1
|
Zuo J, Huesker K, Liu Y, Hocher JG, Zhang X, von Baehr V, Krämer BK, Hocher B. Association of whole blood heavy metal concentrations with kidney function. Sci Rep 2025; 15:8370. [PMID: 40069484 PMCID: PMC11897145 DOI: 10.1038/s41598-025-93548-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/07/2025] [Indexed: 03/15/2025] Open
Abstract
Relatively elevated concentrations of arsenic, lead, and mercury are toxic to the kidneys. However, it is unknown whether kidney function is influenced by these metals in the general population without kidney diseases and without known exposure to these metals. We did a retrospective analysis of data collected from 58,864 outpatients in Germany from January 2014 to October 2022 undergoing measurements of arsenic, lead, and mercury. Routine clinical laboratory parameters were entered into the database if they were analyzed in the same patient within +/- four weeks of the metal analysis. The estimated glomerular filter rate (eGFR) was calculated using the 2021 CKD-EPI equation. The mean age of the study participants was 50.3 ± 17.1, of which 61.8% were women. Complete blood count, CRP, fasting glucose, liver and lipid values, and thyroid function parameters were within the normal range. Median (IQR) eGFR level was 92.14 (79.44-103.85) mL/min/1.73m2. Median (IQR) whole blood values for arsenic were 0.8 (0.4-1.5) µg/l, median (IQR) level for lead was 13.6 (9.5-19.5) µg/l, median (IQR) values for mercury were 0.8 (0.3-1.5) µg/l in whole blood. Arsenic (r= -0.131, p < 0.001, N = 11,211), lead (r = 0.318, p < 0.001, N = 21,733), and mercury (r= -0.149, p < 0.001, N = 22,670) levels correlate all inversely with eGFR. When plotting eGFR against whole blood metal concentrations, no lower safety thresholds were found. Multivariate analysis, considering age, sex, CRP, and fasting glucose as confounding factors, confirmed findings of independent associations of arsenic, lead, and mercury on eGFR. Subgroup analysis revealed that this inverse relationship was particularly pronounced in the lowest age tertile of all study participants. Concentrations of arsenic, lead, and mercury correlated independently inversely with eGFR in a German cohort that largely had a normal kidney function with no known exposure to heavy metals.
Collapse
Affiliation(s)
- Jiao Zuo
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Yvonne Liu
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Johann-Georg Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Xiaoli Zhang
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | | | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology, Pneumology), University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany.
- Institute of Medical Diagnostics, IMD, Berlin, Germany.
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.
- School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
2
|
Chen HH, Huang YL, Wu CY, Chen MC, Shiue HS, Hsu SL, Lin YC, Hsueh YM. Plasma myeloperoxidase interactions with cadmium, lead, arsenic, and selenium and their impact on chronic kidney disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117726. [PMID: 39826409 DOI: 10.1016/j.ecoenv.2025.117726] [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: 07/01/2024] [Revised: 12/21/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Myeloperoxidase (MPO) is an oxidative stress biomarker, with elevated MPO levels linked to chronic kidney disease (CKD) progression. Metal exposure is a risk factor for CKD, and is also correlated to MPO expression, with specific MPO genotypes linked to MPO expression. Therefore, we examined whether MPO plasma levels or MPO polymorphisms were linked to CKD, and explored whether these factors modified associations between CKD and metal concentrations. Accordingly, we recruited 395 age- and sex-matched controls and 215 patients with CKD (persistent estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2 for a minimum of 12 weeks without hemodialysis). We identified no associations between several MPO genotypes and CKD. However, after multivariate adjustment, plasma MPO concentrations were positively correlated with CKD odds ratio (OR) = 5.87 (95 % confidence interval [CI]: 3.14-10.96). Significant additive interactions were observed between high plasma MPO concentrations and elevated blood cadmium (Cd) and lead (Pb) levels, and total urinary arsenic (As), or low plasma selenium (Se) concentrations, leading to increased ORs for CKD, with significant synergy indices recorded. High plasma MPO concentrations also showed multiplicative interactions with elevated blood Pb levels or low plasma Se concentrations, which increased the ORs for CKD (p-values = 0.005 and 0.009, respectively). Our study is the first to show a significant interaction between plasma MPO concentration and metals affecting the OR of CKD.
Collapse
Affiliation(s)
- Hsi-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ya-Li Huang
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Yin Wu
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Chieh Chen
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Horng-Sheng Shiue
- Department of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Sheng-Lun Hsu
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying-Chin Lin
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Geriatric Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Mei Hsueh
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
3
|
Badawy HAE, Osman A, Ahmed TAE, Hincke MT. Evaluation of plant-derived biomaterials for the development of tissue-engineered corneal substitutes. J Biomed Mater Res A 2024; 112:2187-2201. [PMID: 38963322 DOI: 10.1002/jbm.a.37769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
Corneal blindness affects over 10 million patients worldwide. Due to the limited supply of donor corneas and frequent graft failure, bioengineered alternatives are crucial. To overcome drawbacks associated with corneal substitutes from synthetic biomaterials, fabrication from plant-derived biomaterials is a potential alternative. Herein, soy protein and glutenin in combination with different crosslinkers were evaluated for fabrication of corneal substitutes. Optical, mechanical, and biochemical properties of fabricated constructs and control rabbit corneas were evaluated in vitro. Soy protein crosslinked with peroxidase/H202 possessed transparency and mechanical properties comparable to controls, although their water content and biocompatibility were inferior. In contrast, soy protein crosslinked with tannic acid showed similar water content, tensile strength, and biocompatibility as rabbit corneas; however, these constructs displayed significantly lower transparency and higher strain to failure. Finally, glutenin cross-linked using formaldehyde showed excellent transparency, strain to failure, and biocompatibility, however; they exhibited significantly lower water content and tensile strength than controls. This study is the first to establish CIELAB color values for the rabbit cornea, allowing quantitative optical evaluation of tissue-engineered substitutes. Thus, a crosslinking strategy utilizing plant-derived proteins for fabrication of constructs with properties comparable to rabbit corneas is a promising direction for development of tissue-engineered corneal substitutes.
Collapse
Affiliation(s)
- Hadeel A E Badawy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ahmed Osman
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Tamer A E Ahmed
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technology Applications (SRTA-City), Alexandria, Egypt
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Maxwell T Hincke
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Innovation in Medical Education (DIME), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Meine BDM, de Mello JE, Custódio SV, da Silveira LM, Simões WS, Bona NP, Garcia DN, Schneider A, de Souza LP, Domingues WB, Campos VF, Spanevello RM, de Aguiar MSS, Stefanello FM. Tannic acid: A possible therapeutic agent for hypermethioninemia-induced neurochemical changes in young rats. Biochem Biophys Res Commun 2024; 734:150635. [PMID: 39236587 DOI: 10.1016/j.bbrc.2024.150635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
This study explores the therapeutic benefits of tannic acid (TnA) in an experimental protocol of chronic hypermethioninemia in rats. Rats were categorized into four groups: Group I - control, Group II - TnA 30 mg/kg, Group III - methionine (Met) 0.2-0.4 g/kg + methionine sulfoxide (MS) 0.05-0.1 g/kg, Group IV - TnA/Met + MS. Saline was administered by subcutaneous pathway into groups I and II twice daily from postnatal day 6 (P6) to P28, whereas those in groups III and IV received Met + MS. From P28 to P35, groups II and IV received TnA orally. Animals from group III presented cognitive and memory impairment assessed through object recognition and Y-maze tests (p < 0.05). Elevated levels of reactive species, lipid peroxidation, and nitrites followed by a decline in sulfhydryl content, catalase activity, and superoxide dismutase activity were observed in animals treated with Met + MS (p < 0.05). However, TnA treatment reversed all these effects (p < 0.05). In group III, there was an increase in acetylcholinesterase activity and IL-6 levels, coupled with a reduction in Na+/K+-ATPase activity (p < 0.05). TnA was able to protect against these effects (p < 0.05). The gene expression of catalase, brain-derived neurotrophic factor, and nuclear factor erythroid 2-related factor 2 was decreased in the hippocampus and striatum from group III (p < 0.05). TnA reversed almost all of these alterations (p < 0.05). These findings suggest that TnA is a therapeutic target for patients with hypermethioninemia.
Collapse
Affiliation(s)
- Bernardo de Moraes Meine
- Laboratório de Biomarcadores, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Julia Eisenhardt de Mello
- Laboratório de Neuroquímica, Inflamação e Câncer, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Solange Vega Custódio
- Laboratório de Neuroquímica, Inflamação e Câncer, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Larissa Menezes da Silveira
- Laboratório de Biomarcadores, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - William Sanabria Simões
- Laboratório de Biomarcadores, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Natália Pontes Bona
- Laboratório de Biomarcadores, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | | | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Lucas Petitemberte de Souza
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - William Borges Domingues
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Vinicius Farias Campos
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Roselia Maria Spanevello
- Laboratório de Neuroquímica, Inflamação e Câncer, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Laboratório de Neuroquímica, Inflamação e Câncer, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil.
| | - Francieli Moro Stefanello
- Laboratório de Biomarcadores, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil.
| |
Collapse
|
5
|
Lei H, Yu X, Fan D. Nanocomposite Hydrogel for Real-Time Wound Status Monitoring and Comprehensive Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405924. [PMID: 39269428 PMCID: PMC11558094 DOI: 10.1002/advs.202405924] [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: 06/28/2024] [Revised: 08/24/2024] [Indexed: 09/15/2024]
Abstract
Current skin sensors or wound dressings fall short in addressing the complexities and challenges encountered in real-world scenarios, lacking adequate capability to facilitate wound repair. The advancement of methodologies enabling early diagnosis, real-time monitoring, and active regulation of drug delivery for timely comprehensive treatment holds paramount significance for complex chronic wounds. In this study, a nanocomposite hydrogel is devised for real-time monitoring of wound condition and comprehensive treatment. Tannins and siRNA containing matrix metalloproteinase-9 gene siRNA interference are self-assembled to construct a degradable nanogel and modified with bovine serum albumin. The nanogel and pH indicator are encapsulated within a dual-crosslinking hydrogel synthesized with norbornene dianhydride-modified paramylon. The hydrogel exhibited excellent shape adaptability due to borate bonding, and the click polymerization reaction led to rapid in situ curing of the hydrogel. The system not only monitors pH, temperature, wound exudate alterations, and peristalsis during wound healing but also exhibits hemostatic, antimicrobial, anti-inflammatory, and antioxidant properties, modulates macrophage polarization, and facilitates vascular tissue regeneration. This therapeutic approach, which integrates the monitoring of pathological parameters with comprehensive treatment, is anticipated to address the clinical issues and challenges associated with chronic diabetic wounds and infected wounds, offering broad prospects for application.
Collapse
Affiliation(s)
- Huan Lei
- Engineering Research Center of Western Resource Innovation Medicine Green ManufacturingMinistry of EducationSchool of Chemical EngineeringNorthwest UniversityXi'an710069China
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation EngineeringSchool of Chemical EngineeringNorthwest UniversityXi'an710069China
- Biotech. & Biomed. Research InstituteNorthwest UniversityXi'an710069China
| | - Xueqing Yu
- Engineering Research Center of Western Resource Innovation Medicine Green ManufacturingMinistry of EducationSchool of Chemical EngineeringNorthwest UniversityXi'an710069China
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation EngineeringSchool of Chemical EngineeringNorthwest UniversityXi'an710069China
- Biotech. & Biomed. Research InstituteNorthwest UniversityXi'an710069China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green ManufacturingMinistry of EducationSchool of Chemical EngineeringNorthwest UniversityXi'an710069China
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation EngineeringSchool of Chemical EngineeringNorthwest UniversityXi'an710069China
- Biotech. & Biomed. Research InstituteNorthwest UniversityXi'an710069China
| |
Collapse
|
6
|
Maity J, Pal P, Ghosh M, Naskar B, Chakraborty S, Pal R, Mukhopadhyay PK. Molecular Dissection of the Arsenic-Induced Leukocyte Incursion into the Inflamed Thymus and Spleen and Its Amelioration by Co-supplementation of L-Ascorbic Acid and α-Tocopherol. Biol Trace Elem Res 2024:10.1007/s12011-024-04378-z. [PMID: 39325335 DOI: 10.1007/s12011-024-04378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
Arsenic, a surreptitious presence in our environment, perpetuates a persistent global menace with its deleterious impacts. It possesses the capability to trigger substantial immunosuppression by instigating inflammation in critical organs like the thymus and spleen. L-Ascorbic acid (L-AA) exhibits robust immunoregulatory prowess by orchestrating the epigenetic terrain through TET and JHDM pathways. Conversely, α-tocopherol (α-T) demonstrates the capacity to dampen the production of pro-inflammatory cytokines by modulating the PI3K-Akt axis. Given these insights, this inquiry embarks on exploring the mitigative potential of L-AA and α-T co-supplementation at the transcriptome level within leukocytes under arsenic exposure. Concurrently, the research endeavours to unravel the potent anti-inflammatory effects of administering α-T and L-AA, alleviating inflammation within the spleen and thymus amidst arsenic-induced insult and delving deeply into their immunomodulatory mechanisms. The rats were randomly allocated into eight distinct groups for subsequent experimentation: (I) the control group was administered solely with distilled water as the vehicle (control); (II) NaAsO2-treated group (As); (III) NaAsO2 treated along with L-ascorbic acid and α-tocopherol supplemented group (As + L-AA + α-T); (IV) L-ascorbic acid and α-tocopherol supplemented group (L-AA + α-T); (V) NaAsO2 treated along with L-ascorbic acid supplemented group (As + L-AA); (VI) only L-ascorbic acid supplemented group (L-AA); (VII) NaAsO2 treated along with α-tocopherol supplemented group (As + α-T); (VIII) only α-tocopherol supplemented group (α-T). Rats treated with NaAsO2 exhibited an increased neutrophil count in their bloodstream, as revealed by a comprehensive transcriptomic analysis showcasing heightened expressions of ItgaM, MMP9, and Itga4 within circulating leukocytes under arsenic exposure. Concurrently, arsenic heightened the expression of pro-inflammatory cytokines within the thymus and spleen. This elevated cytokine activity promoted the upregulation of ICAM-1 on vascular endothelial cells, facilitating the infiltration of Ly6g + leukocytes into the afflicted thymus and spleen. Remarkably, the combination of L-AA acid and α-T demonstrated substantial therapeutic efficacy, adeptly reducing the influx of Ly6g + leukocytes into these immune sites and subsequent reduction of excessive collagen deposition. The dynamic duo of L-AA and α-T achieved this amelioration by suppressing the expression of ItgaM, MMP9, and Itga4 mRNA within circulating leukocytes and moderating tissue levels of pro-inflammatory cytokines in arsenic-exposed thymus and spleen.
Collapse
Affiliation(s)
- Jeet Maity
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Priyankar Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
- School of Life Science, Department of Biotechnology, Swami Vivekananda University, Barrackpore, India
| | - Madhurima Ghosh
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Bhagyashree Naskar
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Santanu Chakraborty
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Ranjana Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | | |
Collapse
|
7
|
Yang X, Chu F, Jiao Z, Yu H, Yang W, Li Y, Lu C, Ma H, Wang S, Liu Z, Qin S, Sun H. Ellagic acid ameliorates arsenic-induced neuronal ferroptosis and cognitive impairment via Nrf2/GPX4 signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116833. [PMID: 39128446 DOI: 10.1016/j.ecoenv.2024.116833] [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: 04/07/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Arsenic, a neurotoxic metalloid, poses significant health risks. However, ellagic acid, renowned for its antioxidant properties, has shown potential in neuroprotection. This study aimed to investigate the neuroprotective effects of ellagic acid against arsenic-induced neuronal ferroptosis and cognitive impairment and elucidate the underlying mechanisms. Using an arsenic-exposed Wistar rat model and an arsenic-induced HT22 cells model, we assessed cognitive ability, measured serum and brain arsenic levels, and evaluated pathological damage through histological analysis and transmission electron microscopy. Additionally, we examined oxidative stress and iron ion levels using GSH, MDA, ROS and tissue iron biochemical kits, and analyzed the expression of ferroptosis-related markers using western blot and qRT-PCR. Our results revealed that arsenic exposure increased both serum and brain arsenic levels, resulting in hippocampal pathological damage and subsequent decline in learning and memory abilities. Arsenic-induced neuronal ferroptosis was mediated by the inhibition of the xCT/GSH/GPX4/Nrf2 signaling axis and disruption of iron metabolism. Notably, ellagic acid intervention effectively reduced serum and brain arsenic levels, ameliorated neuronal damage, and improved oxidative stress, ferroptosis, and cognitive impairment. These beneficial effects were associated with the activation of the Nrf2/Keap1 signaling pathway, upregulation of GPX4 expression, and enhanced iron ion excretion. In conclusion, ellagic acid demonstrates promising neuroprotective effects against arsenic-induced neurotoxicity by mitigating neuronal ferroptosis and cognitive impairment.
Collapse
Affiliation(s)
- Xiyue Yang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Fang Chu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Zhe Jiao
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Hao Yu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Wenjing Yang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Yang Li
- The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Donghu Distinct, Nanchang, Jiangxi 330006, China
| | - Chunqing Lu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Hao Ma
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Sheng Wang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Zhipeng Liu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Shaoxiao Qin
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China
| | - Hongna Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; NHC Key Laboratory of Etiology and Epidemiology (Harbin Medical University) & Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, China.
| |
Collapse
|
8
|
Rezvankhah S, Zargari F, Sharifi R. Melatonin alleviates arsenic-induced liver injury by regulating protein RKIP and enhancing antioxidant defencse mechanisms. J Biochem Mol Toxicol 2024; 38:e23835. [PMID: 39215756 DOI: 10.1002/jbt.23835] [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: 04/25/2024] [Revised: 08/11/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Arsenic (As) is a highly toxic metal and one of the main factors in cancer development through oxidative stress and production of reactive oxygen species. Prior research has demonstrated melatonin's potential as a free radical scavenger. Raf kinase inhibitory protein (RKIP) is an important regulator of intracellular signaling pathways that has been linked to various types of cancer. The aim of this research was to explore the influence of melatonin's antioxidant properties on the expression of the protein RKIP and the antioxidant status of liver tissue in rats that were exposed to arsenic. Thirty two male Wistar rats were divided into four groups of eight, including control, melatonin-treated (20 mg/Kg of melatonin), sodium arsenite-treated (5.5 mg/Kg of sodium arsenite), and melatonin + sodium arsenite-treated groups (combination) for 4 weeks. The expression level of protein RKIP was measured by Western blot, and malondialdehyde (MDA) content of the liver as well as the activities of antioxidant enzymes were measured. The data analyzed using one-way ANOVA (significance level of p < 0.05) and GraphPad Prism (9) software. Sodium arsenite treatment led to a significant decrease in RKIP protein expression and antioxidant enzyme activity, and an increase in liver MDA levels (p < 0.001). Conversely, melatonin treatment in the combination group resulted in a significant increase in RKIP protein expression and antioxidant enzyme activity and a decrease in liver MDA levels (p < 0.05). These findings suggest that melatonin can attenuate oxidative damage caused by arsenic in liver cells by enhancing RKIP protein expression and antioxidant enzyme activity.
Collapse
Affiliation(s)
- Soheil Rezvankhah
- Department of Biology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Felor Zargari
- Department of Medical Science, Marand Branch, Islamic Azad University, Marand, Iran
| | - Rasoul Sharifi
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
9
|
Abdel-Wahab BA, Zafaar D, Habeeb MS, El-Shoura EAM. Nicorandil mitigates arsenic trioxide-induced lung injury via modulating vital signalling pathways SIRT1/PGC-1α/TFAM, JAK1/STAT3, and miRNA-132 expression. Br J Pharmacol 2024; 181:3215-3231. [PMID: 38741475 DOI: 10.1111/bph.16414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/13/2024] [Accepted: 03/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE Nicorandil, a selective opener of potassium channels, used to treat angina, has drawn attention for its potential in mitigating lung injury, positioning it as a promising therapeutic approach to treat drug-induced lung toxicity. This study aimed to explore the protective role of nicorandil in arsenic trioxide (ATO)-induced lung injury and to elucidate the underlying mechanistic pathways. EXPERIMENTAL APPROACH We assessed the effects of nicorandil (15 mg·kg-1, p.o.) in a rat model of pulmonary injury induced by ATO (5 mg·kg-1, i.p.). The assessment included oxidative stress biomarkers, inflammatory cytokine levels, and other biomarkers, including sirtuin-1, sirtuin-3, STAT3, TFAM, and JAK in lung tissue. Histological examination using H&E staining and molecular investigations using western blotting and PCR techniques were conducted. KEY RESULTS In our model of lung injury, treatment with nicorandil ameliorated pathological changes as seen with H&E staining, reduced tissue levels of toxicity markers, and exerted significant antioxidant and anti-inflammatory actions. On a molecular level, treatment with nicorandil down-regulated JAK, STAT3, PPARγ, Nrf2, VEGF, p53, and micro-RNA 132 while up-regulating Sirt1, 3, TFAM, AMPK, and ERR-α in lung tissue. CONCLUSIONS AND IMPLICATIONS The results presented here show nicorandil as a significant agent in attenuating lung injury induced by ATO in a rodent model. Nonetheless, further clinical studies are warranted to strengthen these findings.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Dalia Zafaar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University of Technology, and Information, Cairo, Egypt
| | | | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| |
Collapse
|
10
|
Ghosh A, Muley A, Bhat S, Ainapure A. Exploring the Renoprotective Potential of Bioactive Nutraceuticals in Chronic Kidney Disease Progression: A Narrative Review. Cureus 2024; 16:e68730. [PMID: 39371767 PMCID: PMC11454842 DOI: 10.7759/cureus.68730] [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] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Chronic kidney disease (CKD) is a condition that is characterized by progressive loss of kidney function over time. A substantial increase in the burden of CKD is evident globally, attributed to multifactorial conditions like an expanding aging population, rising diabetes and hypertension rates, and more significant exposures to risk factors associated with the environment and lifestyle. Nutraceuticals are substances that are usually considered a food or an active part of a food that provides medical or health benefits, including the prevention and treatment of a disease. The aim is to review the positive role of nutraceuticals in managing CKD. A narrative review is generated, extracting the papers from databases like Web of Science, Scopus, ScienceDirect, ResearchGate, and PubMed. Animal and human trials focusing on the effect of different nutraceuticals on the initial stage of kidney disease, i.e., stages 1, 2, and 3 of CKD, were included. The review's outcome is understanding the effectiveness of nutraceuticals that have shown positive results in CKD conditions. Active compounds include ubiquinone, curcumin, nitrates, nitrites, lycopene, and resveratrol. These bioactive components are also beneficial for other comorbid conditions like diabetes, hypertension, and cardiovascular conditions that have an eminent adverse effect on CKD. Lycopene, coenzyme Q10 (CoQ10), resveratrol, curcumin, and flavonoids have positively impacted CKD complications. Nutraceuticals hold great promise for individuals with CKD in the coming years, offering diverse potential benefits. These include delivering vital antioxidant and anti-inflammatory support to alleviate oxidative stress and inflammation, helping to regulate blood pressure and lipid levels for improved cardiovascular health, promoting optimal renal function to sustain kidney health, assisting in maintaining electrolyte balance, warding off complications, influencing gut microbiota for enhanced digestive well-being, and ultimately elevating the overall quality of life, for those managing CKD.
Collapse
Affiliation(s)
- Anindita Ghosh
- Nutritional Sciences and Dietetics, Symbiosis Skills and Professional University, Pune, IND
| | - Arti Muley
- Nutrition and Dietetics, Symbiosis School of Culinary Arts, Pune, IND
| | - Sakshi Bhat
- Nutrition and Dietetics, Symbiosis International (Deemed University), Pune, IND
| | | |
Collapse
|
11
|
Shawky LM, Abo El Wafa SM, Behery M, Bahr MH, Abu Alnasr MT, Morsi AA. Lactobacillus rhamnosus GG and Tannic Acid Synergistically Promote the Gut Barrier Integrity in a Rat Model of Experimental Diarrhea via Selective Immunomodulatory Cytokine Targeting. Mol Nutr Food Res 2024; 68:e2400295. [PMID: 39034291 DOI: 10.1002/mnfr.202400295] [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: 04/26/2024] [Revised: 06/20/2024] [Indexed: 07/23/2024]
Abstract
SCOPE Diarrhea is a common health issue that contributes to a significant annual death rate among children and the elderly worldwide. The anti-diarrheal activity of Lactobacillus rhamnosus GG (LGG) and tannic acid (TA), alone or combined, is examined, in addition to their effect on intestinal barrier integrity. METHODS AND RESULTS Fifty-six adult male Wistar rats are randomly assigned into seven groups: control, LGG alone, TA alone, diarrhea model, diarrhea+LGG, diarrhea+TA, and diarrhea+LGG+TA-treated groups. Diarrhea is induced by high-lactose diet (HLD) consumption. LGG (1x109 CFU/rat) and TA (100 mg Kg-1 d-1) were given orally 4 days after HLD feeding and continued for 10 days. Ileum specimens are processed for biochemical analysis of the local intestinal cytokines, polymerase chain reaction (PCR), and histological study. Also, immunohistochemistry-based identification of Proliferating Cell Nuclear Antigen (PCNA) and zonula occludens 1 (ZO-1) is performed. Compared to the diarrhea model group, both treatments maintain the intestinal mucosal structure and proliferative activity and preserve ZO-1 expression, with the combination group showing the maximal effect. However, LGG-treated diarrheic rats show a remarkable decrease in the intestinal tissue concentrations of tumor necrosis factor-alpha (TNF-α) and nuclear factor Kappa beta (NF-κB); meanwhile, TA treatment leads to a selective decrease of interferon-gamma (INF-γ) and transforming growth factor-beta (TGF-β1). CONCLUSION Individual LGG and TA treatments significantly alleviate diarrhea, probably through a selective immunomodulatory cytokine-dependent mechanism, while the combination of both synergistically maintains the intestinal mucosa by keeping the intestinal epithelial barrier function and regenerative capability.
Collapse
Affiliation(s)
- Lamiaa M Shawky
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| | - Sahar M Abo El Wafa
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| | - Maged Behery
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| | - Mohamed H Bahr
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, 11591, Egypt
- Department of Basic Medical Sciences, Vision Colleges, Riyadh, 11451, Saudi Arabia
| | | | - Ahmed A Morsi
- Department of Histology and Cell Biology, Faculty of Medicine, Fayoum University, Fayoum, 63511, Egypt
| |
Collapse
|
12
|
Maity J, Pal P, Pal R, Mukhopadhyay PK. Co-administration of L-Ascorbic Acid and α-Tocopherol Alleviates Arsenic-Induced Immunotoxicities in the Thymus and Spleen by Dwindling Oxidative Stress-Induced Inflammation. Biol Trace Elem Res 2024; 202:2199-2227. [PMID: 37704839 DOI: 10.1007/s12011-023-03841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023]
Abstract
Herein, we investigated whether L-ascorbic acid (L-AA) and α-tocopherol (α-T) co-administration has the potential to alleviate arsenic-induced immunotoxicities in the thymus, spleen, and circulating leukocytes. Forty-eight adult male Wistar rats were randomly divided into four groups before the treatment: group I (control); group II (sodium arsenite, 3 mg/kg/day/rat); group III (sodium arsenite + L-AA (200 mg/kg/day/rat) and α-T (400 mg/kg/day/rat)); group IV (L-AA and α-T). The result showed that sodium arsenite exposure (consecutive 30 days) caused weight reduction, structural alterations in the thymus and spleen, accompanied by a decrease in thymocyte and splenocyte count. Decreased superoxide dismutase and catalase activity, increased malondialdehyde and protein-carbonyl content, reduced Nrf2 and Bcl2 expression, and increased p-ERK, NF-kβ, Bax, and cleaved-caspase-3 expression were also observed in the thymus and spleen of arsenic-exposed rats. Enhanced plasma ACTH and corticosterone, ROS-induced apoptosis of lymphocytes were also observed. L-AA and α-T co-administration has the potential to abrogate the deleterious impact of arsenic on the thymus, spleen, and circulating lymphocytes. Whole transcriptome analysis of leukocytes revealed that arsenic treatment augmented the expression of Itga4, Itgam, and MMP9 genes, which might help in transient migration of the leukocytes through the endothelial cell layer. Co-administration with L-AA and α-T maintained Itga4, Itgam, and MMP9 gene expression within leukocytes at a lower level.
Collapse
Affiliation(s)
- Jeet Maity
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Priyankar Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Ranjana Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | | |
Collapse
|
13
|
Verma A, Jakhar R, Kumar D, Kumar V, Dhillon T, Dangi M, Chhillar AK. A computational approach to discover antioxidant and anti-inflammatory attributes of silymarin derived from Silybum marianum by comparison with hydroxytyrosol. J Biomol Struct Dyn 2023; 41:11101-11121. [PMID: 36546728 DOI: 10.1080/07391102.2022.2159879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Medicinal plants possess therapeutic potential for reducing reactive oxygen species (ROS)-mediated cellular damage. Hydroxytyrosol is one of the most potent antioxidants that served as control in the current study, including other synthetic antioxidants to computationally identify the antioxidant properties of Silymarin. The sequences of the receptors IκB kinase (IKK), Kelch-like ECH-associated protein 1 (Keap-1) and mitochondrial transcription factor A (Tfam) were retrieved from UniProtKB and homology modeling was performed using Swiss-Model server. Thereof the molecular docking and dynamic simulation studies were performed using Schrödinger's software version 11.5. From the current study, it was reported that on comparison of the binding energy of silymarin, hydroxytyrosol, α-tocopherol, ascorbic acid, butylated hydroxy anisole (BHA) and butylated hydroxytoluene (BHT), Silymarin exhibited better affinities with IKK receptor followed by Hydroxytyrosol suggesting it as the best or comparable of all other known antioxidants that could potentially suppress inflammation and other diseases. Also, Silymarin exhibited poorest binding affinity with Tfam promoting mitochondrial biogenesis, thereby scavenging ROS. However, with Keap-1, Silymarin is ranked 4th in the list, whereas hydroxytyrosol exhibited highest binding affinity to release oxidative stress. The stability of docked complexes made us conclude that Silymarin has comparable antioxidant properties to hydroxytyrosol, better anti-inflammatory potential and mitochondrial biogenesis enhancing properties to ultimately reduce oxidative stress. Now it can be tested further for in vitro or in vivo studies as potential drug against oxidative insult.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Annu Verma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Ritu Jakhar
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Dev Kumar
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Vijay Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Twinkle Dhillon
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Mehak Dangi
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | | |
Collapse
|
14
|
Ren X, Yuan P, Niu J, Liu Y, Li Y, Huang L, Jiang S, Jiao N, Yuan X, Li J, Yang W. Effects of dietary supplementation with microencapsulated Galla chinensis tannins on growth performance, antioxidant capacity, and lipid metabolism of young broiler chickens. Front Vet Sci 2023; 10:1259142. [PMID: 37954663 PMCID: PMC10637619 DOI: 10.3389/fvets.2023.1259142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
This study aimed to investigate the impacts of dietary supplementation with Galla chinensis tannins (GCT) on the growth performance, antioxidant capacity, and lipid metabolism of young broilers. Overall, a total of 216 healthy 1 day-old broilers were randomly allocated to CON group and GCT group, and provided with a basal diet or a basal diet added with 300 mg/kg microencapsulated GCT, respectively, in a 21 days trial. Our findings indicated that dietary GCT addition had no significant effects (p > 0.05) on growth performance. However, GCT supplementation led to a significant reduction in the total cholesterol (TC) concentration in the serum and liver (p < 0.05). Furthermore, GCT supplementation significantly increased the ratios of high-density lipoprotein (HDL) to low-density lipoprotein (LDL) and HDL to TC in the serum, in addition to elevating the activities of enzymes related to lipid metabolism in the liver (p < 0.05). Dietary GCT addition also improved the antioxidant capacity of the broilers, as evidenced by a significant decrease in the concentration of malondialdehyde in serum and liver (p < 0.05). Additionally, the GCT group exhibited significantly increased expressions of hepatic genes associated with antioxidant enzymes (HO-1, GPX1, SOD2, SIRT1, CPT-1, and PPARα) (p < 0.05), while the mRNA expression of SREBP-1 was significantly decreased (p < 0.05) compared with the CON group. In conclusion, dietary addition of 300 mg/kg microencapsulated GCT improved the antioxidant status and lipid metabolism of broilers without affecting their growth performance.
Collapse
Affiliation(s)
- Xiaojie Ren
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- Shandong Taishan Shengliyuan Group Co., Ltd, Tai’an, China
| | - Peng Yuan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Jiaxing Niu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Yang Liu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Yang Li
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- Division of Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Libo Huang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Shuzhen Jiang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Ning Jiao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Xuejun Yuan
- College of Life Sciences, Shandong Agricultural University, Tai’an, China
| | - Junxun Li
- Shandong Taishan Shengliyuan Group Co., Ltd, Tai’an, China
| | - Weiren Yang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| |
Collapse
|
15
|
Abdel-Wahab BA, El-Shoura EAM, Shafiuddin Habeeb M, Zafaar D. Febuxostat alleviates Arsenic Trioxide-Induced renal injury in Rats: Insights on the crosstalk between NLRP3/TLR4, Sirt-1/NF-κB/TGF-β signaling Pathways, and miR-23b-3p, miR-181a-5b expression. Biochem Pharmacol 2023; 216:115794. [PMID: 37689273 DOI: 10.1016/j.bcp.2023.115794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/15/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Febuxostat (FBX), a xanthine oxidase inhibitor, is known to improve renal function and can show promise as a therapeutic agent for preventing drug-induced nephrotoxicity. This study aimed to explore the protective effect of FBX in preventing renal damage caused by arsenic trioxide (ATO) toxicity and uncover the underlying mechanisms. The researchers examined how FBX (10 mg/kg, orally) affected ATO-induced kidney injury (5 mg/kg, intraperitoneally) in rats. Kidney function and toxicity parameters in serum and oxidative stress biomarkers and inflammatory cytokine levels in renal tissue were measured. H&E staining was used to detect histopathological changes in the kidney. Network the molecular mechanisms of FBX in improving kidney injury were investigated using Western blotting and PCR techniques. The findings showed that FBX improved kidney function by inhibiting the pathological changes seen in H&E staining, decreasing levels of probed kidney function and toxicity measures in serum and tissue, and exhibiting antioxidant and anti-inflammatory effects. FBX decreased MDA, MPO, TNF-α, IL-1β, IL-6, COX-II, and NADPH oxidase levels, while increased GSH, GPx, SOD, and IL-10 levels. FBX also reduced the expression of NLRP3, ASC, TLR4, and micro-RNA 181a-5b while increased the expression of IKBα, Sirt-1, and micro-RNA 23b-3p, according to Western blotting and PCR results. In conclusion, FBX can play a vital role in reducing kidney injury in cases of ATO-induced nephrotoxicity, though more clinical research needs to be conducted.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran P.O. Box 1988, Saudi Arabia.
| | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt.
| | | | - Dalia Zafaar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University of Technology, and Information, Cairo, Egypt.
| |
Collapse
|
16
|
Baruah P, Moorthy H, Ramesh M, Padhi D, Govindaraju T. A natural polyphenol activates and enhances GPX4 to mitigate amyloid-β induced ferroptosis in Alzheimer's disease. Chem Sci 2023; 14:9427-9438. [PMID: 37712018 PMCID: PMC10498722 DOI: 10.1039/d3sc02350h] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 08/19/2023] [Indexed: 09/16/2023] Open
Abstract
Ferroptosis, an iron-dependent cell death, plays a crucial role in the pathology of Alzheimer's disease (AD). Several characteristics of AD, including excessive iron accumulation, elevated lipid peroxide and reactive oxygen species (ROS) levels, and decreased glutathione peroxidase 4 (GPX4) levels, align with the features of ferroptosis. While traditional methods of inhibiting ferroptosis have centered on chelating Fe and trapping radicals, therapeutic strategies that modulate the GPX4 axis to mitigate ferroptosis in AD are yet to be explored. This report introduces naturally occurring polyphenols (PPs) as dual-acting therapeutic agents to synergistically alleviate ferroptosis and AD. The mechanisms of action encompass modulation of amyloid and tau cascade, reduction of oxidative stress, mitochondrial rescue, and inhibition of ferroptosis. For the first time, we show that a single multifunctional molecule, tannic acid (TA) binds at the activator site of GPX4, augmenting both its activity and cellular levels, providing a conceptually innovative and integrated approach for treating AD via the GPX4-ferroptosis axis. The ability of TA to enhance GPX4 levels under conditions of AD pathology opens up newer promising therapeutic avenues for combating the crosstalk between ferroptosis and AD.
Collapse
Affiliation(s)
- Prayasee Baruah
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Dikshaa Padhi
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| |
Collapse
|
17
|
Wang M, Cao L. Hydrolysable tannins as a potential therapeutic drug for the human fibrosis-associated disease. Drug Dev Res 2023; 84:1096-1113. [PMID: 37386756 DOI: 10.1002/ddr.22089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
Fibrosis is a pathological change with abnormal tissue regeneration due to a response to persistent injury, which is extensively related to organ damage and failure, leading to high morbidity and mortality worldwide. Although the pathogenesis of fibrosis has been comprehensively elucidated, there are few effective therapies for treating fibrotic diseases. Natural products are increasingly regarded as an effective strategy for fibrosis with numerous favorable functions. Hydrolysable tannins (HT) are a type of natural products that have the potential to treat the fibrotic disease. In this review, we describe some biological activities and the therapeutic prospects of HT in organ fibrosis. Furthermore, the underlying mechanisms of inhibition of HT on fibrotic organs in relation to inflammation, oxidative stress, epithelial-mesenchymal transition, fibroblast activation and proliferation, and extracellular matrix accumulation are discussed. Understanding the mechanism of HT against fibrotic diseases will provide a new strategy for the prevention and attenuation of fibrosis progression.
Collapse
Affiliation(s)
- Meiwei Wang
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Linghui Cao
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
18
|
Yuan P, Xu H, Ma Y, Niu J, Liu Y, Huang L, Jiang S, Jiao N, Yuan X, Yang W, Li Y. Effects of dietary Galla Chinensis tannin supplementation on immune function and liver health in broiler chickens challenged with lipopolysaccharide. Front Vet Sci 2023; 10:1126911. [PMID: 36865438 PMCID: PMC9974168 DOI: 10.3389/fvets.2023.1126911] [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: 12/18/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Herein, Galla Chinensis tannin (GCT) was examined for its influence on preventing lipopolysaccharide (LPS)-induced liver damage in broiler chickens. Approximately 486 one-day-old healthy broilers were randomly allocated to 3 treatment groups (control, LPS, and LPS + GCT). The control and LPS groups were fed a basal diet and the LPS+GCT group was fed the basal diet supplemented with 300 mg/kg GCT. LPS was intraperitoneally injected (1 mg/kg body weight BW) in broilers in the LPS and LPS+GCT groups at 17, 19, and 21 days of age. The results manifested that dietary GCT addition attenuated LPS-induced deleterious effects on serum parameters and significantly increased serum immunoglobulin and complement C3 concentrations relative to the control and LPS groups. Dietary supplementation of GCT inhibited LPS-induced increase in broiler hepatic inflammatory cytokines, caspases activities, and TLR4/NF-κB pathway-related gene mRNA expression. Therefore, 300 mg/kg GCT addition to the diet improved the immune function of broilers and inhibit liver inflammation by blocking the TLR4/NF-κB pathway. Our findings provide support for the application of GCT in poultry production.
Collapse
Affiliation(s)
- Peng Yuan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Haitao Xu
- Animal Husbandry Development Center of Changyi City, Weifang, China
| | - Yuanfei Ma
- Agricultural and Rural Comprehensive Service Center of Bincheng District, Binzhou, China
| | - Jiaxing Niu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Yang Liu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Libo Huang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Shuzhen Jiang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Ning Jiao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Xuejun Yuan
- College of Life Sciences, Shandong Agricultural University, Tai'an, China
| | - Weiren Yang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China,*Correspondence: Weiren Yang ✉
| | - Yang Li
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China,Yang Li ✉
| |
Collapse
|
19
|
Zeng YJ, Liu F, Wu M, Wu XP, Zhang DL, Yuan QQ, Zhou L, Wu ZH. Curcumin combined with arsenic trioxide in the treatment of acute myeloid leukemia: network pharmacology analysis and experimental validation. J Cancer Res Clin Oncol 2023; 149:219-230. [PMID: 36352148 DOI: 10.1007/s00432-022-04463-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE This study aimed to evaluate the effects of curcumin by co-administration of arsenic trioxide (As2O3) in acute myeloid leukemia (AML) treatment, using network pharmacology and experimental validation. METHODS Using Pubchem database, Traditional Chinese Medicine Information Database (TCMID) database, and Swiss target prediction database to predict compound-related targets, AML-associated targets were determined using GeneCards and Online Mendelian Inheritance in Man (OMIM) databases. We identify overlapping common targets by comparing Compounds-related and AML-associated targets and using these targets to perform GO and KEGG functional enrichment analyses. Subsequently, these targets were input into the STRING database, and we used Cytoscape to construct protein-protein interaction (PPI) network. Finally, we used KG1-a cells and the AML mouse model to measure the anti-leukemia effects of curcumin and As2O3 and their combination. RESULTS Compounds and targets screening hinted that 85 intersection targets were predicted in the curcumin treatment of AML, 75 targets in the As2O3 treatment of AML, and 48 targets in the curcumin combined with the As2O3 treatment of AML. GO and KEGG analyses indicated that the top 10 enriched biological processes and top 20 pathways implicated in the therapeutic effects of curcumin and As2O3 on AML, respectively. In addition, network pharmacology screening revealed STAT3, TP53, EP300, MAPK1, and PIK3CA as the top five genes in PPI network of curcumin treatment of AML and TP53, MAPK3, MAPK1, STAT3, and SRC as the top five genes in PPI network of As2O3 treatment of AML. Moreover, the in vitro experiment demonstrated that curcumin combined with As2O3 inhibited proliferation and induced apoptosis in KG1-a cells, and this effect is more substantial than curcumin or As2O3 alone. Mechanistically, the curcumin combined with As2O3 significantly down-regulated the protein expression of JAK2, STAT3, and Bcl-2, and up-regulated the levels of P53, P27, and Bax. In the mouse model, the survival time of mice in each administration group was drawn out to varying degrees, with the most significant prolongation in the curcumin combined with the As2O3 group. CONCLUSION Our results suggested that curcumin and As2O3 combination therapy exerts more significant anti-leukemia effects in the treatment of AML than curcumin or As2O3 monotherapy by up-regulating p53 pathway and down-regulating the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Ying-Jian Zeng
- Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Xinjian District, Nanchang, People's Republic of China.,The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Fan Liu
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Min Wu
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Xin-Ping Wu
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Da-Ling Zhang
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Qiu-Quan Yuan
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Lu Zhou
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China
| | - Zhen-Hui Wu
- Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Xinjian District, Nanchang, People's Republic of China. .,The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People's Republic of China.
| |
Collapse
|
20
|
Chêne C, Rongvaux-Gaïda D, Thomas M, Rieger F, Nicco C, Batteux F. Optimal combination of arsenic trioxide and copper ions to prevent autoimmunity in a murine HOCl-induced model of systemic sclerosis. Front Immunol 2023; 14:1149869. [PMID: 37063915 PMCID: PMC10097895 DOI: 10.3389/fimmu.2023.1149869] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/06/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction Systemic sclerosis (SSc) is a rare chronic autoimmune disease characterized by diffuse fibrosis of the skin and internal organs and vascular abnormalities. The etiology and physiopathology are complex due to the heterogeneity of its overall clinical presentation. Arsenic trioxide (ATO) has been proven to be effective against SSc, sclerodermatous Graft-versus-Host Disease, multiple sclerosis, Crohn's disease or systemic lupus erythematosus animal models and has demonstrated promising effects in human clinical trials. Its efficacy was shown to be related at least in part to the generation of Reactive Oxygen Species (ROS) and the selective deletion of activated immune cells and fibroblasts. However, ATO can induce some adverse effects that must be considered, especially when used for the treatment of a chronic disease. Methods We evaluate here, in vitro and in a mouse model of SSc, the improved efficacy of ATO when associated with a Fenton-like divalent cation, namely copper chloride (CuCl2), also known to trigger the production of ROS. Results In preliminary experiments in vitro, ATO 1 µM + CuCl2 0.5 µM increased ROS production and increased apoptosis of NIH 3T3 murine fibroblasts compared to 1 µM ATO alone. In vivo, in the HOCl-induced mouse model of SSc, co-treatment with ATO 2.5 μg/g + CuCl2 0.5 μg/g significantly alleviated clinical signs such as the thickening of the skin (p<0.01) and cutaneous fibrosis, in a manner equivalent to treatment with ATO 5 µg/g. Our results provide evidence that co-treatment with ATO 2.5 μg/g + CuCl2 0.5 μg/g decreases the number of B cells and the activation of CD4+ T lymphocytes. The co-treatment substantially blocks the NRF2 signaling pathway, increases H2O2 production and results in the improvement of the health status of mice with experimental SSc. Conclusion In conclusion, copper combined with ATO treatment halved the concentration of ATO needed to obtain the same effect as a high dose of ATO alone for the treatment of SSc mice. The strategy of using lower doses of drugs with different mechanisms of action in combination has many potential advantages, the first being to lessen the potential side effects induced by ATO, a drug with side effects quickly increased with dosage.
Collapse
Affiliation(s)
- Charlotte Chêne
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
- R&D Department, MEDSENIC SAS, Strasbourg, France
| | | | - Marine Thomas
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
| | | | - Carole Nicco
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
- *Correspondence: Frédéric Batteux, ; Carole Nicco,
| | - Frédéric Batteux
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
- Service d’immunologie Biologique, AP-HP-Centre Université Paris Cité, Hôpital Cochin, Université Paris Cité, Faculté De Médecine, Paris, France
- *Correspondence: Frédéric Batteux, ; Carole Nicco,
| |
Collapse
|
21
|
Sahakyan G, Vejux A, Sahakyan N. The Role of Oxidative Stress-Mediated Inflammation in the Development of T2DM-Induced Diabetic Nephropathy: Possible Preventive Action of Tannins and Other Oligomeric Polyphenols. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27249035. [PMID: 36558167 PMCID: PMC9786776 DOI: 10.3390/molecules27249035] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Diabetic nephropathy is manifested in more than 10% of people with diabetes. It is a common cause of kidney failure and end-stage kidney disease. Understanding of mechanisms underlying the initiation and development of diabetes-induced kidney injuries will allow for the development of more effective methods of prevention and treatment of the disease. Diabetic nephropathy is a wide-ranging complication of diabetes, and it is necessary to discuss the "weight" of pro-inflammatory pathways and molecules in the progress of renal injuries during the development of the disease. A large spectrum of pro-inflammatory molecules and pathways participate in different stages of the pathophysiological progression of diabetic nephropathy, including pro-inflammatory cytokines, chemokines, their receptors, adhesion molecules, and transcription factors. On the other hand, it is known that one of the consequences of hyperglycemia-induced ROS generation is the up-regulation of pro-inflammatory cascades, which, in turn, activate the transcription of genes encoding cytokines-chemokines, growth factors, and extracellular matrix proteins. It is a proven fact that a variety of plant secondary metabolites, such as tannins, flavonoids, and other polyphenols, demonstrate significant anti-diabetic, redox-modulating properties and effectively modulate the inflammatory response. Thus, this review is discussing the possible role of plant phenols in the prevention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Gohar Sahakyan
- Research Institute of Biology, Yerevan State University, 1 A. Manoogian Str., Yerevan 0025, Armenia
| | - Anne Vejux
- Team “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism”, University Bourgogne Franche-Comté, UFR Sciences Vie Terre et Environnement, 21000 Dijon, France
- Correspondence: (A.V.); (N.S.); Tel.: +33 3-80-39-37-01 (A.V.); Tel.: +374-60-71-05-07 (N.S.)
| | - Naira Sahakyan
- Research Institute of Biology, Yerevan State University, 1 A. Manoogian Str., Yerevan 0025, Armenia
- Research Institute of Biology, Department of Biochemistry, Microbiology & Biotechnology, Yerevan State University, 1 A. Manoogian Str., Yerevan 0025, Armenia
- Correspondence: (A.V.); (N.S.); Tel.: +33 3-80-39-37-01 (A.V.); Tel.: +374-60-71-05-07 (N.S.)
| |
Collapse
|
22
|
Ameliorative effects of Danshensu from the functional food Salvia miltiorrhiza against arsenic trioxide-induced cardiac toxicity in vivo and in vitro: Involvement of inhibiting the AKT/IKK/NF-κB signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
23
|
Sadiku OO, Rodríguez-Seijo A. Metabolic and genetic derangement: a review of mechanisms involved in arsenic and lead toxicity and genotoxicity. Arh Hig Rada Toksikol 2022; 73:244-255. [PMID: 36607725 PMCID: PMC9985351 DOI: 10.2478/aiht-2022-73-3669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/01/2022] [Accepted: 10/01/2022] [Indexed: 01/07/2023] Open
Abstract
Urbanisation and industrialisation are on the rise all over the world. Environmental contaminants such as potentially toxic elements (PTEs) are directly linked with both phenomena. Two PTEs that raise greatest concern are arsenic (As) and lead (Pb) as soil and drinking water contaminants, whether they are naturally occurring or the consequence of human activities. Both elements are potential carcinogens. This paper reviews the mechanisms by which As and Pb impair metabolic processes and cause genetic damage in humans. Despite efforts to ban or limit their use, due to high persistence both continue to pose a risk to human health, which justifies the need for further toxicological research.
Collapse
Affiliation(s)
- Olubusayo Olujimi Sadiku
- University of Lagos, College of Medicine, Faculty of Basic Medical Sciences, Department of Medical Laboratory Science, Lagos, Nigeria
| | - Andrés Rodríguez-Seijo
- University of Porto, Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Matosinhos, Portugal
- University of Porto, Faculty of Sciences, Biology Department, Porto, Portugal
- University of Vigo, Department of Plant Biology and Soil Sciences, Ourense, Spain
| |
Collapse
|
24
|
Tian S, Zhao H, Song H. Shared signaling pathways and targeted therapy by natural bioactive compounds for obesity and type 2 diabetes. Crit Rev Food Sci Nutr 2022; 64:5039-5056. [PMID: 36397728 DOI: 10.1080/10408398.2022.2148090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epidemiological evidence showed that patients suffering from obesity and T2DM are significantly at higher risk for chronic low-grade inflammation, oxidative stress, nonalcoholic fatty liver (NAFLD) and intestinal flora imbalance. Increasing evidence of pathological characteristics illustrates that some common signaling pathways participate in the occurrence, progression, treatment, and prevention of obesity and T2DM. These signaling pathways contain the pivotal players in glucose and lipid metabolism, e.g., AMPK, PI3K/AKT, FGF21, Hedgehog, Notch, and WNT; the inflammation response, for instance, Nrf2, MAPK, NF- kB, and JAK/STAT. Bioactive compounds from plants have emerged as key food components related to healthy status and disease prevention. They can act as signaling molecules to initiate or mediate signaling transduction that regulates cell function and homeostasis to repair and re-functionalize the damaged tissues and organs. Therefore, it is crucial to continuously investigate bioactive compounds as sources of new pharmaceuticals for obesity and T2DM. This review provides comprehensive information of the commonly shared signaling pathways between obesity and T2DM, and we also summarize the therapeutic bioactive compounds that may serve as anti-obesity and/or anti-diabetes therapeutics by regulating these associated pathways, which contribute to improving glucose and lipid metabolism, attenuating inflammation.
Collapse
Affiliation(s)
- Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Haizhen Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
25
|
Wang M, Huang H, Wang L, Yin L, Yang H, Chen C, Zheng Q, He S. Tannic acid attenuates intestinal oxidative damage by improving antioxidant capacity and intestinal barrier in weaned piglets and IPEC-J2 cells. Front Nutr 2022; 9:1012207. [PMID: 36407512 PMCID: PMC9672516 DOI: 10.3389/fnut.2022.1012207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/15/2022] [Indexed: 07/05/2024] Open
Abstract
Tannic acid (TA) has received widespread attention for its beneficial biological function with antioxidant capacity. This study investigated the protective role of TA on the intestinal antioxidant capacity and intestinal barrier in weaned piglets and porcine intestinal epithelial cells (IPEC-J2). A total of 18 weaned piglets were randomly allocated into two groups (n = 9) and fed with a basal diet (control, CON) and a basal diet containing 1,000 mg/kg TA for two weeks. The in vivo results showed that treatment with TA increased both glutathione peroxidase (GSH-PX) activity and the protein expression of ZO-1 in the jejunum of weaned piglets, and reduced the level of malondialdehyde (MDA) in the serum and the mRNA and protein expression of Keap1 in the jejunum of weaned piglets. Furthermore, in vitro results indicated that TA treatment effectively alleviated tert-butyl hydroperoxide (TBH)-induced oxidative stress in IPEC-J2 cells, improved the antioxidant capacity by elevating the cell redox state and activating the Nrf2 pathway, and improved the intestinal barrier by upregulating the mRNA and protein expression of intestinal tight junction proteins and increasing the transepithelial electrical resistance (TEER) value. In conclusion, these results confirmed that TA relieves oxidative injury and improves intestinal barrier function and intestinal antioxidant capacity by activating the Nrf2 signaling pathway. These findings suggest that TA has the potential application in alleviating oxidative stress in the intestine of weaned piglets.
Collapse
Affiliation(s)
- Meiwei Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan Normal University, Changsha City, China
| | - Huijun Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan Normal University, Changsha City, China
| | - Lei Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan Normal University, Changsha City, China
| | - Lanmei Yin
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan Normal University, Changsha City, China
- Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha City, China
| | - Huansheng Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan Normal University, Changsha City, China
- Chinese Academy of Science, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agroecological Processes in Subtropical Region, Scientific Observation and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha City, China
| | - Chiqing Chen
- Wufeng Chicheng Biotechnology Company Limited, Yichang City, China
| | | | - Shanping He
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan Normal University, Changsha City, China
| |
Collapse
|
26
|
Mishra M, Nichols L, Dave AA, Pittman EH, Cheek JP, Caroland AJV, Lotwala P, Drummond J, Bridges CC. Molecular Mechanisms of Cellular Injury and Role of Toxic Heavy Metals in Chronic Kidney Disease. Int J Mol Sci 2022; 23:11105. [PMID: 36232403 PMCID: PMC9569673 DOI: 10.3390/ijms231911105] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive disease that affects millions of adults every year. Major risk factors include diabetes, hypertension, and obesity, which affect millions of adults worldwide. CKD is characterized by cellular injury followed by permanent loss of functional nephrons. As injured cells die and nephrons become sclerotic, remaining healthy nephrons attempt to compensate by undergoing various structural, molecular, and functional changes. While these changes are designed to maintain appropriate renal function, they may lead to additional cellular injury and progression of disease. As CKD progresses and filtration decreases, the ability to eliminate metabolic wastes and environmental toxicants declines. The inability to eliminate environmental toxicants such as arsenic, cadmium, and mercury may contribute to cellular injury and enhance the progression of CKD. The present review describes major molecular alterations that contribute to the pathogenesis of CKD and the effects of arsenic, cadmium, and mercury on the progression of CKD.
Collapse
Affiliation(s)
- Manish Mishra
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Larry Nichols
- Department of Pathology and Clinical Sciences Education, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Aditi A. Dave
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Elizabeth H Pittman
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - John P. Cheek
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Anasalea J. V. Caroland
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Purva Lotwala
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - James Drummond
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Christy C. Bridges
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|
27
|
Jing C, Niu J, Liu Y, Jiao N, Huang L, Jiang S, Yan L, Yang W, Li Y. Tannic Acid Extracted from Galla chinensis Supplementation in the Diet Improves Intestinal Development through Suppressing Inflammatory Responses via Blockage of NF-κB in Broiler Chickens. Animals (Basel) 2022; 12:2397. [PMID: 36139256 PMCID: PMC9495145 DOI: 10.3390/ani12182397] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/11/2022] [Accepted: 09/11/2022] [Indexed: 12/27/2022] Open
Abstract
The objective of this study was to investigate the effects of adding tannic acid (TA) extracted from Galla chinensis to the diet of broiler chickens on intestinal development. A total of 324 healthy 1-day-old broilers were used in a 42 d study, and divided into two treatment groups at random (six replicates per group). Broilers were either received a basal diet or a basal diet supplemented with 300 mg/kg microencapsulated TA extracted from Galla chinensis. The results showed that dietary supplemented with 300 mg/kg TA from Galla chinensis improved intestinal morphology, promoted intestinal mucosal barrier integrity, and elevated mucosal expressions of nutrients transporters and tight junction protein CLDN3 in broilers. Besides, 300 mg/kg TA from Galla chinensis supplementation decreased the concentrations of inflammatory cytokines in serum and intestinal mucosa and reduced the mRNA expression of NF-κB in intestinal mucosa. Above all, supplementation of 300 mg/kg microencapsulated TA extracted from Galla chinensis showed beneficial effects in improving intestinal development, which might be attributed to the suppression of inflammatory responses via blockage of NF-κB in broiler chickens. These findings will support the use of TA sourced from Galla chinensis in poultry industry.
Collapse
Affiliation(s)
- Changwei Jing
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Shizishan Street 1#, Wuhan 430070, China
| | - Jiaxing Niu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Yang Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Ning Jiao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Libo Huang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Shuzhen Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Lei Yan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
- Shandong New Hope Liuhe Group Co., Ltd., Jiudongshui Road 592-26#, Qingdao 266100, China
| | - Weiren Yang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Yang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| |
Collapse
|
28
|
Laha D, Sarkar J, Maity J, Pramanik A, Howlader MSI, Barthels D, Das H. Polyphenolic Compounds Inhibit Osteoclast Differentiation While Reducing Autophagy through Limiting ROS and the Mitochondrial Membrane Potential. Biomolecules 2022; 12:biom12091220. [PMID: 36139058 PMCID: PMC9496366 DOI: 10.3390/biom12091220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/22/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Polyphenolic compounds are a diverse group of natural compounds that interact with various cellular proteins responsible for cell survival, differentiation, and apoptosis. However, it is yet to be established how these compounds interact in myeloid cells during their differentiation and the molecular and intracellular mechanisms involved. Osteoclasts are multinucleated cells that originate from myeloid cells. They resorb cartilage and bone, maintain bone homeostasis, and can cause pathogenesis. Autophagy is a cellular mechanism that is responsible for the degradation of damaged proteins and organelles within cells and helps maintain intracellular homeostasis. Imbalances in autophagy cause various pathological disorders. The current study investigated the role of several polyphenolic compounds, including tannic acid (TA), gallic acid (GA), and ellagic acid (EA) in the regulation of osteoclast differentiation of myeloid cells. We demonstrated that polyphenolic compounds inhibit osteoclast differentiation in a dose-dependent manner. Quantitative real-time PCR, immunocytochemistry, and western blotting revealed that osteoclast markers, such as NFATc1, Cathepsin K, and TRAP were inhibited after the addition of polyphenolic compounds during osteoclast differentiation. In our investigation into the molecular mechanisms, we found that the addition of polyphenolic compounds reduced the number of autophagic vesicles and the levels of LC3B, BECN1, ATG5, and ATG7 molecules through the inactivation of Akt, thus inhibiting the autophagy process. In addition, we found that by decreasing intracellular calcium and decreasing ROS levels, along with decreasing mitochondrial membrane potential, polyphenolic compounds inhibit osteoclast differentiation. Together, this study provides evidence that polyphenolic compounds inhibit osteoclast differentiation by reducing ROS production, autophagy, intracellular Ca2+ level, and mitochondrial membrane potentials.
Collapse
|
29
|
Jing W, Xiaolan C, Yu C, Feng Q, Haifeng Y. Pharmacological effects and mechanisms of tannic acid. Biomed Pharmacother 2022; 154:113561. [PMID: 36029537 DOI: 10.1016/j.biopha.2022.113561] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022] Open
Abstract
In recent years, increasing attention has been paid to the pharmacological efficacy of tannins. Tannic acid (TA), the simplest hydrolysable tannin that has been approved by the FDA as a safe food additive, is one of the most important components of these traditional medicines. Studies have shown that TA displays a wide range of pharmacological activities, such as anti-inflammatory, neuroprotective, antitumor, cardioprotective, and anti-pathogenic effects. Here, we summarize the known pharmacological effects and associated mechanisms of TA. We focus on the effect and mechanism of TA in various animal models of inflammatory disease and organ, brain, and cardiovascular injury. Moreover, we discuss the possible molecular targets and signaling pathways of TA, in addition to the pharmacological effects of TA-based nanoparticles and TA in combination with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Wang Jing
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China.
| | - Chen Xiaolan
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| | - Chen Yu
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| | - Qin Feng
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, PR China
| | - Yang Haifeng
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| |
Collapse
|
30
|
Kim SW, Kim DB, Kim HS. Neuroprotective effects of tannic acid in the postischemic brain via direct chelation of Zn 2+. Anim Cells Syst (Seoul) 2022; 26:183-191. [PMID: 36046027 PMCID: PMC9423855 DOI: 10.1080/19768354.2022.2113915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Seung Woo Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Korea
| | - Da Bin Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Korea
| |
Collapse
|
31
|
Qi X, Tong X, You S, Mao R, Cai E, Pan W, Zhang C, Hu R, Shen J. Mild Hyperthermia-Assisted ROS Scavenging Hydrogels Achieve Diabetic Wound Healing. ACS Macro Lett 2022; 11:861-867. [PMID: 35759676 DOI: 10.1021/acsmacrolett.2c00290] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Excessive reactive oxygen species (ROS) production induces oxidative damage to biomolecules, which can lead to the development of chronic diseases. Biocompatible hydrogel antioxidants composed of natural materials, such as polysaccharides and polyphenols, are of significant option for ROS scavenging. However, rapidly achieving hydrogel antioxidants with convenient, economical, safe, and efficient features remains challenging. Herein, facile synthesis of a physically cross-linked polyphenol/polysaccharide hydrogel by introducing tannic acid microsize particles (TAMP) into a cationic guar gum (CG) matrix is reported. Combining antioxidant/photothermal properties of TAMP and mechanical support from injectable CG, the formulated TAMP/CG is explored for treating diabetic wounds. Both in vitro and in vivo assays verify that TAMP/CG can protect the cells from ROS-induced oxidative damage, which can also be strengthened by the local photothermal heating (42 °C) triggered by near-infrared light. Overall, this study establishes the paradigm of enhanced diabetic wound healing by mild hyperthermia-assisted ROS scavenging hydrogels.
Collapse
Affiliation(s)
- Xiaoliang Qi
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xianqin Tong
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shengye You
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ruiting Mao
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Erya Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wenhao Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chenhao Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Rongdang Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianliang Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| |
Collapse
|
32
|
Han X, Yang Y, Qi J, Zhang M, Xue Y, Chu X, Jia Q, Sun S, Guan S. Protective effects and possible mechanism of 6-gingerol against arsenic trioxide-induced nephrotoxicity based on network pharmacological analysis and experimental validation. Int Immunopharmacol 2022; 110:108926. [PMID: 35728306 DOI: 10.1016/j.intimp.2022.108926] [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: 04/06/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Nephrotoxicity induced by the chemotherapeutic drug arsenic trioxide (ATO) is often overlooked, and the underlying mechanisms remain poorly understood. Based on network pharmacology and experimental validation, this study investigates the protection of 6-gingerol (6G) against ATO-induced nephrotoxicity and the potential mechanisms. METHODS We screened and collected 6G and disease-related targets and then imported the interaction targets into a String database to construct protein-protein interaction (PPI) networks. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). Mice were injected intraperitoneally with ATO (5 mg/kg) for seven days to induce nephrotoxicity, and then the histological morphology of the kidneys, biochemical indices of serum and tissues, and associated protein expressions were observed. RESULTS The network pharmacology results revealed that the effects of 6G against nephrotoxicity are closely related to apoptosis, and the MAPKs pathway was screened for validation. In animal experiments, 6G improved the histopathological morphology of the kidneys, reduced the levels of renal function markers, enhanced antioxidant activity, and decreased the levels of inflammation. Furthermore, 6G reduced apoptotic cells in kidney tissues, decreased the levels of Bax and c-Caspase-3, and increased the level of Bcl-2. The results of immunohistochemistry and western blotting revealed that 6G significantly inhibited the expressions of p-p38, p-ERK, and p-JNK. CONCLUSION The results comprehensively demonstrate the protective effects of 6G against ATO-induced nephrotoxicity. The effects are related to anti-oxidant, anti-inflammatory, and anti-apoptotic properties, possibly through inhibition of the MAPKs pathway.
Collapse
Affiliation(s)
- Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yakun Yang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jiaying Qi
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Muqing Zhang
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China; Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yucong Xue
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xi Chu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qingzhong Jia
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Shijiang Sun
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China.
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China; School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China.
| |
Collapse
|
33
|
Abstract
Arsenic toxicity is a major concern due to its deleterious consequences for human health. Rapid industrialization also has weakened the quality of the environment by introducing pollutants that may disrupt balanced ecosystems, adversely and irreversibly impacting humans, plants, and animals. Arsenic, an important toxicant among all environmental hazards, can lead to several detrimental effects on cells and organs, impacting the overall quality of life. Nevertheless, arsenic also has a rich history as a chemotherapeutic agent used in ancient days for the treatment of diseases such as malaria, cancer, plague, and syphilis when other chemotherapeutic agents were yet to be discovered. Arsenicosis-mediated disorders remain a serious problem due to the lack of effective therapeutic options. Initially, chelation therapy was used to metabolically eliminate arsenic by forming a complex, but adverse effects limited their pharmacological use. More recently, plant-based products have been found to provide significant relief from the toxic effects of arsenic poisoning. They act by different mechanisms affecting various cellular processes. Phytoconstituents such as curcumin, quercetin, diallyl trisulfide, thymoquinone, and others act via various molecular pathways, primarily by attenuating oxidative damage, membrane damage, DNA damage, and proteinopathies. Nonetheless, most of the phytochemicals reviewed here protect against the adverse effects of metal or metalloid exposure, supporting their consideration as alternatives to chelation therapy. These agents, if used prophylactically and in conjunction with other chemotherapeutic agents, may provide an effective approach for management of arsenic toxicity. In a few instances, such strategies like coadministration of phytochemicals with a known chelating agent have led to more pronounced elimination of arsenic from the body with lesser off-site adverse effects. This is possible because combination treatment ensures the use of a reduced dose of chelating agent with a phytochemical without compromising treatment. Thus, these therapies are more practical than conventional therapeutic agents in ameliorating arsenic-mediated toxicity. This review summarizes the potential of phytochemicals in alleviating arsenic toxicity on the basis of available experimental and clinical evidence.
Collapse
Affiliation(s)
- Sabiya Samim Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | - Ankita Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226020, India
| | - Swaran J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226020, India
| |
Collapse
|
34
|
Han X, Yang Y, Zhang M, Chu X, Zheng B, Liu C, Xue Y, Guan S, Sun S, Jia Q. Protective Effects of 6-Gingerol on Cardiotoxicity Induced by Arsenic Trioxide Through AMPK/SIRT1/PGC-1α Signaling Pathway. Front Pharmacol 2022; 13:868393. [PMID: 35571130 PMCID: PMC9096219 DOI: 10.3389/fphar.2022.868393] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/28/2022] [Indexed: 01/08/2023] Open
Abstract
Background and Objective: Arsenic trioxide (As2O3) induced cardiotoxicity to limit the clinical applications of the effective anticancer agent. 6-Gingerol (6G) is the main active ingredient of ginger, a food with many health benefits. The present study aims to investigate the potential pharmacological mechanisms of 6G on As2O3-induced myocardial injury. Methods and Results: Fifty KunMing mice were divided into five groups (n = 10) receiving: 1) physiological saline; 2) 6G (20 mg/kg) alone; 3) As2O3 (5 mg/kg); 4) 6G (10 mg/kg) and As2O3 (5 mg/kg); 5) 6G (20 mg/kg) and As2O3 (5 mg/kg). 6G was given orally and As2O3 was given intraperitoneally once per day for seven consecutive days. Biochemical, histopathological, transmission electron microscopy, ELISA, and western blotting analyses were then performed. Based on the resultant data, As2O3 was found to induce cardiotoxicity in mice. 6G significantly ameliorated As2O3-induced heart injury, histopathological changes, oxidative stress, myocardial mitochondrial damage, inflammation, and cardiomyocyte apoptosis, while reversed As2O3-induced inhibition of the AMPK/SIRT1/PGC-1α pathway. Conclusion: Our experimental results reveal that 6G effectively counteracts As2O3-induced cardiotoxicity including oxidative stress, inflammation and apoptosis, which might be attributed to its activation action on AMPK/SIRT1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yakun Yang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Muqing Zhang
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xi Chu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bin Zheng
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chenxu Liu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yucong Xue
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
- *Correspondence: Shengjiang Guan, ; Shijiang Sun, ; Qingzhong Jia,
| | - Shijiang Sun
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
- *Correspondence: Shengjiang Guan, ; Shijiang Sun, ; Qingzhong Jia,
| | - Qingzhong Jia
- School of Pharmacy, Hebei Medical University, Shijiazhuang, China
- *Correspondence: Shengjiang Guan, ; Shijiang Sun, ; Qingzhong Jia,
| |
Collapse
|
35
|
Basist P, Parveen B, Zahiruddin S, Gautam G, Parveen R, Khan MA, Krishnan A, Shahid M, Ahmad S. Potential nephroprotective phytochemicals: Mechanism and future prospects. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114743. [PMID: 34655670 DOI: 10.1016/j.jep.2021.114743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/24/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kidney disease (KD) is one of the serious health issues, which causes worrisome morbidity and economic burden. Therapeutic strategies are available however majority of them are associated with severe adverse effects and poor patient compliance and adherence. This explorative article was undertaken to provide a holistic review of known nephroprotective (NP) phytoconstituents along with their research-based evidences on mechanism, sources, and clinical trials that may play essential role in prevention and cure of KD. AIM OF THE STUDY The present systematic review aimed to provide in-depth and better evidences of the global burden of KD, phytoconstituents as NP with emphasis on mechanism of action both in vitro and in vivo, their wide biological sources as well as their clinical efficacy in management of kidney disease and its related disorders. MATERIAL AND METHODS Comprehensive information was searched systematically from electronic databases, namely, PubMed, Sciencedirect, Wiley, Scopus, Google scholar and Springer until February 2021 to find relevant data for publication on phytoconstituents with nephroprotective potential. RESULTS In total, 24,327 articles were screened in first search for "phytoconstituents and medicinal plants for nephroprotection and kidney disorder". On the basis of exclusion and inclusion criteria, 24,091 were excluded. Only 236 papers were spotted to have superlative quality data, which is appropriate under titles and sub-titles of the present review. The phytoconstituents having multiple research evidence along with wide number of medicinal plants sources and mechanism reported for nephroprotection have been selected and reviewed. CONCLUSION This review, based on pre-clinical and clinical data of NP phytoconstituents, provides scientific-basis for the rational discovery, development and utilization of these upcoming treatment practices. Further,-more clinical studies are warranted to improve the pharmacodynamic and pharmacokinetic understanding of phytoconstituents. Also, more specific evaluation for natural sources is needed.
Collapse
Affiliation(s)
- Parakh Basist
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Bushra Parveen
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Sultan Zahiruddin
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Gaurav Gautam
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Rabea Parveen
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Human Genetics Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Anuja Krishnan
- Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Shahid
- Department of Pharmaceutical Sciences, Chicago State University College of Pharmacy, Chicago, IL, 60423, USA
| | - Sayeed Ahmad
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
36
|
Sehati F, Ahmadi I, Farivar N, Ranjbaran M, Sadat-Shirazi MS, Nabavizadeh F, Mahla Shavakandi S, Ashabi G. Tannic acid protects aged brain against cerebral hypoperfusion via modulation of Nrf2 and inflammatory pathways. Neurosci Lett 2021; 765:136263. [PMID: 34562517 DOI: 10.1016/j.neulet.2021.136263] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 02/01/2023]
Abstract
Current study purposed to investigate the neuroprotective effects of Tannic Acid (TA) on mild chronic cerebral hypoperfusion model in rats. Male Wistar rats were subjected to permanent Unilateral Common Carotid Artery Occlusion (UCCAO), followed by TA treatment (0.05% w/v) in drinking water for one month. Nuclear factor erythroid 2-related factor 2 (Nrf2), NAD(P)H: quinone oxidoreductase 1 (NQO-1), heme oxygenase-1 (HO-1), factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumor necrosis factor-α (TNF-α), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), caspase-3, blood triglyceride, blood glucose, and liver enzymes' activity were detected after the experimental period. Also, behavioral tests, hematoxylin and eosin (H&E) staining, and PET scan were performed after treatment. Post-treatment of TA improved locomotion and memory function (P < 0.001), and reduced neural cell death (P < 0.001) in the treatment group compared to UCCAO rats. Furthermore, long-term TA treatment significantly increased the levels of Nrf2 (P < 0.001), NQO-1 (P < 0.001), and HO-1 (P < 0.001) in the hippocampus of the treatment group compared to the UCCAO group. TA consumption in the treatment group applied its anti-inflammatory effects via reducing the activity of NF-κB and TNF-α in comparison with the UCCAO group (P < 0.001 for both). Blood triglyceride, blood glucose, and liver enzymes did not change considerably in the groups (P > 0.05). The current results indicate that long-term post-treatment of TA exhibits protective effects against memory deficit and motor dysfunction. The cellular mechanism of TA in hypoperfused rats might be associated with the activation of antioxidant pathways, especially the Nrf2 pathway, and suppressing inflammatory factors like NF-κB and TNF-α.
Collapse
Affiliation(s)
- Fardin Sehati
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ahmadi
- Department of Physiology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Nika Farivar
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra-Sadat Sadat-Shirazi
- Department of Genetic, Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ghorbangol Ashabi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Jia Y, Li J, Liu P, Si M, Jin Y, Wang H, Ma D, Chu L. Based on Activation of p62-Keap1-Nrf2 Pathway, Hesperidin Protects Arsenic-Trioxide-Induced Cardiotoxicity in Mice. Front Pharmacol 2021; 12:758670. [PMID: 34721041 PMCID: PMC8548645 DOI: 10.3389/fphar.2021.758670] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/21/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Hesperidin (HES) is a flavonoid glycoside found in the tangerine peel and has antioxidant properties. Arsenic trioxide (ATO) is an anti-tumour drug; however, its serious cardiotoxicity limits its clinical application. In addition, the protection of HES against ATO-induced cardiotoxicity has not been explored. Objective: The study aims to investigate and identify the underlying effect and mechanism of HES on ATO-induced cardiotoxicity. Methods: Fifty mice were randomly assigned to five groups. Mice were orally given HES:100 or 300 mg/kg/day concurrently and given ATO intraperitoneal injections: 7.5 mg/kg/day for 1 week. Blood and heart tissues were collected for examination. Evaluated in serum was the levels of creatine kinase (CK), lactate dehydrogenase (LDH) and cardiac troponin I (cTnI). In addition, evaluated in heart tissues were the levels of reactive oxygen species (ROS), superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), catalase (CAT), tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6), B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), Caspase-3, cleaved-Caspase-3, p62, Kelch-like ECH-associated protein 1 (Keap1), and nuclear factor erythroid 2-related factor 2 (Nrf2). The heart tissues were also examined for histopathology and mitochondrial ultrastructure. Results: Compared with the ATO group, the HES treatment groups reduced the levels of CK, LDH, cTnI, ROS, MDA, TNF-α, IL-6, Bax, Caspase-3, cleaved-Caspase-3 and Keap1 and enhanced the levels of SOD, GSH, CAT, Bcl-2, p62 and Nrf2. Conclusions: The results demonstrate that HES protects against ATO-induced cardiotoxicity, through inhibiting oxidative stress, and subsequent inflammation and apoptosis. The underlying results are closely related to the regulation of the p62-Keap1-Nrf2 signalling pathway.
Collapse
Affiliation(s)
- Yuxin Jia
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jing Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Panpan Liu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Mingdong Si
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yanyu Jin
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hongfang Wang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Donglai Ma
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China.,Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China.,Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China.,Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
38
|
Molaei E, Molaei A, Abedi F, Hayes AW, Karimi G. Nephroprotective activity of natural products against chemical toxicants: The role of Nrf2/ARE signaling pathway. Food Sci Nutr 2021; 9:3362-3384. [PMID: 34136201 PMCID: PMC8194945 DOI: 10.1002/fsn3.2320] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Nephropathy can occur following exposure of the kidneys to oxidative stress. Oxidative stress is the result of reactive oxygen species (ROS) formation due to intracellular catabolism or exogenous toxicant exposure. Many natural products (NPs) with antioxidant properties have been used to demonstrate that oxidative damage-induced nephrotoxicity can be ameliorated or at least reduced through stimulation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. Nrf2 is a basic leucine zipper (bZip) transcription factor that regulates gene expression of the antioxidant response elements (ARE). Nrf2 is involved in the cellular antioxidant-detoxification machinery. Nrf2 activation is a major mechanism of nephroprotective activity for these NPs, which facilitates its entry into the nucleus, primarily by inhibiting Kelch like-ECH-associated protein 1 (Keap1). The purpose of this article was to review the peer-reviewed literature of NPs that have shown mitigating effects on renal disorder by stimulating Nrf2 and thereby suggesting potential new therapeutic or prophylactic strategies against kidney-damaging xenobiotics.
Collapse
Affiliation(s)
- Emad Molaei
- Faculty of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Ali Molaei
- Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Farshad Abedi
- Faculty of PharmacyMashhad University of Medical SciencesMashhadIran
| | | | - Gholamreza Karimi
- Pharmaceutical Research CenterInstitute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and ToxicologyFaculty of PharmacyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
39
|
Liu M, Zheng B, Liu P, Zhang J, Chu X, Dong C, Shi J, Liang Y, Chu L, Liu Y, Han X. Exploration of the hepatoprotective effect and mechanism of magnesium isoglycyrrhizinate in mice with arsenic trioxide‑induced acute liver injury. Mol Med Rep 2021; 23:438. [PMID: 33846815 PMCID: PMC8060806 DOI: 10.3892/mmr.2021.12077] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
Arsenic trioxide (ATO)-induced hepatotoxicity limits the therapeutic effect of acute myelogenous leukemia treatment. Magnesium isoglycyrrhizinate (MgIG) is a natural compound extracted from licorice and a hepatoprotective drug used in liver injury. It exhibits anti-oxidant, anti-inflammatory and anti-apoptotic properties. The aim of the present study was to identify the protective action and underlying mechanism of MgIG against ATO-induced hepatotoxicity. A total of 50 mice were randomly divided into five groups (n=10/group): Control; ATO; MgIG and high- and low-dose MgIG + ATO. Following continuous administration of ATO for 7 days, the relative weight of the liver, liver enzyme, histological data, antioxidant enzymes, pro-inflammatory cytokines, cell apoptosis and changes in Kelch-like ECH-associated protein 1/nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) signaling pathway were observed. MgIG decreased liver injury, decreased the liver weight and liver index, inhibited oxidative stress and decreased the activity of glutathione, superoxide dismutase and catalase, production of reactive oxygen species and levels of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α. Western blotting showed a decrease in Bax and caspase-3. There was decreased cleaved caspase-3 expression and increased Bcl-2 expression. MgIG notably activated ATO-mediated expression of Keap1 and Nrf2 in liver tissue. MgIG administration was an effective treatment to protect the liver from ATO-induced toxicity. MgIG maintained the level of Nrf2 in the liver and protected the antioxidative defense system to attenuate oxidative stress and prevent ATO-induced liver injury.
Collapse
Affiliation(s)
- Miaomiao Liu
- Department of Pharmacology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Bin Zheng
- Department of Pharmacology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Panpan Liu
- Department of Pharmacology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jianping Zhang
- Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Chunhui Dong
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jing Shi
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yingran Liang
- Department of Pharmacology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Li Chu
- Department of Pharmacology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yanshuang Liu
- Hebei Key Laboratory of Integrative Medicine on Liver‑Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xue Han
- Department of Pharmacology, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
40
|
Zheng B, Yang Y, Li J, Li J, Zuo S, Chu X, Xu S, Ma D, Chu L. Magnesium Isoglycyrrhizinate Alleviates Arsenic Trioxide-Induced Cardiotoxicity: Contribution of Nrf2 and TLR4/NF-κB Signaling Pathway. Drug Des Devel Ther 2021; 15:543-556. [PMID: 33603344 PMCID: PMC7886103 DOI: 10.2147/dddt.s296405] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Magnesium isoglycyrrhizinate (MgIG), a single stereoisomer magnesium salt of glycyrrhizic acid, has beneficial effects on the cardiovascular system through anti-inflammatory, anti-oxidation, and anti-apoptotic actions. However, MgIG has not been shown to provide protection against cardiotoxicity induced by arsenic trioxide (ATO). This study aims to demonstrate the protection of MgIG against ATO-induced cardiac toxicity in mice and to investigate the underlying mechanism. METHODS A mouse cardiotoxicity model was established by administering 5 mg/kg ATO for 7 days. MgIG used in conjunction with the ATO to assess its cardioprotection. RESULTS MgIG administration could significantly reduce reactive oxygen species generation and the changes in tissue morphology. Also, MgIG administration increased the activity of antioxidase, such as superoxide dismutase, catalase, and glutathione peroxidase, and reduced malondialdehyde content and pro-inflammatory cytokine levels. Western blotting showed decreased expression of Bcl-2 associated X protein and Caspase-3, with increased expression of B-cell lymphoma 2. Importantly, MgIG administration increased nuclear factor-erythroid-2-related factor 2 (Nrf2) expression, while the expressions of nuclear factor kappa-B (NF-κB) and toll-like receptor-4 (TLR4) were significantly decreased. CONCLUSION Our data showed that MgIG alleviates ATO-induced cardiotoxicity, which is associated to the anti-inflammation, anti-oxidation, and anti-apoptosis action, potentially through activation of the Nrf2 pathway and suppression of the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Bin Zheng
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Yakun Yang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Jinghan Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Jing Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Saijie Zuo
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| | - Shan Xu
- Hebei Province Hospital of Chinese Medicine, Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Donglai Ma
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People’s Republic of China
| |
Collapse
|
41
|
Zhao Z, Li J, Zheng B, Liang Y, Shi J, Zhang J, Han X, Chu L, Chu X, Gao Y. Ameliorative effects and mechanism of crocetin in arsenic trioxide‑induced cardiotoxicity in rats. Mol Med Rep 2020; 22:5271-5281. [PMID: 33173984 PMCID: PMC7646993 DOI: 10.3892/mmr.2020.11587] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/20/2020] [Indexed: 01/15/2023] Open
Abstract
Arsenic trioxide (ATO) is commonly used to treat patients with acute promyelocytic leukemia since it was authorized by the U.S. Food and Drug Administration in the 1970s, but its applicability has been limited by its cardiotoxic effects. Therefore, the aim of the present study was to investigate the cardioprotective effects and underlying mechanism of crocetin (CRT), the critical ingredient of saffron. Sprague-Dawley rats were then randomly divided into four groups (n=10/group): i) Control group; ii) ATO group, iii) CRT-low (20 mg/kg) group; and iv) CRT-high (40 mg/kg) group. Rats in the Control and ATO groups were intraperitoneally injected with equal volumes of 0.9% sodium chloride solution, and CRT groups were administered with either 20 and 40 mg/kg CRT. Following 6 h, all groups except the Control group were intraperitoneally injected with 5 mg/kg ATO over 10 days. Cardiotoxicity was indicated by changes in electrocardiographic (ECG) patterns, morphology and marker enzymes. Histomorphological changes in the heart tissue were observed by pathological staining. The levels of superoxide dismutase, glutathione peroxidase, malondialdehyde and catalase in the serum were analyzed using colometric commercial assay kits, and the levels of reactive oxygen species in the heart tissue were detected using the fluorescent probe dihydroethidium. The expression levels of inflammatory factors and activities of apoptosis-related proteins were analyzed using immunohistochemistry. The protein expression levels of silent information regulator of transcription 1 were measured using western blotting. Cardiotoxicity was induced in male Sprague-Dawley rats with ATO (5 mg/kg). CRT (20 and 40 mg/kg) and ATO were co-administered to evaluate possible cardioprotective effects. CRT significantly reduced the heart rate and J-point elevation induced by ATO in rats. Histological changes were evaluated via hematoxylin and eosin staining. CRT decreased the levels of creatine kinase and lactate dehydrogenase, increased the activities of superoxide dismutase, glutathione-peroxidase and catalase, and decreased the levels of malondialdehyde and reactive oxygen species. Moreover, CRT downregulated the expression levels of the pro-inflammatory factors IL-1, TNF-α, IL-6, Bax and p65, as well as increased the expression of Bcl-2. It was also identified that CRT enhanced silent information regulator of transcription 1 protein expression. Thus, the present study demonstrated that CRT treatment effectively ameliorated ATO-induced cardiotoxicity. The protective effects of CRT can be attributed to the inhibition of oxidative stress, inflammation and apoptosis. Therefore, CRT represents a promising therapeutic method for improving the cardiotoxic side effects caused by ATO treatment, and additional clinical applications are possible, but warrant further investigation.
Collapse
Affiliation(s)
- Zhifeng Zhao
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jinghan Li
- Department of Preventive Medicine, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Bin Zheng
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yingran Liang
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jing Shi
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jianping Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver‑Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xue Han
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Li Chu
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yonggang Gao
- Department of Preventive Medicine, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
42
|
Xue Y, Li M, Xue Y, Jin W, Han X, Zhang J, Chu X, Li Z, Chu L. Mechanisms underlying the protective effect of tannic acid against arsenic trioxide‑induced cardiotoxicity in rats: Potential involvement of mitochondrial apoptosis. Mol Med Rep 2020; 22:4663-4674. [PMID: 33173965 PMCID: PMC7646850 DOI: 10.3892/mmr.2020.11586] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022] Open
Abstract
Arsenic trioxide (ATO) is a frontline chemotherapy drug used in the therapy of acute promyelocytic leukemia. However, the clinical use of ATO is hindered by its cardiotoxicity. The present study aimed to observe the potential effects and underlying mechanisms of tannic acid (TA) against ATO-induced cardiotoxicity. Male rats were intraperitoneally injected with ATO (5 mg/kg/day) to induce cardiotoxicity. TA (20 and 40 mg/kg/day) was administered to evaluate its cardioprotective efficacy against ATO-induced heart injury in rats. Administration of ATO resulted in pathological damage in the heart and increased oxidative stress as well as levels of serum cardiac biomarkers creatine kinase and lactate dehydrogenase and the inflammatory marker NF-κB (p65). Conversely, TA markedly reversed this phenomenon. Additionally, TA treatment caused a notable decrease in the expression levels of cleaved caspase-3/caspase-3, Bax, p53 and Bad, while increasing Bcl-2 expression levels. Notably, the application of TA decreased the expression levels of cytochrome c, second mitochondria-derived activator of caspases and high-temperature requirement A2, which are apoptosis mitochondrial-associated proteins. The present findings indicated that TA protected against ATO-induced cardiotoxicity, which may be associated with oxidative stress, inflammation and mitochondrial apoptosis.
Collapse
Affiliation(s)
- Yucong Xue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Mengying Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yurun Xue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Weiyue Jin
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jianping Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Ziliang Li
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
43
|
Liu P, Xue Y, Zheng B, Liang Y, Zhang J, Shi J, Chu X, Han X, Chu L. Crocetin attenuates the oxidative stress, inflammation and apoptosisin arsenic trioxide-induced nephrotoxic rats: Implication of PI3K/AKT pathway. Int Immunopharmacol 2020; 88:106959. [PMID: 32919218 DOI: 10.1016/j.intimp.2020.106959] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/13/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
Abstract
Arsenic trioxide (ATO)-induced renal toxicity through oxidative stress and apoptosis restricts the therapeutic action of acute myelogenous leukemia. Crocetin (Crt) possesses antioxidant and antiapoptosis properties, and has certain renal protective effects, but it has not been reported that it has protective effect on renal injury caused by ATO. The current study explored the effects and mechanisms of Crt on kidney damage induced by ATO. Fifty Sprague-Dawley rats were randomly divided into five groups. Adult rats were given Crt concurrently with ATO for 1 week. On the 8th day, rats were killed and blood and kidney tissues were collected. Histopathological changes were measured, and kidneytissues and serum were used to determine renal function and antioxidant enzyme activity. In addition, the protein expression levels of P-PI3K, PI3K, P-AKT, AKT, CytC, Bax, Bcl-2 and Caspase-3 were determined via western blot analysis. Results revealed ATO induced renal morphological alterations and activated serum BUN and CRE. Compared with the control group, ROS, MDA, IL-1β, TNF-α, protein carbonyls (PC), lipid hydroperoxides (LOOH) and arsenic concentration levels were found to be significantly increased and SOD, CAT, GSH-Px, GSH and total sulphydryl groups (TSH) levels were attenuated in the ATO group. Crt markedly reduced oxidative stress in ATO-induced nephrotoxicity. Further, ATO induced apoptosis by significantly enhancing CytC, Bax and Caspase-3 and inhibiting Bcl-2. Administration with Crt markedly improved the expression of apoptosis factor. Moreover, Crt treatment stimulated the expressions of P-PI3K, PI3K, P-AKT, AKT induced by ATO. This study indicates Crt could prevent renal injury caused by ATO through inhibiting oxidative stress, inflammation and apoptosis, and its mechanism may be related to activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Panpan Liu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Yurun Xue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Bin Zheng
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Yingran Liang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Jianping Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Jing Shi
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Xi Chu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China.
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China.
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China.
| |
Collapse
|
44
|
Hesperidin protects against cadmium-induced pancreatitis by modulating insulin secretion, redox imbalance and iNOS/NF-ĸB signaling in rats. Life Sci 2020; 259:118268. [PMID: 32800830 DOI: 10.1016/j.lfs.2020.118268] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Abstract
AIM Cadmium is a persistent ubiquitous environmental toxicant that elicits several biological defects on delicate body organs. Growing evidence suggests that cadmium (Cd) may perturb signaling pathways to induce oxidative pancreatitis. Thus, we explored whether hesperidin, a flavonone, could mitigate Cd-induced oxidative stress-mediated inflammation and pancreatitis in Wistar rats. MAIN METHODS Forty (40) rats randomly assigned to 5 groups (n = 8) were administered normal saline or hesperidin (Hsp) followed by Cd intoxication for 28 days. KEY FINDINGS Cadmium accumulated in the pancreas of rats, and markedly decreased insulin, pancreatic superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activities and glutathione (GSH) level. Cadmium considerably increased malondialdehyde (MDA), serum lipase and amylase activities. Cadmium induced pancreatic pro-inflammation via over-expression of inducible nitric oxide synthase (iNOS), nuclear factor-ĸB (NF-κB), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), along with histopathological alterations. Hesperidin prominently decreased serum amylase and lipase activities, and markedly increased insulin level, pancreatic antioxidant defense mechanism, whereas iNOS, NF-κB, IL-6 and TNF-α levels significantly decreased. Changes in histology confirmed our biochemical findings. SIGNIFICANCE Our findings suggest that Cd induced pancreatitis via pro-inflammation and oxidative stress; Hsp, thus, protects against Cd-induced pancreatitis via attenuation of oxidative stress and proinflammatory responses in pancreas.
Collapse
|