimately lead to HCC; however, inflammation also plays a role in decreasing tumor growth. Some data indicate that NLRP3 inflammasome activation plays an important role in autoimmune hepatitis, but the evidence is scarce. Most researchers have reported that NLRP3 inflammasome activation is essential in liver injury induced by a variety of drugs and hepatotropic virus infection; however, few reports indicate that this pathway can play a beneficial role by inducing liver regeneration. Modulation of the NLRP3 inflammasome appears to be a suitable strategy to treat liver diseases.
Collapse
Affiliation(s)
- Erika Ramos-Tovar
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Apartado Postal 11340, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México.
| |
Collapse
|
10
|
Villalva M, Martínez-García JJ, Jaime L, Santoyo S, Pelegrín P, Pérez-Jiménez J. Polyphenols as NLRP3 inflammasome modulators in cardiometabolic diseases: a review of in vivo studies. Food Funct 2023; 14:9534-9553. [PMID: 37855750 DOI: 10.1039/d3fo03015f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
The nucleotide-binding domain and leucine-rich repeat containing receptors (NLRs) are components of the innate immune system, important in coordinating the inflammatory response. Among them, NLRP3 can form inflammasomes, multiprotein complexes activating the inflammatory caspase-1 and leading, through a cell death-mediated signaling cascade, to the release of several proinflammatory cytokines. Dietary polyphenols, plant secondary metabolites, have been reported to exhibit anti-inflammatory properties, although studies have focused most on their effect on the expression of the final circulating cytokines rather than on the upstream signals activating the NLRP3 inflammasome. The present review explores current knowledge on the potential of dietary polyphenols to regulate the whole NLRP3 inflammasome pathway, in the context of cardiometabolic pathologies (obesity, cardiovascular diseases, type 2 diabetes and non-alcoholic fatty liver disease), based on in vivo studies. A clear tendency towards a decrease in the expression of the whole NLRP3 inflammasome signaling pathway when several animal models were supplemented with polyphenols was observed, commonly showing a dose-response effect; these modifications were concomitant with clinical improvements in the pathologies. Nevertheless, the diversity of doses used, the disparity in polyphenol structures tested and, particularly, the scarce clinical trials and exploration of mechanisms of action show the need to develop further research on the topic.
Collapse
Affiliation(s)
- Marisol Villalva
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), Madrid, Spain.
- Institute of Food Science Research (CIAL). Universidad Autónoma de Madrid (CEI UAM+CSIC), 28049, Madrid, Spain
| | - Juan José Martínez-García
- Molecular Inflammation Group, University Clinical Hospital Virgen de la Arrixaca, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Murcia, Spain
- Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Laura Jaime
- Institute of Food Science Research (CIAL). Universidad Autónoma de Madrid (CEI UAM+CSIC), 28049, Madrid, Spain
| | - Susana Santoyo
- Institute of Food Science Research (CIAL). Universidad Autónoma de Madrid (CEI UAM+CSIC), 28049, Madrid, Spain
| | - Pablo Pelegrín
- Molecular Inflammation Group, University Clinical Hospital Virgen de la Arrixaca, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Murcia, Spain
- Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Jara Pérez-Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), Madrid, Spain.
- CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM), ISCIII, Madrid, Spain
| |
Collapse
|
11
|
Kanika, Khan R. Functionalized nanomaterials targeting NLRP3 inflammasome driven immunomodulation: Friend or Foe. NANOSCALE 2023; 15:15906-15928. [PMID: 37750698 DOI: 10.1039/d3nr03857b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The advancement in drug delivery systems in recent times has significantly enhanced therapeutic effects by enabling site-specific targeting through nanocarriers. These nanocarriers serve as invaluable tools for pharmacotherapeutic advancements against various disorders that enhance the effectiveness of encapsulated drugs by reducing their toxicity and increasing the efficacy of less potent drugs, thereby improving the therapeutic index. Inflammasomes, protein complexes located in the activated immune cell cytoplasm, regulate the activation of caspases involved in inflammation. However, aberrant activation of inflammasomes can result in uncontrolled tissue responses, contributing to the development of various diseases. Therefore, achieving a precise balance between inflammasome inhibition and activation is crucial for effectively treating inflammatory disorders through targeted functionalized nanocarriers. Despite the wealth of available data on the relevance of functionalized nanocarriers in inflammatory disorders, the nanotechnological potential to modulate inflammasomes has not been adequately explored. In this comprehensive review, we highlight the latest research on the modulation of the inflammasome cascade, both upregulating and downregulating its function, using nanocarriers in the context of inflammatory disorders. The utilization of nanocarriers as a therapeutic strategy holds immense potential for researchers aiming to effectively target and modulate inflammasomes in the treatment of inflammatory disorders, thus improving disease severity outcomes.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| |
Collapse
|
12
|
Xue JC, Yuan S, Hou XT, Meng H, Liu BH, Cheng WW, Zhao M, Li HB, Guo XF, Di C, Li MJ, Zhang QG. Natural products modulate NLRP3 in ulcerative colitis. Front Pharmacol 2023; 14:1265825. [PMID: 37849728 PMCID: PMC10577194 DOI: 10.3389/fphar.2023.1265825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/21/2023] [Indexed: 10/19/2023] Open
Abstract
Ulcerative colitis (UC) is a clinically common, progressive, devastating, chronic inflammatory disease of the intestine that is recurrent and difficult to treat. Nod-like receptor protein 3 (NLRP3) is a protein complex composed of multiple proteins whose formation activates cysteine aspartate protease-1 (caspase-1) to induce the maturation and secretion of inflammatory mediators such as interleukin (IL)-1β and IL-18, promoting the development of inflammatory responses. Recent studies have shown that NLRP3 is associated with UC susceptibility, and that it maintains a stable intestinal environment by responding to a wide range of pathogenic microorganisms. The mainstay of treatment for UC is to control inflammation and relieve symptoms. Despite a certain curative effect, there are problems such as easy recurrence after drug withdrawal and many side effects associated with long-term medication. NLRP3 serves as a core link in the inflammatory response. If the relationship between NLRP3 and gut microbes and inflammation-associated factors can be analyzed concerning its related inflammatory signaling pathways, its expression status as well as specific mechanism in the course of IBD can be elucidated and further considered for clinical diagnosis and treatment of IBD, it is expected that the development of lead compounds targeting the NLRP3 inflammasome can be developed for the treatment of IBD. Research into the prevention and treatment of UC, which has become a hotbed of research in recent years, has shown that natural products are rich in therapeutic means, and multi-targets, with fewer adverse effects. Natural products have shown promise in treating UC in numerous basic and clinical trials over the past few years. This paper describes the regulatory role of the NLRP3 inflammasome in UC and the mechanism of recent natural products targeting NLRP3 against UC, which provides a reference for the clinical treatment of this disease.
Collapse
Affiliation(s)
- Jia-Chen Xue
- Department of Nuclear Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
- Department of Immunology and Pathogenic Biology, Yanbian University College of Basic Medicine, Yanji, Jilin, China
| | - Shuo Yuan
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Xiao-Ting Hou
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Huan Meng
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Bao-Hong Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Wen-Wen Cheng
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Ming Zhao
- Department of Nuclear Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Hong-Ben Li
- Department of Nuclear Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xue-Fen Guo
- Department of Nuclear Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Chang Di
- Department of Nuclear Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Min-Jie Li
- Department of Nuclear Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Qing-Gao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
- Department of Immunology and Pathogenic Biology, Yanbian University College of Basic Medicine, Yanji, Jilin, China
| |
Collapse
|
13
|
Lu B, Li C, Jing L, Zhuang F, Xiang H, Chen Y, Huang B. Rosmarinic acid nanomedicine for rheumatoid arthritis therapy: Targeted RONS scavenging and macrophage repolarization. J Control Release 2023; 362:631-646. [PMID: 37708976 DOI: 10.1016/j.jconrel.2023.09.012] [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: 05/28/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
The infiltration of inflammatory cells, especially macrophages, integrated with the production of reactive oxygen and nitrogen species (RONS) and the release of inflammatory cytokines play a crucial role in the pathogenesis of rheumatoid arthritis (RA). Synergistic combination of RONS scavenging and macrophage repolarization from pro-inflammatory M1 phenotype towards anti-inflammatory M2 phenotype, provides a promising strategy for efficient RA treatment. Herein, this study reported a unique self-assembly strategy to construct distinct rosmarinic acid nanoparticles (RNPs) for efficient RA treatment using the naturally occurring polyphenol-based compound, rosmarinic acid (RosA). The designed RNPs exhibited favorable capability in scavenging RONS and pro-inflammatory cytokines produced by macrophages. Attributing to the widened vascular endothelial-cell gap at inflammation sites, RNPs could target and accumulate at the inflammatory joints of collagen-induced arthritis (CIA) rats for guaranteeing therapeutic effect. In vivo investigation demonstrated that RNPs alleviated the symptoms of RA, including joint swelling, synovial hyperplasia, cartilage degradation, and bone erosion in CIA rats. Additionally, the designed RNPs promoted macrophage polarization from M1 phenotype towards M2 phenotype, resulting in the suppressed progression of RA. Therefore, this research represents the representative paradigm for RA therapy using antioxidative nanomedicine deriving from the natural polyphenol-based compound.
Collapse
Affiliation(s)
- Beilei Lu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China; Shanghai Institute of Medical Imaging, Shanghai 200032, PR China; Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai 200032, PR China
| | - Cuixian Li
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China; Shanghai Institute of Medical Imaging, Shanghai 200032, PR China; Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai 200032, PR China
| | - Luxia Jing
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China; Shanghai Institute of Medical Imaging, Shanghai 200032, PR China; Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai 200032, PR China
| | - Fan Zhuang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China; Shanghai Institute of Medical Imaging, Shanghai 200032, PR China; Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai 200032, PR China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| | - Beijian Huang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China; Shanghai Institute of Medical Imaging, Shanghai 200032, PR China; Institute of Medical Ultrasound and Engineering, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
14
|
Pratelli G, Tamburini B, Carlisi D, De Blasio A, D’Anneo A, Emanuele S, Notaro A, Affranchi F, Giuliano M, Seidita A, Lauricella M, Di Liberto D. Foodomics-Based Approaches Shed Light on the Potential Protective Effects of Polyphenols in Inflammatory Bowel Disease. Int J Mol Sci 2023; 24:14619. [PMID: 37834065 PMCID: PMC10572570 DOI: 10.3390/ijms241914619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and progressive inflammatory disorder affecting the gastrointestinal tract (GT) caused by a wide range of genetic, microbial, and environmental factors. IBD is characterized by chronic inflammation and decreased gut microbial diversity, dysbiosis, with a lower number of beneficial bacteria and a concomitant increase in pathogenic species. It is well known that dysbiosis is closely related to the induction of inflammation and oxidative stress, the latter caused by an imbalance between reactive oxygen species (ROS) production and cellular antioxidant capacity, leading to cellular ROS accumulation. ROS are responsible for intestinal epithelium oxidative damage and the increased intestinal permeability found in IBD patients, and their reduction could represent a potential therapeutic strategy to limit IBD progression and alleviate its symptoms. Recent evidence has highlighted that dietary polyphenols, the natural antioxidants, can maintain redox equilibrium in the GT, preventing gut dysbiosis, intestinal epithelium damage, and radical inflammatory responses. Here, we suggest that the relatively new foodomics approaches, together with new technologies for promoting the antioxidative properties of dietary polyphenols, including novel delivery systems, chemical modifications, and combination strategies, may provide critical insights to determine the clinical value of polyphenols for IBD therapy and a comprehensive perspective for implementing natural antioxidants as potential IBD candidate treatment.
Collapse
Affiliation(s)
- Giovanni Pratelli
- Department of Physics and Chemistry (DiFC) Emilio Segrè, University of Palermo, 90128 Palermo, Italy;
| | - Bartolo Tamburini
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (B.T.); (D.C.); (S.E.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90127 Palermo, Italy;
| | - Daniela Carlisi
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (B.T.); (D.C.); (S.E.)
| | - Anna De Blasio
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy; (A.D.B.); (A.D.); (A.N.); (F.A.); (M.G.)
| | - Antonella D’Anneo
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy; (A.D.B.); (A.D.); (A.N.); (F.A.); (M.G.)
| | - Sonia Emanuele
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (B.T.); (D.C.); (S.E.)
| | - Antonietta Notaro
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy; (A.D.B.); (A.D.); (A.N.); (F.A.); (M.G.)
| | - Federica Affranchi
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy; (A.D.B.); (A.D.); (A.N.); (F.A.); (M.G.)
| | - Michela Giuliano
- Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy; (A.D.B.); (A.D.); (A.N.); (F.A.); (M.G.)
| | - Aurelio Seidita
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90127 Palermo, Italy;
| | - Marianna Lauricella
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (B.T.); (D.C.); (S.E.)
| | - Diana Di Liberto
- Section of Biochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (B.T.); (D.C.); (S.E.)
| |
Collapse
|
15
|
Taravat M, Asadpour R, Jozani RJ, Fattahi A, Khordadmehr M. Enhanced anti-inflammatory effect of Rosmarinic acid by encapsulation and combination with the exosome in mice with LPS-induced endometritis through suppressing the TLR4-NLRP3 signaling pathway. J Reprod Immunol 2023; 159:103992. [PMID: 37451160 DOI: 10.1016/j.jri.2023.103992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/16/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
The TLR4-NLRP3 signaling pathway plays an essential role in the development of inflammation and especially endometritis. Rosmarinic acid (RA) can have potent anti-inflammatory effects in the drug-loading system. The purpose of this was to evaluate the anti-inflammatory effects of RA loaded to exosomes (RLE) on lipopolysaccharide (LPS)-induced endometritis in mice. RA was loaded into serum-derived exosome, using sonication methods. Animals in the treatment groups were subjected to uterine horn injection of RA, exosome, RA combination with exosome (R+E), and RA loaded to exosome (RLE) in uterine horn by two dosages in each group (5 and 10 mg/kg of RA or exosome), 24 h after inducing endometritis. Histopathological analysis, MPO production, immunohistochemistry, and qPCR were used to determine whether the treatment groups were adequate in controlling inflammation. The results showed that treatment groups, and mainly RLE10 and R10 +E10 groups, could modulate pathological changes, inhibit myeloperoxidase (MPO) activity, and significantly reduce the gene and protein expression of TLR4, NLRP3, inflammatory cytokines such as IL-1β, IL-18, and TNF-α, and lastly, GSDM-D as a pyroptosis factor. In conclusion, RA loaded and combination with exosomes at a dosage of 10 mg/kg (RLE10 and R10 +E10) improved endometritis in mice through a suppressing TLR4-NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Morteza Taravat
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Reza Asadpour
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Razi Jafari Jozani
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monireh Khordadmehr
- Department of Basic science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
16
|
Nani A, Tehami W. Targeting inflammasome pathway by polyphenols as a strategy for pancreatitis, gastrointestinal and liver diseases management: an updated review. Front Nutr 2023; 10:1157572. [PMID: 37743919 PMCID: PMC10513047 DOI: 10.3389/fnut.2023.1157572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Obesity, pancreatitis, cardiovascular, gastrointestinal (GI), and liver diseases have all been linked to the Western lifestyle, characterized by increased unhealthy food consumption and decreased physical activity. Besides obesity and pancreatitis, many GI and liver diseases are associated with inflammation. Inflammasomes are multi-protein complexes that mediate acute and restorative inflammatory pathways. However, many aberrations in inflammasome activity originate from shifts in dietary habits. Evidence reveals that dietary polyphenols effectively modulate inflammasome-associated dysfunctions. With a focus on pancreatitis, GI, and liver disorders, this review set out to provide the most relevant evidence for the therapeutic impact of polyphenols via the regulation of the inflammasome pathway. Overall, flavonoid and non-flavonoid polyphenols maintain intestinal eubiosis, downregulate NLRP3 inflammasome canonical pathway, and restore redox status via upregulating Nrf2/HO-1 signaling. These effects at the level of the intestine, the liver, and the pancreas are associated with decreased systemic levels of key pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6.
Collapse
Affiliation(s)
- Abdelhafid Nani
- Laboratory of Saharan Natural Resources, University of Ahmed Draia, Adrar, Algeria
| | | |
Collapse
|
17
|
Fulgheri F, Aroffu M, Ramírez M, Román-Álamo L, Peris JE, Usach I, Nacher A, Manconi M, Fernàndez-Busquets X, Manca ML. Curcumin or quercetin loaded nutriosomes as oral adjuvants for malaria infections. Int J Pharm 2023; 643:123195. [PMID: 37394159 DOI: 10.1016/j.ijpharm.2023.123195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Artemisinin, curcumin or quercetin, alone or in combination, were loaded in nutriosomes, special phospholipid vesicles enriched with Nutriose FM06®, a soluble dextrin with prebiotic activity, that makes these vesicles suitable for oral delivery. The resulting nutriosomes were sized between 93 and 146 nm, homogeneously dispersed, and had slightly negative zeta potential (around -8 mV). To improve their shelf life and storability over time, vesicle dispersions were freeze-dried and stored at 25 °C. Results confirmed that their main physico-chemical characteristics remained unchanged over a period of 12 months. Additionally, their size and polydispersity index did not undergo any significant variation after dilution with solutions at different pHs (1.2 and 7.0) and high ionic strength, mimicking the harsh conditions of the stomach and intestine. An in vitro study disclosed the delayed release of curcumin and quercetin from nutriosomes (∼53% at 48 h) while artemisinin was quickly released (∼100% at 48 h). Cytotoxicity assays using human colon adenocarcinoma cells (Caco-2) and human umbilical vein endothelial cells (HUVECs) proved the high biocompatibility of the prepared formulations. Finally, in vitro antimalarial activity tests, assessed against the 3D7 strain of Plasmodium falciparum, confirmed the effectiveness of nutriosomes in the delivery of curcumin and quercetin, which can be used as adjuvants in the antimalaria treatment. The efficacy of artemisinin was also confirmed but not improved. Overall results proved the possible use of these formulations as an accompanying treatment of malaria infections.
Collapse
Affiliation(s)
- Federica Fulgheri
- Dept. of Life and Environmental Sciences of the University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, Monserrato 09042, CA, Italy
| | - Matteo Aroffu
- Dept. of Life and Environmental Sciences of the University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, Monserrato 09042, CA, Italy
| | - Miriam Ramírez
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, ES-08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Lucía Román-Álamo
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, ES-08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - José Esteban Peris
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain
| | - Iris Usach
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain
| | - Amparo Nacher
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - Maria Manconi
- Dept. of Life and Environmental Sciences of the University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, Monserrato 09042, CA, Italy.
| | - Xavier Fernàndez-Busquets
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, ES-08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Maria Letizia Manca
- Dept. of Life and Environmental Sciences of the University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, Monserrato 09042, CA, Italy
| |
Collapse
|
18
|
Alves-Silva JM, Pedreiro S, Cruz MT, Salgueiro L, Figueirinha A. Exploring the Traditional Uses of Thymbra capitata Infusion in Algarve (Portugal): Anti-Inflammatory, Wound Healing, and Anti-Aging. Pharmaceuticals (Basel) 2023; 16:1202. [PMID: 37765010 PMCID: PMC10538188 DOI: 10.3390/ph16091202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
Inflammation plays a pivotal role in the resolution of infection or tissue damage. In addition, inflammation is considered a hallmark of aging, which in turn compromises wound healing. Thymbra capitata is an aromatic plant, whose infusion is traditionally used as an anti-inflammatory and wound-healing agent. In this study, a T. capitata infusion was prepared and characterized by HPLC-PDA-ESI-MSn and its safety profile determined by the resazurin metabolic assay. The anti-inflammatory potential was revealed in lipopolysaccharide (LPS)-stimulated macrophages by assessing nitric oxide (NO) release and levels of inducible nitric oxide synthase (iNOS) and the interleukin-1β pro-form (pro-IL-1β). Wound-healing capacity was determined using the scratch assay. The activity of senescence-associated β-galactosidase was used to unveil the anti-senescent potential, along with the nuclear accumulation of yH2AX and p21 levels. The antiradical potential was assessed by DPPH and ABTS scavenging assays. The infusion contains predominantly rosmarinic acid and salvianolic acids. The extract decreased NO, iNOS, and pro-IL-1β levels. Interestingly, the extract promoted wound healing and decreased β-galactosidase activity, as well as yH2AX and p21 levels. The present work highlights strong antiradical, anti-inflammatory, and wound healing capacities, corroborating the traditional uses ascribed to this plant. We have described, for the first time for this extract, anti-senescent properties.
Collapse
Affiliation(s)
- Jorge Miguel Alves-Silva
- Univ Coimbra, Institute for Clinical and Biomedical Research, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
| | - Sónia Pedreiro
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Maria Teresa Cruz
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Univ Coimbra, Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, 3030-790 Coimbra, Portugal
| | - Artur Figueirinha
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| |
Collapse
|
19
|
Fan YM, Wei YY, Wang HR, Yu-Ga, Zhang YN, Hao Z. Inhibitory effect of Portulaca oleracea L. aqueous extract and juice on NLRP3 inflammasome activation in an ulcerative colitis mouse model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:86380-86394. [PMID: 37402916 DOI: 10.1007/s11356-023-28365-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
Portulaca oleracea L. (PO) is an edible and medicinal plant used for treating gastrointestinal diseases. However, the effects of PO on ulcerative colitis (UC) and underlying mechanisms remain unclear. This study investigated the effects of PO aqueous extract (POE) and PO juice (PJ) on dextran sulfate sodium (DSS)-induced UC in a mouse model and attempted to unravel their underlying mechanisms. The results revealed that PJ contains more bioactive compounds and has more overlapping targets with UC than POE. Both POE and PJ effectively reduced Disease Activity Index scores and inflammatory cell infiltration in the UC mouse model, but PJ had a better effect than POE. Furthermore, PJ inhibited pyroptosis by decreasing the expression of the NLRP3 inflammasome, while also repairing the dysfunction of the intestinal barrier by upregulating the expression of tight junction proteins. Therefore, based on the study findings, we concluded that PJ can improve DSS-induced UC and may suppress pyroptosis by interfering with the activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Yi-Meng Fan
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Yuan-Yuan Wei
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Hui-Ru Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Yu-Ga
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Yan-Nan Zhang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultura University, Beijing, 100193, China.
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, People's Republic of China.
- National Center of Technology Innovation for Medicinal function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China.
| |
Collapse
|
20
|
Liu A, Chai X, Zhu S, Chin PT, He M, Xu YJ, Liu Y. Effects of N-succinyl-chitosan coating on properties of astaxanthin-loaded PEG-liposomes: Environmental stability, antioxidant/antibacterial activities, and in vitro release. Int J Biol Macromol 2023:125311. [PMID: 37302627 DOI: 10.1016/j.ijbiomac.2023.125311] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/22/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Astaxanthin (AST) has outstanding antioxidant and anti-inflammation bioactivities, but the low biocompatibility and stability limit its application in foods. In this study, N-succinyl-chitosan (NSC)-coated AST polyethylene glycol (PEG)-liposomes were constructed to enhance the biocompatibility, stability, and intestinal-targeted migration of AST. The AST NSC/PEG-liposomes were uniform in size, had larger particles, greater encapsulation efficiency, and better storage, pH, and temperature stability than the AST PEG-liposomes. AST NSC/PEG-liposomes exerted stronger antibacterial and antioxidant activities against Escherichia coli and Staphylococcus aureus than AST PEG-liposomes. The NSC coating not only protects AST PEG-liposomes from gastric acid but also prolongs the retention and sustained release of AST NSC/PEG-liposomes depending on the intestinal pH. Moreover, caco-2 cellular uptake studies showed that AST NSC/PEG-liposomes had higher cellular uptake efficiency than AST PEG-liposomes. And AST NSC/PEG-liposomes were taken up by caco-2 cells through clathrin mediated endocytic, macrophage pathways and paracellular transport pathway. These results further proved that AST NSC/PEG-liposomes delayed the release and promoted the intestinal absorption of AST. Hence, AST PEG-liposomes coated with NSC could potentially be used as an efficient delivery system for therapeutic AST.
Collapse
Affiliation(s)
- Aiyang Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xiuhang Chai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Shuang Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Ping-Tan Chin
- Department of Food Technology, Faculty of Food Science and Technology, University Putra Malaysia, Selangor 410500, Malaysia
| | - Mengxue He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
21
|
Direito R, Barbalho SM, Figueira ME, Minniti G, de Carvalho GM, de Oliveira Zanuso B, de Oliveira Dos Santos AR, de Góes Corrêa N, Rodrigues VD, de Alvares Goulart R, Guiguer EL, Araújo AC, Bosso H, Fornari Laurindo L. Medicinal Plants, Phytochemicals and Regulation of the NLRP3 Inflammasome in Inflammatory Bowel Diseases: A Comprehensive Review. Metabolites 2023; 13:728. [PMID: 37367886 DOI: 10.3390/metabo13060728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
Ongoing research explores the underlying causes of ulcerative colitis and Crohn's disease. Many experts suggest that dysbiosis in the gut microbiota and genetic, immunological, and environmental factors play significant roles. The term "microbiota" pertains to the collective community of microorganisms, including bacteria, viruses, and fungi, that reside within the gastrointestinal tract, with a particular emphasis on the colon. When there is an imbalance or disruption in the composition of the gut microbiota, it is referred to as dysbiosis. Dysbiosis can trigger inflammation in the intestinal cells and disrupt the innate immune system, leading to oxidative stress, redox signaling, electrophilic stress, and inflammation. The Nod-like Receptor (NLR) Family Pyrin Domain Containing 3 (NLRP3) inflammasome, a key regulator found in immunological and epithelial cells, is crucial in inducing inflammatory diseases, promoting immune responses to the gut microbiota, and regulating the integrity of the intestinal epithelium. Its downstream effectors include caspase-1 and interleukin (IL)-1β. The present study investigated the therapeutic potential of 13 medicinal plants, such as Litsea cubeba, Artemisia anomala, Piper nigrum, Morus macroura, and Agrimonia pilosa, and 29 phytocompounds such as artemisitene, morroniside, protopine, ferulic acid, quercetin, picroside II, and hydroxytyrosol on in vitro and in vivo models of inflammatory bowel diseases (IBD), with a focus on their effects on the NLRP3 inflammasome. The observed effects of these treatments included reductions in IL-1β, tumor necrosis factor-alpha, IL-6, interferon-gamma, and caspase levels, and increased expression of antioxidant enzymes, IL-4, and IL-10, as well as regulation of gut microbiota. These effects could potentially provide substantial advantages in treating IBD with few or no adverse effects as caused by synthetic anti-inflammatory and immunomodulated drugs. However, additional research is necessary to validate these findings clinically and to develop effective treatments that can benefit individuals who suffer from these diseases.
Collapse
Affiliation(s)
- Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical & Regulatory Science, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, São Paulo, Brazil
| | - Maria Eduardo Figueira
- Laboratory of Systems Integration Pharmacology, Clinical & Regulatory Science, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Gabriel Magno de Carvalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Bárbara de Oliveira Zanuso
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Ana Rita de Oliveira Dos Santos
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Natália de Góes Corrêa
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Avenida Monte Carmelo, 800, Marília 17519-030, São Paulo, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Henrique Bosso
- Medical Department, School of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Avenida Monte Carmelo, 800, Marília 17519-030, São Paulo, Brazil
| |
Collapse
|
22
|
Caban M, Lewandowska U. Encapsulation of Polyphenolic Compounds Based on Hemicelluloses to Enhance Treatment of Inflammatory Bowel Diseases and Colorectal Cancer. Molecules 2023; 28:molecules28104189. [PMID: 37241929 DOI: 10.3390/molecules28104189] [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: 04/19/2023] [Revised: 05/13/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammatory bowel diseases (IBD) and colorectal cancer (CRC) are difficult to cure, and available treatment is associated with troubling side effects. In addition, current therapies have limited efficacy and are characterized by high costs, and a large segment of the IBD and CRC patients are refractive to the treatment. Moreover, presently used anti-IBD therapies in the clinics are primarily aimed on the symptomatic control. That is why new agents with therapeutic potential against IBD and CRC are required. Currently, polyphenols have received great attention in the pharmaceutical industry and in medicine due to their health-promoting properties. They may exert anti-inflammatory, anti-oxidative, and anti-cancer activity, via inhibiting production of pro-inflammatory cytokines and enzymes or factors associated with carcinogenesis (e.g., matrix metalloproteinases, vascular endothelial growth factor), suggesting they may have therapeutic potential against IBD and CRC. However, their use is limited under both processing conditions or gastrointestinal interactions, reducing their stability and hence their bioaccessibility and bioavailability. Therefore, there is a need for more effective carriers that could be used for encapsulation of polyphenolic compounds. In recent years, natural polysaccharides have been proposed for creating carriers used in the synthesis of polyphenol encapsulates. Among these, hemicelluloses are particularly noteworthy, being characterized by good biocompatibility, biodegradation, low immunogenicity, and pro-health activity. They may also demonstrate synergy with the polyphenol payload. This review discusses the utility and potential of hemicellulose-based encapsulations of polyphenols as support for treatment of IBD and CRC.
Collapse
Affiliation(s)
- Miłosz Caban
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215 Lodz, Poland
| | - Urszula Lewandowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215 Lodz, Poland
| |
Collapse
|
23
|
Sun HX, Zhu Y. Progress on Regulation of NLRP3 Inflammasome by Chinese Medicine in Treatment of Ulcerative Colitis. Chin J Integr Med 2023:10.1007/s11655-023-3551-1. [PMID: 37148482 DOI: 10.1007/s11655-023-3551-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 05/08/2023]
Abstract
Ulcerative colitis (UC) is a chronic, non-specific intestinal disease that not only affects the quality of life of patients and their families but also increases the risk of colorectal cancer. The nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome is an important component of inflammatory response system, and its activation induces an inflammatory cascade response that is involved in the development and progression of UC by releasing inflammatory cytokines, damaging intestinal epithelial cells, and disrupting the intestinal mucosal barrier. Chinese medicine (CM) plays a vital role in the prevention and treatment of UC and is able to regulate NLRP3 inflammasome. Many experimental studies on the regulation of NLRP3 inflammasome mediated by CM have been carried out, demonstrating that CM formulae with main effects of clearing heat, detoxifying toxicity, drying dampness, and activating blood circulation. Flavonoids and phenylpropanoids can effectively regulate NLRP3 inflammasome. Other active components of CM can interfere with the process of NLRP3 inflammasome assembly and activation, leading to a reduction in inflammation and UC symptoms. However, the reports are relatively scattered and lack systematic reviews. This paper reviews the latest findings regarding the NLRP3 inflammasome activation-related pathways associated with UC and the potential of CM in treating UC through modulation of NLRP3 inflammasome. The purpose of this review is to explore the possible pathological mechanisms of UC and suggest new directions for development of therapeutic tools.
Collapse
Affiliation(s)
- Hao-Xian Sun
- Department of Gastroenterology, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ying Zhu
- Department of Gastroenterology, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China.
| |
Collapse
|
24
|
Alves-Silva JM, Pedreiro S, Cavaleiro C, Cruz MT, Figueirinha A, Salgueiro L. Effect of Thymbra capitata (L.) Cav. on Inflammation, Senescence and Cell Migration. Nutrients 2023; 15:nu15081930. [PMID: 37111149 PMCID: PMC10146686 DOI: 10.3390/nu15081930] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/30/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Aromatic plants are reported to display pharmacological properties, including anti-aging. This work aims to disclose the anti-aging effect of the essential oil (EO) of Thymbra capitata (L.) Cav., an aromatic and medicinal plant widely used as a spice, as well as of the hydrodistillation residual water (HRW), a discarded by-product of EO hydrodistillation. The phytochemical characterization of EO and HRW was assessed by GC-MS and HPLC-PDA-ESI-MSn, respectively. The DPPH, ABTS, and FRAP assays were used to disclose the antioxidant properties. The anti-inflammatory potential was evaluated using lipopolysaccharide-stimulated macrophages by assessing NO production, iNOS, and pro-IL-1β protein levels. Cell migration was evaluated using the scratch wound assay, and the etoposide-induced senescence was used to assess the modulation of senescence. The EO is mainly characterized by carvacrol, while the HRW is predominantly characterized by rosmarinic acid. The HRW exerts a stronger antioxidant effect in the DPPH and FRAP assays, whereas the EO was the most active sample in the ABTS assay. Both extracts reduce NO, iNOS, and pro-IL-1β. The EO has no effect on cell migration and presents anti-senescence effects. In opposition, HRW reduces cell migration and induces cellular senescence. Overall, our study highlights interesting pharmacological properties for both extracts, EO being of interest as an anti-aging ingredient and HRW relevant in cancer therapy.
Collapse
Affiliation(s)
- Jorge M Alves-Silva
- Institute for Clinical and Biomedical Research, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
| | - Sónia Pedreiro
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Carlos Cavaleiro
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
- Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| |
Collapse
|
25
|
Wang Q, Xu K, Cai X, Wang C, Cao Y, Xiao J. Rosmarinic Acid Restores Colonic Mucus Secretion in Colitis Mice by Regulating Gut Microbiota-Derived Metabolites and the Activation of Inflammasomes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4571-4585. [PMID: 36883243 DOI: 10.1021/acs.jafc.2c08444] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Maintaining a steady state of mucus barrier is an important potential target for polyphenol to exert its anticolitis activity. This study elucidates the pivotal role of polyphenol rosmaric acid (RA) in regulating the mucus barrier function and alleviating inflammation by identifying its gut microbiota-derived metabolites and evaluating its inhibitory effect on inflammasomes in colitis mice. Results demonstrated that RA treatment promoted the proliferation of goblet cells and restored the level of mucus secretion, especially Muc2. RA reshaped the microbiota of colitis mice, particularly the boost of core probiotics, such as p. Bacteroidaceae, f. Muribaculaceae, g. Muribaculaceae, g. Alistipes, and g. Clostridia_UCG-014. Nontargeted metabonomics and targeted metabonomics confirmed a significant increase in the bile acids and their metabolites (7-sulfocholic acid, stercobilin, chenodeoxycholic acid 3-sulfate, chenodeoxycholic acid sulfate, and ursodeoxycholic acid 3-sulfate), indole metabolites ((R)-2,3-dihydro-3,5-dihydroxy-2-oxo-3-indoleacetic acid, frovatriptan, 3-formyl-6-hydroxyindole, and brassicanal A), and short-chain fatty acids (SCFAs) (acetic acid, butyric acid, isobutyric acid, isovaleric acid, and valeric acid) that contributed to the strengthened mucus barrier function. In addition, being absorbed mainly in the lower digestive tract, RA inhibited the overexpression of inflammasomes (especially NLRP6) that occurred in colitis mice to promote the mucus secretion of goblet cells. These data confirmed that RA, as a promising candidate to enhance gut health, restored colonic mucus secretion in colitis mice by mediating the production of gut microbiota-derived metabolites and the overexpression of inflammasomes. The presented study provides scientific evidence explaining the apparent paradox of low bioavailability and high bioactivity in polyphenols.
Collapse
Affiliation(s)
- Qun Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Kangjie Xu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xu Cai
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Chujing Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
26
|
Wang T, Xu H, Dong R, Wu S, Guo Y, Wang D. Effectiveness of targeting the NLRP3 inflammasome by using natural polyphenols: A systematic review of implications on health effects. Food Res Int 2023; 165:112567. [PMID: 36869555 DOI: 10.1016/j.foodres.2023.112567] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/13/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Globally, inflammation and metabolic disorders pose serious public health problems and are major health concerns. It has been shown that natural polyphenols are effective in the treatment of metabolic diseases, including anti-inflammation, anti-diabetes, anti-obesity, neuron-protection, and cardio-protection. NLRP3 inflammasome, which are multiprotein complexes located within the cytosol, play an important role in the innate immune system. However, aberrant activation of the NLRP3 inflammasome were discovered as essential molecular mechanisms in triggering inflammatory processes as well as implicating it in several major metabolic diseases, such as type 2 diabetes mellitus, obesity, atherosclerosis or cardiovascular disease. Recent studies indicate that natural polyphenols can inhibit NLRP3 inflammasome activation. In this review, the progress of natural polyphenols preventing inflammation and metabolic disorders via targeting NLRP3 inflammasome is systemically summarized. From the viewpoint of interfering NLRP3 inflammasome activation, the health effects of natural polyphenols are explained. Recent advances in other beneficial effects, clinical trials, and nano-delivery systems for targeting NLRP3 inflammasome are also reviewed. NLRP3 inflammasome is targeted by natural polyphenols to exert multiple health effects, which broadens the understanding of polyphenol mechanisms and provides valuable guidance to new researchers in this field.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000 Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China
| | - Ruixia Dong
- College of Horticulture, Jinling Institute of Technology, 211169 Nanjing, China
| | - Shanshan Wu
- College of Agriculture & Biotechnology, Zhejiang University, 310058 Hanzhou, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China.
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China.
| |
Collapse
|
27
|
Ali FE, Ibrahim IM, Ghogar OM, Abd-alhameed EK, Althagafy HS, Hassanein EH. Therapeutic interventions target the NLRP3 inflammasome in ulcerative colitis: Comprehensive study. World J Gastroenterol 2023; 29:1026-1053. [PMID: 36844140 PMCID: PMC9950862 DOI: 10.3748/wjg.v29.i6.1026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/29/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
One of the significant health issues in the world is the prevalence of ulcerative colitis (UC). UC is a chronic disorder that mainly affects the colon, beginning with the rectum, and can progress from asymptomatic mild inflammation to extensive inflammation of the entire colon. Understanding the underlying molecular mechanisms of UC pathogenesis emphasizes the need for innovative therapeutic approaches based on identifying molecular targets. Interestingly, in response to cellular injury, the NLR family pyrin domain containing 3 (NLRP3) inflammasome is a crucial part of the inflammation and immunological reaction by promoting caspase-1 activation and the release of interleukin-1β. This review discusses the mechanisms of NLRP3 inflammasome activation by various signals and its regulation and impact on UC.
Collapse
Affiliation(s)
- Fares E.M Ali
- Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Islam M. Ibrahim
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Osama M Ghogar
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Esraa K. Abd-alhameed
- Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 12345, Egypt
| | - Hanan S. Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah 12345, Saudi Arabia
| | - Emad H.M. Hassanein
- Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| |
Collapse
|
28
|
Laurindo LF, de Maio MC, Minniti G, de Góes Corrêa N, Barbalho SM, Quesada K, Guiguer EL, Sloan KP, Detregiachi CRP, Araújo AC, de Alvares Goulart R. Effects of Medicinal Plants and Phytochemicals in Nrf2 Pathways during Inflammatory Bowel Diseases and Related Colorectal Cancer: A Comprehensive Review. Metabolites 2023; 13:243. [PMID: 36837862 PMCID: PMC9966918 DOI: 10.3390/metabo13020243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are related to nuclear factor erythroid 2-related factor 2 (Nrf2) dysregulation. In vitro and in vivo studies using phytocompounds as modulators of the Nrf2 signaling in IBD have already been published. However, no existing review emphasizes the whole scenario for the potential of plants and phytocompounds as regulators of Nrf2 in IBD models and colitis-associated colorectal carcinogenesis. For these reasons, this study aimed to build a review that could fill this void. The PubMed, EMBASE, COCHRANE, and Google Scholar databases were searched. The literature review showed that medicinal plants and phytochemicals regulated the Nrf2 on IBD and IBD-associated colorectal cancer by amplifying the expression of the Nrf2-mediated phase II detoxifying enzymes and diminishing NF-κB-related inflammation. These effects improve the bowel environment, mucosal barrier, colon, and crypt disruption, reduce ulceration and microbial translocation, and consequently, reduce the disease activity index (DAI). Moreover, the modulation of Nrf2 can regulate various genes involved in cellular redox, protein degradation, DNA repair, xenobiotic metabolism, and apoptosis, contributing to the prevention of colorectal cancer.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Avenida Monte Carmelo, 800, Marília 17519-030, São Paulo, Brazil
| | - Mariana Canevari de Maio
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Avenida Monte Carmelo, 800, Marília 17519-030, São Paulo, Brazil
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Natália de Góes Corrêa
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, São Paulo, Brazil
| | - Karina Quesada
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, São Paulo, Brazil
| | | | - Claudia R. P. Detregiachi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
29
|
Xue JC, Yuan S, Meng H, Hou XT, Li J, Zhang HM, Chen LL, Zhang CH, Zhang QG. The role and mechanism of flavonoid herbal natural products in ulcerative colitis. Biomed Pharmacother 2023; 158:114086. [PMID: 36502751 DOI: 10.1016/j.biopha.2022.114086] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine that presents clinically with abdominal pain, mucopurulent stools, and posterior urgency. The lesions of UC are mainly concentrated in the rectal and colonic mucosa and submucosa. For patients with mild to moderate UC, the best pharmacological treatment includes glucocorticoids, immunosuppressants, antibiotics, and biologics, but the long-term application can have serious toxic side effects. Currently, nearly 40% of UC patients are treated with herbal natural products in combination with traditional medications to reduce the incidence of toxic side effects. Flavonoid herbal natural products are the most widely distributed polyphenols in plants and fruits, which have certain antioxidant and anti-inflammatory activities. Flavonoid herbal natural products have achieved remarkable efficacy in the treatment of UC. The pharmacological mechanisms are related to anti-inflammation, promotion of mucosal healing, maintenance of intestinal immune homeostasis, and regulation of intestinal flora. In this paper, we summarize the flavonoid components of anti-ulcerative colitis and their mechanisms reported in the past 10 years, to provide a basis for rational clinical use and the development of new anti-ulcerative colitis drugs.
Collapse
Affiliation(s)
- Jia-Chen Xue
- Department of Immunology and Pathogenic Biology, Yanbian University College of Basic Medicine, Yanji, Jilin Province 133002, China
| | - Shuo Yuan
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning Province 116622, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Huan Meng
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning Province 116622, China
| | - Xiao-Ting Hou
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning Province 116622, China
| | - Jiao Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning Province 116622, China
| | - Hua-Min Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning Province 116622, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Li-Li Chen
- Jinan People's Hospital, Jinan, Shandong Province 271100, China
| | - Cheng-Hao Zhang
- Department of Oral Teaching and Research, Yanbian University, Yanji, Jilin Province 133000, China.
| | - Qing-Gao Zhang
- Department of Immunology and Pathogenic Biology, Yanbian University College of Basic Medicine, Yanji, Jilin Province 133002, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning Province 116622, China.
| |
Collapse
|
30
|
Li J, Duan Q, Wei X, Wu J, Yang Q. Kidney-Targeted Nanoparticles Loaded with the Natural Antioxidant Rosmarinic Acid for Acute Kidney Injury Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204388. [PMID: 36253133 DOI: 10.1002/smll.202204388] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/24/2022] [Indexed: 06/16/2023]
Abstract
Acute kidney injury (AKI) is a common clinical disease with high morbidity and mortality, and with a lack of effective drugs for treatment. Oxidative stress is very important in the occurrence and progression of AKI, and antioxidants use is one of the promising treatments. Rosmarinic acid (RA) is a ubiquitous natural polyphenol with powerful antioxidant and anti-inflammatory activities. Due to its inherent characteristic with poor water solubility and inferior bioavailability, its clinical application is impeded. Hence, the authors design a nanoparticle for effectively delivering RA, which is a chemical complex of RA and fourth-generation poly-amidoamine-based amphiphilic polymer (G4-PAMAM). The nanoparticle is modified with l-serine due to the specific interaction between kidney injury molecule-1 (Kim-1) and serine, which eventually generates a promising AKI kidney-targeting nanoparticle (S-G-R). The S-G-R is rapidly cumulated and long-term retained in ischemia-reperfusion-induced AKI kidneys, especially in the damaged renal tubular cells. The S-G-R exhibits more excellent antioxidative and antiapoptotic effects in vitro and has a more outstanding ability to improve the renal function, repair damaged renal tissue, and decrease oxidative stress, inflammatory response and apoptosis of tubular cells in vivo. Overall, this study might develop a safe and effective targeting strategy for the therapy of AKI.
Collapse
Affiliation(s)
- Jiajia Li
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Qijia Duan
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xiaona Wei
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Jianping Wu
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Qiongqiong Yang
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
31
|
Cyclodextrin Derivatives as Promising Solubilizers to Enhance the Biological Activity of Rosmarinic Acid. Pharmaceutics 2022; 14:pharmaceutics14102098. [PMID: 36297533 PMCID: PMC9611598 DOI: 10.3390/pharmaceutics14102098] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Rosmarinic acid (RA) is a natural antioxidant with neuroprotective properties; however, its preventive and therapeutic use is limited due to its slight solubility and poor permeability. This study aimed to improve RA physicochemical properties by systems formation with cyclodextrins (CDs): hydroxypropyl-α-CD (HP-α-CD), HP-β-CD, and HP-γ-CD, which were prepared by the solvent evaporation (s.e.) method. The interactions between components were determined by X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC) and Fourier Transform infrared spectroscopy (FTIR). The sites of interaction between RA and CDs were suggested as a result of in silico studies focused on assessing the interaction between molecules. The impact of amorphous systems formation on water solubility, dissolution rate, gastrointestinal (GIT) permeability, and biological activity was studied. RA solubility was increased from 5.869 mg/mL to 113.027 mg/mL, 179.840 mg/mL, and 194.354 mg/mL by systems formation with HP-α-CD, HP-β-CD, and HP-γ-CD, respectively. During apparent solubility studies, the systems provided an acceleration of RA dissolution. Poor RA GIT permeability at pH 4.5 and 5.8, determined by parallel artificial membrane permeability assay (PAMPA system), was increased; RA–HP-γ-CD s.e. indicated the greatest improvement (at pH 4.5 from Papp 6.901 × 10−7 cm/s to 1.085 × 10−6 cm/s and at pH 5.8 from 5.019 × 10−7 cm/s to 9.680 × 10−7 cm/s). Antioxidant activity, which was determined by DPPH, ABTS, CUPRAC, and FRAP methods, was ameliorated by systems; the greatest results were obtained for RA–HP-γ-CD s.e. The inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) was increased from 36.876% for AChE and 13.68% for BChE to a maximum inhibition of the enzyme (plateau), and enabled reaching IC50 values for both enzymes by all systems. CDs are efficient excipients for improving RA physicochemical and biological properties. HP-γ-CD was the greatest one with potential for future food or dietary supplement applications.
Collapse
|
32
|
Chen L, Liu D, Mao M, Liu W, Wang Y, Liang Y, Cao W, Zhong X. Betaine ameliorates acute sever ulcerative colitis by inhibiting oxidative stress induced inflammatory pyroptosis. Mol Nutr Food Res 2022; 66:e2200341. [PMID: 36069237 DOI: 10.1002/mnfr.202200341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/13/2022] [Indexed: 11/10/2022]
Abstract
SCOPE Betaine rich in beet is used as an important source of human nutrition. Here we aimed to explore whether betaine supplementation can protect against acute sever ulcerative colitis (ASUC) and the underlying mechanism METHODS AND RESULTS: : ASUC model was induced by dextran sulfate sodium (DSS), and effects of betaine as a methyl donor on ASUC were evaluated. Betaine mitigated the changes, e.g., elevated DAI, weight lose, spleen enlargement, colon shortening and disordered colonic mucosa. We then verified the protective effects of betaine on colonic barrier integrity in ASUC through examining tight junction proteins by western blot and immunofluorescence. Spectrophotometry method and western blot confirmed that betaine can decrease levels of oxidative markers (MDA, MPO, NOS and COX2), and promote expressions of antioxidant proteins (GSH, NRF2, CAT and SOD1). Further, betaine prevented colonic inflammatory pyroptosis by blocking expressions of NLRP3 inflammasome complex (NLRP3, ASC and cleaved-caspase 1), N terminal-GSDMD, and release of relevant inflammatory factors. CONCLUSION Betaine inhibits colonic oxidative stress induced inflammatory pyroptosis to alleviate ASUC, which shows therapeutic potential against colitis and other acute inflammatory disorder. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ling Chen
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Dandan Liu
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Mingli Mao
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wenjia Liu
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yajuan Wang
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yue Liang
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xiaolin Zhong
- The First Affiliated Hospital of University of South China, Department of Endocrinology and Metabolism, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
33
|
Xiao J, Tian W, Abdullah, Wang H, Chen M, Huang Q, Zhang M, Lu M, Song M, Cao Y. Updated design strategies for oral delivery systems: maximized bioefficacy of dietary bioactive compounds achieved by inducing proper digestive fate and sensory attributes. Crit Rev Food Sci Nutr 2022; 64:817-836. [PMID: 35959723 DOI: 10.1080/10408398.2022.2109583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Interest in the application of dietary bioactive compounds (DBC) in healthcare and pharmaceutical industries has motivated researchers to develop functional delivery systems (FDS) aiming to maximize their bioefficacy. As the direct and indirect health benefiting effects of DBC are acknowledged, traditional design principle of FDS aiming at improving the bioavailability of intact DBC is challenged by the updated one, where the maximized bioefficacy of DBC delivered by FDS will be achieved via rationally absorbed at target sites with proper metabolism pathways. This article briefly summarized the absorption and metabolic fates of orally digested DBC along with their direct and indirect mechanisms to perform health benefiting effects. Current strategies in designing the next generation FDS with an emphasis on their modulation effects on the distribution portion between the upper and lower digestive tract, portal vein and lymphatic absorption, human digestive and gut microbiota enzymatic mediated metabolism were highlighted. Updated research progresses of FDS in adjusting sensory attributes of food end products and inducing synergistic effects rooting from matrix materials and co-delivered cargos were also discussed. Challenges as well as future perspectives concerning the precise nutrition and the critical role of delivery systems in dietary intervention were proposed.
Collapse
Affiliation(s)
- Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Wenni Tian
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Abdullah
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Haonan Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Meimiao Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Qingrong Huang
- Department of Food Science, Rutgers, the State University of New Jersey, New Jersey, New Brunswick, USA
| | - Man Zhang
- Department of Food Science, Rutgers, the State University of New Jersey, New Jersey, New Brunswick, USA
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
34
|
Yao Y, Li R, Liu D, Long L, He N. Rosmarinic acid alleviates acetaminophen-induced hepatotoxicity by targeting Nrf2 and NEK7-NLRP3 signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113773. [PMID: 35753269 DOI: 10.1016/j.ecoenv.2022.113773] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/02/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
Rosmarinic acid (RA) is a natural polyphenol with various biological activities, such as anti-oxidative, anti-fibrotic, and hepatoprotective properties. The objective of this study was to investigate the protective effect of RA against acetaminophen (APAP)-induced hepatotoxicity (AILI) and explore the underlying mechanisms. Kunming mice were treated with RA (20, 40, or 80 mg/kg, i.g) for 7d, followed by an intraperitoneal injection of APAP (500 mg/kg). The liver injury was evaluated by serum biochemical and liver histopathological examinations. Human HepG2 cells were pre-treated with RA (20, 40, or 80 μmol/L) and then incubated with APAP (25 mmol/L) for 24 h. The MTT assay, wound healing assay, transwell migration assay, flow cytometry, and western blotting were employed to further evaluate RA's protective effects on AILI and explore the mechanisms. The results indicated that RA pre-treatment lowered the serum ALT and AST levels, ameliorated the histological damage to the liver, and reduced ROS generation and the production of IL-1β and IL-18 in the liver tissues in APAP-treated mice. Moreover, pre-treatment with RA could promote the cell viability and migration ability and inhibit apoptosis in APAP-treated HepG2 cells. Mechanistically, RA could significantly suppress the APAP-induced activation of the NEK7-NLRP3 signaling pathway. Notably, depletion of Nrf2 by short hairpin RNA (shRNA) partly eliminated the protective effects of RA on AILI and the suppression of NEK7-NLRP3 signaling by RA. In summary, these results indicate that RA has a protective role against AILI through Nrf2-mediated inhibition of ROS production and suppression of the NEK7-NLRP3 pathway.
Collapse
Affiliation(s)
- Yang Yao
- Department of Central Laboratory, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, PR China.
| | - Rong Li
- Department of Central Laboratory, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, PR China
| | - Dan Liu
- Department of Immunology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, PR China
| | - Lihui Long
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, PR China
| | - Na He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, PR China
| |
Collapse
|
35
|
Chen S, Zhu H, Luo Y. Chitosan-based oral colon-specific delivery systems for polyphenols: recent advances and emerging trends. J Mater Chem B 2022; 10:7328-7348. [PMID: 35766297 DOI: 10.1039/d2tb00874b] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oral colon-targeted delivery systems (OCDSs) have attracted great attention in the delivery of active compounds targeted to the colon for the treatment of colon and non-colon diseases with the advantages of enhanced efficacy and reduced side effects. Chitosan, the second-most abundant biopolymer next to cellulose, has great biocompatibility, is non-toxic, is sensitive to colonic flora and shows strong adhesion to colonic mucus, making it an ideal biomaterial candidate for the construction of OCDSs. Being rich in functional groups, the chitosan structure is easily modified, both physically and chemically, for the fabrication of delivery systems with diverse geometries, including nanoparticles, microspheres/microparticles, and hydrogels, that are resistant to the harsh environment of the upper gastrointestinal tract (GIT). This review offers a detailed overview of the preparation of chitosan-based delivery systems as the basis for building OCDSs. A variety of natural polyphenols with potent biological activities are used to treat diseases of the colon, or to be metabolized as active ingredients by colonic microorganisms to intervene in remote organ diseases after absorption into the circulation. However, the poor solubility of polyphenols limits their application, and the acidic environment of the upper GIT and various enzymes in the small intestine disrupt their structure and activity. As a result, the development of OCDSs for polyphenols has become an emerging and popular area of current research in the past decade. Thus, the second objective of this review is to systematically summarize the most recent research findings in this area and shed light on the future development of chitosan-based OCDSs for nutritional and biomedical applications.
Collapse
Affiliation(s)
- Sunni Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Honglin Zhu
- Nanotechnology and Biodelivery Laboratory, Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | - Yangchao Luo
- Nanotechnology and Biodelivery Laboratory, Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
36
|
Noor S, Mohammad T, Rub MA, Raza A, Azum N, Yadav DK, Hassan MI, Asiri AM. Biomedical features and therapeutic potential of rosmarinic acid. Arch Pharm Res 2022; 45:205-228. [PMID: 35391712 PMCID: PMC8989115 DOI: 10.1007/s12272-022-01378-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/18/2022] [Indexed: 12/17/2022]
Abstract
For decades, the use of secondary metabolites of various herbs has been an attractive strategy in combating human diseases. Rosmarinic acid (RA) is a bioactive phenolic compound commonly found in plants of Lamiaceae and Boraginaceae families. RA is biosynthesized using amino acids tyrosine and phenylalanine via enzyme-catalyzed reactions. However, the chemical synthesis of RA involves an esterification reaction between caffeic acid and 3,4-dihydroxy phenyl lactic acid contributing two phenolic rings to the structure of RA. Several studies have ascertained multiple therapeutic benefits of RA in various diseases, including cancer, diabetes, inflammatory disorders, neurodegenerative disorders, and liver diseases. Many previous scientific papers indicate that RA can be used as an anti-plasmodic, anti-viral and anti-bacterial drug. In addition, due to its high anti-oxidant capacity, this natural polyphenol has recently gained attention for its possible application as a nutraceutical compound in the food industry. Here we provide state-of-the-art, flexible therapeutic potential and biomedical features of RA, its implications and multiple uses. Along with various valuable applications in safeguarding human health, this review further summarizes the therapeutic advantages of RA in various human diseases, including cancer, diabetes, neurodegenerative diseases. Furthermore, the challenges associated with the clinical applicability of RA have also been discussed.
Collapse
Affiliation(s)
- Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Malik Abdul Rub
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ali Raza
- Department of Medical Biochemistry, Jawahar Lal Nehru Medical College, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Naved Azum
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsugu, Incheon, 21924, Korea.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| | - Abdullah M Asiri
- Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
37
|
da Silva GB, Yamauchi MA, Zanini D, Bagatini MD. Novel possibility for cutaneous melanoma treatment by means of rosmarinic acid action on purinergic signaling. Purinergic Signal 2022; 18:61-81. [PMID: 34741236 PMCID: PMC8570242 DOI: 10.1007/s11302-021-09821-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer cases have increased significantly in Brazil and worldwide, with cutaneous melanoma (CM) being responsible for nearly 57,000 deaths in the world. Thus, this review article aims at exploring and proposed hypotheses with respect to the possibility that RA can be a promising and alternative compound to be used as an adjuvant in melanoma treatment, acting on purinergic signaling. The scarcity of articles evidencing the action of this compound in this signaling pathway requires further studies. Considering diverse evidence found in the literature, we hypothesize that RA can be an effective candidate for the treatment of CM acting as a modulating molecule of purinergic cellular pathway through P2X7 blocking, mitigating the Warburg effect, and as antagonic molecule of the P2Y12 receptor, reducing the formation of adhesive molecules that prevent adherence in tumor cells. In this way, our proposals for CM treatment based on targeting purinergic signaling permeate the integral practice, going from intracell to extracell. Undoubtedly, much is still to be discovered and elucidated about this promising compound, this paper being an interesting work baseline to support more research studies.
Collapse
Affiliation(s)
- Gilnei Bruno da Silva
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Milena Ayumi Yamauchi
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Daniela Zanini
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Margarete Dulce Bagatini
- Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Fronteira Sul, Chapecó, SC, 89815-899, Brazil.
| |
Collapse
|
38
|
Advancements in Fabrication and Application of Chitosan Composites in Implants and Dentistry: A Review. Biomolecules 2022; 12:biom12020155. [PMID: 35204654 PMCID: PMC8961661 DOI: 10.3390/biom12020155] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 02/05/2023] Open
Abstract
Chitosan is a biopolymer that is found in nature and is produced from chitin deacetylation. Chitosan has been studied thoroughly for multiple applications with an interdisciplinary approach. Antifungal antibacterial activities, mucoadhesion, non-toxicity, biodegradability, and biocompatibility are some of the unique characteristics of chitosan-based biomaterials. Moreover, chitosan is the only widely-used natural polysaccharide, and it is possible to chemically modify it for different applications and functions. In various fields, chitosan composite and compound manufacturing has acquired much interest in developing several promising products. Chitosan and its derivatives have gained attention universally in biomedical and pharmaceutical industries as a result of their desired characteristics. In the present mini-review, novel methods for preparing chitosan-containing materials for dental and implant engineering applications along with challenges and future perspectives are discussed.
Collapse
|
39
|
Koprivica I, Jonić N, Diamantis D, Gajić D, Saksida T, Pejnović N, Tzakos AG, Stojanović I. Phenethyl ester of rosmarinic acid attenuates autoimmune responses during type 1 diabetes development in mice. Life Sci 2022; 288:120184. [PMID: 34838848 DOI: 10.1016/j.lfs.2021.120184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022]
Abstract
AIMS Rosmarinic acid (RA) is a polyphenol that occurs in plants of the Lamiaceae family. Phenethyl ester of RA (PERA), a novel RA derivative, has been developed and evaluated in vivo in an animal model of type 1 diabetes (T1D). METHODS T1D was induced in male C57BL/6 mice using multiple low doses of streptozotocin (STZ) administered intraperitoneally for 5 consecutive days. Intraperitoneal administration of PERA (2.5 mg/kg bw) began from the first STZ injection and continued for 20 days. KEY FINDINGS PERA-treated mice exhibited lower incidence of T1D (monitored up to 38 days from the disease induction), and fluorescent histochemical analysis showed that their pancreatic islets expressed more insulin. PERA treatment significantly down-regulated the proportions of CD11b+ and CD11c+ myeloid cells in the immune cell infiltrates in the pancreatic islets early during T1D pathogenesis (on day 9 after T1D induction), while on day 15, PERA significantly reduced the proportions of CD11c+, CD8+, Th1 and Th17 cells. Simultaneously, it was found that the cells from the pancreatic infiltrates of PERA-treated mice produced significantly less reactive oxygen species than cells from the control group. SIGNIFICANCE These findings suggest that PERA efficiently prevented T1D development in mice. Interestingly, PERA attenuated the inflammatory process in the islets through temporally specific interference with the innate and adaptive immune response and therefore shows great promise for further clinical evaluation as a novel T1D therapeutic.
Collapse
Affiliation(s)
- Ivan Koprivica
- Institute for Biological Research "Siniša Stanković" National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Natalija Jonić
- Institute for Biological Research "Siniša Stanković" National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dimitris Diamantis
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Dragica Gajić
- Institute for Biological Research "Siniša Stanković" National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Institute for Biological Research "Siniša Stanković" National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nada Pejnović
- Institute for Biological Research "Siniša Stanković" National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Andreas G Tzakos
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Ivana Stojanović
- Institute for Biological Research "Siniša Stanković" National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
40
|
Liu P, Li Y, Wang R, Ren F, Wang X. Oxidative Stress and Antioxidant Nanotherapeutic Approaches for Inflammatory Bowel Disease. Biomedicines 2021; 10:biomedicines10010085. [PMID: 35052764 PMCID: PMC8773244 DOI: 10.3390/biomedicines10010085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress, caused by the accumulation of reactive species, is associated with the initiation and progress of inflammatory bowel disease (IBD). The investigation of antioxidants to target overexpressed reactive species and modulate oxidant stress pathways becomes an important therapeutic option. Nowadays, antioxidative nanotechnology has emerged as a novel strategy. The nanocarriers have shown many advantages in comparison with conventional antioxidants, owing to their on-site accumulation, stability of antioxidants, and most importantly, intrinsic multiple reactive species scavenging or catalyzing properties. This review concludes an up-to-date summary of IBD nanomedicines according to the classification of the delivered antioxidants. Moreover, the concerns and future perspectives in this study field are also discussed.
Collapse
Affiliation(s)
- Ping Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (P.L.); (Y.L.); (R.W.); (F.R.)
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (P.L.); (Y.L.); (R.W.); (F.R.)
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (P.L.); (Y.L.); (R.W.); (F.R.)
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (P.L.); (Y.L.); (R.W.); (F.R.)
| | - Xiaoyu Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (P.L.); (Y.L.); (R.W.); (F.R.)
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Correspondence: ; Tel.: +86-010-62738589
| |
Collapse
|
41
|
Boldura OM, Marc S, Otava G, Hutu I, Balta C, Tulcan C, Mircu C. Utilization of Rosmarinic and Ascorbic Acids for Maturation Culture Media in Order to Increase Sow Oocyte Quality Prior to IVF. Molecules 2021; 26:7215. [PMID: 34885797 PMCID: PMC8659116 DOI: 10.3390/molecules26237215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 11/25/2022] Open
Abstract
The beneficial effect of antioxidant supplementation in maturation culture media of sow oocytes was evaluated by the expression quantification of apoptotic genes and the genes that ensure stability of germ cells during fertilization. The oocytes were cultivated for 44 h in conventional medium (C) or in medium supplemented with 105 µM rosmarinic acid (R) and 0.5 mM ascorbic acid (A) and classified into three quality classes by morphological observation from which the total RNA was isolated. The gene expression of Ptx3 and the apoptotic regulator p53, Bax and BCL-2 were evaluated by quantitative PCR technique. The decreased expression of the Bax gene in the A and R groups, compared to the control, indicates a protective role of antioxidants in the cells. Cell homeostasis was maintained, as reflected in the ratio of Bax/Bcl-2 in class I COCs (cumulus-oocyte complex) regardless of the experimental group, indicating minimum cellular stress. The expression of p53 genes was higher in all class III COC, but in A1 and R1 the expression was lower than in C1, and a similar Ptx-3 gene decreased significantly in groups A1, A2, A3 and R1 compared with control groups. Antioxidant supplementation showed beneficial effects on all morphological classes of pig COCs.
Collapse
Affiliation(s)
- Oana-Maria Boldura
- Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I” Timisoara, 300645 Timişoara, Romania; (O.-M.B.); (S.M.); (G.O.); (I.H.); (C.M.)
- BUASVM’s Research Institute for Biosecurity and Bioengineering, University of Agricultural Sciences and Veterinary Medicine ”King Michael I of Romania” from Timisoara, 300645 Timişoara, Romania
| | - Simona Marc
- Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I” Timisoara, 300645 Timişoara, Romania; (O.-M.B.); (S.M.); (G.O.); (I.H.); (C.M.)
- BUASVM’s Research Institute for Biosecurity and Bioengineering, University of Agricultural Sciences and Veterinary Medicine ”King Michael I of Romania” from Timisoara, 300645 Timişoara, Romania
| | - Gabriel Otava
- Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I” Timisoara, 300645 Timişoara, Romania; (O.-M.B.); (S.M.); (G.O.); (I.H.); (C.M.)
- BUASVM’s Research Institute for Biosecurity and Bioengineering, University of Agricultural Sciences and Veterinary Medicine ”King Michael I of Romania” from Timisoara, 300645 Timişoara, Romania
| | - Ioan Hutu
- Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I” Timisoara, 300645 Timişoara, Romania; (O.-M.B.); (S.M.); (G.O.); (I.H.); (C.M.)
- BUASVM’s Research Institute for Biosecurity and Bioengineering, University of Agricultural Sciences and Veterinary Medicine ”King Michael I of Romania” from Timisoara, 300645 Timişoara, Romania
| | - Cornel Balta
- Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Camelia Tulcan
- Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I” Timisoara, 300645 Timişoara, Romania; (O.-M.B.); (S.M.); (G.O.); (I.H.); (C.M.)
- BUASVM’s Research Institute for Biosecurity and Bioengineering, University of Agricultural Sciences and Veterinary Medicine ”King Michael I of Romania” from Timisoara, 300645 Timişoara, Romania
| | - Calin Mircu
- Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I” Timisoara, 300645 Timişoara, Romania; (O.-M.B.); (S.M.); (G.O.); (I.H.); (C.M.)
- BUASVM’s Research Institute for Biosecurity and Bioengineering, University of Agricultural Sciences and Veterinary Medicine ”King Michael I of Romania” from Timisoara, 300645 Timişoara, Romania
| |
Collapse
|
42
|
Teixeira-Costa BE, Andrade CT. Chitosan as a Valuable Biomolecule from Seafood Industry Waste in the Design of Green Food Packaging. Biomolecules 2021; 11:1599. [PMID: 34827597 PMCID: PMC8615795 DOI: 10.3390/biom11111599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 12/21/2022] Open
Abstract
Chitosan is a versatile biomolecule with a broad range of applications in food and pharmaceutical products. It can be obtained by the alkaline deacetylation of chitin. This biomolecule can be extracted using conventional or green methods from seafood industry residues, e.g., shrimp shells. Chitin has limited applications because of its low solubility in organic solvents. Chitosan is soluble in acidified solutions allowing its application in the food industry. Furthermore, biological properties, such as antioxidant, antimicrobial, as well as its biodegradability, biocompatibility and nontoxicity have contributed to its increasing application as active food packaging. Nevertheless, some physical and mechanical features have limited a broader range of applications of chitosan-based films. Green approaches may be used to address these limitations, leading to well-designed chitosan-based food packaging, by employing principles of a circular and sustainable economy. In this review, we summarize the properties of chitosan and present a novel green technology as an alternative to conventional chitin extraction and to design environmentally friendly food packaging based on chitosan.
Collapse
Affiliation(s)
- Barbara E. Teixeira-Costa
- Programa de Pós-Graduação em Ciência de Alimentos, Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Moniz Aragão 360, Bloco 8G/CT2, Rio de Janeiro 21941-594, RJ, Brazil;
- Faculdade de Ciências Agrárias, Universidade Federal do Amazonas, Avenida General Rodrigo Otávio 6200, Manaus 69077-000, AM, Brazil
| | - Cristina T. Andrade
- Programa de Pós-Graduação em Ciência de Alimentos, Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Moniz Aragão 360, Bloco 8G/CT2, Rio de Janeiro 21941-594, RJ, Brazil;
| |
Collapse
|
43
|
Ahamad N, Kar A, Mehta S, Dewani M, Ravichandran V, Bhardwaj P, Sharma S, Banerjee R. Immunomodulatory nanosystems for treating inflammatory diseases. Biomaterials 2021; 274:120875. [PMID: 34010755 DOI: 10.1016/j.biomaterials.2021.120875] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/26/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023]
Abstract
Inflammatory disease (ID) is an umbrella term encompassing all illnesses involving chronic inflammation as the central manifestation of pathogenesis. These include, inflammatory bowel diseases, hepatitis, pulmonary disorders, atherosclerosis, myocardial infarction, pancreatitis, arthritis, periodontitis, psoriasis. The IDs create a severe burden on healthcare and significantly impact the global socio-economic balance. Unfortunately, the standard therapies that rely on a combination of anti-inflammatory and immunosuppressive agents are palliative and provide only short-term relief. In contrast, the emerging concept of immunomodulatory nanosystems (IMNs) has the potential to address the underlying causes and prevent reoccurrence, thereby, creating new opportunities for treating IDs. The IMNs offer exquisite ability to precisely modulate the immune system for a therapeutic advantage. The nano-sized dimension of IMNs allows them to efficiently infiltrate lymphatic drainage, interact with immune cells, and subsequently to undergo rapid endocytosis by hyperactive immune cells (HICs) at inflamed sites. Thus, IMNs serve to restore dysfunctional or HICs and alleviate the inflammation. We identified that different IMNs exert their immunomodulatory action via either of the seven mechanisms to modulate; cytokine production, cytokine neutralization, cellular infiltration, macrophage polarization, HICs growth inhibition, stimulating T-reg mediated tolerance and modulating oxidative-stress. In this article, we discussed representative examples of IMNs by highlighting their rationalization, design principle, and mechanism of action in context of treating various IDs. Lastly, we highlighted technical challenges in the application of IMNs and explored the future direction of research, which could potentially help to overcome those challenges.
Collapse
Affiliation(s)
- Nadim Ahamad
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Abhinanda Kar
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Sourabh Mehta
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India; IITB-Monash Research Academy IIT Bombay, Powai, Mumbai, 400076, India
| | - Mahima Dewani
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Vasanthan Ravichandran
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Prateek Bhardwaj
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shivam Sharma
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rinti Banerjee
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
44
|
Xu Q, Zhou X, Strober W, Mao L. Inflammasome Regulation: Therapeutic Potential for Inflammatory Bowel Disease. Molecules 2021; 26:molecules26061725. [PMID: 33808793 PMCID: PMC8003415 DOI: 10.3390/molecules26061725] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammasomes are multiprotein complexes formed to regulate the maturation of pro-inflammatory caspases, in response to intracellular or extracellular stimulants. Accumulating studies showed that the inflammasomes are implicated in the pathogenesis of inflammatory bowel disease (IBD), although their activation is not a decisive factor for the development of IBD. Inflammasomes and related cytokines play an important role in the maintenance of gut immune homeostasis, while its overactivation might induce excess immune responses and consequently cause tissue damage in the gut. Emerging studies provide evidence that some genetic abnormalities might induce enhanced NLRP3 inflammasome activation and cause colitis. In these cases, the colonic inflammation can be ameliorated by blocking NLRP3 activation or its downstream cytokine IL-1β. A number of natural products were shown to play a role in preventing colon inflammation in various experimental colitis models. On the other hand, lack of inflammasome function also causes intestinal abnormalities. Thus, an appropriate regulation of inflammasomes might be a promising therapeutic strategy for IBD intervention. This review aims at summarizing the main findings in these studies and provide an outline for further studies that might contribute to our understanding of the role of inflammasomes in the pathogenesis and therapeutic treatment of IBD.
Collapse
Affiliation(s)
- Qiuyun Xu
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Q.X.); (X.Z.)
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Q.X.); (X.Z.)
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence: (W.S.); (L.M.)
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226019, China; (Q.X.); (X.Z.)
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
- Correspondence: (W.S.); (L.M.)
| |
Collapse
|