1
|
Liu S, Wang Y, Ying L, Li H, Zhang K, Liang N, Luo G, Xiao L. Quercetin Mitigates Lysophosphatidylcholine (LPC)-Induced Neutrophil Extracellular Traps (NETs) Formation through Inhibiting the P2X7R/P38MAPK/NOX2 Pathway. Int J Mol Sci 2024; 25:9411. [PMID: 39273358 PMCID: PMC11395007 DOI: 10.3390/ijms25179411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are three-dimensional reticular structures that release chromatin and cellular contents extracellularly upon neutrophil activation. As a novel effector mechanism of neutrophils, NETs possess the capacity to amplify localized inflammation and have been demonstrated to contribute to the exacerbation of various inflammatory diseases, including cardiovascular diseases and tumors. It is suggested that lysophosphatidylcholine (LPC), as the primary active component of oxidized low-density lipoprotein, represents a significant risk factor for various inflammatory diseases, such as cardiovascular diseases and neurodegenerative diseases. However, the specific mechanism of NETs formation induced by LPC remains unclear. Quercetin has garnered considerable attention due to its anti-inflammatory properties, serving as a prevalent flavonoid in daily diet. However, little is currently known about the underlying mechanisms by which quercetin inhibits NETs formation and alleviates associated diseases. In our study, we utilized LPC-treated primary rat neutrophils to establish an in vitro model of NETs formation, which was subsequently subjected to treatment with a combination of quercetin or relevant inhibitors/activators. Compared to the control group, the markers of NETs and the expression of P2X7R/P38MAPK/NOX2 pathway-associated proteins were significantly increased in cells treated with LPC alone. Quercetin intervention decreased the LPC-induced upregulation of the P2X7R/P38MAPK/NOX2 pathway and effectively reduced the expression of NETs markers. The results obtained using a P2X7R antagonist/activator and P38MAPK inhibitor/activator support these findings. In summary, quercetin reversed the upregulation of the LPC-induced P2X7R/P38MAPK/NOX2 pathway, further mitigating NETs formation. Our study investigated the potential mechanism of LPC-induced NETs formation, elucidated the inhibitory effect of quercetin on NETs formation, and offered new insights into the anti-inflammatory properties of quercetin.
Collapse
Affiliation(s)
- Si Liu
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Yan Wang
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Linyao Ying
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Hao Li
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Keyi Zhang
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Na Liang
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Gang Luo
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Lin Xiao
- Xiangya School of Public Health, Central South University, Changsha 410013, China
| |
Collapse
|
2
|
Jeong SY, Choi WS, Kwon OS, Lee JS, Son SY, Lee CH, Lee S, Song JY, Lee YJ, Lee JY. Extract of Pinus densiflora needles suppresses acute inflammation by regulating inflammatory mediators in RAW264.7 macrophages and mice. PHARMACEUTICAL BIOLOGY 2022; 60:1148-1159. [PMID: 35695008 PMCID: PMC9196672 DOI: 10.1080/13880209.2022.2079679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/25/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Pinus densiflora Siebold & Zucc. (Pinaceae) needle extracts ameliorate oxidative stress, but research into their anti-inflammatory effects is limited. OBJECTIVE To investigate antioxidant and anti-inflammatory effects of a Pinus densiflora needles (PINE) ethanol extract in vitro and in vivo. MATERIALS AND METHODS We measured levels of reactive oxygen species (ROS), superoxide dismutase (SOD) and inflammatory mediators in lipopolysaccharide (LPS)-stimulated RAW264.7 cells at various PINE concentrations (25, 50 and 100 μg/mL; but 6.25, 12.5 and 25 μg/mL for interleukin-1β and prostaglandin E2 (PGE2)). Thirty ICR mice were randomized to six groups: vehicle, control, PINE pre-treatment (0.1, 0.3 and 1 mg/left ear for 10 min followed by arachidonic acid treatment for 30 min) and dexamethasone. The posttreatment ear thickness and myeloperoxidase (MPO) activity were measured. RESULTS PINE 100 μg/mL significantly decreased ROS (IC50, 70.93 μg/mL, p < 0.01), SOD (IC50, 30.99 μg/mL, p < 0.05), malondialdehyde (p < 0.01), nitric oxide (NO) (IC50, 27.44 μg/mL, p < 0.01) and tumour necrosis factor-alpha (p < 0.05) levels. Interleukin-1β (p < 0.05) and PGE2 (p < 0.01) release decreased significantly with 25 μg/mL PINE. PINE 1 mg/ear inhibited LPS-stimulated expression of cyclooxygenase-2 and inducible NO synthase in RAW264.7 macrophages and significantly inhibited ear oedema (36.73-15.04% compared to the control, p < 0.01) and MPO activity (167.94-105.59%, p < 0.05). DISCUSSION AND CONCLUSIONS PINE exerts antioxidant and anti-inflammatory effects by inhibiting the production of inflammatory mediators. Identified flavonoids such as taxifolin and quercetin glucoside can be attributed to effect of PINE.
Collapse
Affiliation(s)
- Seul-Yong Jeong
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Won Seok Choi
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Oh Seong Kwon
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Jong Seok Lee
- National Institute of Biological Resources, Incheon, Republic of Korea
| | - Su Young Son
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
- Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul, Republic of Korea
| | - Sarah Lee
- National Institute of Biological Resources, Incheon, Republic of Korea
| | - Jin Yong Song
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Yeon Jin Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Ji-Yun Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Recombinant Human Annexin A5 Alleviated Traumatic-Brain-Injury Induced Intestinal Injury by Regulating the Nrf2/HO-1/HMGB1 Pathway. Molecules 2022; 27:molecules27185755. [PMID: 36144494 PMCID: PMC9501944 DOI: 10.3390/molecules27185755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/26/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Aims: Annexin A5 (ANXA5) exhibited potent antithrombotic, antiapoptotic, and anti-inflammatory properties in a previous study. The role of ANXA5 in traumatic brain injury (TBI)-induced intestinal injury is not fully known. Main methods: Recombinant human ANXA5 (50 µg/kg) or vehicle (PBS) was administered to mice via the tail vein 30 min after TBI. Mouse intestine tissue was gathered for hematoxylin and eosin staining 0.5 d, 1 d, 2 d, and 7 d after modeling. Intestinal Western blotting, immunofluorescence, TdT-mediated dUTP nick-end labeling staining, and enzyme-linked immunosorbent assays were performed 2 days after TBI. A series of kits were used to assess lipid peroxide indicators such as malonaldehyde, superoxide dismutase activity, and catalase activity. Key findings: ANXA5 treatment improved the TBI-induced intestinal mucosa injury at different timepoints and significantly increased the body weight. It significantly reduced apoptosis and matrix metalloproteinase-9 and inhibited the degradation of tight-junction-associated protein in the small intestine. ANXA5 treatment improved intestinal inflammation by regulating inflammation-associated factors. It also mitigated the lipid peroxidation products 4-HNE, 8-OHDG, and malonaldehyde, and enhanced the activity of the antioxidant enzymes, superoxide dismutase and catalase. Lastly, ANXA5 significantly enhanced nuclear factor E2-related factor 2 (Nrf2) and hemeoxygenase-1, and decreased high mobility group box 1 (HMGB1). Significance: Collectively, the results suggest that ANXA5 inhibits TBI-induced intestinal injury by restraining oxidative stress and inflammatory responses. The mechanisms involved sparking the Nrf2/hemeoxygenase-1-induced antioxidant system and suppressing the HMGB1 pathway. ANXA5 may be an attractive therapeutic candidate for protecting against TBI-induced intestinal injury.
Collapse
|
4
|
Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-κB Signaling Pathway. Int J Mol Sci 2022; 23:ijms23136939. [PMID: 35805952 PMCID: PMC9266441 DOI: 10.3390/ijms23136939] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/05/2023] Open
Abstract
TLR4/NF-κB is a key inflammatory signaling transduction pathway, closely involved in cell differentiation, proliferation, apoptosis, and pro-inflammatory response. Toll like receptor 4 (TLR4), the first mammalian TLR to be characterized, is the innate immune receptor that plays a key role in inflammatory signal transductions. Nuclear factor kappa B (NF-κB), the TLR4 downstream, is the key to accounting for the expression of multiple genes involved in inflammatory responses, such as pro-inflammatory cytokines. Inflammatory bowel disease (IBD) in humans is a chronic inflammatory disease with high incidence and prevalence worldwide. Targeting the TLR4/NF-κB signaling pathway might be an effective strategy to alleviate intestinal inflammation. Polyphenol phytochemicals have shown noticeable alleviative effects by acting on the TLR4/NF-κB signaling pathway in intestinal inflammation. This review summarizes the pharmacological effects of more than 20 kinds of polyphenols on intestinal inflammation via targeting the TLR4/NF-κB signaling pathway. We expected that polyphenol phytochemicals targeting the TLR4/NF-κB signaling pathway might be an effective approach to treat IBD in future clinical research applications.
Collapse
|
5
|
Feng YD, Ye W, Tian W, Meng JR, Zhang M, Sun Y, Zhang HN, Wang SJ, Wu KH, Liu CX, Liu SY, Cao W, Li XQ. Old targets, new strategy: Apigenin-7-O-β-d-(-6″-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through HO-1 and MAO-B inhibition. Free Radic Biol Med 2022; 184:74-88. [PMID: 35398494 DOI: 10.1016/j.freeradbiomed.2022.03.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022]
Abstract
With the increasing morbidity and mortality, intestinal ischemia/reperfusion injury (IIRI) has attracted more and more attention, but there is no efficient therapeutics at present. Apigenin-7-O-β-D-(-6″-p-coumaroyl)-glucopyranoside (APG) is a new flavonoid glycoside isolated from Clematis tangutica that has strong antioxidant abilities in previous studies. However, the pharmacodynamic function and mechanism of APG on IIRI remain unknown. This study aimed to investigate the effects of APG on IIRI both in vivo and in vitro and identify the potential molecular mechanism. We found that APG could significantly improve intestinal edema and increase Chiu's score. MST analysis suggested that APG could specifically bind to heme oxygenase 1 (HO-1) and monoamine oxidase b (MAO-B). Simultaneously, APG could attenuate ROS generation and Fe2+ accumulation, maintain mitochondria function thus inhibit ferroptosis with a dose-dependent manner. Moreover, we used siRNA silencing technology to confirm that knocking down both HO-1 and MAO-B had a positive effect on intestine. In addition, we found the HO-1 and MAO-B inhibitors also could reduce endothelial cell loss and protect vascular endothelial after reperfusion. We demonstrate that APG plays a protective role on decreasing activation of HO-1 and MAO-B, attenuating IIRI-induced ROS generation and Fe2+ accumulation, maintaining mitochondria function thus inhibiting ferroptosis.
Collapse
Affiliation(s)
- Ying-Da Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Ye
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Jing-Ru Meng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Meng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Hui-Nan Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shou-Jia Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Ke-Han Wu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chen-Xu Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shao-Yuan Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
6
|
Lucena Périco L, de Cássia Dos Santos R, Peixoto Rodrigues V, Vasti Alfieri Nunes V, Vilegas W, Machado da Rocha LR, Dos Santos C, Hiruma-Lima CA. Role of the antioxidant pathway in the healing of peptic ulcers induced by ischemia-reperfusion in male and female rats treated with Eugenia punicifolia. Inflammopharmacology 2022; 30:1383-1394. [PMID: 35445989 DOI: 10.1007/s10787-022-00946-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/14/2022] [Indexed: 11/09/2022]
Abstract
Ischaemia and reperfusion (I/R)-induced gastrointestinal disorders are caused by free radicals, resulting in organ damage and functional disarrangement. This study aimed to investigate the healing effects of hydroalcoholic extracts from the leaves of Eugenia punicifolia (Kunth) DC. (HEEP) in male and female Wistar rats with I/R-induced peptic injuries, and the role of antioxidants in improving this response. After I/R-induced gastric and duodenal injuries, male and female [intact (INT) and ovariectomized (OVZ)] rats were orally treated with HEEP for 6 days. Biochemical analysis was used to determine the catalase (CAT), superoxide dismutase (SOD), and myeloperoxidase (MPO) activities, as well as malondialdehyde and reduced glutathione levels, to measure the gastric and duodenal healing process. Six days of HEEP treatment significantly decreased the I/R-induced gastric [male (73.68%), INT (52.83%), and OVZ (43.13%)] and duodenal damage [male (57.03%), INT (56.04%), and OVZ (54.83%)] in all groups. In OVZ rats, the healing effect of HEEP occurred because of the increased activity of SOD (2x) and CAT (1.16x) in the gastric mucosa. In the duodenal mucosa of INT rats, the extract reduced MPO (20.83%) activity. The 6-day HEEP treatment improved the healing of I/R-induced peptic ulcer injury, with the system acting differently in males and females. The antioxidant system is an important component of the HEEP activity during post-I/R mucosal recovery. This result revealed the importance of antioxidant compounds in minimizing the severity of I/R-related events.
Collapse
Affiliation(s)
- Larissa Lucena Périco
- Department of Structural and Functional Biology (Physiology), Biosciences Institute, UNESP-São Paulo State University, Botucatu, São Paulo, CEP 18618-689, Brazil. .,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Raquel de Cássia Dos Santos
- Laboratory of Pharmacology and Molecular Biology, São Francisco University, CEP 12916-900, Bragança Paulista, São Paulo, Brazil
| | - Vinícius Peixoto Rodrigues
- Department of Structural and Functional Biology (Physiology), Biosciences Institute, UNESP-São Paulo State University, Botucatu, São Paulo, CEP 18618-689, Brazil
| | - Vânia Vasti Alfieri Nunes
- Department of Structural and Functional Biology (Physiology), Biosciences Institute, UNESP-São Paulo State University, Botucatu, São Paulo, CEP 18618-689, Brazil
| | - Wagner Vilegas
- Biosciences Institute, UNESP-São Paulo State University, São Vicente, São Paulo, CEP 11330-900, Brazil
| | - Lúcia Regina Machado da Rocha
- Department of Structural and Functional Biology (Physiology), Biosciences Institute, UNESP-São Paulo State University, Botucatu, São Paulo, CEP 18618-689, Brazil
| | - Catarina Dos Santos
- Department of Biological Science, Faculty of Sciences and Languages, UNESP-São Paulo State University, Assis, São Paulo, CEP 19806-900, Brazil
| | - Clélia Akiko Hiruma-Lima
- Department of Structural and Functional Biology (Physiology), Biosciences Institute, UNESP-São Paulo State University, Botucatu, São Paulo, CEP 18618-689, Brazil
| |
Collapse
|
7
|
Itou H, Toyota R, Takeda M. Phytochemical quercetin alleviates hyperexcitability of trigeminal nociceptive neurons associated with inflammatory hyperalgesia comparable to NSAIDs. Mol Pain 2022; 18:17448069221108971. [PMID: 35734996 PMCID: PMC9234920 DOI: 10.1177/17448069221108971] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Quercetin is a flavonoid that is widely found in fruits and vegetables. Quercetin inhibits cyclooxygenase-2 and modulates voltage-gated ion channels, however, its effect on nociceptive neuron-associated inflammatory hyperalgesia remains unknown. The present study investigated under in vivo conditions whether systemic administration of quercetin attenuates the inflammation-induced hyperexcitability of trigeminal spinal nucleus caudalis (SpVc) neurons associated with mechanical hyperalgesia and compared its effect to the non-steroidal anti-inflammatory drug, diclofenac. Complete Freund's adjuvant was injected into the whisker pads of rats to induce inflammation, and then mechanical stimulation was applied to the orofacial area to assess the threshold of escape. The mechanical threshold was significantly lower in inflamed rats compared to uninjected naïve rats, and this lowered threshold returned to control levels 2 days after administration of quercetin or diclofenac. The mean discharge frequency of SpVc wide-dynamic range (WDR) neurons to both non-noxious and noxious mechanical stimuli in inflamed rats was significantly decreased after quercetin or diclofenac administration under combination of three anesthetic agents (medetomidine, midazolam and butorphanol). In addition, the increased mean spontaneous discharge of SpVc WDR neurons in inflamed rats significantly decreased after quercetin or diclofenac administration. Similarly, quercetin or diclofenac restored the expanded mean receptive field size in inflamed rats to control levels. In this study, the combination of three anesthetic agents did not result in any obvious "noxious pinch-evoked after discharges" in CFA inflamed day 2 rat as described previously in pentobarbital-anesthetized rats. Together, these results suggest that administration of quercetin attenuates inflammatory hyperalgesia associated with hyperexcitability of nociceptive SpVc WDR neurons via inhibition of the peripheral cyclooxygenase-2 signaling cascade and voltage-gated ion channels. These findings support the proposed potential of quercetin as a therapeutic agent in complementary alternative medicine strategies for preventing trigeminal inflammatory mechanical hyperalgesia.
Collapse
Affiliation(s)
- Haruka Itou
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, Kanagawa, Japan
| | - Ryou Toyota
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, Kanagawa, Japan
| | - Mamoru Takeda
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, Kanagawa, Japan
| |
Collapse
|
8
|
Li M, Zheng Z. Protective effect of parecoxib sodium against ischemia reperfusion‑induced intestinal injury. Mol Med Rep 2021; 24:776. [PMID: 34498709 PMCID: PMC8436217 DOI: 10.3892/mmr.2021.12416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/18/2021] [Indexed: 01/03/2023] Open
Abstract
Ischemia reperfusion (I/R)-induced intestinal injury is a pathophysiological process leading to oxidative stress and inflammatory responses, and revealing its underlying mechanisms is essential for developing therapeutic strategies. Cyclooxygenase (COX) has been reported to be involved in I/R injury. Parecoxib sodium, a selective inhibitor for COX-2, exerts protective effects, such as reducing I/R-induced injuries in the heart, kidney and brain. However, the potential role of parecoxib sodium in protecting the small intestine against I/R-induced injury has rarely been investigated. Therefore, the aim of the present study was to elucidate the effects and potential mechanisms of parecoxib sodium in I/R-induced intestinal injury. In total, 60 Sprague-Dawley rats were randomly divided into four groups: Control (sham operation) group, intestinal I/R group, 10 mg/kg parecoxib sodium-pre-treated I/R (I/R + Pare/10) group and the 20 mg/kg parecoxib sodium-pre-treated I/R (I/R + Pare/20) group. A regular I/R model was established to induce the intestinal injury in rats. Parecoxib sodium at 10 or 20 mg/kg was intraperitoneally administered into rats in both I/R + Pare groups once daily for 5 consecutive days prior to ischemia. Blood samples and small intestinal tissues were collected at 2 h after reperfusion. Changes in the levels of malondialdehyde, nitric oxide, interleukin (IL)-1β, IL-8, intercellular cell adhesion molecule-1 and IL-10, as well as the total antioxidant capacity were determined using ELISA, as were the activities of superoxidase dismutase and myeloperoxidase. Furthermore, the protein expression levels of total caspase-3, cleaved caspase-3, Bcl-2 and Bax were examined via western blot analysis. In addition, the daily survival rate post-reperfusion was examined for 7 days. It was revealed that parecoxib sodium increased the levels of antioxidants and suppressed the intestinal oxidative injury induced by I/R. Moreover, parecoxib sodium downregulated the expression levels of the proinflammatory factors, but upregulated the expression levels of anti-inflammatory factors. The results also demonstrated that parecoxib sodium attenuated I/R-induced apoptosis and increased the survival rate of rats. Thus, administration of parecoxib sodium prior to intestinal I/R attenuated intestinal injury and increased the rat survival rate by inhibiting I/R-induced inflammation, oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Mei Li
- Department of Anesthesiology, Huangyan Hospital Affiliated to Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, Zhejiang 318020, P.R. China
| | - Zhi Zheng
- Department of Anesthesiology, Huangyan Hospital Affiliated to Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, Zhejiang 318020, P.R. China
| |
Collapse
|
9
|
Aldhahrani A. Protective effects of guarana ( Paullinia cupana) against methotrexate-induced intestinal damage in mice. Food Sci Nutr 2021; 9:3397-3404. [PMID: 34262701 PMCID: PMC8269666 DOI: 10.1002/fsn3.2101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/31/2022] Open
Abstract
This study aimed to examine the effects of guarana (Paullinia cupana) on intestinal damage induced by MTX in mice. Mice were classified into four groups: control, MTX, guarana (Paullinia cupana), and guarana (Paullinia cupana) together with MTX. Total antioxidant capacity together with glutathione, superoxide dismutase, MDA, ALT, AST, myeloperoxidase, total protein and IL-1β were detected in the serum. Bax and Bcl2 expressions were detected in intestine together with histopathological examination and immunohistochemical examination of caspase-9. Intoxication with MTX inhibited antioxidant and promoted myeloperoxidase activity in experimental mouse models but pre-administration of guarana ameliorated this effect by inhibiting IL-1β. Real-time quantitative PCR (qRT-PCR) analysis found that MTX intoxication upregulated BAX expression, causing apoptosis, and downregulated Bcl2 expression. These were also brought under control following guarana pre-administration. Histological examination of intestine indicated hyperplasia and desquamation of superficial epithelium of villi in the MTX-administered group, as well as round cell infiltration in the lamina propria. Pre-administration of guarana protected against these effects. The MTX group showed that caspase-9 expression was upregulated, increasing immune-reactivity in comparison to the guarana experimental groups. These combined effects lead to the conclusion that guarana has a preventative or protective effect against MTX-induced oxidative stress in the intestinal tissue.
Collapse
Affiliation(s)
- Adil Aldhahrani
- Department of clinical laboratory sciencesTurabah University CollegeTaif UniversityTaifSaudi Arabia
| |
Collapse
|
10
|
Cui QK, Li H, Li Z, Li J, Song L. Study on the mechanism of the Modified Ginseng-Schisandra Decoction (MGSD) in the treatment of recurrent respiratory tract infection (RRTI) based on network pharmacology. Transl Pediatr 2021; 10:1701-1711. [PMID: 34295785 PMCID: PMC8261594 DOI: 10.21037/tp-21-240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/17/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate the mechanism of Modified Ginseng-Schisandra Decoction in the treatment of recurrent respiratory tract infection (RRTI) using network pharmacology. METHODS To screen the active ingredients of A Modified Ginseng-Schisandra Decoction, TCMSP, TCMID, Batman-TCM and PubChem database were applied. To predict the targets of active ingredients on RRTI, TCMSP, Pubmed, OMIM, Drug Bank, GAD and TTD database were used. The compounds-therapeutic target network was constructed with Cytoscape 3.7.2 software. The STRING database was used to construct a protein-protein interaction (PPI) network, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was used to identify potential signal pathways. RESULTS The 3 main active ingredients of Modified Ginseng-Schisandra Decoction obtained by screening were quercetin, kaempferol, and isoflavone; the main therapeutic targets were PTGS2, ESR1, AR, PPARG, NOS2, and others. Based on the PPI network, we found that the targets of Modified Ginseng-Schisandra Decoction were significantly enriched in (FDR <0.01) cancer pathway, tumor necrosis factor (TNF) signaling pathway, hypoxia-inducible factor (HIF-1) signaling pathway, and others. CONCLUSIONS Modified Ginseng-Schisandra Decoction can treat RRTI primarily through acting in the signal transduction of some key nodes of cancer pathway and TNF pathway. It exerts a direct or indirect influence on multiple signaling pathways, and has the characteristics of multicomponent, multitarget, and multichannel action.
Collapse
Affiliation(s)
- Qing-Ke Cui
- Department of Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Li
- Department of Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhan Li
- Department of Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Li
- Department of Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Liqi Song
- Jilin Province Jilin Hospital of Integrated Chinese and Western Medicine, Jilin, China
| |
Collapse
|
11
|
Gonçalves GFB, Silva MEM, Sampaio FJB, Pereira-Sampaio MA, de Souza DB. Quercetin as a nephroprotector after warm ischemia: histomorphometric evaluation in a rodent model. Int Braz J Urol 2021; 47:796-802. [PMID: 33848072 PMCID: PMC8321496 DOI: 10.1590/s1677-5538.ibju.2020.0358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/20/2020] [Indexed: 01/20/2023] Open
Abstract
Purpose: To quantitatively evaluate the possible long-term protective effects of quercetin during renal warm ischemia. Materials and Methods: Male rats were allocated into 4 groups: sham (S), sham quercetin (SQ), ischemia (I), and ischemia quercetin (IQ). Groups SQ and IQ received quercetin (50mg/kg) before and after surgery. Groups I and IQ had their left renal vessels clamped for 60 minutes. All animals were euthanized four weeks after the procedure, and serum urea and creatinine levels were measured. Renal weight and volume, cortex-non-cortex area ratio (C-NC), cortical volume (CV), glomerular volumetric density (Vv[glom]), volume-weighted glomerular volume (VWGV) and number of glomeruli per kidney (N[glom]) were evaluated by stereological methods. Results were considered statistically significant when p <0.05. Results: Serum urea levels in group I increased by 10.4% in relation to group S, but no differences were observed among the other groups. The C-NC of group I was lower than those of all other groups, and group IQ had similar results to sham groups. The Vv[glom] and N[glom] of group I were lower than those of group S (33.7% and 28.3%, respectively) and group IQ had no significant difference compared to the S group. Conclusions: Quercetin was effective as a nephroprotective agent in preventing the glomerular loss observed when the kidney was subjected to warm ischemia. This suggests that this flavonoid may be used preventively in kidney surgery, when warm ischemia is necessary, such as partial nephrectomy.
Collapse
Affiliation(s)
- Gabriela F Buys Gonçalves
- Unidade de Pesquisa Urogenital, Universidade do Estado do Rio de Janeiro - UERJ, Rio de Janeiro, RJ, Brasil
| | - Maria Eduarda M Silva
- Departamento de Medicina Veterinária, Fundação Educacional Serra dos Órgãos, Teresópolis, RJ, Brasil
| | - Francisco J B Sampaio
- Unidade de Pesquisa Urogenital, Universidade do Estado do Rio de Janeiro - UERJ, Rio de Janeiro, RJ, Brasil
| | - Marco A Pereira-Sampaio
- Unidade de Pesquisa Urogenital, Universidade do Estado do Rio de Janeiro - UERJ, Rio de Janeiro, RJ, Brasil.,Departamento de Morfologia, Universidade Federal Fluminense - UFF, Niterói, RJ, Brasil
| | - Diogo Benchimol de Souza
- Unidade de Pesquisa Urogenital, Universidade do Estado do Rio de Janeiro - UERJ, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
12
|
Mechanism of Action of Bu-Fei-Yi-Shen Formula in Treating Chronic Obstructive Pulmonary Disease Based on Network Pharmacology Analysis and Molecular Docking Validation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9105972. [PMID: 33313323 PMCID: PMC7718855 DOI: 10.1155/2020/9105972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
Objective To explore the mechanism of action of Bu-Fei-Yi-Shen formula (BFYSF) in treating chronic obstructive pulmonary disease (COPD) based on network pharmacology analysis and molecular docking validation. Methods First of all, the pharmacologically active ingredients and corresponding targets in BFYSF were mined by the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, the analysis platform, and literature review. Subsequently, the COPD-related targets (including the pathogenic targets and known therapeutic targets) were identified through the TTD, CTD, DisGeNet, and GeneCards databases. Thereafter, Cytoscape was employed to construct the candidate component-target network of BFYSF in the treatment of COPD. Moreover, the cytoHubba plug-in was utilized to calculate the topological parameters of nodes in the network; then, the core components and core targets of BFYSF in the treatment of COPD were extracted according to the degree value (greater than or equal to the median degree values for all nodes in the network) to construct the core network. Further, the Autodock vina software was adopted for molecular docking study on the core active ingredients and core targets, so as to verify the above-mentioned network pharmacology analysis results. Finally, the Omicshare database was applied in enrichment analysis of the biological functions of core targets and the involved signaling pathways. Results In the core component-target network of BFYSF in treating COPD, there were 30 active ingredients and 37 core targets. Enrichment analysis suggested that these 37 core targets were mainly involved in the regulation of biological functions, such as response to biological and chemical stimuli, multiple cellular life processes, immunity, and metabolism. Besides, multiple pathways, including IL-17, Toll-like receptor (TLR), TNF, and HIF-1, played certain roles in the effect of BFYSF on treating COPD. Conclusion BFYSF can treat COPD through the multicomponent, multitarget, and multipathway synergistic network, which provides basic data for intensively exploring the mechanism of action of BFYSF in treating COPD.
Collapse
|
13
|
Dong Y, Hou Q, Lei J, Wolf PG, Ayansola H, Zhang B. Quercetin Alleviates Intestinal Oxidative Damage Induced by H 2O 2 via Modulation of GSH: In Vitro Screening and In Vivo Evaluation in a Colitis Model of Mice. ACS OMEGA 2020; 5:8334-8346. [PMID: 32309744 PMCID: PMC7161027 DOI: 10.1021/acsomega.0c00804] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 03/24/2020] [Indexed: 05/13/2023]
Abstract
The gastrointestinal tract is exposed to pro-oxidants from food, host immune factors, and microbial pathogens, which may induce oxidative damage. Oxidative stress has been shown to play an important role in the onset of inflammatory bowel disease. This study aimed to use a novel model to evaluate the effects of a screened natural component and explore its possible mechanism. An in vitro oxidative stress Caco2 cell model induced by H2O2 was established using a real-time cellular analysis system and verified by addition of glutathione (GSH). A variety of plant components were chosen for the screening. Quercetin was the most effective phytochemical to alleviate the decreased cell index caused by H2O2 among the tested plant components. Furthermore, quercetin ameliorated dextran sulfate sodium salt (DSS)-induced colitis and further increased the serum GSH. The mechanism of quercetin protection was explored in Caco2. Reversed H2O2-induced cell damage and decreased reactive oxygen species and apoptosis ratio were observed in quercetin-treated cells. Also, quercetin increased expression of the glutamate-cysteine ligase catalytic subunit (GCLC), the first rate-limiting enzyme of glutathione synthesis, and increased intracellular GSH concentration under H2O2 treatment. This effect was abolished by the GCLC inhibitor buthionine sulfoximine. These results indicated that quercetin can improve cell proliferation and increase intracellular GSH concentrations by upregulating transcription of GCLC to eliminate excessive reactive oxygen species (ROS). Increased extracellular H2O2 concentration induced by quercetin under oxidative stress was related to the inhibition of AQP3 and upregulation of NOX1/2, which may contribute to the observed protective effects of quercetin. Moreover, the novel H2O2-induced oxidative stress cell model based on the real-time cellular analysis system was an effective model to screen natural products to deal with intestinal oxidative damage and help accelerate the discovery of new drugs for inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Yuanyang Dong
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Qihang Hou
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Jiaqi Lei
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Patricia G. Wolf
- Division
of Nutritional Sciences, University of Illinois
at Urbana-Champaign, 1207 W. Gregory Avenue, Urbana, Illinois 61801, United
States
| | - Hammed Ayansola
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Bingkun Zhang
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
14
|
Identifying Synergistic Mechanisms of Multiple Ingredients in Shuangbai Tablets against Proteinuria by Virtual Screening and a Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1027271. [PMID: 32025234 PMCID: PMC6984745 DOI: 10.1155/2020/1027271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/08/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
Abstract
Shuangbai Tablets (SBT), a traditional herbal mixture, has shown substantial clinical efficacy. However, a systematic mechanism of its active ingredients and pharmacological mechanisms of action against proteinuria continues being lacking. A network pharmacology approach was effectual in discovering the relationship of multiple ingredients and targets of the herbal mixture. This study aimed to identify key targets, major active ingredients, and pathways of SBT against proteinuria by network pharmacology approach combined with thin layer chromatography (TLC). Human phenotype (HP) disease analysis, gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and molecular docking were used in this study. To this end, a total of 48 candidate targets of 118 active ingredients of SBT were identified. Network analysis showed PTGS2, ESR1, and NOS2 to be the three key targets, and beta-sitosterol, quercetin, and berberine were the three major active ingredients; among them one of the major active ingredients, quercetin, was discriminated by TLC. These results of the functional enrichment analysis indicated that the most relevant disease including these 48 candidate proteins is proteinuria, SBT treated proteinuria by sympathetically regulating multiple biological pathways, such as the HIF-1, RAS, AGE-RAGE, and VEGF signaling pathways. Additionally, molecular docking validation suggested that major active ingredients of SBT were capable of binding to HIF-1A and VEGFA of the main pathways. Consequently, key targets, major active ingredients, and pathways based on data analysis of SBT against proteinuria were systematically identified confirming its utility and providing a new drug against proteinuria.
Collapse
|
15
|
Toth S, Jonecova Z, Maretta M, Curgali K, Kalpakidis T, Pribula M, Kusnier M, Fagova Z, Fedotova J, La Rocca G, Rodrigo L, Caprnda M, Zulli A, Ciccocioppo R, Mechirova E, Kruzliak P. The effect of Betanin parenteral pretreatment on Jejunal and pulmonary tissue histological architecture and inflammatory response after Jejunal ischemia-reperfusion injury. Exp Mol Pathol 2019; 110:104292. [DOI: 10.1016/j.yexmp.2019.104292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/10/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
|
16
|
Périco LL, Rodrigues VP, Ohara R, Nunes VVA, da Rocha LRM, Vilegas W, Dos Santos C, Hiruma-Lima CA. Can the gastric healing effect of Eugenia punicifolia be the same in male and female rats? JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:268-278. [PMID: 30763697 DOI: 10.1016/j.jep.2019.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/20/2019] [Accepted: 02/06/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eugenia punicifolia (Kunth) DC. (Myrtaceae), an Amazonian medicinal plant known as "pedra-ume-caá," is popularly used as a natural remedy for inflammation, wounds, infections, diabetes, fever, and flu. Its anti-inflammatory, antinociceptive, and gastroprotective effects have already been characterized. We evaluated the gastric healing effect of the hydroalcoholic extract of the leaves of E. punicifolia (HEEP) in male and female Wistar rats against nonsteroidal anti-inflammatory drugs (NSAIDs) and ethanol. MATERIALS AND METHODS The healing effect of HEEP on the gastric mucosa of adult male and female Wistar rats was measured after the chronic application of aggressive factors such as NSAIDs or 80% ethanol. Male, and intact and ovariectomized (OVZ) female rats were treated with HEEP for two days (NSAIDs) or one, two, four, and six days (80% ethanol). The stomachs were analyzed macroscopically for ulcerative lesions (mm2), and the healing process was measured using biochemical analysis with anti-inflammatory and antioxidant parameters. RESULTS Macroscopic evaluation of the gastric mucosa showed that gastric lesions induced by NSAIDs were significantly healed (66%) and pro-inflammatory interleukin 5 cytokine level was decreased after two-day oral treatment with HEEP compared with those in the negative control group (p < 0.05). However, the gastric lesions induced by NSAIDs did not heal in HEEP-treated female rats (p > 0.05). In addition, four-day treatment with HEEP significantly healed the gastric lesions induced by ethanol in male and female rats (63% and 78%, respectively) compared to those of the negative control group (p < 0.05). However, the OVZ group required six days of HEEP treatment to heal gastric ulcers (67% compared to the control group). HEEP exerts the healing effect against ethanol by significantly reducing neutrophil infiltration into the gastric mucosa by decreasing myeloperoxidase activity in male and OVZ rats after four and six days of treatment, respectively (p < 0.05). Four-day treatment with HEEP also increased the level of a non-enzymatic antioxidant, reduced glutathione in intact females compared to that of the negative control group (p < 0.05). CONCLUSION These findings indicated that HEEP was effective in promoting the healing of gastric ulcers induced by NSAIDs or ethanol. The gastric healing effects of this extract could be affected by female sex hormone interference; in future, comprehensive studies should be performed by considering sex differences.
Collapse
Affiliation(s)
- Larissa Lucena Périco
- Department of Physiology, Biosciences Institute, UNESP-São Paulo State University, CEP 18618-689 Botucatu, São Paulo, Brazil
| | - Vinícius Peixoto Rodrigues
- Department of Physiology, Biosciences Institute, UNESP-São Paulo State University, CEP 18618-689 Botucatu, São Paulo, Brazil
| | - Rie Ohara
- Department of Physiology, Biosciences Institute, UNESP-São Paulo State University, CEP 18618-689 Botucatu, São Paulo, Brazil
| | - Vânia Vasti Alfieri Nunes
- Department of Physiology, Biosciences Institute, UNESP-São Paulo State University, CEP 18618-689 Botucatu, São Paulo, Brazil
| | - Lúcia Regina Machado da Rocha
- Department of Physiology, Biosciences Institute, UNESP-São Paulo State University, CEP 18618-689 Botucatu, São Paulo, Brazil
| | - Wagner Vilegas
- Biosciences Institute, UNESP-São Paulo State University, CEP 11330-900 São Vicente, São Paulo, Brazil
| | - Catarina Dos Santos
- Department of Biological Science, Faculty of Sciences and Languages, UNESP-São Paulo State University, CEP 19806-900 Assis, São Paulo, Brazil
| | - Clélia Akiko Hiruma-Lima
- Department of Physiology, Biosciences Institute, UNESP-São Paulo State University, CEP 18618-689 Botucatu, São Paulo, Brazil.
| |
Collapse
|
17
|
Bertoni S, Ballabeni V, Barocelli E, Tognolini M. Mesenteric ischemia-reperfusion: an overview of preclinical drug strategies. Drug Discov Today 2018; 23:1416-1425. [DOI: 10.1016/j.drudis.2018.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/27/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
|
18
|
Sherif IO. Uroprotective mechanism of quercetin against cyclophosphamide-induced urotoxicity: Effect on oxidative stress and inflammatory markers. J Cell Biochem 2018; 119:7441-7448. [PMID: 29775228 DOI: 10.1002/jcb.27053] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/23/2018] [Indexed: 12/27/2022]
Abstract
The urotoxicity is a common complication associated with patients receiving cyclophosphamide (CYP). This study was designed to investigate the uroprotective mechanism of quercetin (Quer) flavonoid against CYP induced urotoxicity via determination of oxidative stress markers as well as inflammatory mediators in bladder tissue. Forty male Wistar rats were divided into four groups; Normal group: received saline for 10 days. Quer control group: received quercetin 50 mg/kg/day for 10 days. CYP group: received saline for 10 days and injected with a single dose of 150 mg/kg CYP intraperitoneal (i.p) at day 8. The Quer + CYP group: received Quer 50 mg/kg/day for 10 days plus CYP 150 mg/kg i.p. injection at day 8. The CYP injection produced a significant elevation in bladder contents of malondialdehyde (MDA), and nitric oxide (NO), and bladder protein levels and expressions of tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) in addition to the upregulation of cyclooxygenase-2 (COX-2) bladder gene expression. Also, CYP injection showed a marked reduction in bladder levels of catalase, superoxide dismutase (SOD), and IL-10 when compared with normal group. Moreover, histopathological examination of the bladder showed degenerative alterations, severe edema, and inflammation following CYP injection. Quer attenuated the biochemical markers and histopathological changes induced by CYP. The uroprotective effect of Quer was exerted by restoring the balance between oxidative/antioxidative status and pro-/anti-inflammatory cytokines via its antioxidant and anti-inflammatory activities.
Collapse
Affiliation(s)
- Iman O Sherif
- Emergency Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
19
|
Curgali K, Toth S, Jonecova Z, Maretta M, Kalpakidis T, Petriskova I, Kusnier M, Soltes J, Svana M, Caprnda M, Delev D, Rodrigo L, Mechirova E, Kruzliak P. Quercetin protects jejunal mucosa from experimental intestinal ischemia reperfusion injury by activation of CD68 positive cells. Acta Histochem 2018; 120:28-32. [PMID: 29129327 DOI: 10.1016/j.acthis.2017.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
The aim of our study was to analyse the possible protective effect of quercetin application during the jejunal ischemia-reperfusion injury (IRI) in rats. Quercetin was administered intraperitoneally 30min before 1h ischemia of superior mesenteric artery with following 24h lasting reperfusion period. The male specific pathogen-free (SPF) Charles River Wistar rats were used. In the group with applied quercetin, the significantly increased (p<0.001) levels of anti-inflammatory cytokine IL10 were observed both in the blood serum and jejunal tissue. The improvement of the mucosal tissue morphology and proliferating and DNA repairing cell number measured by PCNA activity were recorded by more than 30% higher in the quercetin group. Simultaneously, significant elongation of the intestinal glands (p<0.001) and increase in the number of CD68-positive cells in the lamina propria mucosae (p<0.001) in comparison with control group were found. Based on our results, the preventive application of quercetin before induction of jejunal IRI stimulates faster jejunal mucosa restoration and it seems to have immunomodulatory and anti-inflammatory effects as well. CD68-positive macrophages could have crucial role in this process since they work as both growth factor and cytokine producers.
Collapse
|