1
|
He Y, Yang Z, Guo D, Luo C, Liu Q, Xian L, Yang F, Huang C, Wei Q. The multifaceted nature of SUMOylation in heart disease and its therapeutic potential. Mol Cell Biochem 2025:10.1007/s11010-025-05286-z. [PMID: 40287894 DOI: 10.1007/s11010-025-05286-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/06/2025] [Indexed: 04/29/2025]
Abstract
SUMOylation (SUMO), a crucial post-translational modification, is implicated in the regulation of diverse biological processes and plays a pivotal role in both the maintenance of cardiac function and progression and treatment of heart disease. Here, we reviewed the mechanisms by which SUMO-related various aspects of cardiac function and disease, including cardiac hypertrophy, heart failure, ischemia-reperfusion injury, and myocardial infarction. Furthermore, we highlight its potential as a therapeutic target.
Collapse
Affiliation(s)
- Ying He
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhijie Yang
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Dan Guo
- Medical College, Guangxi University, Nanning, Guangxi, China
| | - Cheng Luo
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Qiaoqiao Liu
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Lei Xian
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fan Yang
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China.
- Liuzhou Key Laboratory of Primary Cardiomyopathy in Prevention and Treatment, Liuzhou, Guangxi, China.
| | - Chusheng Huang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Qingjun Wei
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
2
|
Gunter NV, Teh SS, Jantan I, Law KP, Morita H, Mah SH. Natural xanthones as modulators of the Nrf2/ARE signaling pathway and potential gastroprotective agents. Phytother Res 2025; 39:1721-1734. [PMID: 38372084 DOI: 10.1002/ptr.8160] [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: 10/31/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024]
Abstract
Oxidative stress is implicated in the initiation, pathogenesis, and progression of various gastric inflammatory diseases (GID). The prevalence of these diseases remains a concern along with the increasing risks of adverse effects in current clinical interventions. Hence, new gastroprotective agents capable of inhibiting oxidative stress by modulating cellular defense systems such as the nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway are critically needed to address these issues. A candidate to solve the present issue is xanthone, a natural compound that reportedly exerts gastroprotective effects via antioxidant, anti-inflammatory, and cytoprotective mechanisms. Moreover, xanthone derivatives were shown to modulate the Nrf2/ARE signaling pathway to counter oxidative stress in both in vitro and in vivo models. Thirteen natural xanthones have demonstrated the ability to modulate the Nrf2/ARE signaling pathway and have high potential as lead compounds for GID as indicated by their in vivo gastroprotective action-particularly mangiferin (2), α-mangostin (3), and γ-mangostin (4). Further studies on these compounds are recommended to validate the Nrf2 modulatory ability in relation to their gastroprotective action.
Collapse
Affiliation(s)
- Natalie Vivien Gunter
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Soek Sin Teh
- Energy and Environment Unit, Engineering and Processing Division, Malaysian Palm Oil Board, Kajang, Malaysia
| | - Ibrahim Jantan
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Kung Pui Law
- School of Pre-University Studies, Taylor's College, Subang Jaya, Malaysia
| | - Hiroyuki Morita
- Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Siau Hui Mah
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
3
|
El-Sayed NM, Menze ET, Tadros MG, Hanna DMF. Mangiferin mitigates methotrexate-induced liver injury and suppresses hepatic stellate cells activation in rats: Imperative role of Nrf2/NF-κB/NLRP3 signaling axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119296. [PMID: 39732297 DOI: 10.1016/j.jep.2024.119296] [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: 08/18/2024] [Revised: 11/16/2024] [Accepted: 12/26/2024] [Indexed: 12/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mangifera indica (family Anacardiaceae), often acknowledged as mango and renowned for being a plant of diverse ethnopharmacological background since ancient times, harbors the polyphenolic bioactive constituent, mangiferin (MNG). MNG is a major phytochemical of Mangifera indica and other plants with a wide range of reported pharmacological activities, including antioxidant, anti-inflammatory, neuroprotective and hepatoprotective effects. MNG has also been utilized in traditional medicine; it is reportedly a major bioactive element in over 40 polyherbal products in traditional Chinese medicine (TCM), and two prominent anti-inflammatory, immunomodulatory and antiviral Cuban formulations. Despite the availability of evidence in support of MNG hepatoprotective properties, its hepatoprotective potential against MTX-induced liver injury and fibrosis has not been explored yet. AIM To unravel the hepatoprotective potential of MNG against MTX-induced hepatic injury and fibrosis and elucidate the possible underlying molecular mechanisms. MATERIALS AND METHODS Male Sprague-Dawley rats were, randomly, distributed into five groups; two of which were administered MNG 50 mg/kg and MNG 100 mg/kg intraperitoneally (i.p.) for ten days, and a single i.p. injection of MTX 40 mg/kg on the seventh day to establish hepatotoxicity. Blood and liver tissue samples were retrieved from all study groups and analyzed for liver functions, histopathological alterations, and oxidative stress, inflammatory, and fibrotic biomarkers. RESULTS MNG restored the MTX-induced degenerations in hepatic architecture and function. Moreover, it combated the MTX-elicited oxidative stress evidently by the significantly attenuated hepatic tissue levels of malondialdehyde, and the significantly elevated reduced glutathione and Nrf2 levels. MNG also halted inflammation depicted by the downregulation of the NF-κB/NLRP3 inflammasome axis. It further demonstrated anti-fibrogenic potential as evidenced by the significant reduction in fibrous tissue deposition and hepatic expression of α-SMA. CONCLUSION The current study proved the hepatoprotective, and anti-fibrogenic effects of MNG against MTX-induced hepatotoxicity via the downregulation of NF-κB/NLRP3 inflammasome signaling axis, preceded by the amelioration of oxidative stress and Nrf2 signaling upregulation.
Collapse
Affiliation(s)
- Nada M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt; School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Diana M F Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt.
| |
Collapse
|
4
|
El-Shitany NA, El-Saidy EA, El-Naggar ME, Sokar SS. Cilostazol protects against gastric ulcers by regulating PPAR-γ, HO-1, PECAM-1, pErk-1, NF-κB, Bcl-2, and cleaved caspase-3 protein expression. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9033-9050. [PMID: 38884677 PMCID: PMC11522149 DOI: 10.1007/s00210-024-03176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
Millions of individuals worldwide, across all age groups, suffer from the widespread health issue of gastric ulcers. In many experiments, cilostazol (Cls), a phosphodiesterase-3 inhibitor, was recently shown to have anti-ulcer activity. Notably, Cls increases the expression and transcriptional activity of PPAR-γ in vitro and in vivo. This study aimed to evaluate the protective effect of Cls against ethanol-induced gastric ulcers and clarify the possible underlying mechanisms with an emphasis on the role of PPAR-γ. Male albino rats were treated with ethanol to induce gastric ulcers, or they were pretreated with Cls, omeprazole (Omp), GW9662, or Cls + GW9662 for 14 consecutive days before receiving ethanol. Cls protects against ethanol-induced gastric ulcers. Cls treatment significantly reduced ethanol-induced upregulation of the pro-inflammatory markers (IL-1β, IL-6, TNF-α, and NF-κB), MDA (a marker of lipid peroxidation), and caspase-3 and cleaved caspase-3 (apoptotic markers). On the other hand, Cls treatment counteracted ethanol-induced downregulation of PPAR-γ, pErk-1, HO-1 and GSH (antioxidant markers), PECAM-1 and NO (healing markers), and Bcl-2 (antiapoptotic marker). However, when combined with GW9662, a potent antagonist of PPAR-γ, Cls loses its effects. In conclusion, these results suggest that PPAR-γ and pErk-1 are essential for Cls's protective effects against ethanol-induced gastric ulcers.
Collapse
Affiliation(s)
- Nagla A El-Shitany
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Eman A El-Saidy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Mostafa E El-Naggar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Samia S Sokar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
5
|
Elbaz EM, Abdel Rahman AAS, El-Gazar AA, Ali BM. Protective effect of dimethyl fumarate against ethanol-provoked gastric ulcers in rats via regulation of HMGB1/TLR4/NF-κB, and PPARγ/SIRT1/Nrf2 pathways: Involvement of miR-34a-5p. Arch Biochem Biophys 2024; 759:110103. [PMID: 39053682 DOI: 10.1016/j.abb.2024.110103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/07/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Aberration of the gastric mucosal barrier homeostasis circuit is one of the key features linked to the onset of gastric ulcers (GU). This work aimed to inspect the gastroprotective influence of dimethyl fumarate (DMF) on ethanol-induced GU in rats and to decipher the possible mechanisms entailed. Rats were pretreated with either DMF (80 mg/kg) or omeprazole (OMP) (20 mg/kg) by oral gavage for 2 weeks. After 24 h of starvation, ethanol (5 ml/kg, oral) was employed to trigger GU in rats, while carboxymethyl cellulose (CMC) was used as a control. Ethanol notably elevated both macroscopic and microscopic gastric damage. DMF and OMP exhibited similar effects on gastric ulcer healing. DMF intervention led to a substantial improvement in gastric insults. DMF significantly reduced ethanol-triggered gastric lesions, as manifested by decreased gastric secretion, acidity, ulcer surface area percent, reduced leukocyte incursion, and increased mucus percent. DMF upregulated miR-34a-5p expression concomitant with the suppression of high mobility group box1 (HMGB1) and inflammatory responses in gastric mucosal homogenate. DMF improved GU by restoring reduced antioxidant defense mechanisms through the coactivation of nuclear factor erythroid 2-related factor-2 (Nrf2), peroxisome proliferator-activated receptor gamma (PPARγ), and sirtuin1 (SIRT1), indicating the protective role of the PPARγ/SIRT1/Nrf2 pathway. Intriguingly, DMF mitigated apoptosis in ethanol-elicited GU. Taken together, this research implies the potential for the repurposing of DMF as an innovative gastroprotective medication to reestablish the balance of the gastric mucosal barrier via the attenuation of gastric inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Eman M Elbaz
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Amina A S Abdel Rahman
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Amira A El-Gazar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| | - Bassam Mohamed Ali
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| |
Collapse
|
6
|
Vilas-Boas C, Sousa J, Lima E, Running L, Resende D, Ribeiro ARL, Sousa E, Santos MM, Aga DS, Tiritan ME, Ruivo R, Atilla-Gokcumen GE, Correia-da-Silva M. Preliminary hazard assessment of a new nature-inspired antifouling (NIAF) agent. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 933:172824. [PMID: 38688370 DOI: 10.1016/j.scitotenv.2024.172824] [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: 08/29/2023] [Revised: 03/29/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
A recently synthesized aminated 3,4-dioxygenated xanthone (Xantifoul2) was found to have promising antifouling (AF) effects against the settlement of the macrofouler Mytilus galloprovincialis larvae. Preliminary assessment indicated that Xantifoul2 has reduced ecotoxicological impacts: e.g., being non-toxic to the marine crustacea Artemia salina (<10 % mortality at 50 μM) and showing low bioconcentration factor in marine organisms. In order to meet the EU Biocidal Product Regulation, a preliminary hazard assessment of this new nature-inspired antifouling (NIAF) agent was conducted in this work. Xantifoul2 did not affect the swimming ability of the planktonic crustacean Daphnia magna, the growth of the diatom Phaeodactylum tricornutum, and the cellular respiration of luminescent Gram-negative bacteria Vibrio fischeri, supporting the low toxicity towards several non-target marine species. Regarding human cytotoxicity, Xantifoul2 did not affect the cell viability of retinal human cells (hTERT-RPE-1) and lipidomic studies revealed depletion of lipids involved in cell death, membrane modeling, lipid storage, and oxidative stress only at a high concentration (10 μM). Accelerated degradation studies in water were conducted under simulated sunlight to allow the understanding of putative transformation products (TPs) that could be generated in the aquatic ecosystems. Both Xantifoul2 and photolytic-treated Xantifoul2 in the aqueous matrix were therefore evaluated on several nuclear receptors (NRs). The results of this preliminary hazard assessment of Xantifoul2, combined with the high degradation rates in water, provide strong evidence of the safety of this AF agent under the evaluated conditions, and provide the support for future validation studies before this compound can be introduced in the market.
Collapse
Affiliation(s)
- Cátia Vilas-Boas
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - João Sousa
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Erica Lima
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Logan Running
- Chemistry Department, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Diana Resende
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Ana Rita L Ribeiro
- LSRE-LCM - Laboratory of Separation and Reaction Engineering - Laboratory of Catalysis and Materials, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Miguel M Santos
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal; Department of Biology, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Diana S Aga
- Chemistry Department, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Maria Elizabeth Tiritan
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal
| | - Raquel Ruivo
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal.
| | | | - Marta Correia-da-Silva
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, 4450-208 Matosinhos, Portugal.
| |
Collapse
|
7
|
Blagov AV, Summerhill VI, Sukhorukov VN, Zhigmitova EB, Postnov AY, Orekhov AN. Potential use of antioxidants for the treatment of chronic inflammatory diseases. Front Pharmacol 2024; 15:1378335. [PMID: 38818374 PMCID: PMC11137403 DOI: 10.3389/fphar.2024.1378335] [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: 01/29/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
The excessive production of various reactive oxidant species over endogenous antioxidant defense mechanisms leads to the development of a state of oxidative stress, with serious biological consequences. The consequences of oxidative stress depend on the balance between the generation of reactive oxidant species and the antioxidant defense and include oxidative damage of biomolecules, disruption of signal transduction, mutation, and cell apoptosis. Accumulating evidence suggests that oxidative stress is involved in the physiopathology of various debilitating illnesses associated with chronic inflammation, including cardiovascular diseases, diabetes, cancer, or neurodegenerative processes, that need continuous pharmacological treatment. Oxidative stress and chronic inflammation are tightly linked pathophysiological processes, one of which can be simply promoted by another. Although, many antioxidant trials have been unsuccessful (some of the trials showed either no effect or even harmful effects) in human patients as a preventive or curative measure, targeting oxidative stress remains an interesting therapeutic approach for the development of new agents to design novel anti-inflammatory drugs with a reliable safety profile. In this regard, several natural antioxidant compounds were explored as potential therapeutic options for the treatment of chronic inflammatory diseases. Several metalloenzymes, such as superoxide dismutase, catalase, and glutathione peroxidase, are among the essential enzymes that maintain the low nanomolar physiological concentrations of superoxide (O2•-) and hydrogen peroxide (H2O2), the major redox signaling molecules, and thus play important roles in the alteration of the redox homeostasis. These enzymes have become a striking source of motivation to design catalytic drugs to enhance the action of these enzymes under pathological conditions related to chronic inflammation. This review is focused on several major representatives of natural and synthetic antioxidants as potential drug candidates for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | | | - Anton Y. Postnov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - Alexander N. Orekhov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Institute for Atherosclerosis Research, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| |
Collapse
|
8
|
Gong H, Zhao N, Zhu C, Luo L, Liu S. Treatment of gastric ulcer, traditional Chinese medicine may be a better choice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117793. [PMID: 38278376 DOI: 10.1016/j.jep.2024.117793] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/30/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastric ulcer (GU) is the injury of the gastric mucosa caused by the stimulation of various pathogenic factors penetrating the deep mucosal muscle layer. An increasing number of studies have shown that traditional Chinese medicine (TCM) is highly effective in treating GU due to its multitarget, multilevel, and multi-pathway effects. AIM OF THE STUDY To review the latest research progress in the treatment of GU by TCM, including clinical and experimental studies, focusing on the target and mechanism of action of drugs and providing a theoretical basis for the treatment of GU by natural herbs. MATERIALS AND METHODS Electronic databases (PubMed, Elsevier, Springer, Web of Science, and CNKI) were searched using the keywords "gastric ulcer", "gastric mucosal lesion", "TCM" and or paired with "peptic ulcer" and "natural drugs" for studies published in the last fifteen years until 2023. RESULTS TCM, including single components of natural products, Chinese patent medicines (CPM), and TCM decoction, is expected to treat GU by regulating various mechanisms, such as redox balance, inflammatory factors, angiogenesis, gastric mucosal protective factors, intestinal flora, apoptosis, and autophagy. CONCLUSIONS We discussed and summarized the mechanism of TCM in the treatment of GU, which provided a sufficient basis for TCM treatment of GU.
Collapse
Affiliation(s)
- Haiying Gong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Conglei Zhu
- Department of Pharmacy, Fuyang People's Hospital, Fuyang, Anhui, China
| | - Lin Luo
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Ghareeb MA, Mohammed HS, Aboushousha T, Lotfy DM, El-Shazly MAM, Sobeh M, Taha EFS. Ipomoea carnea mitigates ethanol-induced ulcers in irradiated rats via Nrf2/HO -1 pathway: an in vivo and in silico study. Sci Rep 2024; 14:3469. [PMID: 38342928 PMCID: PMC10859386 DOI: 10.1038/s41598-024-53336-1] [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: 11/01/2023] [Accepted: 01/31/2024] [Indexed: 02/13/2024] Open
Abstract
The aim of this study was to investigate the potential of Ipomoea carnea flower methanolic extract (ICME) as a natural gastroprotective therapy against ethanol-induced gastric ulcers, particularly in individuals exposed to ionizing radiation (IR). The study focused on the Nrf2/HO-1 signaling pathway, which plays a crucial role in protecting the gastrointestinal mucosa from oxidative stress and inflammation. Male Wistar rats were divided into nine groups, the control group received distilled water orally for one week, while other groups were treated with ethanol to induce stomach ulcers, IR exposure, omeprazole, and different doses of ICME in combination with ethanol and/or IR. The study conducted comprehensive analyses, including LC-HRESI-MS/MS, to characterize the phenolic contents of ICME. Additionally, the Nrf2/HO-1 pathway, oxidative stress parameters, gastric pH, and histopathological changes were examined. The results showed that rats treated with IR and/or ethanol exhibited histopathological alterations, increased lipid peroxidation, decreased antioxidant enzyme activity, and reduced expression levels of Nrf2 and HO-1. However, pretreatment with ICME significantly improved these parameters. Phytochemical analysis identified 39 compounds in ICME, with flavonoids, hydroxybenzoic acids, and fatty acids as the predominant compounds. Virtual screening and molecular dynamics simulations suggested that ICME may protect against gastric ulceration by inhibiting oxidative stress and inflammatory mediators. In conclusion, this study demonstrates the potential of ICME as a natural gastroprotective therapy for preventing gastric ulcers. These findings contribute to the development of novel interventions for gastrointestinal disorders using natural plant extracts particularly in individuals with a history of radiation exposure.
Collapse
Affiliation(s)
- Mosad A Ghareeb
- Medicinal Chemistry Department, Theodor Bilharz Research Institute, Kornaish El-Nile, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Hala Sh Mohammed
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11311, Egypt
| | - Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, Kornaish El-Nile, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Dina M Lotfy
- Drug Radiation Research Department, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Maha A M El-Shazly
- Medicinal Chemistry Department, Theodor Bilharz Research Institute, Kornaish El-Nile, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Mansour Sobeh
- AgroBioSciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150, Ben-Guerir, Morocco
| | - Eman F S Taha
- Health Radiation Research Department, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|
10
|
Zivković J, Kumar KA, Rushendran R, Ilango K, Fahmy NM, El-Nashar HAS, El-Shazly M, Ezzat SM, Melgar-Lalanne G, Romero-Montero A, Peña-Corona SI, Leyva-Gomez G, Sharifi-Rad J, Calina D. Pharmacological properties of mangiferin: bioavailability, mechanisms of action and clinical perspectives. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:763-781. [PMID: 37658210 DOI: 10.1007/s00210-023-02682-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/20/2023] [Indexed: 09/03/2023]
Abstract
This review aims to provide an in-depth analysis of the pharmacological properties of mangiferin, focusing primarily on its bioavailability and mechanisms of action, and its potential therapeutic applications, especially in the context of chronic diseases. We conducted a comprehensive examination of in vitro and in vivo studies, as well as clinical trials involving mangiferin or plant extracts containing mangiferin. The primary source of mangiferin is Mangifera indica, but it's also found in other plant species from the families Anacardiaceae, Gentianaceae, and Iridaceae. Mangiferin has exhibited a myriad of therapeutic properties, presenting itself as a promising candidate for treating various chronic conditions including neurodegenerative disorders, cardiovascular diseases, renal and pulmonary diseases, diabetes, and obesity. Despite the promising results showcased in many in vitro studies and certain animal studies, the application of mangiferin has been limited due to its poor solubility, absorption, and overall bioavailability. Mangiferin offers significant therapeutic potential in treating a spectrum of chronic diseases, as evidenced by both in vitro and clinical trials. However, the challenges concerning its bioavailability necessitate further research, particularly in optimizing its delivery and absorption, to harness its full medicinal potential. This review serves as a comprehensive update on the health-promoting and therapeutic activities of mangiferin.
Collapse
Affiliation(s)
- Jelena Zivković
- Institute for Medicinal Plants Research "Dr. Josif Pančić", Tadeuša Košćuška 1, Belgrade, Serbia.
| | - Kammala Ananth Kumar
- Department of Obstetrics and Gynecology, Division of Basic Sciences and Translational Medicine, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Rapuru Rushendran
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Kattankulatur, 603203, Tamil Nadu, India
| | - Kaliappan Ilango
- School of Pharmacy, Hindustan Institute Technology and Science, Padur, Chennai, 603 103, India
| | - Nouran M Fahmy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
- Center of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
- Center of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Shahira M Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
- Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | - Guiomar Melgar-Lalanne
- Instituto de Ciencias Básicas, Universidad Veracruzana, Avda. Castelazo Ayala S/N, 91190, Xalapa, Veracruz, Mexico
| | - Alejandra Romero-Montero
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico
| | - Gerardo Leyva-Gomez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
11
|
Tureyen A, Demirel HH, Demirkapi EN, Eryavuz AM, Ince S. Tubuloside A, a phenylethanoid glycoside, alleviates diclofenac induced hepato-nephro oxidative injury via Nrf2/HO-1. J Cell Mol Med 2023; 27:3404-3413. [PMID: 37772986 PMCID: PMC10623516 DOI: 10.1111/jcmm.17968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 09/30/2023] Open
Abstract
The most prominent adverse effects of nonsteroidal anti-inflammatory drugs (NSAIDs) such as diclofenac (DF) are hepato-renal damage. Natural antioxidants can be preferred as an alternative and/or combination to improve this damage. This present study was conducted to evaluate the protective effect of Tubuloside A (TA) against diclofenac (DF)-induced hepato-renal damage. TA (1 mg/kg, ip) was administered to male Sprague-Dawley rats for 5 days, and DF (50 mg/kg, ip) was administered on Days 4 and 5. Plasma aspartate amino transferase, alanine amino transferase, alkaline phosphatase, blood urea nitrogen and creatinine were measured to evaluate liver and kidney functions. Additionally, oxidative stress parameters (malondialdehyde, glutathione, superoxide dismutase, catalase, and 8-oxo-7,8-dihydro-2'-deoxyguanosine) in blood, liver, and kidney tissues, changes in mRNA expression of genes involved in the Nrf2/HO-1 signalling pathway (Nrf2, HO-1, NQO-1, IL-6, iNOS, Cox-2, TNF-α, IL1-β and NFκB) and apoptotic process (Bcl-2, Cas-3 and Bax) in liver and kidney tissues were determined. Additionally, tissue sections were evaluated histopathologically. Biochemical, histopathological, and molecular results demonstrated the hepato-renal toxic effects of DF, and TA treatment protected the liver and kidney from DF-induced damage. This provides an explanation for the hepato-nephro damage caused by DF and offers new ideas and drug targets together with TA for the prevention and treatment of DF injury.
Collapse
Affiliation(s)
- Ali Tureyen
- Department of GastroenterologyMinistry of Health Eskisehir City HospitalEskisehirTurkey
| | | | - Ezgi Nur Demirkapi
- Faculty of Veterinary Medicine, Department of PhysiologyAfyon Kocatepe UniversityAfyonkarahisarTurkey
| | - Azra Mila Eryavuz
- Department of Biochemistry, Faculty of Veterinary MedicineAfyon Kocatepe UniversityAfyonkarahisarTurkey
| | - Sinan Ince
- Department of Pharmacology and Toxicology, Faculty of Veterinary MedicineAfyon Kocatepe UniversityAfyonkarahisarTurkey
| |
Collapse
|
12
|
El-Beheiry KM, El-Sayed El-Sayad M, El-Masry TA, Elsisi AE. Combination of metformin and hesperidin mitigates cyclophosphamide-induced hepatotoxicity. Emerging role of PPAR-γ/Nrf-2/NF-κB signaling pathway. Int Immunopharmacol 2023; 117:109891. [PMID: 36812672 DOI: 10.1016/j.intimp.2023.109891] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
Cyclophosphamide (CP) is widely used as an immunosuppressive and chemotherapeutic drug. However, its therapeutic application is restricted by its adverse effects, particularly hepatotoxicity. Both metformin (MET) and hesperidin (HES) have promising antioxidant, anti-inflammatory, and anti-apoptotic effects. Therefore, the principal aim of the current study is to investigate the hepatoprotective effects of MET, HES, and their combinations on the CP-induced hepatotoxicity model. Hepatotoxicity was evoked by a single (I.P) injection of CP (200 mg/kg) on day 7. For this study, 64 albino rats were randomly categorized into eight equal groups; naïve, control vehicle, untreated CP (200 mg/kg, IP), and CP 200 groups treated with MET 200, HES 50, HES 100 or a combination of MET 200 with HES 50 and HES 100 respectively orally daily for 12 days. At the end of the study, the liver function biomarkers, oxidative stress, inflammatory parameters, histopathological and immunohistochemical analysis of PPAR-γ, Nrf-2, NF-κB, Bcl-2, and caspase3 were assessed. CP significantly increased serum ALT, AST, total bilirubin, hepatic MDA, NO content, NF-κB, and TNF-α. Otherwise, albumin, hepatic GSH content, Nrf-2, and PPAR-γ expression decreased considerably compared to the control vehicle group. The combinations of MET200 with HES50 or HES100 induced pronounced hepatoprotective, anti-oxidative, anti-inflammatory, and anti-apoptotic effects on CP-treated rats. The possible explanation of such hepatoprotective effects may be mediated via upregulation of Nrf-2, PPAR-γ, Bcl-2 expression, hepatic GSH content, and marked suppression of TNF-α and NF-κB expression. In conclusion, the current study showed that combining MET and HES revealed a remarkable hepatoprotective effect against CP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Kareman M El-Beheiry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Magda El-Sayed El-Sayad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
13
|
Possible Implication of Nrf2, PPAR-γ and MAPKs Signaling in the Protective Role of Mangiferin against Renal Ischemia/Reperfusion in Rats. Pharmaceuticals (Basel) 2022; 16:ph16010006. [PMID: 36678503 PMCID: PMC9863472 DOI: 10.3390/ph16010006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Mangiferin (Mang) is a known glucosylxanthone that has proven its shielding effect against ischemia/reperfusion (Is/R). However, its full underlying mechanistic perspective against renal Is/R induced lesions is not fully revealed. Consequently, the purpose of this study is to track further non-investigated modulatory signals of Mang against the renal Is/R model involving nuclear factor erythroid 2-related factor (Nrf)2/heme oxygenase (HO)-1, peroxisome proliferator-activated receptor (PPAR)-γ/nuclear factor (NF)-κB, p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (JNK) signaling. To ratify our aim, Mang was administrated (20 mg/kg, i.p for seven days) before the induction of bilateral Is/R. Mechanistic maneuver revealed that Mang balanced oxidative state via increasing the expression of the antioxidant Nrf2/HO-1 cue with subsequent enhancement of GSH besides MDA lessening. Additionally, Mang enhanced PPAR-γ mRNA expression and declined p-p38 MAPK and p-JNK expression with concomitant NF-κB downsizing leading to iNOS/NOx and TNF-α rebating. Furthermore, the Mang anti-apoptotic trait was affirmed by enriching Bcl-2 expression as well as decreasing Bax and caspase-3 expression. All these potentials were in the line with the molecular docking results and the improved histopathological findings and renal function biomarkers. Consequently, Mang provided plausible protective mechanisms against renal Is/R-related events, possibly by amending oxidative status, inflammatory mediators, and apoptotic cell death through the involvement of Nrf2, PPAR-γ, MAPK, JNK, and NF-κB signaling.
Collapse
|
14
|
Mazumder S, Bindu S, De R, Debsharma S, Pramanik S, Bandyopadhyay U. Emerging role of mitochondrial DAMPs, aberrant mitochondrial dynamics and anomalous mitophagy in gut mucosal pathogenesis. Life Sci 2022; 305:120753. [PMID: 35787999 DOI: 10.1016/j.lfs.2022.120753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022]
Abstract
Gastroduodenal inflammation and ulcerative injuries are increasing due to expanding socio-economic stress, unhealthy food habits-lifestyle, smoking, alcoholism and usage of medicines like non-steroidal anti-inflammatory drugs. In fact, gastrointestinal (GI) complications, associated with the prevailing COVID-19 pandemic, further, poses a challenge to global healthcare towards safeguarding the GI tract. Emerging evidences have discretely identified mitochondrial dysfunctions as common etiological denominators in diseases. However, it is worth realizing that mitochondrial dysfunctions are not just consequences of diseases. Rather, damaged mitochondria severely aggravate the pathogenesis thereby qualifying as perpetrable factors worth of prophylactic and therapeutic targeting. Oxidative and nitrosative stress due to endogenous and exogenous stimuli triggers mitochondrial injury causing production of mitochondrial damage associated molecular patterns (mtDAMPs), which, in a feed-forward loop, inflicts inflammatory tissue damage. Mitochondrial structural dynamics and mitophagy are crucial quality control parameters determining the extent of mitopathology and disease outcomes. Interestingly, apart from endogenous factors, mitochondria also crosstalk and in turn get detrimentally affected by gut pathobionts colonized during luminal dysbiosis. Although mitopathology is documented in various pre-clinical/clinical studies, a comprehensive account appreciating the mitochondrial basis of GI mucosal pathologies is largely lacking. Here we critically discuss the molecular events impinging on mitochondria along with the interplay of mitochondria-derived factors in fueling mucosal pathogenesis. We specifically emphasize on the potential role of aberrant mitochondrial dynamics, anomalous mitophagy, mitochondrial lipoxidation and ferroptosis as emerging regulators of GI mucosal pathogenesis. We finally discuss about the prospect of mitochondrial targeting for next-generation drug discovery against GI disorders.
Collapse
Affiliation(s)
- Somnath Mazumder
- Department of Zoology, Raja Peary Mohan College, 1 Acharya Dhruba Pal Road, Uttarpara, West Bengal 712258, India
| | - Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal 736101, India
| | - Rudranil De
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata, West Bengal 700135, India
| | - Subhashis Debsharma
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, West Bengal 700032, India
| | - Saikat Pramanik
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, West Bengal 700032, India
| | - Uday Bandyopadhyay
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, West Bengal 700032, India; Division of Molecular Medicine, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata, West Bengal 700091, India.
| |
Collapse
|
15
|
Attia SH, Elshazly SM, Abdelaal MM, Soliman E. Reno-protective effect of mangiferin against methotrexate-induced kidney damage in male rats: PPARγ-mediated antioxidant activity. Saudi Pharm J 2022; 30:1252-1261. [PMID: 36249937 PMCID: PMC9561181 DOI: 10.1016/j.jsps.2022.06.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Methotrexate (MTX) is an immunosuppressant used for the treatment of cancer and autoimmune diseases. MTX has a major adverse effect, acute kidney injury, which limits its use. Mangiferin (MF) is a natural bioactive xanthonoid used as a traditional herbal supplement to boost the immune system due to its potent anti-inflammatory and antioxidant activity. The present study evaluates the protective effect of MF against MTX-induced kidney damage. Male Wistar rats received MTX to induce nephrotoxicity or were pretreated with MF for 10 constitutive days before MTX administration. MF dose-dependently improved renal functions of MTX-treated rats and this activity was correlated with increased renal expression of PPARγ, a well-known transcriptional regulator of the immune response. Pretreating rats with PPARγ inhibitor, BADGE, reduced the reno-protective activity of MF. Furthermore, MF treatment significantly reduced MTX-induced upregulation of the pro-inflammatory (NFκB, interleukin-1ß, TNF-α, and COX-2), oxidative stress (Nrf-2, hemoxygenase-1, glutathione, and malondialdehyde), and nitrosative stress (nitric oxide and iNOS) markers in the kidney. Importantly, BADGE treatment significantly reduced the anti-inflammatory and antioxidant activity of MF. Therefore, our data suggest that the reno-protective effect of MF against MTX-induced nephrotoxicity is due to inhibition of inflammation and oxidative stress in a PPAR-γ-dependent manner.
Collapse
Affiliation(s)
- Seba Hassan Attia
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Egypt
| | | | | | - Eman Soliman
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Egypt
- Corresponding author.
| |
Collapse
|
16
|
Abu-Elfotuh K, Hamdan AME, Abbas AN, Alahmre ATS, Elewa MAF, Masoud RAE, Ali AA, Othman M, Kamal MM, Hassan FAM, Khalil MG, El-Sisi AM, Abdel Hady MMM, Abd-Elhaleim El Azazy MK, Awny MM, Wahid A. Evaluating the neuroprotective activities of vinpocetine, punicalagin, niacin and vitamin E against behavioural and motor disabilities of manganese-induced Parkinson's disease in Sprague Dawley rats. Biomed Pharmacother 2022; 153:113330. [PMID: 35780621 DOI: 10.1016/j.biopha.2022.113330] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 01/22/2023] Open
Abstract
The current study investigated the neuroprotective activity of some drugs and nutriceuticals with antioxidant and anti-inflammatory potential on the pathogenesis of Parkinson's disease (PD). Rats were categorized into seven groups: Rats received tween80 daily for 5 weeks as a control group, MnCl2 (10 mg/kg, i.p) either alone (group II) or in combination with vinpocetine (VIN) (20 mg/kg) (group III), punicalagin (PUN) (30 mg/kg) (group IV), niacin (85 mg/kg) (group V), vitamin E (Vit E) (100 mg/kg) (group VI) or their combination (group VII). Motor activities was examined using open-field and catalepsy. Striatal monamines, acetylcholinesterase, excitatory/inhibitory neurotransmitters, redox status, pro-oxidant content, brain inflammatory, apoptotic and antioxidant biomarkers levels were assessed. Besides, histopathological investigations of different brain regions were determined. Groups (IV -GVII) showed improved motor functions of PD rats. Applied drugs significantly increased the brain levels of monoamines with the strongest effect to PUN. Meanwhile, they significantly decreased levels of acetylcholinesterase with a strongest effect to PUN. Moreover, they exhibited significant neuronal protection and anti-inflammatory abilities through significant reduction of the brain levels of COX2, TNF-α and Il-1β with a strongest effect to the PUN. Interestingly; groups (IV - GVII) showed restored glutamate/GABA balance and exhibited a pronounced decrease in caspase-3 content and GSK-3β protein expression levels. In addition, they significantly increased Bcl2 mRNA expression levels with a strongest effect for PUN. All these findings were further confirmed by the histopathological examinations. As a conclusion, we propose VIN and PUN to mitigate the progression of PD via their antioxidant, anti-inflammatory, anti-apoptotic, neurotrophic and neurogenic activities.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Pharmacology and Toxicology Department (Girls), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | | | | | | | - Mohammed A F Elewa
- Biochemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Rehab Ali Elsayed Masoud
- Forensic Medicine and Clinical Toxicology Department, Faculty of medicine for girls, Al-Azhar University, Cairo, Egypt
| | - Azza A Ali
- Pharmacology and Toxicology Department (Girls), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohamed Othman
- Lecturer, Department of anatomy, Faculty of Medicine, King Salman International University, El-Tur Campus, Saini, Egypt
| | - Mona M Kamal
- Pharmacology and Toxicology Department (Girls), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Fatma Alzahraa M Hassan
- Biochemistry and molecular biology Department, Faculty of Pharmacy, Al-Azhar, University, Cairo, Egypt
| | - Mona G Khalil
- Pharmacology and Toxicology Department, Modern University for Technology and Information, Cairo, Egypt
| | - Ahmed M El-Sisi
- Biochemistry and Molecular Biology Department (boys), Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Nahda University (NUB), Beni-Suef, Egypt
| | - Manal M M Abdel Hady
- Department of Pharmacology, Faculty of Pharmacy, Qantra University, Sinai, Egypt
| | | | - Magdy M Awny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Cairo, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
17
|
Heme Oxygenase 1/Peroxisome Proliferator-Activated Receptor Gamma Pathway Protects Intimal Hyperplasia and Mitigates Arteriovenous Fistula Dysfunction by Regulating Oxidative Stress and Inflammatory Response. Cardiovasc Ther 2022; 2022:7576388. [PMID: 35812724 PMCID: PMC9207017 DOI: 10.1155/2022/7576388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/28/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose An arteriovenous fistula (AVF) is the preferred vascular access mode for maintenance hemodialysis, and access stenosis and thrombosis are the primary causes of AVF dysfunction. This study is aimed at exploring the molecular mechanisms underlying AVF development and the roles of the heme oxygenase 1/peroxisome proliferator-activated receptor gamma (HO-1/PPAR-γ) pathway in AVF. Method AVF model mice were established, and the vascular tissues from the arteriovenous anastomosis site were sent for mRNA sequencing. Differentially expressed mRNAs (DEmRNAs) were screened and subjected to functional analysis. Thereafter, the mice with HO-1 knockdown and coprotoporphyrin IX chloride (COPP) pretreatment were used to investigate the roles of the HO-1/PPAR-γ pathway in AVF. Results By sequencing, 2514 DEmRNAs, including 1323 upregulated and 1191 downregulated genes, were identified. These DEmRNAs were significantly enriched in the PPAR signaling pathway, AMPK signaling pathway, glucagon signaling pathway, IL-17 signaling pathway, and Toll-like receptor signaling pathway. High expression of HO-1 and PPAR-γ reduced endothelial damage and intimal hyperplasia during AVF maturation. After AVF was established, the levels of transforming growth factor-β (TGF-β), interleukin-1β (IL-1β), interleukin-18 (IL-18), and reactive oxygen species (ROS) were significantly increased (P < 0.05), and HO-1 normal expression and COPP pretreatment evidently decreased their levels in AVF (P < 0.05). Additionally, AVF significantly upregulated HO-1 and PPAR-γ and downregulated MMP9, and COPP pretreatment and HO-1 normal expression further upregulated and downregulated their expression. Conclusion The HO-1/PPAR-γ pathway may suppress intimal hyperplasia induced by AVF and protect the intima of blood vessels by regulating MMP9 and ROS, thus mitigating AVF dysfunction.
Collapse
|
18
|
Prabhakar P, Pavankumar GS, Raghu SV, Rao S, Prasad K, George T, Baliga MS. Utility of Indian fruits in cancer prevention and treatment: Time to undertake translational and bedside studies. Curr Pharm Des 2022; 28:1543-1560. [PMID: 35652402 DOI: 10.2174/1381612828666220601151931] [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: 12/16/2021] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
The World Health Organization predicts a 70% increase in cancer incidents in developing nations over the next decade, and it will be the second leading cause of death worldwide. Traditional plant-based medicine systems play an important role against various diseases and provide health care to a large section of the population in developing countries. Indigenous fruits and their bioactive compounds with beneficial effects like antioxidant, antiproliferative, and immunomodulatory are shown to be useful in preventing the incidence of cancer. India is one of the biodiversity regions and is native to numerous flora and fauna in the world. Of the many fruiting trees indigenous to India, Mango (Mangifera indica), Black plum (Eugenia jambolana or Syzygium jambolana), Indian gooseberry (Emblica officinalis or Phyllanthus emblica), kokum (Garcinia indica or Brindonia indica), stone apple or bael (Aegle marmelos), Jackfruit (Artocarpus heterophyllus), Karaunda (Carissa carandas) and Phalsa (Grewia asiatica), Monkey Jackfruit (Artocarpus lakoocha) and Elephant apple (Dillenia indica) have been shown to be beneficial in preventing cancer and in the treatment of cancer in validated preclinical models of study. In this review, efforts are also made to collate the fruits' anticancer effects and the important phytochemicals. Efforts are also made at emphasizing the underlying mechanism/s responsible for the beneficial effects in cancer prevention and treatment. These fruits have been a part of the diet, are non-toxic, and easily acceptable for human application. The plants and some of their phytochemicals possess diverse medicinal properties. The authors propose that future studies should be directed at detailed studies with various preclinical models of study with both composite fruit extract/juice and the individual phytochemicals. Additionally, translational studies should be planned with the highly beneficial, well-investigated and pharmacologically multifactorial amla to understand its usefulness as a cancer preventive in the high-risk population and as a supportive agent in cancer survivors. The outcome of both preclinical and clinical studies will be useful for patients, the healthcare fraternity, pharmaceutical, and agro-based sectors.
Collapse
Affiliation(s)
- Pankaj Prabhakar
- Department of Pharmacology, Indira Gandhi Institute of Medical Sciences (IGIMS), Sheikhpura, Patna, Bihar, 800014, India
| | - Giriyapura Srikantachar Pavankumar
- Department of Biotechnology, Kuvempu University, India.,Sri Lakshmi Group of Institution, Magadi Main Road, Sunkadakatte, Bengaluru, Karnataka, India
| | - Shamprasad Varija Raghu
- Department of Applied Zoology, Mangalore University, Mangalagangotri, Konaje, Karnataka India
| | - Suresh Rao
- Radiation Oncology, c Pumpwell, Mangalore, Karnataka, India
| | - Krishna Prasad
- Medical Oncology, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka, India
| | - Thomas George
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka, India
| | | |
Collapse
|
19
|
da Mata AMOF, Paz MFCJ, de Menezes AAPM, dos Reis AC, da Silva Souza B, de Carvalho Sousa CD, Machado SA, Medeiros TSG, Sarkar C, Islam MT, Sharifi-Rad J, Daştan SD, Alshehri MM, de Castro e Sousa JM, de Carvalho Melo Cavalcante AA. Evaluation of mutagenesis, necrosis and apoptosis induced by omeprazole in stomach cells of patients with gastritis. Cancer Cell Int 2022; 22:154. [PMID: 35436881 PMCID: PMC9016981 DOI: 10.1186/s12935-022-02563-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/30/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Gastritis is a superficial and prevalent inflammatory lesion that is considered a public health concern once can cause gastric ulcers and gastric cancer, especially when associated with Helicobacter pylori infection. Proton pump inhibitors, such as omeprazole, are the most widely used drugs to treat this illness. The aim of the study was evaluate cytogenetic effects of omeprazole in stomach epithelial cells of patients with gastritis in presence and absence of H. pylori, through cytogenetic biomarkers and catalse and superoxide dismutase analysis. METHODS The study included 152 patients from the Gastroenterology Outpatient Clinic of Hospital Getúlio Vargas, Teresina-Brazil, that reported continuous and prolonged omeprazole use in doses of 20, 30 and 40 mg/kg. The participants were divided into groups: (1) patients without gastritis (n = 32); (2) patients without gastritis but with OME use (n = 24); (3) patients with gastritis (n = 26); (4) patients with gastritis undergoing OME therapy (n = 26); (5) patients with gastritis and H. pylori (n = 22) and (6) patients with gastritis and H. pylori on OME therapy (n = 22). RESULTS OME induced cytogenetic imbalance in the stomach epithelium through the formation of micronuclei (group 6 > 1, 2, 3, 4, 5; group 5 > 1, 2, 3; group 4 > 1, 2, 3); bridges (groups 4 and 6 > 1, 2, 3, 5 and group 2 > 3, 5); buds (groups 2,4,6 > , 1, 3, 5); binucleated cells (group 6 > 1, 2, 3, 4, 5; group 4 > 1, 2, 3); (groups 2 and 3 > 1); picnoses (group 6 > 1, 2, 3, 4, 5), groups 2 and 5 > 1, 3; group 4 > 1, 2, 3, 5); cariorrexis (groups 6 and 4 > 1, 2, 3, 5; groups 2, 3, 5 > 1) and karyolysis (groups 2, 4, and 6 > 1, 3, 5; groups 3 and 5 > 1). The OME cytogenetic instability was associated with H. pylori infection, indicating clastogenic/aneugenic effects, chromosomes alterations, gene expression changes, cytotoxicity and apoptosis. CONCLUSIONS The cytogenetic changescan be attributed to several mechanisms that are still unclear, including oxidative damage, as observed by increased catalase and superoxide dismutase expresion. Positive correlations between antioxidant enzymes were found with micronuclei formation, and were negative for picnoses. Thus, the continuous and prolonged omeprazole use induces genetic instability, which can be monitored through cytogenetic analyzes, as precursor for gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chandan Sarkar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University Bangladesh, Gopalganj, 8100 Bangladesh
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University Bangladesh, Gopalganj, 8100 Bangladesh
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Sevgi Durna Daştan
- Department of Biology, Faculty of Science, Sivas Cumhuriyet University, 58140 Sivas, Turkey
- Beekeeping Development Application and Research Center, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | | | | |
Collapse
|
20
|
Awny MM, Al-Mokaddem AK, Ali BM. Mangiferin mitigates di-(2-ethylhexyl) phthalate-induced testicular injury in rats by modulating oxidative stress-mediated signals, inflammatory cascades, apoptotic pathways, and steroidogenesis. Arch Biochem Biophys 2021; 711:108982. [PMID: 34400143 DOI: 10.1016/j.abb.2021.108982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/13/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is an endocrine disruptor that causes reproductive defects in male animal models. This study was conducted to explore the plausible modulatory effects of mangiferin (MF) against DEHP-induced testicular injury in rats. Thirty-two adult male albino rats were allocated into four groups. Two groups were given DEHP (2 g/kg/day, p.o) for 14 days. One of these groups was treated with MF (20 mg/kg/day, i.p) for 7 days before and 14 days after DEHP administration. A vehicle-treated control was included, and another group of rats was given MF only. Results revealed that MF treatment suppressed oxidative testicular injury by amplifying the mRNA expression of nuclear factor-erythroid 2 related factor-2 (Nrf2) and increasing hemoxygenase-1 (HO-1), glutathione, and total antioxidant capacity (TAC) levels. This treatment also enhanced superoxide dismutase activity, but it decreased malondialdehyde and nitric oxide levels. MF had an anti-inflammatory characteristic, as demonstrated by the downregulation of the mRNA of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The content of tumor necrosis factor-alpha also decreased. MF modulated the apoptotic pathway by suppressing the mRNA of cytochrome c (Cyt c), Fas ligand content, Bax IHC expression, caspase-3 activity and cleaved caspase-3 IHC expression. It also upregulated the expression levels of heat-shock protein 70 (HSP70) and B-cell lymphoma 2. Moreover, MF upregulated the mRNA expression levels of HSP70 and c-kit and enriched the content of steroidogenic acute regulatory (StAR) protein, which were reflected in serum testosterone levels. This result indicated that MF played crucial roles in steroidogenesis and spermatogenesis. Besides, the activities of testicular marker enzymes, namely, acid and alkaline phosphatases, and lactate dehydrogenase, significantly increased. Histopathological observations provided evidence supporting the biochemical and molecular measurements. In conclusion, MF provided protective mechanisms against the DEHP-mediated deterioration of testicular functions partially through its antioxidant, anti-inflammatory, and anti-apoptotic properties. It also involved the restoration of steroidogenesis and spermatogenesis through the modulation of Nrf2/HO-1, NF-κB/Cyt c/HSP70, and c-Kit signaling cascades.
Collapse
Affiliation(s)
- Magdy M Awny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Cairo, Egypt.
| | - Asmaa K Al-Mokaddem
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Bassam Mohamed Ali
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Cairo, Egypt
| |
Collapse
|
21
|
Zhao W, Zhang X, Rong J. SUMOylation as a Therapeutic Target for Myocardial Infarction. Front Cardiovasc Med 2021; 8:701583. [PMID: 34395563 PMCID: PMC8355363 DOI: 10.3389/fcvm.2021.701583] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/01/2021] [Indexed: 12/23/2022] Open
Abstract
Myocardial infarction is a prevalent and life-threatening cardiovascular disease. The main goal of existing interventional therapies is to restore coronary reperfusion while few are designed to ameliorate the pathology of heart diseases via targeting the post-translational modifications of those critical proteins. Small ubiquitin-like modifier (SUMO) proteins are recently discovered to form a new type of protein post-translational modifications (PTM), known as SUMOylation. SUMOylation and deSUMOylation are dynamically balanced in the maintenance of various biological processes including cell division, DNA repair, epigenetic transcriptional regulation, and cellular metabolism. Importantly, SUMOylation plays a critical role in the regulation of cardiac functions and the pathology of cardiovascular diseases, especially in heart failure and myocardial infarction. This review summarizes the current understanding on the effects of SUMOylation and SUMOylated proteins in the pathophysiology of myocardial infarction and identifies the potential treatments against myocardial injury via targeting SUMO. Ultimately, this review recommends SUMOylation as a key therapeutic target for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Wei Zhao
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, University of Hong Kong, Hong Kong, China.,Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuying Zhang
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Jianhui Rong
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, University of Hong Kong, Hong Kong, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
22
|
Mannan A, Garg N, Singh TG, Kang HK. Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ɣ): Molecular Effects and Its Importance as a Novel Therapeutic Target for Cerebral Ischemic Injury. Neurochem Res 2021; 46:2800-2831. [PMID: 34282491 DOI: 10.1007/s11064-021-03402-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Cerebral ischemic injury is a leading cause of death and long-term disability throughout the world. Peroxisome proliferator-activated receptor gamma (PPAR-ɣ) is a ligand-activated nuclear transcription factor that is a member of the PPAR family. PPAR-ɣ has been shown in several in vitro and in vivo models to prevent post-ischemic inflammation and neuronal damage by negatively controlling the expression of genes modulated by cerebral ischemic injury, indicating a neuroprotective effect during cerebral ischemic injury. A extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the mechanistic role of Peroxisome proliferator activated receptor gamma and its modulation in Cerebral ischemic injury. PPAR-ɣ can interact with specific DNA response elements to control gene transcription and expression when triggered by its ligand. It regulates lipid metabolism, improves insulin sensitivity, modulates antitumor mechanisms, reduces oxidative stress, and inhibits inflammation. This review article provides insights on the current state of research into the neuroprotective effects of PPAR-ɣ in cerebral ischemic injury, as well as the cellular and molecular mechanisms by which these effects are modulated, such as inhibition of inflammation, reduction of oxidative stress, suppression of pro-apoptotic production, modulation of transcription factors, and restoration of injured tissue through neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Harmeet Kaur Kang
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
23
|
El Badawy SA, Ogaly HA, Abd-Elsalam RM, Azouz AA. Benzyl isothiocyanates modulate inflammation, oxidative stress, and apoptosis via Nrf2/HO-1 and NF-κB signaling pathways on indomethacin-induced gastric injury in rats. Food Funct 2021; 12:6001-6013. [PMID: 34037056 DOI: 10.1039/d1fo00645b] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The present study investigated the gastroprotective activity of benzyl isothiocyanates (BITC) on indomethacin (IND)-induced gastric injury in a rat model and explicated the possible involved biochemical, cellular, and molecular mechanisms. The rat model with gastric ulcers was established by a single oral dose of IND (30 mg per kg b.wt). BITC (0.75 and 1.5 mg kg-1) and esomeprazole (20 mg per kg b.wt) were orally administered for 3 weeks to rats before the induction of gastric injury. Compared with the IND group, BITC could diminish both the macroscopic and microscopic pathological morphology of gastric mucosa. BITC significantly preserved the antioxidants (glutathione GSH, superoxide dismutase SOD), nitric oxide (NO), and prostaglandin E2 (PGE2) contents, while decreasing the gastric mucosal malondialdehyde (MDA), tumor necrosis factor alpha (TNFα), and myeloperoxidase (MPO) contents. Moreover, BITC remarkably upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), hemoxygenase-1 (HO-1), and NAD(P)H : quinone oxidoreductase (NQO1). In addition, BITC activates the expression of heat shock protein 70 (HSP-70) and downregulated the expression of nuclear factor-κB (NF-κB) and caspase-3 to promote gastric mucosal cell survival. To the best of our knowledge, this study is the first published report to implicate the suppression of inflammation, oxidative stress, and Nrf2 signaling pathway as a potential mechanism for the gastroprotective activity of BITC.
Collapse
Affiliation(s)
- Shymaa A El Badawy
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hanan A Ogaly
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia. and Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Reham M Abd-Elsalam
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Asmaa A Azouz
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
24
|
Rahman Z, Dwivedi DK, Jena GB. The intervention of tert-butylhydroquinone protects ethanol-induced gastric ulcer in type II diabetic rats: the role of Nrf2 pathway. Can J Physiol Pharmacol 2021; 99:522-535. [PMID: 33095998 DOI: 10.1139/cjpp-2020-0173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ethanol consumption increases the prevalence of gastric ulcer (GU) in rats with type II diabetes (T2D). Induction of GU by absolute ethanol (5 mL/kg or 3.94 g/kg) in the animal model resembles human ulcer characteristics. The aim was to investigate the role of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in the treatment of GU in diabetic condition. The rats were exposed to absolute ethanol 1 h before sacrifice and T2D was induced by combined exposure of high-fat diet and low dose streptozotocin. Pretreatment of tert-butylhydroquinone (tBHQ) (25 and 50 mg/kg), metformin (500 mg/kg), and omeprazole (20 mg/kg) were given once daily for last three consecutive weeks. In ethanol-exposed diabetic rats, pretreatment with tBHQ, omeprazole, and metformin reduced gastric mucosal lesion, ulcer index, histological alterations, malondialdehyde level, and apoptosis. Furthermore, the intervention of tBHQ, omeprazole, and metformin improved the integrity of the stomach mucosa, glutathione, gastric pH, collagen, and goblet cells. tBHQ treatment improved ethanol-induced alterations of Nrf2, catalase, heat shock protein 70 (HSP70), NF-κB, and endothelin-1 expressions in diabetic rats. In diabetic conditions, the incidence of GU is increased due to elevated levels of reactive oxygen species, inflammatory mediators, depleted levels of cellular antioxidants, and altered gastric parameters. The tBHQ intervention could be a rational strategy to protect these changes.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Durgesh Kumar Dwivedi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - G B Jena
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| |
Collapse
|
25
|
Huang R, Zhang C, Wang X, Hu H. PPARγ in Ischemia-Reperfusion Injury: Overview of the Biology and Therapy. Front Pharmacol 2021; 12:600618. [PMID: 33995008 PMCID: PMC8117354 DOI: 10.3389/fphar.2021.600618] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a complex pathophysiological process that is often characterized as a blood circulation disorder caused due to various factors (such as traumatic shock, surgery, organ transplantation, burn, and thrombus). Severe metabolic dysregulation and tissue structure destruction are observed upon restoration of blood flow to the ischemic tissue. Theoretically, IRI can occur in various tissues and organs, including the kidney, liver, myocardium, and brain, among others. The advances made in research regarding restoring tissue perfusion in ischemic areas have been inadequate with regard to decreasing the mortality and infarct size associated with IRI. Hence, the clinical treatment of patients with severe IRI remains a thorny issue. Peroxisome proliferator-activated receptor γ (PPARγ) is a member of a superfamily of nuclear transcription factors activated by agonists and is a promising therapeutic target for ameliorating IRI. Therefore, this review focuses on the role of PPARγ in IRI. The protective effects of PPARγ, such as attenuating oxidative stress, inhibiting inflammatory responses, and antagonizing apoptosis, are described, envisaging certain therapeutic perspectives.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xing Wang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Mei S, Ma H, Chen X. Anticancer and anti-inflammatory properties of mangiferin: A review of its molecular mechanisms. Food Chem Toxicol 2021; 149:111997. [DOI: 10.1016/j.fct.2021.111997] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
|
27
|
Morozkina SN, Nhung Vu TH, Generalova YE, Snetkov PP, Uspenskaya MV. Mangiferin as New Potential Anti-Cancer Agent and Mangiferin-Integrated Polymer Systems-A Novel Research Direction. Biomolecules 2021; 11:79. [PMID: 33435313 PMCID: PMC7827323 DOI: 10.3390/biom11010079] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
For a long time, the pharmaceutical industry focused on natural biologically active molecules due to their unique properties, availability and significantly less side-effects. Mangiferin is a naturally occurring C-glucosylxantone that has substantial potential for the treatment of various diseases thanks to its numerous biological activities. Many research studies have proven that mangiferin possesses antioxidant, anti-infection, anti-cancer, anti-diabetic, cardiovascular, neuroprotective properties and it also increases immunity. It is especially important that it has no toxicity. However, mangiferin is not being currently applied to clinical use because its oral bioavailability as well as its absorption in the body are too low. To improve the solubility, enhance the biological action and bioavailability, mangiferin integrated polymer systems have been developed. In this paper, we review molecular mechanisms of anti-cancer action as well as a number of designed polymer-mangiferin systems. Taking together, mangiferin is a very promising anti-cancer molecule with excellent properties and the absence of toxicity.
Collapse
Affiliation(s)
- Svetlana N. Morozkina
- Institute BioEngineering, ITMO University, Kronverkskiy Prospekt, 49A, 197101 Saint-Petersburg, Russia; (T.H.N.V.); (P.P.S.); (M.V.U.)
| | - Thi Hong Nhung Vu
- Institute BioEngineering, ITMO University, Kronverkskiy Prospekt, 49A, 197101 Saint-Petersburg, Russia; (T.H.N.V.); (P.P.S.); (M.V.U.)
| | - Yuliya E. Generalova
- Department of Analytical Chemistry, Faculty of Industrial Technology of Dosage Forms, Saint Petersburg State Chemical Pharmaceutical University, Prof. Popova Street 14A, 197022 Saint-Petersburg, Russia;
| | - Petr P. Snetkov
- Institute BioEngineering, ITMO University, Kronverkskiy Prospekt, 49A, 197101 Saint-Petersburg, Russia; (T.H.N.V.); (P.P.S.); (M.V.U.)
| | - Mayya V. Uspenskaya
- Institute BioEngineering, ITMO University, Kronverkskiy Prospekt, 49A, 197101 Saint-Petersburg, Russia; (T.H.N.V.); (P.P.S.); (M.V.U.)
| |
Collapse
|
28
|
Gunter NV, Teh SS, Lim YM, Mah SH. Natural Xanthones and Skin Inflammatory Diseases: Multitargeting Mechanisms of Action and Potential Application. Front Pharmacol 2020; 11:594202. [PMID: 33424605 PMCID: PMC7793909 DOI: 10.3389/fphar.2020.594202] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of skin inflammatory diseases such as atopic dermatitis, acne, psoriasis, and skin cancers generally involve the generation of oxidative stress and chronic inflammation. Exposure of the skin to external aggressors such as ultraviolet (UV) radiation and xenobiotics induces the generation of reactive oxygen species (ROS) which subsequently activates immune responses and causes immunological aberrations. Hence, antioxidant and anti-inflammatory agents were considered to be potential compounds to treat skin inflammatory diseases. A prime example of such compounds is xanthone (xanthene-9-one), a class of natural compounds that possess a wide range of biological activities including antioxidant, anti-inflammatory, antimicrobial, cytotoxic, and chemotherapeutic effects. Many studies reported various mechanisms of action by xanthones for the treatment of skin inflammatory diseases. These mechanisms of action commonly involve the modulation of various pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor α (TNF-α), as well as anti-inflammatory cytokines such as IL-10. Other mechanisms of action include the regulation of NF-κB and MAPK signaling pathways, besides immune cell recruitment via modulation of chemokines, activation, and infiltration. Moreover, disease-specific activity contributed by xanthones, such as antibacterial action against Propionibacterium acnes and Staphylococcus epidermidis for acne treatment, and numerous cytotoxic mechanisms involving pro-apoptotic and anti-metastatic effects for skin cancer treatment have been extensively elucidated. Furthermore, xanthones have been reported to modulate pathways responsible for mediating oxidative stress and inflammation such as PPAR, nuclear factor erythroid 2-related factor and prostaglandin cascades. These pathways were also implicated in skin inflammatory diseases. Xanthones including the prenylated α-mangostin (2) and γ-mangostin (3), glucosylated mangiferin (4) and the caged xanthone gambogic acid (8) are potential lead compounds to be further developed into pharmaceutical agents for the treatment of skin inflammatory diseases. Future studies on the structure-activity relationships, molecular mechanisms, and applications of xanthones for the treatment of skin inflammatory diseases are thus highly recommended.
Collapse
Affiliation(s)
| | - Soek Sin Teh
- Engineering and Processing Division, Energy and Environment Unit, Malaysian Palm Oil Board, Kajang, Malaysia
| | - Yang Mooi Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Malaysia.,Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Malaysia
| | - Siau Hui Mah
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia.,Centre for Drug Discovery and Molecular Pharmacology, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
29
|
|
30
|
Geck MS, Cristians S, Berger-González M, Casu L, Heinrich M, Leonti M. Traditional Herbal Medicine in Mesoamerica: Toward Its Evidence Base for Improving Universal Health Coverage. Front Pharmacol 2020; 11:1160. [PMID: 32848768 PMCID: PMC7411306 DOI: 10.3389/fphar.2020.01160] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/16/2020] [Indexed: 01/28/2023] Open
Abstract
The quality of health care in Mesoamerica is influenced by its rich cultural diversity and characterized by social inequalities. Especially indigenous and rural communities confront diverse barriers to accessing formal health services, leading to often conflicting plurimedical systems. Fostering integrative medicine is a fundamental pillar for achieving universal health coverage (UHC) for marginalized populations. Recent developments toward health sovereignty in the region are concerned with assessing the role of traditional medicines, and particularly herbal medicines, to foster accessible and culturally pertinent healthcare provision models. In Mesoamerica, as in most regions of the world, a wealth of information on traditional and complementary medicine has been recorded. Yet these data are often scattered, making it difficult for policy makers to regulate and integrate traditionally used botanical products into primary health care. This critical review is based on a quantitative analysis of 28 survey papers focusing on the traditional use of botanical drugs in Mesoamerica used for the compilation of the "Mesoamerican Medicinal Plant Database" (MAMPDB), which includes a total of 12,537 use-records for 2188 plant taxa. Our approach presents a fundamental step toward UHC by presenting a pharmacological and toxicological review of the cross-culturally salient plant taxa and associated botanical drugs used in traditional medicine in Mesoamerica. Especially for native herbal drugs, data about safety and effectiveness are limited. Commonly used cross-culturally salient botanical drugs, which are considered safe but for which data on effectiveness is lacking constitute ideal candidates for treatment outcome studies.
Collapse
Affiliation(s)
- Matthias S. Geck
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- Biovision – Foundation for Ecological Development, Zurich, Switzerland
| | - Sol Cristians
- Botanical Garden, Institute of Biology, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mónica Berger-González
- Centro de Estudios en Salud, Universidad del Valle de Guatemala, Guatemala, Guatemala
- Department of Epidemiology and Public Heath, Swiss TPH, University of Basel, Basel, Switzerland
| | - Laura Casu
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Michael Heinrich
- Pharmacognosy and Phytotherapy, UCL School of Pharmacy, London, United Kingdom
| | - Marco Leonti
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
31
|
Mangiferin Alleviates Ovalbumin-Induced Allergic Rhinitis via Nrf2/HO-1/NF-κB Signaling Pathways. Int J Mol Sci 2020; 21:ijms21103415. [PMID: 32408566 PMCID: PMC7279452 DOI: 10.3390/ijms21103415] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Mangiferin (MF), extracted from mango trees, is considered to have anti-inflammatory, anti-apoptotic, and antioxidant effects. However, its effects on allergic rhinitis (AR), remain unclear. We investigated the mechanisms underlying the protective action of MF in ovalbumin (OVA)-induced AR models. AR was induced by OVA challenge in BALB/c mice. Prior to this, MF and dexamethasone were administered. Mice were examined for nasal mucosal inflammation, the generation of allergen-specific cytokine response, and histopathological changes in the nasal mucosa and lung tissue. MF ameliorated nasal symptoms and nasal mucosa inflammation in OVA-induced AR and reduced inflammatory cell infiltration and epithelial disruption in these tissues. MF inhibited the overproduction of Th2/Th17 cytokines and transcription factors. MF downregulated the HO-1/Nrf2 pathways, reduced oxidative stress biomarker levels, and the NF-κB signaling pathways were inhibited. MF exerts protective effects in AR by inhibiting NF-κB and activating HO-1/Nrf2 pathways. MF could be used for the treatment of AR.
Collapse
|
32
|
Rahman Z, Dwivedi DK, Jena GB. Ethanol-induced gastric ulcer in rats and intervention of tert-butylhydroquinone: Involvement of Nrf2/HO-1 signalling pathway. Hum Exp Toxicol 2020; 39:547-562. [PMID: 31876185 DOI: 10.1177/0960327119895559] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gastric ulcer (GU) is the most common health concern that occurs due to alcohol consumption, smoking and physiological stress. Ethanol-induced GU in animal model resembles the pathophysiology of human ulcer. The present study was designed to investigate the cytoprotective and anti-inflammatory properties of tert-butylhydroquinone (tBHQ), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, against gastric mucosal damage induced by acute exposure of ethanol (5 ml/kg). The intervention of tBHQ (25 and 50 mg/kg, per os (po)) and omeprazole (20 mg/kg, po) was done for 10 consecutive days. Omeprazole was chosen as a standard drug because it is prescribed for the treatment of GU. Pretreatment of tBHQ decreased gastric mucosal lesion, ulcer index, apoptotic cells and lipid peroxidation level induced by ethanol. Furthermore, the intervention of tBHQ increased gastric mucosa integrity, pH, reduced glutathione, collagen and mucus-producing goblet cells. Intervention of tBHQ increased the expression of antioxidant markers such as Nrf2, haeme oxygenase-1 and catalase and decreased the expressions of inflammatory markers such as nuclear factor kappa-light-chain-enhancer of activated B cells and cyclooxygenase-2. The cytoprotective potential of tBHQ against gastric mucosal damage might be due to its ability to enhance cellular antioxidants and anti-inflammatory responses.
Collapse
Affiliation(s)
- Z Rahman
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - D K Dwivedi
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - G B Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| |
Collapse
|
33
|
Paz MFCJ, de Alencar MVOB, de Lima RMP, Sobral ALP, do Nascimento GTM, dos Reis CA, Coêlho MDPSDS, do Nascimento MLLB, Gomes Júnior AL, Machado KDC, de Menezes AAPM, de Lima RMT, de Oliveira Filho JWG, Dias ACS, dos Reis AC, da Mata AMOF, Machado SA, Sousa CDDC, da Silva FCC, Islam MT, de Castro e Sousa JM, Melo Cavalcante AADC. Pharmacological Effects and Toxicogenetic Impacts of Omeprazole: Genomic Instability and Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3457890. [PMID: 32308801 PMCID: PMC7146093 DOI: 10.1155/2020/3457890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/19/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022]
Abstract
Omeprazole (OME) is commonly used to treat gastrointestinal disorders. However, long-term use of OME can increase the risk of gastric cancer. We aimed to characterize the pharmacological effects of OME and to correlate its adverse effects and toxicogenetic risks to the genomic instability mechanisms and cancer-based on database reports. Thus, a search (till Aug 2019) was made in the PubMed, Scopus, and ScienceDirect with relevant keywords. Based on the study objective, we included 80 clinical reports, forty-six in vitro, and 76 in vivo studies. While controversial, the findings suggest that long-term use of OME (5 to 40 mg/kg) can induce genomic instability. On the other hand, OME-mediated protective effects are well reported and related to proton pump blockade and anti-inflammatory activity through an increase in gastric flow, anti-inflammatory markers (COX-2 and interleukins) and antiapoptotic markers (caspases and BCL-2), glycoprotein expression, and neutrophil infiltration reduction. The reported adverse and toxic effects, especially in clinical studies, were atrophic gastritis, cobalamin deficiencies, homeostasis disorders, polyp development, hepatotoxicity, cytotoxicity, and genotoxicity. This study highlights that OME may induce genomic instability and increase the risk of certain types of cancer. Therefore, adequate precautions should be taken, especially in its long-term therapeutic strategies and self-medication practices.
Collapse
Affiliation(s)
- Márcia Fernanda Correia Jardim Paz
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, PI, Brazil
- Laboratory of Genetic Toxicity, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, PI, Brazil
| | | | | | - André Luiz Pinho Sobral
- Laboratory of Genetic Toxicity, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, PI, Brazil
- University Hospital, Teresina, PI, Brazil
| | | | | | | | | | - Antonio Luiz Gomes Júnior
- Laboratory of Genetic Toxicity, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, PI, Brazil
- University Centre UNINOVAFAPI, Teresina, PI, Brazil
| | | | | | - Rosália Maria Torres de Lima
- Laboratory of Genetic Toxicity, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, PI, Brazil
| | | | - Ana Carolina Soares Dias
- Laboratory of Genetics and Molecular Biology, Federal University of Maranhão, São Luís, MA, Brazil
| | - Antonielly Campinho dos Reis
- Laboratory of Genetic Toxicity, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, PI, Brazil
| | | | | | | | - Felipe Cavalcanti Carneiro da Silva
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, PI, Brazil
- Department of Biological Sciences, Federal University of Piauí, Picos, PI, Brazil
| | - Muhammad Torequl Islam
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | | | - Ana Amélia de Carvalho Melo Cavalcante
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, PI, Brazil
- Laboratory of Genetic Toxicity, Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, PI, Brazil
| |
Collapse
|
34
|
Ko IG, Jin JJ, Hwang L, Kim SH, Kim CJ, Han JH, Kwak MS, Yoon JY, Jeon JW. Evaluating the mucoprotective effect of polydeoxyribonucleotide against indomethacin-induced gastropathy via the MAPK/NF-κB signaling pathway in rats. Eur J Pharmacol 2020; 874:172952. [PMID: 31996319 DOI: 10.1016/j.ejphar.2020.172952] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 12/23/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) cause gastric mucosal damage and gastric ulceration. Among the most commonly used NSAIDs, indomethacin upregulates mucosal tumor necrosis factor-α, which activates nuclear factor-kappa B (NF-κB), and mitogen-activated protein kinases (MAPK) to induce various pro-inflammatory mediators. Polydeoxyribonucleotide (PDRN) is an adenosine A2A receptor agonist that exerts anti-inflammatory effects. In this study, we evaluated the efficacy of PDRN in the initial treatment of gastropathy against that of ecabet sodium and irsoglandin maleate, which are commonly used medications. The rats were administrated indomethacin once a day for 7 days after 24 h of fasting to induce gastropathy. Rats in the drug-treated groups were orally administrated 500 μl of distilled water containing the drug once daily for 7 days 1 h after indomethacin administration. Indomethacin administration caused mucosal damage and increased pro-inflammatory cytokine release. Both NF-κB and MAPK cascade factors were increased by indomethacin administration. PDRN therapy more potently suppressed the expressions of NF-κB and MAPK cascade factors compared to other drugs. The expression of cyclic adenosine-3',5'-monophosphate was also increased by PDRN treatment in the indomethacin-induced gastropathy rats. These changes led to a reduction in pro-inflammatory cytokines and apoptotic factors, which ultimately promote recovery of damaged gastric tissue. Therefore, PDRN may serve as a new therapeutic option in the initial treatment of NSAIDs-induced gastropathy.
Collapse
Affiliation(s)
- Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Lakkyong Hwang
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sang-Hoon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jin Hee Han
- Department of Anesthesiology and Pain Medicine, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Min Seop Kwak
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, 05278, Republic of Korea
| | - Jin Young Yoon
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, 05278, Republic of Korea
| | - Jung Won Jeon
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, 05278, Republic of Korea.
| |
Collapse
|
35
|
Garrido-Suárez BB, Garrido G, Piñeros O, Delgado-Hernández R. Mangiferin: Possible uses in the prevention and treatment of mixed osteoarthritic pain. Phytother Res 2019; 34:505-525. [PMID: 31755173 DOI: 10.1002/ptr.6546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 10/07/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) pain has been proposed to be a mixed pain state, because in some patients, central nervous system factors are superimposed upon the more traditional peripheral factors. In addition, a considerable amount of preclinical and clinical evidence has shown that, accompanying the central neuroplasticity changes and partially driven by a peripheral nociceptive input, a real neuropathic component occurs that are particularly linked to disease severity and progression. Hence, innovative strategies targeting neuroprotection and particularly neuroinflammation to prevent and treat OA pain could be introduced. Mangiferin (MG) is a glucosylxanthone that is broadly distributed in higher plants, such as Mangifera indica L. Previous studies have documented its analgesic, anti-inflammatory, antioxidant, neuroprotective, and immunomodulatory properties. In this paper, we propose its potential utility as a multitargeted compound for mixed OA pain, even in the context of multimodal pharmacotherapy. This hypothesis is supported by three main aspects: the cumulus of preclinical evidence around this xanthone, some preliminary clinical results using formulations containing MG in clinical musculoskeletal or neuropathic pain, and by speculations regarding its possible mechanism of action according to recent advances in OA pain knowledge.
Collapse
Affiliation(s)
- Bárbara B Garrido-Suárez
- Laboratorio de Farmacología y Toxicología, Centro de Investigación y Desarrollo de Medicamentos, Havana, Cuba
| | - Gabino Garrido
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Universidad Católica del Norte, Antofagasta, Chile
| | - Octavio Piñeros
- Departamento de Investigaciones, Universidad de Santiago de Cali, Cali, Colombia
| | - René Delgado-Hernández
- Centro de Estudio para las Investigaciones y Evaluaciones Biológicas, Instituto de Farmacia y Alimentos, Universidad de La Habana, Havana, Cuba
| |
Collapse
|
36
|
Wang Z, Chen Z, Jiang Z, Luo P, Liu L, Huang Y, Wang H, Wang Y, Long L, Tan X, Liu D, Jin T, Wang Y, Wang Y, Liao F, Zhang C, Chen L, Gan Y, Liu Y, Yang F, Huang C, Miao H, Chen J, Cheng T, Fu X, Shi C. Cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents. Nat Commun 2019; 10:2538. [PMID: 31182708 PMCID: PMC6557849 DOI: 10.1038/s41467-019-10386-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 05/08/2019] [Indexed: 12/13/2022] Open
Abstract
The pathological mechanisms of radiation ulcer remain unsolved and there is currently no effective medicine. Here, we demonstrate that persistent DNA damage foci and cell senescence are involved in radiation ulcer development. Further more, we identify cordycepin, a natural nucleoside analogue, as a potent drug to block radiation ulcer (skin, intestine, tongue) in rats/mice by preventing cell senescence through the increase of NRF2 nuclear expression (the assay used is mainly on skin). Finally, cordycepin is also revealed to activate AMPK by binding with the α1 and γ1 subunit near the autoinhibitory domain of AMPK, then promotes p62-dependent autophagic degradation of Keap1, to induce NRF2 dissociate from Keap1 and translocate to the nucleus. Taken together, our findings identify cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents, and activation of AMPK or NRF2 may thus represent therapeutic targets for preventing cell senescence and radiation ulcer.
Collapse
Affiliation(s)
- Ziwen Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Zelin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Zhongyong Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Peng Luo
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Lang Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Yu Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Huilan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Institute of Clinical Medicine, Southwest Medical University, 646000, Luzhou, China
| | - Yu Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Lei Long
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xu Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Dengqun Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Taotao Jin
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yawei Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Fengying Liao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Long Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yibo Gan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yunsheng Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Fan Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chunji Huang
- College of Basic Medical Sciences, Third Military Medical University, 400038, Chongqing, China
| | - Hongming Miao
- College of Basic Medical Sciences, Third Military Medical University, 400038, Chongqing, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University, 40038, Chongqing, China
| | - Tianmin Cheng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, the First Affiliated Hospital, Chinese PLA General Hospital, Trauma Center of Postgraduate Medical College, 100000, Beijing, China.
| | - Chunmeng Shi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| |
Collapse
|
37
|
Byrsonima intermedia A. Juss partitions promote gastroprotection against peptic ulcers and improve healing through antioxidant and anti-inflammatory activities. Biomed Pharmacother 2019; 111:1112-1123. [DOI: 10.1016/j.biopha.2018.12.132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/21/2018] [Accepted: 12/30/2018] [Indexed: 12/16/2022] Open
|
38
|
Xie B, Liu X, Yang J, Cheng J, Gu J, Xue S. PIAS1 protects against myocardial ischemia-reperfusion injury by stimulating PPARγ SUMOylation. BMC Cell Biol 2018; 19:24. [PMID: 30419807 PMCID: PMC6233564 DOI: 10.1186/s12860-018-0176-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/25/2018] [Indexed: 01/07/2023] Open
Abstract
Background Myocardial ischemia-reperfusion injury (IRI) has become one of the most serious complications after reperfusion therapy in patients with acute myocardial infarction. Small ubiquitin-like modification (SUMOylation) is a reversible process, including SUMO E1-, E2-, and E3-mediated SUMOylation and SUMO-specific protease-mediated deSUMOylation, with the latter having been shown to play a vital role in myocardial IRI previously. However, little is known about the function and regulation of SUMO E3 ligases in myocardial IRI. Results In this study, we found dramatically decreased expression of PIAS1 after ischemia/reperfusion (I/R) in mouse myocardium and H9C2 cells. PIAS1 deficiency aggravated apoptosis and inflammation of cardiomyocytes via activating the NF-κB pathway after I/R. Mechanistically, we identified PIAS1 as a specific E3 ligase for PPARγ SUMOylation. Moreover, H9C2 cells treated with hypoxia/reoxygenation (H/R) displayed reduced PPARγ SUMOylation as a result of down-regulated PIAS1, and act an anti-apoptotic and anti-inflammatory function through repressing NF-κB activity. Finally, overexpression of PIAS1 in H9C2 cells could remarkably ameliorate I/R injury. Conclusions Collectively, our findings demonstrate the crucial role of PIAS1-mediated PPARγ SUMOylation in protecting against myocardial IRI. Electronic supplementary material The online version of this article (10.1186/s12860-018-0176-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Xie
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Xinyu Liu
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 201700, 1158 East Gongyuan Road, Shanghai, China
| | - Jie Yang
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, 200025, 280 South Chongqing Road, Shanghai, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory for Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, 280 South Chongqing Road, Shanghai, China
| | - Jianmin Gu
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China.
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
39
|
Garrido-Suárez BB, Garrido G, Castro-Labrada M, Pardo-Ruíz Z, Bellma Menéndez A, Spencer E, Godoy-Figueiredo J, Ferreira SH, Delgado-Hernández R. Anti-allodynic Effect of Mangiferin in Rats With Chronic Post-ischemia Pain: A Model of Complex Regional Pain Syndrome Type I. Front Pharmacol 2018; 9:1119. [PMID: 30333751 PMCID: PMC6176059 DOI: 10.3389/fphar.2018.01119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 09/13/2018] [Indexed: 12/29/2022] Open
Abstract
The present study reproduces chronic post-ischemia pain (CPIP), a model of complex regional pain syndrome type I (CRPS-I), in rats to examine the possible transient and long-term anti-allodynic effect of mangiferin (MG); as well as its potential beneficial interactions with some standard analgesic drugs and sympathetic-mediated vasoconstriction and vasodilator agents during the earlier stage of the pathology. A single dose of MG (50 and 100 mg/kg, p.o.) decreased mechanical allodynia 72 h post-ischemia-reperfusion (I/R). MG 100 mg/kg, i.p. (pre- vs. post-drug) increased von Frey thresholds in a yohimbine and naloxone-sensitive manner. Sub-effective doses of morphine, amitriptyline, prazosin, clonidine and a NO donor, SIN-1, in the presence of MG were found to be significantly anti-allodynic. A long-term anti-allodynic effect at 7 and 13 days post-I/R after repeated oral doses of MG (50 and 100 mg/kg) was also observed. Further, MG decreased spinal and muscle interleukin-1β concentration and restored muscle redox status. These results indicate that MG has a transient and long-term anti-allodynic effect in CPIP rats that appears to be at least partially attributable to the opioid and α2 adrenergic receptors. Additionally, its anti-inflammatory and antioxidant mechanisms could also be implicated in this effect. The association of MG with sub-effective doses of these drugs enhances the anti-allodynic effect; however, an isobolographic analysis should be performed to define a functional interaction between them. These findings suggest the possible clinical use of MG in the treatment of CRPS-I in both early sympathetically maintained pain and long-term sympathetically independent pain.
Collapse
Affiliation(s)
- Bárbara B. Garrido-Suárez
- Laboratorio de Farmacología y Toxicología, Centro de Investigación y Desarrollo de Medicamentos, Havana, Cuba
| | - Gabino Garrido
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Universidad Católica del Norte, Antofagasta, Chile
| | - Marian Castro-Labrada
- Laboratorio de Farmacología y Toxicología, Centro de Investigación y Desarrollo de Medicamentos, Havana, Cuba
| | - Zenia Pardo-Ruíz
- Laboratorio de Farmacología y Toxicología, Centro de Investigación y Desarrollo de Medicamentos, Havana, Cuba
| | - Addis Bellma Menéndez
- Laboratorio de Farmacología y Toxicología, Centro de Investigación y Desarrollo de Medicamentos, Havana, Cuba
| | - Evelyn Spencer
- Laboratorio de Farmacología y Toxicología, Centro de Investigación y Desarrollo de Medicamentos, Havana, Cuba
| | - Jozi Godoy-Figueiredo
- Department of Pharmacology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Sergio H. Ferreira
- Department of Pharmacology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - René Delgado-Hernández
- Centro de Estudio para las Investigaciones y Evaluaciones Biológicas, Instituto de Farmacia y Alimentos, Universidad de La Habana, Havana, Cuba
| |
Collapse
|
40
|
A modified pectic polysaccharide from turmeric (Curcuma longa) with antiulcer effects via anti–secretary, mucoprotective and IL–10 mediated anti–inflammatory mechanisms. Int J Biol Macromol 2018; 118:864-880. [PMID: 29924982 DOI: 10.1016/j.ijbiomac.2018.06.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/03/2018] [Accepted: 06/11/2018] [Indexed: 12/15/2022]
|
41
|
Mangiferin attenuates cisplatin-induced acute kidney injury in rats mediating modulation of MAPK pathway. Mol Cell Biochem 2018; 452:141-152. [DOI: 10.1007/s11010-018-3420-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/28/2018] [Indexed: 02/08/2023]
|
42
|
Pal R, Chaudhary MJ, Tiwari PC, Nath R, Pant KK. Pharmacological and biochemical studies on protective effects of mangiferin and its interaction with nitric oxide (NO) modulators in adjuvant-induced changes in arthritic parameters, inflammatory, and oxidative biomarkers in rats. Inflammopharmacology 2018; 27:10.1007/s10787-018-0507-8. [PMID: 29934863 DOI: 10.1007/s10787-018-0507-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/09/2018] [Indexed: 10/28/2022]
Abstract
Current study was designed to evaluate protective effect of mangiferin and its interaction with low dose of nitric oxide (NO) modulators in complete Freund's adjuvant (CFA) inoculated rats. Male wistar rats (200-300 g, n = 8 per group) were used in the study. On day ''0'' of study arthritis was induced in rats by injecting 0.2 ml CFA in sub-planter region of right hind paw of animals. Treatment with methotrexate (5 mg/kg), mangiferin (10-30 mg/kg) alone and in combination with NO modulators was given (i.p.) from days 14 to 28. After 28 days, blood and joint synovial fluid was collected for biochemical analysis and rat paws were excised to estimate MDA and SOD in tissue (paw) homogenates. CFA inoculation significantly increases (1) arthritic index, (2) ankle diameter, (3) paw volume, and (4) serum TNF-α, IL-6, IL-1β, and synovial TNF-α levels (p < 0.001). The serum Th1 (IFN-γ) and Th2 (IL-4) cytokine levels, MDA levels in rat paw tissue homogenates and serum NF-κB levels were also found significantly increased. Significant decrease in serum IL-10 levels and SOD activity was found after CFA inoculation. These CFA-induced arthritic changes, cytokine profile, and oxidative stress markers were significantly reversed by mangiferin (10-30 mg/kg) treatment alone and in combination with L-arginine and L-NAME nitric oxide modulators (p < 0.05). Treatment with methotrexate (5 mg/kg) also significantly reversed these adjuvant changes (p < 0.05). However, effect of methotrexate was less marked as compared to mangiferin (30 mg/kg) alone and in combination with L-NAME (10 mg/kg), but was comparable or slightly better than mangiferin (10 and 20 mg/kg). Thus, on the basis of our findings, we can suggest that interaction of mangiferin with nitric oxide modulators may have therapeutic value for chronic inflammatory disease such as RA.
Collapse
Affiliation(s)
- Rishi Pal
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India.
| | - Manju J Chaudhary
- Department of Physiology, Government Medical College, Tirwa Road, Kannauj, UP, India
| | - Prafulla Chandra Tiwari
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| | - Rajendra Nath
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| | - Kamlesh Kumar Pant
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| |
Collapse
|
43
|
Sadhukhan P, Saha S, Dutta S, Sil PC. Mangiferin Ameliorates Cisplatin Induced Acute Kidney Injury by Upregulating Nrf-2 via the Activation of PI3K and Exhibits Synergistic Anticancer Activity With Cisplatin. Front Pharmacol 2018; 9:638. [PMID: 29967581 PMCID: PMC6015878 DOI: 10.3389/fphar.2018.00638] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Occurrence of oxidative stress is the principal cause of acute kidney injury induced by cisplatin. Mangiferin, a naturally occurring antioxidant molecule, is found to ameliorate several oxidative stress mediated pathophysiological conditions including cancer. Cisplatin induced cytotoxicity was measured in NKE cells by MTT assay and microscopic analysis. Induction of oxidative stress and regulation of proapoptotic molecules were subsequently investigated by using different spectrophotometric analyses, FACS and immunocytochemistry. Induction of nephrotoxicity was determined by analyzing different serum biomarkers and histological parameters in vivo using swiss albino mice. Activation of NF-κB mediated pro-inflammatory and caspase dependent signaling cascades were investigated by semi-quantitative RT-PCR and immunoblotting. Mangiferin was found to ameliorate cisplatin induced nephrotoxicity in vitro and in vivo by attenuating the induction of oxidative stress and upregulating Nrf-2 mediated pro-survival signaling cascades via the activation of PI3K. Additionally, mangiferin showed synergistic anticancer activity with cisplatin in cancer cell lines (MCF-7 and SKRC-45) and EAC cell induced solid tumor bearing experimental mice. The ameliorative effect of mangiferin is primarily attributed to its anti-oxidant and anti-inflammatory properties. It acts differentially in normal tissue cells and tumor cells by modulating different cell survival regulatory signaling molecules. For the first time, the study reveals a mechanistic basis of mangiferin action against cisplatin induced nephrotoxicity. Since Mangiferin shows synergistic anticancer activity with cisplatin, it can be considered as a promising drug candidate, to be used in combination with cisplatin.
Collapse
Affiliation(s)
| | | | | | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
44
|
Piao X, Li S, Sui X, Guo L, Liu X, Li H, Gao L, Cai S, Li Y, Wang T, Liu B. 1-Deoxynojirimycin (DNJ) Ameliorates Indomethacin-Induced Gastric Ulcer in Mice by Affecting NF-kappaB Signaling Pathway. Front Pharmacol 2018; 9:372. [PMID: 29725297 PMCID: PMC5917448 DOI: 10.3389/fphar.2018.00372] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/03/2018] [Indexed: 12/27/2022] Open
Abstract
Gastric ulcer (GU) is a main threat to public health. 1-Deoxynojirimycin (DNJ) has antioxidant and anti-inflammatory properties and may prevent GU but related mechanism remains unclear. DNJ was extracted from the supernatants of Bacillus subtilis by using ethanol and purified by using CM-Sepharose chromatography. A GU mouse model was induced by indomethacin. The functional role of DNJ in GU mice was explored by measuring the main molecules in the NF-KappaB pathway. After the model establishment, 40 GU mice were evenly assigned into five categories: IG (received vehicle control), LG (10 μg DNJ daily), MG (20 μg DNJ daily), HG (40 μg DNJ daily), and RG (0.5 mg ranitidine daily). Meanwhile, eight healthy mice were assigned as a control group (CG). After 1-month therapy, weight and gastric volume were investigated. The levels of serum inflammatory cytokines (IL-6 and TNF-α), antioxidant indices [superoxide dismutase (SOD), catalase (CAT), and reduced glutathione (GSH)], and oxidant biomarker malondialdehyde (MDA) were examined via ELISA. Meanwhile, inflammatory cytokine (IL-6 and TNF-α) levels, and key molecules (NF-κB p65), cyclooxygenase 1 (COX-1 and COX2) involved in NF-κB pathway, were analyzed by using Western Blot. COX-1 and COX-2 levels were further measured by immunohistochemistry. The effects of DNJ on gastric functions were explored by measuring the changes of Motilin (MOT), Substance P (SP), Somatostatin (SS), and Vasoactive intestinal peptide (VIP) in GU mouse models with ELISA Kits. The results indicated that DNJ prevented indomethacin-caused increase of gastric volume. DNJ improved histopathology of GU mice when compared with the mice from IG group (P < 0.05). DNJ consumption decreased the levels of IL-6 and TNF-α (P < 0.05). DNJ increased antioxidant indices of GU mice by improving the activities of SOD, CAT and reduced GSH, and reduced MDA levels (P < 0.05). DNJ increased the levels of prostaglandin E2, COX-1, COX2, and reduced the levels of and NF-κB p65 (P < 0.05). DNJ showed protection for gastric functions of GU mice by reducing the levels of MOT and SP, and increasing the levels of SS and VIP. DNJ treatment inactivates NF-κB signaling pathway, and increases anti-ulceration ability of the models.
Collapse
Affiliation(s)
- Xuehua Piao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Shuangdi Li
- Heart Disease Center, The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xiaodan Sui
- Department of Hepatology, The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Lianyi Guo
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xingmei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hongmei Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Leming Gao
- School of Stomatology, 2nd Dental Center, Peking University, Beijing, China
| | - Shusheng Cai
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanrong Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Tingting Wang
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Baohai Liu
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
45
|
Burton-Freeman BM, Sandhu AK, Edirisinghe I. Mangos and their bioactive components: adding variety to the fruit plate for health. Food Funct 2018; 8:3010-3032. [PMID: 28612853 DOI: 10.1039/c7fo00190h] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The diet is an essential factor affecting the risk for development and progression of modern day chronic diseases, particularly those with pathophysiological roots in inflammation and oxidative stress-induced damage. The potential impact of certain foods and their bioactive compounds to reverse or prevent destructive dysregulated processes leading to disease has attracted intense research attention. The mango (Mangifera indica Linn.) is a tropical fruit with distinctive nutritional and phytochemical composition. Notably, the mango contains several essential water- and lipid-soluble micronutrients along with the distinguishing phytochemicals gallotannins and mangiferin. In vitro and in vivo studies reveal various mechanisms through which mangos or their associated compounds reduce risk or reverse metabolic- and inflammation-associated diseases. Health benefits of isolated individual mango compounds and extracts from mango by-products are well described in the literature with less attention devoted to the whole fruit. Here, we review and summarize the available literature assessing the health promoting potential of mango flesh, the edible portion contributing to dietary fruit intake, focusing specifically on modern day health issues of obesity and the risk factors and diseases it precipitates, including diabetes and cardiovascular disease. Additionally, this review explores new insights on the benefits of mango for brain, skin and intestinal health. Overall, the foundation of research supporting the potential role of mangos in reducing risk for inflammation- and metabolically-based chronic diseases is growing.
Collapse
Affiliation(s)
- Britt M Burton-Freeman
- Center for Nutrition Research, Institute for Food Safety and Health, Illinois Institute of Technology, IL, USA.
| | | | | |
Collapse
|
46
|
Panneerselvam L, Raghunath A, Perumal E. Differential expression of myocardial heat shock proteins in rats acutely exposed to fluoride. Cell Stress Chaperones 2017; 22:743-750. [PMID: 28451878 PMCID: PMC5573692 DOI: 10.1007/s12192-017-0801-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/28/2017] [Accepted: 04/16/2017] [Indexed: 01/21/2023] Open
Abstract
Acute fluoride (F-) toxicity is known to cause severe cardiac complications and leads to sudden heart failure. Previously, we reported that increased myocardial oxidative damage, apoptosis, altered cytoskeleton and AMPK signaling proteins associated with energy deprivation in acute F- induced cardiac dysfunction. The present study was aimed to decipher the status of myocardial heat shock proteins (Hsps-Hsp27, Hsp32, Hsp40, Hsp60, Hsp70, Hsp90) and heat shock transcription factor 1 (Hsf1) in acute F--intoxicated rats. In order to study the expression of myocardial Hsps, male Wistar rats were treated with single oral doses of 45 and 90 mg/kg F- for 24 h. The expression levels of myocardial Hsps were determined using RT-PCR, western blotting, and immunohistochemical studies. Acute F--intoxicated rats showed elevated levels of both the transcripts and protein expression of Hsf1, Hsp27, Hsp32, Hsp60, and Hsp70 when compared to control. In addition, the expression levels of Hsp40 and Hsp90 were significantly declined in a dose-dependent fashion in F--treated animals. Our result suggests that differential expression of Hsps in the rat myocardium could serve as a balance between pro-survival and death signal during acute F--induced heart failure.
Collapse
Affiliation(s)
- Lakshmikanthan Panneerselvam
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, 641 046, India
| | - Azhwar Raghunath
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, 641 046, India.
| |
Collapse
|
47
|
Sandoval-Gallegos EM, Ramírez-Moreno E, Lucio JGD, Arias-Rico J, Cruz-Cansino N, Ortiz MI, Cariño-Cortés R. In Vitro Bioaccessibility and Effect of Mangifera indica (Ataulfo) Leaf Extract on Induced Dyslipidemia. J Med Food 2017; 21:47-56. [PMID: 28850305 DOI: 10.1089/jmf.2017.0042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death in the world, and epidemiological evidence points to dietary habits, stress, and obesity as major risk factors promoting pathological conditions like atherosclerosis, hypertension, and thrombosis. Current therapeutic approaches for CVDs rely on lifestyle changes and/or the use of drug agents. However, since the efficacy of such interventions is often limited by poor compliance and/or significant side effects, continued research on new preventive and therapeutic approaches is much needed. Our study is aimed to determine the bioaccessibility, total content of phenolic compounds, and antioxidant capacity (DPPH·, ABTS·+) of a methanolic extract from Mangifera indica L. leaves (MEM), and its lipid-lowering effect on an induced dyslipidemia model in Wistar rats. Our results showed that mangiferin is the main component of MEM. The extract showed a total content of polyphenol compounds of 575.28 gallic acid equivalents per dry matter basis (GAE/g db), antioxidant activity 77.68 μmol Trolox equivalents per gram (TE/g) db as measured by DPPH· and 20,630 μmol TE/g db by ABTS·+, and 12% of phenolic compounds were bioaccessible, and 100 mg/kg of MEM reduced hyperlipidemia levels induced in Wistar rats. Further study on the potential use of MEM as a nutraceutical to prevent CVDs in high-fat diet consumers is required.
Collapse
Affiliation(s)
| | - Esther Ramírez-Moreno
- 1 Interdisciplinary Research Center, Academic Area of Nutrition, Institute of Health Sciences , Pachuca, México
| | - Juan Gayosso De Lucio
- 2 Academic Area of Pharmacy, Institute of Health Sciences, Autonomous University of Hidalgo State , Pachuca, México
| | - José Arias-Rico
- 3 Academic Area of Nurse, Institute of Health Sciences, Autonomous University of Hidalgo State , Pachuca, México
| | - Nelly Cruz-Cansino
- 1 Interdisciplinary Research Center, Academic Area of Nutrition, Institute of Health Sciences , Pachuca, México
| | - Mario I Ortiz
- 4 Academic Area of Medicine, Autonomous University of Hidalgo State , Pachuca, México
| | - Raquel Cariño-Cortés
- 4 Academic Area of Medicine, Autonomous University of Hidalgo State , Pachuca, México
| |
Collapse
|
48
|
Collaborative Power of Nrf2 and PPAR γ Activators against Metabolic and Drug-Induced Oxidative Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1378175. [PMID: 28928902 PMCID: PMC5591982 DOI: 10.1155/2017/1378175] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/25/2017] [Indexed: 12/30/2022]
Abstract
Mammalian cells have evolved a unique strategy to protect themselves against oxidative damage induced by reactive oxygen species (ROS). Especially, two transcription factors, nuclear factor erythroid 2p45-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor γ (PPARγ), have been shown to play key roles in establishing this cellular antioxidative defense system. Recently, several researchers reported ameliorating effects of pharmacological activators for these Nrf2 and PPARγ pathways on the progression of various metabolic disorders and drug-induced organ injuries by oxidative stress. In this review, general features of Nrf2 and PPARγ pathways in the context of oxidative protection will be summarized first. Then, a number of successful applications of natural and synthetic Nrf2 and PPARγ activators to the alleviation of pathological and drug-related oxidative damage will be discussed later.
Collapse
|
49
|
Yu Q, Li X, Cao X. Linarin could protect myocardial tissue from the injury of Ischemia-reperfusion through activating Nrf-2. Biomed Pharmacother 2017; 90:1-7. [DOI: 10.1016/j.biopha.2017.03.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/21/2017] [Accepted: 03/09/2017] [Indexed: 10/19/2022] Open
|
50
|
Lauricella M, Emanuele S, Calvaruso G, Giuliano M, D'Anneo A. Multifaceted Health Benefits of Mangifera indica L. (Mango): The Inestimable Value of Orchards Recently Planted in Sicilian Rural Areas. Nutrients 2017; 9:E525. [PMID: 28531110 PMCID: PMC5452255 DOI: 10.3390/nu9050525] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 02/05/2023] Open
Abstract
Historically, Mangifera indica L. cultivations have been widely planted in tropical areas of India, Africa, Asia, and Central America. However, at least 20 years ago its spreading allowed the development of some cultivars in Sicily, an island to the south of Italy, where the favourable subtropical climate and adapted soils represent the perfect field to create new sources of production for the Sicilian agricultural supply chain. Currently, cultivations of Kensington Pride, Keitt, Glenn, Maya, and Tommy Atkins varieties are active in Sicily and their products meet the requirements of local and European markets. Mango plants produce fleshy stone fruits rich in phytochemicals with an undisputed nutritional value for its high content of polyphenolics and vitamins. This review provides an overview of the antioxidant, anti-inflammatory, and anticancer properties of mango, a fruit that should be included in everyone's diet for its multifaceted biochemical actions and health-enhancing properties.
Collapse
Affiliation(s)
- Marianna Lauricella
- Department of Experimental Biomedicine and Clinical Neurosciences, Laboratory of Biochemistry, University of Palermo, via del Vespro 129, 90127 Palermo, Italy.
| | - Sonia Emanuele
- Department of Experimental Biomedicine and Clinical Neurosciences, Laboratory of Biochemistry, University of Palermo, via del Vespro 129, 90127 Palermo, Italy.
| | - Giuseppe Calvaruso
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, Laboratory of Biochemistry, University of Palermo, via del Vespro 129, 90127 Palermo, Italy.
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, Laboratory of Biochemistry, University of Palermo, via del Vespro 129, 90127 Palermo, Italy.
| | - Antonella D'Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, Laboratory of Biochemistry, University of Palermo, via del Vespro 129, 90127 Palermo, Italy.
| |
Collapse
|