1
|
Jang H, Han SC, Lee J, Shin HY, Hwang JH, Ha JH. Anti-inflammatory effects of rutin in lipopolysaccharide-stimulated canine macrophage cells. Nutr Res Pract 2025; 19:143-153. [PMID: 39959750 PMCID: PMC11821770 DOI: 10.4162/nrp.2025.19.1.143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND/OBJECTIVES Inflammatory responses are key pathological factors in various canine diseases, making the control of inflammatory responses vital for canine health. This study examined the anti-inflammatory effects of rutin on DH82 cells, a type of canine macrophage, against lipopolysaccharide (LPS)-induced inflammatory responses. MATERIALS/METHODS The inflammatory in vitro experimental model was established by stimulating canine macrophage DH82 cells with LPS. To evaluate the inflammation-preventative effects of rutin, analyses were conducted using enzyme-linked immunosorbent assay, western blot, and real-time quantitative reverse transcription polymerase chain reaction. RESULTS Rutin inhibited the LPS-induced increase in the protein and gene levels of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, tumor necrosis factor-α), while anti-inflammatory cytokines (IL-10, transforming growth factor-β1) levels remained unchanged. Furthermore, rutin suppressed the LPS-induced activation of phosphorylated extracellular signal-regulated kinase, Jun N-terminal kinase, inhibitor of nuclear factor kappa B, and nuclear factor kappa B (NF-κB) in DH82 cells. CONCLUSION Rutin exerts anti-inflammatory effects by inhibiting the mitogen-activated protein kinase-NF-κB signaling pathway and reducing the production of pro-inflammatory cytokines in DH82 cells.
Collapse
Affiliation(s)
- Hyunsoo Jang
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Su-Cheol Han
- Companion Animal New Drug Development Center, Korea Institute of Toxicology, Jeongeup 56212, Korea
| | - Jisu Lee
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Ha-Young Shin
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup 56212, Korea
| | - Jeong Ho Hwang
- Companion Animal New Drug Development Center, Korea Institute of Toxicology, Jeongeup 56212, Korea
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup 56212, Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
- Research Center for Industrialization of Natural Neutralization, Dankook University, Yongin 16890, Korea
| |
Collapse
|
2
|
Thakur P, Mittal N, Chaudhary J, Kamboj S, Jain A. Unveiling the substantial role of rutin in the management of drug-induced nephropathy using network pharmacology and molecular docking. Int Immunopharmacol 2025; 146:113911. [PMID: 39733639 DOI: 10.1016/j.intimp.2024.113911] [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: 06/18/2024] [Revised: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 12/31/2024]
Abstract
INTRODUCTION Flavonoids including quercetin, kaempferol, myricetin, rutin etc. have always been a part of traditional Chinese medicine for the treatment of several ailments. Rutin (RT), also known as rutoside, sophorin is one of the flavanol glycoside having structure resemblance with quercetin. It is found to exhibit several biological activities viz. anti-inflammatory, anticancer, antioxidant, cardioprotective, antidepressant, neuroprotective etc. but the mechanisms by which it exhibits these effects is still under research. AIM The protective effects of rutin against drug induced nephropathy have already been discovered. Therefore, in this study, the main focus is to explore the mechanism by which rutin provides protection against drug-induced nephropathy using modern method like network pharmacology and molecular docking. MATERIALS AND METHODS Genes linked to drug-induced nephropathy and targets connected with rutin were obtained by searching through a number of extensive databases, including David software, Venn plot database, Swiss target prediction database, String database, Gene card & OMIM database, and Pubchem. In order to locate mapping targets, the acquired targets were examined and intersected. A protein-protein interaction (PPI) network was then built to find potential targets. RESULTS From the KEGG pathway, the target pathway responsible for drug-induced nephropathy were found to be XDH, HSD17B2, MET, PRKCB, CD38, ALDH2, CDK1, PTK2, CYP19A1, TNF, F2, PTGS2, ESR1, GSK3B, GLO1, ALOX12, MMP3, PRKCZ, CXCR1, CA4, EGFR, PDE5A, F10, AKR1B1, DRD4, TERT, CA3, PLG, TP53, PRKCH, PIK3R1, PRKACA, CYP1B1, ALOX5, PLK1, CHEK1, KCNH2, PRKCD, MAPT, MPO, NOX4, AVPR2, ACHE, MCL1, KDR, ABCG2, CCR1, PIK3CG, FLT3, ADORA1, IL2, SYK, IGF1R, CA2, SERPINE1, INSR, PRKCA, APP, MMP9. From these identified targets, the 14 selected pathways which have major role in providing protection in drug-induced nephropathy have been discussed. CONCLUSION As RT can inhibit various metabolic and proinflammatory pathways involved, it can help in prevention and treatment of drug-induced nephropathy. FUTURE ASPECTS The revelation of mode of action of bioactive constituent rutin against drug-induced nephropathy provides a theoretical basis for designing more promising compounds in future for treatment of nephropathy.
Collapse
Affiliation(s)
- Prashant Thakur
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University) Mullana, Ambala, Haryana, India
| | - Nitish Mittal
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University) Mullana, Ambala, Haryana, India
| | - Jasmine Chaudhary
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University) Mullana, Ambala, Haryana, India
| | - Sonia Kamboj
- Ch. Devi Lal College of Pharmacy, Jagadhri, Haryana, India
| | - Akash Jain
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University) Mullana, Ambala, Haryana, India.
| |
Collapse
|
3
|
Petkova-Parlapanska K, Stefanov I, Ananiev J, Georgiev T, Hadzhibozheva P, Petrova-Tacheva V, Kaloyanov N, Georgieva E, Nikolova G, Karamalakova Y. Sambucus nigra-Lyophilized Fruit Extract Attenuated Acute Redox-Homeostatic Imbalance via Mutagenic and Oxidative Stress Modulation in Mice Model on Gentamicin-Induced Nephrotoxicity. Pharmaceuticals (Basel) 2025; 18:85. [PMID: 39861148 PMCID: PMC11768164 DOI: 10.3390/ph18010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Gentamicin (GM) administration is associated with decreased metabolism, increased oxidative stress, and induction of nephrotoxicity. Sambucus nigra L., containing flavonoids, anthocyanins, and phytosterols, possesses antioxidant and anti-inflammatory potential. Objectives: The present study aimed to investigate the nephroprotective and anti-inflammatory potential of lyophilized Sambucus nigra fruit extract (S. nigra extract) to reduce acute oxidative stress and residual toxicity of GM in a 7-day experimental model in Balb/c rodents. Methods: The S. nigra extract was lyophilized (300 rpm; 10 min; -45 °C) to improve pharmacological properties. Balb/c mice were divided into four (n = 6) groups: controls; S. nigra extract per os (120 mg kg-1 day-1 bw); GM (200 mg kg-1 day-1 bw) (4); and GM + S. nigra therapy. The activities of antioxidant and renal enzymes, cytokines, and levels of oxidative stress biomarkers-Hydroxiproline, CysC, GST, KIM-1, PGC-1α, MDA, GSPx-were analyzed by ELISA tests. The ROS and RNS levels, as well as 5-MSL-protein oxidation, were measured by EPR spectroscopy. Results: The antioxidant-protective effect of S. nigra extract (120 mg kg-1) was demonstrated by reduced MDA, ROS, and RNS and increased activation of endogenous enzymes. Furthermore, S. nigra extract significantly reduced the expression of IL-1β, IL-6, IL-10, TNF-α, IFN-γ, and KIM-1 and regulated collagen/protein (PGC-1α and albumin) deposition in renal tissues. Conclusions: Histological evaluation confirmed that S. nigra (120 mg kg-1) attenuated renal dysfunction and structural damage by modulating oxidative stress and acute inflammation and could be used as an anti-fibrotic alternative in GM nephrotoxicity.
Collapse
Affiliation(s)
- Kamelia Petkova-Parlapanska
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| | - Ivaylo Stefanov
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Faculty of Medicine, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (I.S.); (J.A.); (E.G.)
| | - Julian Ananiev
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Faculty of Medicine, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (I.S.); (J.A.); (E.G.)
| | - Tsvetelin Georgiev
- Department “Physiology, Pathophysiology and Pharmacology” Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (T.G.); (P.H.)
| | - Petya Hadzhibozheva
- Department “Physiology, Pathophysiology and Pharmacology” Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (T.G.); (P.H.)
| | - Veselina Petrova-Tacheva
- Department of Molecular Biology, Immunology and Medical Genetics, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Nikolay Kaloyanov
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 St. Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| | - Ekaterina Georgieva
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Faculty of Medicine, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria; (I.S.); (J.A.); (E.G.)
| | - Galina Nikolova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| | - Yanka Karamalakova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, 11 Armeiska Str., 6000 Stara Zagora, Bulgaria;
| |
Collapse
|
4
|
Zhao X, Chen X, Yue C. Rutin Ameliorates Inflammation and Oxidative Stress in Ulcerative Colitis by Inhibiting NLRP3 Inflammasome Signaling Pathway. Cell Biochem Biophys 2024; 82:3715-3726. [PMID: 39138797 PMCID: PMC11576901 DOI: 10.1007/s12013-024-01459-7] [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] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory disease. We intend to explore the mechanism of Rutin in the therapy of UC. Disease activity index (DAI) and hematoxylin-eosin staining were employed to assess therapeutic effect of Rutin on dextran sulfate sodium-stimulated mice. The proliferation was detected by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay. Oxidative stress (OS) was assessed by measuring reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD). Inflammatory factors were detected using enzyme-linked immunosorbent assay and immunofluorescence staining. mRNA and protein expressions were detected by real-time quantitative polymerase chain reaction and immunoblotting assay. Rutin decreased DAI scores and ameliorated pathological damage in UC mice with decreased levels of inflammatory factors. Rutin recovered the inhibited proliferation of fetal human colon cells caused by lipopolysaccharide. Rutin inhibited OS by reducing ROS and MDA, while enhancing SOD activity in LPS-induced fetal human colon cells. Rutin inhibited NLRP3 inflammasome in UC mice and cell model. Silencing NLRP3 enhanced the inhibitory effect of Rutin on OS in lipopolysaccharide-induced fetal human colon cells. Conversely, NLRP3 overexpression reversed the restraining role of Rutin in OS. Rutin ameliorates UC by inhibiting inflammation and OS through suppressing NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiangdong Zhao
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, No. 1, Fuhua Road, Nanyuan Street, Futian District, Shenzhen, 518003, Guangdong, China
| | - Xiaochao Chen
- Department of Anorectal, Chengdu Anorectal Hospital, No.152, Daqiang East Street, Qingyang District, Chengdu, 610015, Sichuan, China
| | - Chaochi Yue
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
5
|
Yang XR, Wen R, Yang N, Zhang TN. Role of sirtuins in sepsis and sepsis-induced organ dysfunction: A review. Int J Biol Macromol 2024; 278:134853. [PMID: 39163955 DOI: 10.1016/j.ijbiomac.2024.134853] [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: 04/29/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes a high mortality rate and current treatment focuses on supportive therapies but lacks specific therapeutic targets. Notably, sirtuins (SIRTs) shows potential clinical application in the treatment of sepsis. It has been demonstrated that SIRTs, the nicotinamide adenine dinucleotide+(NAD+)-dependent deacetylases that regulate key signaling pathways in eukaryotes and prokaryotes, are involved in a variety of biological processes. To date, seven mammalian yeast Sir2 homologs have been identified. SIRTs can regulate inflammation, oxidative stress, apoptosis, autophagy, and other pathways that play important roles in sepsis-induced organ dysfunction. However, the existing studies on SIRTs in sepsis are too scattered, and there is no relevant literature to integrate them. This review innovatively summarizes the different mechanisms of SIRTs in sepsis organ dysfunction according to the different systems, and focuses on SIRT agonists, inhibitors, and targeted drugs that have been proved to be effective in the treatment of sepsis, so as to integrate the clinical research and basic research closely. We searched PubMed for all literature related to SIRTs and sepsis since its inception using the following medical subject headings: sirtuins, SIRTs, and sepsis. Data on the mechanisms of SIRTs in sepsis-induced organ damage and their potential as targets for disease treatment were extracted.
Collapse
Affiliation(s)
- Xin-Ru Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
6
|
Silva AP, Cordeiro MLDS, Aquino-Martins VGDQ, de Moura Melo LF, Paiva WDS, Naliato GFDS, Theodoro RC, Meneses CHSG, Rocha HAO, Scortecci KC. Prospecting of the Antioxidant Activity from Extracts Obtained from Chañar ( Geoffroea decorticans) Seeds Evaluated In Vitro and In Vivo Using the Tenebrio molitor Model. Nutrients 2024; 16:2813. [PMID: 39275132 PMCID: PMC11396818 DOI: 10.3390/nu16172813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/16/2024] Open
Abstract
Geoffroea decorticans, commonly known as Chañar, is a native Chilean plant widely used in folk medicine for its expectorant, pain relief, and antinociceptive properties. This study explored the antioxidant, cytotoxic, and protective effects of its ethanolic (EE) and aqueous (EA) seed extracts against oxidative stress induced by copper sulfate, using both in vitro and in vivo approaches. Phytochemical analyses revealed the presence of phenolic compounds and flavonoids in the extracts. High-Performance Liquid Chromatography (HPLC) coupled with Gas Chromatography-Mass Spectrometry/Mass Spectrometry (GC-MS/MS) identified significant components such as phytol, alpha-tocopherol, vitexin, and rutin, with the EE being particularly rich in phytol and vitexin. Antioxidant assays-measuring the total antioxidant capacity (TAC), reducing power, DPPH radical scavenging, and copper and iron chelation-confirmed their potent antioxidant capabilities. Both extracts were non-cytotoxic and provided protection against CuSO4-induced oxidative stress in the 3T3 cell line. Additionally, the use of Tenebrio molitor as an invertebrate model underscored the extracts' antioxidant and protective potentials, especially that of the EE. In conclusion, this study highlights the significant antioxidant and protective properties of Chañar seed extracts, particularly the ethanolic extract, in both in vitro and in vivo models.
Collapse
Affiliation(s)
- Ariana Pereira Silva
- Laboratório de Transformação de Plantas e Análise em Microscopia (LTPAM), Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Maria Lucia da Silva Cordeiro
- Laboratório de Transformação de Plantas e Análise em Microscopia (LTPAM), Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Verônica Giuliani de Queiroz Aquino-Martins
- Laboratório de Transformação de Plantas e Análise em Microscopia (LTPAM), Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Luciana Fentanes de Moura Melo
- Laboratório de Transformação de Plantas e Análise em Microscopia (LTPAM), Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Weslley de Souza Paiva
- Laboratório de Biotecnologia de Polímeros Naturais (BIOPOL), Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Georggia Fatima da Silva Naliato
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Instituto de Medicina Tropical, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59077-080, RN, Brazil
| | - Raquel Cordeiro Theodoro
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Instituto de Medicina Tropical, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59077-080, RN, Brazil
| | - Carlos Henrique Salvino Gadelha Meneses
- Laboratório de Biotecnologia Vegetal (LBV), Departamento de Biologia, Centro de Ciências Biológicas e da Saúde, Universidade Estadual da Paraiba (UEPB), Campina Grande 58429-500, PB, Brazil
| | - Hugo Alexandre Oliveira Rocha
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Laboratório de Biotecnologia de Polímeros Naturais (BIOPOL), Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Katia Castanho Scortecci
- Laboratório de Transformação de Plantas e Análise em Microscopia (LTPAM), Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Programa de Pós-Graduação em Bioquímica e Biologia Molecular, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| |
Collapse
|
7
|
Darvishzadeh Mahani F, Raji-Amirhasani A, Khaksari M, Mousavi MS, Bashiri H, Hajializadeh Z, Alavi SS. Caloric and time restriction diets improve acute kidney injury in experimental menopausal rats: role of silent information regulator 2 homolog 1 and transforming growth factor beta 1. Mol Biol Rep 2024; 51:812. [PMID: 39007943 DOI: 10.1007/s11033-024-09716-x] [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: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Estrogen has a protective impact on acute kidney injury (AKI); moreover, reducing the daily intake of calories impedes developing diseases. The present study aimed to determine the effects of calorie restriction (CR) and time restriction (TR) diets on the expression of silent information regulator 2 homolog 1 (SIRT1), transforming growth factor beta 1 (TGF-β1), and other indicators in the presence and absence of ovaries in AKI female rats. METHODS The female rats were divided into two groups, ovariectomized (OVX) and sham, and were placed on CR and TR diets for eight weeks; afterward, AKI was induced by injecting glycerol, and kidney injury indicators and biochemical parameters were measured before and after AKI. RESULTS After AKI, the levels of urine albumin excretion rate, urea, and creatinine in serum, and TGF-β1 increased, while creatinine clearance and SIRT1 decreased in kidney tissue. CR improved kidney indicators and caused a reduction in TGF-β1 and an increase in SIRT1 in ovary-intact rats. Moreover, CR prevented total antioxidant capacity (TAC) decrease and malondialdehyde (MDA) increase resulting from AKI. Before AKI, an increase in body weight, fasting blood sugar (FBS), low-density lipoprotein (LDL), triglyceride (TG), and total cholesterol (TC), and a decrease in high-density lipoprotein (HDL) were observed in OVX rats compared to sham rats, but CR prevented these changes. The effects of TR were similar to those of CR in all indicators except for TGF-β1, SIRT1, urea, creatinine, and albumin. CONCLUSION The present study indicated that CR is more effective than TR in preventing AKI, probably by increasing SIRT1 and decreasing TGF-β1 in ovary-intact animals.
Collapse
Affiliation(s)
- Fatemeh Darvishzadeh Mahani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Raji-Amirhasani
- Endocrinology and Metabolism Research Center, Kerman University of Medical SciencesKerman, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical SciencesKerman, Kerman, Iran.
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Physiology Research Center, Department of Physiology and Pharmacology, 22 Bahman Blvd, Kerman, Iran.
| | - Maryam Sadat Mousavi
- Clinical Research Development Unit, Shafa Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Samaneh Sadat Alavi
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Chen S, Li Z, Xiao Y, Zhou Z, Zhan Q, Yu L. Rutin targets AKT to inhibit ferroptosis in ventilator-induced lung injury. Phytother Res 2024; 38:3401-3416. [PMID: 38666397 DOI: 10.1002/ptr.8212] [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: 01/12/2024] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 07/12/2024]
Abstract
Our previous research confirmed that rutin reduced ventilator-induced lung injury (VILI) in mice. Ferroptosis has been reported to participate in the pathogenic process of VILI. We will explore whether rutin inhibits ferroptosis to alleviate VILI. A mouse model of VILI was constructed with or without rutin pretreatment to perform a multiomics analysis. Hematoxylin-eosin (HE) staining and transmission electron microscopy were used to evaluate lung injury in VILI mice. Dihydroethidium (DHE) staining and the malondialdehyde (MDA) and superoxide dismutase (SOD) levels were detected. Molecular docking was performed to determine the binding affinity between rutin and ferroptosis-related proteins. Western blot analysis, real-time PCR (RT-PCR) and immunohistochemical (IHC) staining were conducted to detect the expression levels of GPX4, XCT, ACSL4, FTH1, AKT and p-AKT in lung tissues. Microscale thermophoresis (MST) was used to evaluate the binding between rutin and AKT1. Transcriptomic and proteomic analyses showed that ferroptosis may play a key role in VILI mice. Metabolomic analysis demonstrated that rutin may affect ferroptosis via the AKT pathway. Molecular docking analysis indicated that rutin may regulate the expression of ferroptosis-related proteins. Moreover, rutin upregulated GPX4 expression and downregulated the expression of XCT, ACSL4 and FTH1 in the lung tissues. Rutin also increased the ratio of p-AKT/AKT and p-AKT expression. MST analysis showed that rutin binds to AKT1. Rutin binds to AKT to activate the AKT signaling pathway, contributing to inhibit ferroptosis, thus preventing VILI in mice. Our study elucidated a possible novel strategy of involving the use of rutin for preventing VILI.
Collapse
Affiliation(s)
- Shengsong Chen
- Department of Pulmonary and Critical Care Medicine, National Regional Center for Respiratory Medicine, Jiangxi Hospital of China-Japan Friendship Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhonghao Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Yuhong Xiao
- Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhaobin Zhou
- Department of Pulmonary and Critical Care Medicine, National Regional Center for Respiratory Medicine, Jiangxi Hospital of China-Japan Friendship Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, National Regional Center for Respiratory Medicine, Jiangxi Hospital of China-Japan Friendship Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Lingling Yu
- Department of Rehabilitation Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
You J, Li Y, Chong W. The role and therapeutic potential of SIRTs in sepsis. Front Immunol 2024; 15:1394925. [PMID: 38690282 PMCID: PMC11058839 DOI: 10.3389/fimmu.2024.1394925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by the host's dysfunctional response to infection. Abnormal activation of the immune system and disturbance of energy metabolism play a key role in the development of sepsis. In recent years, the Sirtuins (SIRTs) family has been found to play an important role in the pathogenesis of sepsis. SIRTs, as a class of histone deacetylases (HDACs), are widely involved in cellular inflammation regulation, energy metabolism and oxidative stress. The effects of SIRTs on immune cells are mainly reflected in the regulation of inflammatory pathways. This regulation helps balance the inflammatory response and may lessen cell damage and organ dysfunction in sepsis. In terms of energy metabolism, SIRTs can play a role in immunophenotypic transformation by regulating cell metabolism, improve mitochondrial function, increase energy production, and maintain cell energy balance. SIRTs also regulate the production of reactive oxygen species (ROS), protecting cells from oxidative stress damage by activating antioxidant defense pathways and maintaining a balance between oxidants and reducing agents. Current studies have shown that several potential drugs, such as Resveratrol and melatonin, can enhance the activity of SIRT. It can help to reduce inflammatory response, improve energy metabolism and reduce oxidative stress, showing potential clinical application prospects for the treatment of sepsis. This review focuses on the regulation of SIRT on inflammatory response, energy metabolism and oxidative stress of immune cells, as well as its important influence on multiple organ dysfunction in sepsis, and discusses and summarizes the effects of related drugs and compounds on reducing multiple organ damage in sepsis through the pathway involving SIRTs. SIRTs may become a new target for the treatment of sepsis and its resulting organ dysfunction, providing new ideas and possibilities for the treatment of this life-threatening disease.
Collapse
Affiliation(s)
- Jiaqi You
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| | - Yilin Li
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Chong
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Ghosian-Moghaddam MH, Mohseni-Moghaddam P, Roghani M. Therapeutic Potential of Diosgenin in Amelioration of Carbon Tetrachloride-Induced Murine Liver Injury. Drug Res (Stuttg) 2024; 74:156-163. [PMID: 38458224 DOI: 10.1055/a-2263-1329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Diosgenin is a sapogenin with antidiabetic, antioxidant, and anti-inflammatory properties. The current study investigated whether diosgenin could ameliorate carbon tetrachloride (CCL4)-induced liver injury. To cause liver injury, CCL4 was injected intraperitoneally twice a week for 8 weeks. Daily oral administration of diosgenin at doses of 20, 40, and 80 mg/kg was started one day before CCL4 injection and continued for 8 weeks. Finally, serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and also albumin were assessed. Catalase and superoxide dismutase (SOD) activities in addition to glutathione (GSH) and malondialdehyde (MDA) levels were also quantified in the liver homogenate and routine histological evaluation was also conducted. Elevated serum levels of liver enzymes and decreased serum level of albumin caused by CCL4 were significantly restored following diosgenin administration at doses of 40 and 80 mg/kg. Long-term administration of CCL4 increased inflammatory and apoptotic factors such as IL-1β, caspase 3, TNF-α, and IL-6 and decreased SOD and catalase activities as well as GSH level in liver homogenates; while MDA level was increased. Treatment with diosgenin increased SOD and catalase activities and GSH levels in the liver of injured animals. In addition, liver MDA, IL-1β, caspase 3, TNF-α, and IL-6 level or activity decreased by diosgenin treatment. Additionally, diosgenin aptly prevented aberrant liver histological changes. According to obtained results, diosgenin can dose-dependently diminish CCl4-induced liver functional deficits and histological changes in a dose-dependent manner, possibly due to its antioxidant and anti-inflammation properties, and its beneficial effect is comparable to known hepatoprotective agent silymarin.
Collapse
Affiliation(s)
| | - Parvaneh Mohseni-Moghaddam
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
11
|
Lombardo GE, Russo C, Maugeri A, Navarra M. Sirtuins as Players in the Signal Transduction of Citrus Flavonoids. Int J Mol Sci 2024; 25:1956. [PMID: 38396635 PMCID: PMC10889095 DOI: 10.3390/ijms25041956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Sirtuins (SIRTs) belong to the family of nicotine adenine dinucleotide (NAD+)-dependent class III histone deacetylases, which come into play in the regulation of epigenetic processes through the deacetylation of histones and other substrates. The human genome encodes for seven homologs (SIRT1-7), which are localized into the nucleus, cytoplasm, and mitochondria, with different enzymatic activities and regulatory mechanisms. Indeed, SIRTs are involved in different physio-pathological processes responsible for the onset of several human illnesses, such as cardiovascular and neurodegenerative diseases, obesity and diabetes, age-related disorders, and cancer. Nowadays, it is well-known that Citrus fruits, typical of the Mediterranean diet, are an important source of bioactive compounds, such as polyphenols. Among these, flavonoids are recognized as potential agents endowed with a wide range of beneficial properties, including antioxidant, anti-inflammatory, hypolipidemic, and antitumoral ones. On these bases, we offer a comprehensive overview on biological effects exerted by Citrus flavonoids via targeting SIRTs, which acted as modulator of several signaling pathways. According to the reported studies, Citrus flavonoids appear to be promising SIRT modulators in many different pathologies, a role which might be potentially evaluated in future therapies, along with encouraging the study of those SIRT members which still lack proper evidence on their support.
Collapse
Affiliation(s)
- Giovanni Enrico Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| | - Caterina Russo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| | - Alessandro Maugeri
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| |
Collapse
|
12
|
Ma X, Ren X, Zhang X, Wang G, Liu H, Wang L. Rutin ameliorate PFOA induced renal damage by reducing oxidative stress and improving lipid metabolism. J Nutr Biochem 2024; 123:109501. [PMID: 37890710 DOI: 10.1016/j.jnutbio.2023.109501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/09/2023] [Accepted: 10/22/2023] [Indexed: 10/29/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a persistent environmental pollutant that can accumulate in the kidneys and eventually cause kidney damage. Rutin (RUTIN) is a natural flavonoid with multiple biological activities, and its use in against kidney damage has been widely studied in recent years. It is not yet known whether rutin protects against kidney damage caused by PFOA. In this study, 30 ICR mice were randomly divided into three groups: CTRL group, PFOA group and PFOA+RUTIN group. The mice were fed continuously by gavage for 28 days. Renal pathological changes were assessed by HE and PASM staining, and serum renal function and lipid indicators were measured. RNA-seq and enrichment analysis using GO, KEGG and PPI to detect differential expression of genes in treatment groups. Kidney tissue protein expression was determined by Western blot. Research has shown that rutin can improve glomerular and tubular structural damage, and increase serum CREA, HDL-C levels and decrease LDH, LDL-C levels. The expression of AQP1 and ACOT1 was up-regulated after rutin treatment. Transcriptomic analysis indicated that PFOA and rutin affect the transcriptional expression of genes related to lipid metabolism and oxidative stress, and may affected by PI3K-Akt, PPAR, NRF2/KEAP1 signaling pathways. In conclusion, rutin ameliorated renal damage caused by PFOA exposure, and this protective effect may be exerted by ameliorating oxidative stress and regulating lipid metabolism.
Collapse
Affiliation(s)
- Xinzhuang Ma
- School of Public Health, Bengbu Medical College, Bengbu, PR China
| | - Xijuan Ren
- School of Public Health, Bengbu Medical College, Bengbu, PR China
| | - Xuemin Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, PR China
| | - Guangyin Wang
- School of Public Health, Bengbu Medical College, Bengbu, PR China
| | - Hui Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, PR China.
| | - Li Wang
- School of Public Health, Bengbu Medical College, Bengbu, PR China.
| |
Collapse
|
13
|
Laranjeira IM, Dias ACP, Pinto-Ribeiro FL. Genista tridentata Phytochemical Characterization and Biological Activities: A Systematic Review. BIOLOGY 2023; 12:1387. [PMID: 37997986 PMCID: PMC10669091 DOI: 10.3390/biology12111387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/25/2023]
Abstract
Genista tridentata (L.) Willk., known as "prickled broom", is a Leguminosae (Fabaceae) species native to the Iberian Peninsula, Morocco, Algeria, and Tunisia. It is used in folk medicine as an anti-inflammatory, for gastrointestinal and respiratory disorders, rheumatism, and headaches, to lower blood pressure, against hypercholesterolemia and hyperglycemia. This study aimed to systematically review the literature on the bioactivities and phytochemical profile of Genista tridentata to understand its pharmacological potential. For this, four electronic databases (PubMed, GoogleScholar, Repositórios Cientificos de Acesso Aberto de Portugal (RCCAP), and ScienceDirect) were searched from inception up to 31 December 2022. From a total of 264 potentially eligible studies considered for screening, 34 papers were considered eligible for this systematic review. The sampling included 71 extracts, collected mainly in Portugal. Genista tridentata extracts present a high level of flavonoids and phenolic compounds. The flowers and aerial parts of the plant were the most studied, and aqueous extracts were the most used. The results predict a high potential for the application of Genista tridentata as a new source of natural antioxidants and preservatives for the food industry with subsequent health benefits, such as the production of nutraceuticals. Moreover, the results indicate that the plant can be collected at all seasons of the year, which represents a benefit for the industry.
Collapse
Affiliation(s)
- Inês Martins Laranjeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus of Gualtar, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Centre of Molecular and Environmental Biology (CBMA), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Alberto Carlos Pires Dias
- Centre of Molecular and Environmental Biology (CBMA), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Filipa Lacerda Pinto-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus of Gualtar, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
| |
Collapse
|
14
|
Yi X, Xu C, Yang J, Zhong C, Yang H, Tang L, Song S, Yu J. Tiliroside Protects against Lipopolysaccharide-Induced Acute Kidney Injury via Intrarenal Renin-Angiotensin System in Mice. Int J Mol Sci 2023; 24:15556. [PMID: 37958538 PMCID: PMC10648967 DOI: 10.3390/ijms242115556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 11/15/2023] Open
Abstract
Tiliroside, a natural flavonoid, has various biological activities and improves several inflammatory diseases in rodents. However, the effect of Tiliroside on lipopolysaccharide (LPS)-induced acute kidney injury (AKI) and the underlying mechanisms are still unclear. This study aimed to evaluate the potential renoprotective effect of Tiliroside on LPS-induced AKI in mice. Male C57BL/6 mice were intraperitoneally injected with LPS (a single dose, 3 mg/kg) with or without Tiliroside (50 or 200 mg/kg/day for 8 days). Tiliroside administration protected against LPS-induced AKI, as reflected by ameliorated renal dysfunction and histological alterations. LPS-stimulated renal expression of inflammatory cytokines, fibrosis markers, and kidney injury markers in mice was significantly abolished by Tiliroside. This flavonoid also stimulated autophagy flux but inhibited oxidative stress and tubular cell apoptosis in kidneys from LPS-injected mice. Mechanistically, our study showed the regulation of Tiliroside on the intrarenal renin-angiotensin system in LPS-induced AKI mice. Tiliroside treatment suppressed intrarenal AGT, Renin, ACE, and Ang II, but upregulated intrarenal ACE2 and Ang1-7, without affecting plasma Ang II and Ang1-7 levels. Collectively, our data highlight the renoprotective action of Tiliroside on LPS-induced AKI by suppressing inflammation, oxidative stress, and tubular cell apoptosis and activating autophagy flux via the shift towards the intrarenal ACE2/Ang1-7 axis and away from the intrarenal ACE/Ang II axis.
Collapse
Affiliation(s)
- Xiaoli Yi
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Jing Yang
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Chao Zhong
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Huiru Yang
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Le Tang
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Shanshan Song
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330002, China; (X.Y.); (J.Y.); (C.Z.); (H.Y.); (L.T.); (S.S.)
| | - Jun Yu
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
15
|
Chen S, Bai Y, Xia J, Zhang Y, Zhan Q. Rutin alleviates ventilator-induced lung injury by inhibiting NLRP3 inflammasome activation. iScience 2023; 26:107866. [PMID: 37817937 PMCID: PMC10561045 DOI: 10.1016/j.isci.2023.107866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/24/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Whether rutin relieves ventilator-induced lung injury (VILI) remains unclear. Here, we used network pharmacology, bioinformatics, and molecular docking to predict the therapeutic targets and potential mechanisms of rutin in the treatment of VILI. Subsequently, a mouse model of VILI was established to confirm the effects of rutin on VILI. HE staining showed that rutin alleviated VILI. TUNEL staining showed that rutin reduced apoptosis in the lung tissue of mice with VILI, and the same change was observed in the ratio of Bax/Bcl2. Furthermore, rutin reduced the expression of NLRP3, ASC, Caspase1, IL1β, and IL18 in the lung tissues of mice with VILI. Mechanistically, rutin suppressed the TLR4/NF-κB-P65 pathway, which promoted the M1 to M2 macrophage transition and alleviated inflammation in mice with VILI. Rutin relieved NLRP3 inflammasome activation by regulating M1/M2 macrophage polarization and inhibiting the activation of the TLR4/NF-κB-P65 pathway, resulting in the amelioration of VILI in mice.
Collapse
Affiliation(s)
- Shengsong Chen
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Peking Union Medical College, Chinese Academy of Medical Sciences, No 9 Dongdan Santiao, Dongcheng District, Beijing 100730, P.R.China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- WHO Collaborating Center for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
| | - Yu Bai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Peking Union Medical College, Chinese Academy of Medical Sciences, No 9 Dongdan Santiao, Dongcheng District, Beijing 100730, P.R.China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- WHO Collaborating Center for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- WHO Collaborating Center for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- WHO Collaborating Center for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Peking Union Medical College, Chinese Academy of Medical Sciences, No 9 Dongdan Santiao, Dongcheng District, Beijing 100730, P.R.China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
- WHO Collaborating Center for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, Beijing 100029, P.R.China
| |
Collapse
|
16
|
Malla BA, Ali A, Maqbool I, Dar NA, Ahmad SB, Alsaffar RM, Rehman MU. Insights into molecular docking and dynamics to reveal therapeutic potential of natural compounds against P53 protein. J Biomol Struct Dyn 2023; 41:8762-8781. [PMID: 36281711 DOI: 10.1080/07391102.2022.2137241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/11/2022] [Indexed: 10/31/2022]
Abstract
P53 is eminent tumour suppressor protein that plays a prominent role in cell cycle arrest, DNA repair, senescence, differentiation and initiation of apoptosis. P53 is an attractive drug target and the high toxicity of some cancer chemotherapy drugs increase the demand for new anti-cancer drugs from natural products. In this current scenario, identification of promising anticancer compounds from natural sources by repurposing approach is still relevant for the early prevention and effective management of cancer. In present study, we docked natural compounds like podophyllotoxin, quercetin and rutin along standard drugs (MG-132 and Bay 61-3606) against p53 protein. ADME/T analysis predicted toxicity of phytochemicals and drugs. In silico docking analysis of podophyllotoxin, quercetin and rutin gave HDOCK docking scores of -187.87, -148. 97 and -143.85, whereas control drugs MG-132 and Bay 61-3606 showed docking scores of -159.59 and -140.71 against p53 respectively. AutoDock analysis of rutin and MG-132 showed highest binding affinity scores of -7.3 and -6.8 kcal/mol against p53. Molecular dynamic simulation for p53 protein displayed stable conformation and convergence. In this study, P53-rutin complex showed free binding energy score of 11.84 kcal/mol and P53-MG-132 complex reported free energy score of 16.3 kcal/mol. Protein contacts atlas gives non-covalent contacts framework by exploring interfaces of individual subunits and protein-ligand interactions. STRING tool predicts physical and functional interactions between proteins. The results of this study revealed that rutin and MG-132 could be promising inhibitors against targeted p53 protein and this could prove detrimental for molecular therapeutics and drug-designing strategies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bashir Ahmad Malla
- Department of Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, J&K, India
| | - Aarif Ali
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, J&K, India
| | - Irfan Maqbool
- Department of Clinical Biochemistry, SKIMS Soura, Srinagar, J&K, India
| | - Nazir Ahmad Dar
- Department of Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, J&K, India
| | - Sheikh Bilal Ahmad
- Division of Veterinary Biochemistry, SKUAST-K, Shuhama Alusteng, J&K, India
| | - Rana M Alsaffar
- Department Of Pharmacology & Toxicology, College Of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Zhang W, Chen H, Xu Z, Zhang X, Tan X, He N, Shen J, Dong J. Liensinine pretreatment reduces inflammation, oxidative stress, apoptosis, and autophagy to alleviate sepsis acute kidney injury. Int Immunopharmacol 2023; 122:110563. [PMID: 37392573 DOI: 10.1016/j.intimp.2023.110563] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Liensinine is mainly derived from alkaloids extracted and isolated from lotus seeds (Nelumbo nucifera Gaertn). It possesses anti-inflammatory, and antioxidant, according to contemporary pharmacological investigations. However, the effects and therapeutic mechanisms of liensinine on acute kidney injury (AKI) models of sepsis are unclear. To gain insight into these mechanisms, we established a sepsis kidney injury model by LPS injection of mice treated with liensinine, and stimulation of HK-2 with LPS in vitro and treated with liensinine and inhibitors of p38 MAPK, JNK MAPK. We first found that liensinine significantly reduced kidney injury in sepsis mice, while suppressing excessive inflammatory responses, restoring renal oxidative stress-related biomarkers, reducing increased apoptosis in TUNEL-positive cells and excessive autophagy, and that this process was accompanied by an increase in JNK/ p38-ATF 2 axis. In vitro experiments further demonstrated that lensinine reduced the expression of KIM-1, NGAL, inhibited pro- and anti-inflammatory secretion disorders, regulated the activation of the JNK/p38-ATF 2 axis, and reduced the accumulation of ROS, as well as the reduction of apoptotic cells detected by flow cytometry, and that this process played the same role as that of p38 MAPK, JNK MAPK inhibitors. We speculate that liensinine and p38 MAPK, JNK MAPK inhibitors may act on the same targets and could be involved in the mechanism of alleviating sepsis kidney injury in part through modulation of the JNK/p38-ATF 2 axis. Our study demonstrates that lensinine is a potential drug and thus provides a potential avenue for the treatment of AKI.
Collapse
Affiliation(s)
- Wei Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Huizhen Chen
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang 222000, China
| | - Zhaoyun Xu
- Blood Transfusion Department, Ganyu District People's Hospital of Lianyungang City, Lianyungang 222100, China
| | - Xiao Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xuelian Tan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Nana He
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jinyang Shen
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
18
|
Ramalho CEL, Reis DDS, Caixeta GAB, Oliveira MCD, Silva DMFD, Cruvinel WDM, Teófilo MNG, Gomes CM, Sousa PAD, Soares LF, Melo AMD, Rocha JD, Bailão EFLC, Amaral VCS, Paula JAMD. Genotoxicity and maternal-fetal safety of the dried extract of leaves of Azadirachta indica A. Juss (Meliaceae) in Wistar rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116403. [PMID: 36963474 DOI: 10.1016/j.jep.2023.116403] [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: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Azadirachta indica A. Juss (Meliaceae), popularly known as "neem", is used for the treatment of rheumatism, cancer, ulcers, diabetes, respiratory problems, among others. This species is present on six continents and contains more than 400 bioactive compounds. Practically all parts of the plant are used in the treatment of diseases. Although it is widely used, no study has evaluated the safety of this species throughout the gestational period in Wistar rats. AIM OF THE STUDY To evaluate the genotoxicity and the effect of treatment with dried extract of leaves of Azadirachta indica on maternal toxicity and fetal development. MATERIALS AND METHODS The dried extract of leaves of A. indica was obtained by spray drying after percolation of the plant material in 30% ethanol (w/w). The total flavonoids and rutin contents of the extract were determined by spectrophotometric method and HPLC-DAD, respectively. Pregnant Wistar rats (n = 40) were divided into four groups (n = 10/group): one control and three groups treated with dried extract of leaves of A. indica at doses of 300, 600 or 1200 mg/kg. Treatments were carried out from gestational day (GD) 0-20. During gestation, clinical signs of toxicity, weight gain, feed and water consumption of the dams were evaluated. On GD 21, rats were euthanized and cardiac blood was collected. Liver, kidneys, lung, heart, uterus, ovaries and bone marrow were collected. Reproductive performance parameters, histopathological analysis, biochemistry and genotoxicity were evaluated. Fetuses were evaluated for external morphology, skeletal and visceral changes. RESULTS The total flavonoid content of the extract ranged from 2.64 to 3.01%, and the rutin content was 1.07%. There was no change in body mass gain, food and water consumption between the evaluated groups. There was also no difference between the groups in terms of biochemical parameters, reproductive performance, histopathological analysis of the mother's organs and genotoxicity. Supernumerary ossification sites of the sternum were observed, and other skeletal and visceral alterations were not significant. CONCLUSIONS The treatment did not induce maternal toxicity, it was neither embryotoxic nor fetotoxic. The extract was not potentially genotoxic, and at a dose of 1200 mg/kg, it caused changes in the ossification of the sternum.
Collapse
Affiliation(s)
- Carlos Eduardo Lacerda Ramalho
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde (CAPS). Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil; Laboratório de Pesquisa, Desenvolvimento & Inovação de Produtos da Biodiversidade. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Diego Dos Santos Reis
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais e Sintéticos. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Graziele Alícia Batista Caixeta
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde (CAPS). Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil; Laboratório de Farmacologia e Toxicologia de Produtos Naturais e Sintéticos. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Micaelle Cristina de Oliveira
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais e Sintéticos. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Danielle Milany Fernandes da Silva
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais e Sintéticos. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Wilson de Melo Cruvinel
- Escola de Ciências Médicas e da Vida, Pontifícia Universidade Católica de Goiás, Goiânia, Goiás, Brazil
| | | | - Clayson Moura Gomes
- Escola de Ciências Médicas e da Vida, Pontifícia Universidade Católica de Goiás, Goiânia, Goiás, Brazil
| | | | - Leiza Fagundes Soares
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde (CAPS). Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil; Laboratório de Pesquisa, Desenvolvimento & Inovação de Produtos da Biodiversidade. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Anielly Monteiro de Melo
- Laboratório de Pesquisa, Desenvolvimento & Inovação de Produtos da Biodiversidade. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Jamira Dias Rocha
- Laboratório de Biotecnologia. Universidade Estadual de Goiás, Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Elisa Flávia Luiz Cardoso Bailão
- Laboratório de Biotecnologia. Universidade Estadual de Goiás, Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Vanessa Cristiane Santana Amaral
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde (CAPS). Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil; Laboratório de Farmacologia e Toxicologia de Produtos Naturais e Sintéticos. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil
| | - Joelma Abadia Marciano de Paula
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde (CAPS). Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil; Laboratório de Pesquisa, Desenvolvimento & Inovação de Produtos da Biodiversidade. Universidade Estadual de Goiás. Campus Anápolis de Ciências Exatas e Tecnológicas, Anápolis, Goiás, Brazil.
| |
Collapse
|
19
|
Dong R, Zhang X, Liu Y, Zhao T, Sun Z, Liu P, Xiang Q, Xiong J, Du X, Yang X, Gui D, Xu Y. Rutin alleviates EndMT by restoring autophagy through inhibiting HDAC1 via PI3K/AKT/mTOR pathway in diabetic kidney disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154700. [PMID: 36774842 DOI: 10.1016/j.phymed.2023.154700] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/09/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is a primary microvascular complication of diabetes. However, a complete cure for DKD has not yet been found. Although there is evidence that Rutin can delay the onset of DKD, the underlying mechanism remains unclear. PURPOSE To investigate the renoprotective effect of Rutin in the process of DKD and to explore its potential molecular mechanisms. METHODS Db/db mice and high glucose (HG)-induced human renal glomerular endothelial cells (GEnCs) were used as in vivo and in vitro models, respectively. Western blot (WB), Immunohistochemistry (IHC)and Immunofluorescence (IF) staining were used to identify the expression level of proteins associated with endothelial-to-mesenchymal transition (EndMT) and autophagy. Tandem Mass Tag (TMT)-based proteomics analysis was utilized to reveal the mechanism of Rutin in DKD. Transfection with small interfering RNA (siRNA) to reveal the role of histone deacetylase 1 (HDAC1) in HG-induced GEnCs. RESULTS Following 8 weeks of Rutin administration, db/db mice's kidney function and structure significantly improved. In HG-induced GEnCs, activation of autophagy attenuates cellular EndMT. Rutin could alleviate EndMT and restore autophagy in vivo and in vitro models. Proteomics analysis results showed that HDAC1 significantly downregulated in the 200 mg/kg/d Rutin group compared with the db/db group. Transfection with si-HDAC1 in GEnCs partially blocked HG-induced EndMT and restored autophagy. Furthermore, Rutin inhibits the phosphorylation of the PI3K / AKT/ mTOR pathway. HDAC1 overexpression was suppressed in HG-induced GEnCs after using Rapamycin, a specific mTOR inhibitor, verifying the correlation between mTOR and HDAC1. CONCLUSION Rutin alleviates EndMT by restoring autophagy through inhibiting HDAC1 via the PI3K/AKT/mTOR pathway in DKD.
Collapse
Affiliation(s)
- Ruixue Dong
- Faculty of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Xi Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Yadi Liu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Tingting Zhao
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Zhongyan Sun
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Peiyu Liu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Qian Xiang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Jianfeng Xiong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Xinwen Du
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Dingkun Gui
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Youhua Xu
- Faculty of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China; Department of Endocrinology, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China; Zhuhai MUST Science and Technology Research Institute, Macau University of Science and Technology, Hengqin, Zhuhai, China.
| |
Collapse
|
20
|
Li K, Gao L, Zhou S, Ma YR, Xiao X, Jiang Q, Kang ZH, Liu ML, Liu TX. Erythropoietin promotes energy metabolism to improve LPS-induced injury in HK-2 cells via SIRT1/PGC1-α pathway. Mol Cell Biochem 2023; 478:651-663. [PMID: 36001204 DOI: 10.1007/s11010-022-04540-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 08/08/2022] [Indexed: 11/25/2022]
Abstract
Acute kidney injury (AKI) is one of frequent complications of sepsis with high mortality. Mitochondria is the center of energy metabolism participating in the pathogenesis of sepsis-associated AKI, and SIRT1/PGC1-α signaling pathway plays a crucial role in the modulation of energy metabolism. Erythropoietin (EPO) exerts protective functions on chronic kidney disease. We aimed to assess the effects of EPO on cell damage and energy metabolism in a cell model of septic AKI. Renal tubular epithelial cells HK-2 were treated with LPS and human recombinant erythropoietin (rhEPO). Cell viability was detected by CCK-8 and mitochondrial membrane potential was determined using JC-1 fluorescent probe. Then the content of ATP, ADP and NADPH, as well as lactic acid, were measured for the assessment of energy metabolism. Oxidative stress was evaluated by detecting the levels of ROS, MDA, SOD and GSH. Pro-inflammatory cytokines, including TNF-α, IL-6, and IL-1β, were measured with ELISA. Moreover, qRT-PCR and western blot were performed to detect mRNA and protein expressions. shSIRT1 was used to knockdown SIRT1, while EX527 and SR-18292 were applied to inhibit SIRT1 and PGC1-α, respectively, to investigate the regulatory mechanism of rhEPO on inflammatory injury and energy metabolism. In LPS-exposed HK-2 cells, rhEPO attenuated cell damage, inflammation and abnormal energy metabolism, as indicated by the elevated cell viability, the inhibited oxidative stress, cell apoptosis and inflammation, as well as the increased mitochondrial membrane potential and energy metabolism. However, these protective effects induced by rhEPO were reversed after SIRT1 or PGC1-α inhibition. EPO activated SIRT1/PGC1-α pathway to alleviate LPS-induced abnormal energy metabolism and cell damage in HK-2 cells. Our study suggested that rhEPO played a renoprotective role through SIRT1/PGC1-α pathway, which supported its therapeutic potential in septic AKI.
Collapse
Affiliation(s)
- Kan Li
- Department of Nephrology, The First Hospital of Lanzhou University, No.1 Donggangxi Road, Chengguan District, Lanzhou, 730000, Gansu Province, China
| | - Li Gao
- Department of Gynaecology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Sen Zhou
- Department of Nephrology, The First Hospital of Lanzhou University, No.1 Donggangxi Road, Chengguan District, Lanzhou, 730000, Gansu Province, China
| | - Yan-Rong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Xiao Xiao
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Qian Jiang
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Zhi-Hong Kang
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Ming-Long Liu
- Department of Nephrology, The First Hospital of Lanzhou University, No.1 Donggangxi Road, Chengguan District, Lanzhou, 730000, Gansu Province, China
| | - Tian-Xi Liu
- Department of Nephrology, The First Hospital of Lanzhou University, No.1 Donggangxi Road, Chengguan District, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
21
|
Rahmani S, Naraki K, Roohbakhsh A, Hayes AW, Karimi G. The protective effects of rutin on the liver, kidneys, and heart by counteracting organ toxicity caused by synthetic and natural compounds. Food Sci Nutr 2023; 11:39-56. [PMID: 36655104 PMCID: PMC9834893 DOI: 10.1002/fsn3.3041] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/21/2022] [Accepted: 08/25/2022] [Indexed: 01/21/2023] Open
Abstract
Rutin is a flavonoid present in many plant species. Because of its antioxidant, anti-inflammatory, and antiapoptotic properties, rutin is of interest for its potential protective effects against toxic agents. The hepatoprotective, renoprotective, and cardioprotective effects of rutin are reviewed. The antioxidant effects of rutin are elicited by enhancing antioxidant enzymes such as GST, GGT, CAT, GPx, SOD, and GR, activating the Nrf2/HO-1 pathway, elevating GSH content, and the reduction in MDA. The anti-inflammatory effects of rutin are mediated by the inhibition of IL-1β, IL-6, TGF-β1, COX-2, iNOS, TLR4, and XO. Rutin exerted its antiapoptotic effects by inhibition of free radicals, caspase-3/-7/-9, hsp70, HMGB1, and p53, and the elevation of the antiapoptotic protein Bcl-2. Rutin has potential therapeutic effectiveness against several toxicants, and its beneficial effects are more than likely mediated by its antioxidant, anti-inflammatory, and/or antiapoptotic property.
Collapse
Affiliation(s)
- Sohrab Rahmani
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Karim Naraki
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Institute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
| | - A. Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public HealthUniversity of South FloridaTampaFloridaUSA
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMichiganUSA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
- Pharmaceutical Research Center, Institute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
22
|
Shahmohammadi A, Golchoobian R, Mirahmadi SMS, Rousta AM, Ansari F, Sharayeli M, Baluchnejadmojarad T, Roghani M. Scutellarin alleviates lipopolysaccharide-provoked septic nephrotoxicity via attenuation of inflammatory and oxidative events and mitochondrial dysfunction. Immunopharmacol Immunotoxicol 2022; 45:295-303. [DOI: 10.1080/08923973.2022.2141644] [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]
Affiliation(s)
| | - Ravieh Golchoobian
- Department of Physiology and Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | | | - Fariba Ansari
- Department of Physiology, School of Medicine, Shahed University, Tehran, Iran
| | - Maryam Sharayeli
- Department of Pathology, School of Medicine, Shahed University, Tehran, Iran
| | | | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
23
|
Hebert JF, Burfeind KG, Malinoski D, Hutchens MP. Molecular Mechanisms of Rhabdomyolysis-Induced Kidney Injury: From Bench to Bedside. Kidney Int Rep 2022; 8:17-29. [PMID: 36644345 PMCID: PMC9831947 DOI: 10.1016/j.ekir.2022.09.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 01/18/2023] Open
Abstract
Rhabdomyolysis-induced acute kidney injury (RIAKI) occurs following damage to the muscular sarcolemma sheath, resulting in the leakage of myoglobin and other metabolites that cause kidney damage. Currently, the sole recommended clinical treatment for RIAKI is aggressive fluid resuscitation, but other potential therapies, including pretreatments for those at risk for developing RIAKI, are under investigation. This review outlines the mechanisms and clinical significance of RIAKI, investigational treatments and their specific targets, and the status of ongoing research trials.
Collapse
Affiliation(s)
- Jessica F. Hebert
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA,Correspondence: Jessica F. Hebert, Oregon Health and Science University, Department of Anesthesiology and Perioperative Medicine, Portland, Oregon, USA.
| | - Kevin G. Burfeind
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Darren Malinoski
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA,Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| | - Michael P. Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA,Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| |
Collapse
|
24
|
Zaghloul RA, Abdelghany AM, Samra YA. Rutin and selenium nanoparticles protected against STZ-induced diabetic nephropathy in rats through downregulating Jak-2/Stat3 pathway and upregulating Nrf-2/HO-1 pathway. Eur J Pharmacol 2022; 933:175289. [PMID: 36122758 DOI: 10.1016/j.ejphar.2022.175289] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 12/01/2022]
Abstract
Diabetic nephropathy (DN) is a renal complication of diabetic hyperglycemia. The Signal transducer and activator of transcription 3 (Stat3) is a center molecule of the chronic inflammation causing DN progression. Therefore, the study investigated the possible inhibitory effects of Rutin (Ru) and Selenium (Se), formulated as nanoparticles (SeNPs), on Stat3 pathway in streptozotocin (STZ)-induced DN in Sprague-Dawley rats. Ru (100 mg/kg/orally) and SeNPs (equivalent to 5 mg of Se/kg/orally) were given as treatment for eight weeks. An assessment of fasting blood glucose, renal function biomarkers, GSH, and MDA was carried out spectrophotometrically. ELISA assessment of renal IL-6, NF-κB, TNF-α, Jak-2, and p-Stat3 was performed. Sirt-1, Nrf-2, and HO-1 were assessed immunohistochemically. DN group receiving Ru + SeNPs showed a decrease in fasting blood glucose, serum creatinine, and urea (163.8 ± 22.8, 0.54 ± 0.1, and 53.6 ± 25.7 mg/dl, respectively), compared to the DN group (443.8 ± 42.72, 1.58 ± 0.4, and 281.8 ± 47.35 mg/dl, respectively). In addition, it exhibited elevation in the levels of Sirt-1, Nrf-2 and HO-1 compared to the DN group. Finally, Ru + SeNPs exhibited a significant reduction in IL-6, NF-κB, TNF-α, Jak-2, and p-Stat3 (42.8 ± 10.3, 1.2 ± 0.1, 53.4 ± 3.87, 0.8 ± 0.06 and 1.1 ± 0.2 U/g tissue, respectively) when compared to the DN group (155.3 ± 13.97, 2.8 ± 0.3, 105.5 ± 32.84, 2.03 ± 0.2 and 2.56 ± 0.15 U/g tissue, respectively). Therefore, combining Ru with SeNPs has a potential renoprotective effect against DN by upregulating Nrf-2/HO-1 and downregulating Jak-2/Stat3 Pathways.
Collapse
Affiliation(s)
- Randa A Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Amr M Abdelghany
- Spectroscopy Department, Physics Research Institute, National Research Centre, 33 Elbehouth St., Dokki, 12311, Egypt; Basic Science Department, Horus University, New Damietta, Damietta, Egypt
| | - Yara A Samra
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Biochemistry Dept., Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt
| |
Collapse
|
25
|
He FF, Wang YM, Chen YY, Huang W, Li ZQ, Zhang C. Sepsis-induced AKI: From pathogenesis to therapeutic approaches. Front Pharmacol 2022; 13:981578. [PMID: 36188562 PMCID: PMC9522319 DOI: 10.3389/fphar.2022.981578] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a heterogenous and highly complex clinical syndrome, which is caused by infectious or noninfectious factors. Acute kidney injury (AKI) is one of the most common and severe complication of sepsis, and it is associated with high mortality and poor outcomes. Recent evidence has identified that autophagy participates in the pathophysiology of sepsis-associated AKI. Despite the use of antibiotics, the mortality rate is still at an extremely high level in patients with sepsis. Besides traditional treatments, many natural products, including phytochemicals and their derivatives, are proved to exert protective effects through multiple mechanisms, such as regulation of autophagy, inhibition of inflammation, fibrosis, and apoptosis, etc. Accumulating evidence has also shown that many pharmacological inhibitors might have potential therapeutic effects in sepsis-induced AKI. Hence, understanding the pathophysiology of sepsis-induced AKI may help to develop novel therapeutics to attenuate the complications of sepsis and lower the mortality rate. This review updates the recent progress of underlying pathophysiological mechanisms of sepsis-associated AKI, focuses specifically on autophagy, and summarizes the potential therapeutic effects of phytochemicals and pharmacological inhibitors.
Collapse
|
26
|
Youssef SSM, Ibrahim NK, El-Sonbaty SM, El-Din Ezz MK. Rutin Suppresses DMBA Carcinogenesis in the Breast Through Modulating IL-6/NF-κB, SRC1/HSP90 and ER-α. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221118213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rutin dietary supplements may offer pharmacological benefits as anticancer and antiinflammatory properties. This study aimed to investigate the inhibitory and protective effect of rutin on signaling pathways of mammary gland carcinogenesis expermintally induced in female rats by 7,12-di-methyl benz (a) anthracene (DMBA). Results showed that rutin administration ameliorated DMBA toxicity and carcinogic effect on kidney and liver revealed by a significant decrease of urea and creatinine levels, and the activity of the liver enzymes alanine aminotransferase (ALT) and alkaline phosphatase (ALP). The antioxidant state indicated by the total antioxidant capacity (TAC) was significantly increased accompanied by a reduction in the inflammatory markers of interleukin-1β (IL-1B), interleukin-6 (IL-6), and tumor necrosis factor (TNF-α) with induction of apoptosis indicated by a significant increase in caspase-3 level. Rutin significantly reduced the levels of the tumor markers carcinoma antigen 15-3 (CA 15-3) and proto-oncogene tyrosine-protein kinase Src1 (Src1). along with downregulation of nuclear factor-kB (NF-κB), heat shock protein 90 (HSP 90), and inducible nitric oxide synthase (iNOS) gene expression. The present study demonstrated the beneficial anticancer activity of rutin as a protective and therapeutic agent. Rutin induces its antitumor activity through elevation of the antioxidant state, inhibition of inflammatory cytokines, downregulation of oncogenes expression, and stimulation of apoptosis.
Collapse
Affiliation(s)
| | - Nashwa K Ibrahim
- National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Egypt
| | - Sawsan M El-Sonbaty
- National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Egypt
| | | |
Collapse
|
27
|
Zhao JL, Qiao XH, Mao JH, Liu F, Fu HD. The interaction between cellular senescence and chronic kidney disease as a therapeutic opportunity. Front Pharmacol 2022; 13:974361. [PMID: 36091755 PMCID: PMC9459105 DOI: 10.3389/fphar.2022.974361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/03/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasingly serious public health problem in the world, but the effective therapeutic approach is quite limited at present. Cellular senescence is characterized by the irreversible cell cycle arrest, senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Renal senescence shares many similarities with CKD, including etiology, mechanism, pathological change, phenotype and outcome, however, it is difficult to judge whether renal senescence is a trigger or a consequence of CKD, since there is a complex correlation between them. A variety of cellular signaling mechanisms are involved in their interactive association, which provides new potential targets for the intervention of CKD, and then extends the researches on senotherapy. Our review summarizes the common features of renal senescence and CKD, the interaction between them, the strategies of senotherapy, and the open questions for future research.
Collapse
Affiliation(s)
- Jing-Li Zhao
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiao-Hui Qiao
- Department of Pediatric Internal Medicine, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Jian-Hua Mao
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- *Correspondence: Jian-Hua Mao,
| | - Fei Liu
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hai-Dong Fu
- Department of Nephrology, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
28
|
The Correlation between the Level of Skin Advanced Glycation End Products in Type 2 Diabetes Mellitus and the Stages of Diabetic Retinopathy and the Types of Traditional Chinese Medicine Syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5193944. [PMID: 35845597 PMCID: PMC9286975 DOI: 10.1155/2022/5193944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 02/06/2022] [Accepted: 05/09/2022] [Indexed: 12/20/2022]
Abstract
Objective We aimed to analyze the correlation between the level of skin advanced glycation end products (AGEs) in type 2 diabetes mellitus (T2DM) patients and the diabetic retinopathy (DR) staging in different traditional Chinese medicine (TCM) syndromes. Methods 416 T2DM patients were divided into normal group, nonproliferative diabetic retinopathy (NPDR) group (mild, moderate, and severe), and proliferative diabetic retinopathy (PDR) group according to the DR grade. Patients' height, weight, fasting blood glucose (FBG), hemoglobin A1C (HbA1c), blood lipid, renal function, and skin AGEs were measured. According to TCM syndrome differentiation criteria, 230 patients with T2DM and DR were divided into I. qi and yin deficiency, collateral stasis group; II. liver and kidney deficiency, eye collaterals loss group; and III. yin and yang deficiency, blood stasis, and phlegm coagulation group. Results The skin AGEs levels of different DR staging groups were statistically significant (P < 0.05), and the skin AGEs levels in the mild and moderate NPDR groups were significantly higher (P < 0.05) than those of the normal group. It was significantly higher (P < 0.05) in the severe NPDR group than in the normal group, mild and moderate NPDR groups. The skin AGEs levels of the PDR group were significantly higher (P < 0.05) than the normal group, mild and moderate NPDR groups. It was positively correlated with DR stage, HbA1c, total cholesterol (TC), low-density lipoprotein (LDL), and urine metal analysis (UMA) (r = 0.467, 0.411, 0.413, 0.503, 0.424, P < 0.05). The skin AGEs levels of the qi and yin deficiency and collaterals stasis syndrome group were significantly higher (P < 0.05) than in the liver and kidney deficiency and eye collaterals loss groups. It was also significantly higher (P < 0.05) in yin and yang deficiency, blood stasis, and phlegm coagulation syndrome groups than in qi and yin deficiency and collaterals stasis syndrome groups. Conclusion There is a positive correlation between skin AGEs and DR staging in T2DM patients. Skin AGEs level is predictive for the risk of DR complications in T2DM patients and is vital in assessing DR degree per TCM syndrome type.
Collapse
|
29
|
Salari S, Ghorbanpour A, Marefati N, Baluchnejadmojarad T, Roghani M. Therapeutic effect of lycopene in lipopolysaccharide nephrotoxicity through alleviation of mitochondrial dysfunction, inflammation, and oxidative stress. Mol Biol Rep 2022; 49:8429-8438. [DOI: 10.1007/s11033-022-07661-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/30/2022] [Indexed: 12/29/2022]
|
30
|
Ma X, Wang F, Hang T, Dramou P. Degradation study of rutin as template from magnetic composite molecularly imprinted polymer supernatant samples by liquid chromatography–mass spectrometry. J Chromatogr A 2022; 1673:463199. [DOI: 10.1016/j.chroma.2022.463199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/03/2022] [Accepted: 06/03/2022] [Indexed: 12/11/2022]
|
31
|
Cheng L, Shi L, He C, Wang C, Lv Y, Li H, An Y, Dai H, Duan Y, Zhang H, Huang Y, Fu W, Meng Y, Zhao B. Rutin-activated adipose tissue thermogenesis is correlated with increased intestinal short-chain fatty acid levels. Phytother Res 2022; 36:2495-2510. [PMID: 35445769 DOI: 10.1002/ptr.7462] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
The activation of thermogenic programs in brown adipose tissue (BAT) and white adipose tissue (WAT) provides a promising approach to increasing energy expenditure during obesity and diabetes treatment. Although evidence has been found that rutin activates BAT against obesity and type 2 diabetes mellitus (T2DM), its potential mechanism is not completely understood. In this study, we focused on the potential modulating effect of rutin on short-chain fatty acids (SCFAs) and the thermogenesis of BAT and WAT, aiming to elucidate the molecular mechanism of rutin in the treatment of obesity and T2DM. The results showed that rutin could significantly reduce the body weight and fasting blood glucose, inhibit fat accumulation, relieve hepatic steatosis and ameliorate the disorder of glycolipid metabolism in db/db mice. Moreover, rutin also increased the expression of uncoupling protein 1 (Ucp1) and other thermogenic genes and proteins in BAT and inguinal WAT (IWAT), indicating that rutin activated BAT and induced browning of IWAT. Importantly, rutin markedly enhanced the concentration of SCFAs (acetate, propionate and butyrate) and SCFA-producing enzymes (acetate kinase (ACK), methylmalonyl-CoA decarboxylase (MMD) and butyryl-CoA (BUT)) in feces of db/db mice. In addition, rutin significantly increased the mRNA expression of monocarboxylate transporter 1 (Mct1), catabolic enzyme acyl-CoA medium-chain synthetase 3 (Acsm3), carnitine palmitoyl transferase 1α (Cpt-1α) and Cpt-1β genes in BAT and IWAT of db/db mice, which is conducive to inducing adipocyte thermogenesis. In summary, our findings revealed that rutin played a variety of regulatory roles in improving glucose and lipid metabolism disorders, reducing hepatic steatosis, inducing browning of IWAT and activating BAT, which has potential therapeutic significance for the treatment of obesity and T2DM. Mechanistically, rutin activates the thermogenesis of BAT and IWAT, which may be associated with increasing the concentration of SCFAs.
Collapse
Affiliation(s)
- Long Cheng
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Shi
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Changhao He
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chen Wang
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yinglan Lv
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huimin Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yongcheng An
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hongyu Dai
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhui Duan
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huilin Zhang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Huang
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Wanxin Fu
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yanyan Meng
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Hispidulin Ameliorates Endotoxin-Induced Acute Kidney Injury in Mice. Molecules 2022; 27:molecules27062019. [PMID: 35335387 PMCID: PMC8948942 DOI: 10.3390/molecules27062019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/13/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023] Open
Abstract
Lipopolysaccharide (LPS) is an endotoxin that plays a crucial role in septic acute kidney injury (AKI). Hispidulin is a natural flavonoid that possesses various biological activities. Recent studies have shown that hispidulin administration alleviates various inflammatory diseases in animal models. This study aimed to investigate the renoprotective effect of hispidulin on LPS-induced AKI. Male C57BL/6 mice were administered LPS (10 mg/kg) with or without hispidulin (50 mg/kg). Hispidulin administration attenuated renal dysfunction, histological alterations, and the upregulation of neutrophil gelatinase-associated lipocalin. This flavonoid also reduced cytokine production and Toll-like receptor 4 expression, inhibited nuclear factor-κB and mitogen-activated protein kinase cascades, and alleviated immune cell infiltration. The oxidation of lipids and DNA was also inhibited by hispidulin administration. This antioxidant effect of hispidulin was associated with the downregulation of NADPH oxidase 4, the activation of catalase and superoxide dismutase activities, and the restoration of glutathione levels. Moreover, hispidulin administration attenuated tubular cell apoptosis by inhibiting caspase-3 pathway. These data suggest that hispidulin ameliorates endotoxin-induced kidney injury by suppressing inflammation, oxidative stress, and tubular cell death.
Collapse
|
33
|
Potential of Polyphenols to Restore SIRT1 and NAD+ Metabolism in Renal Disease. Nutrients 2022; 14:nu14030653. [PMID: 35277012 PMCID: PMC8837945 DOI: 10.3390/nu14030653] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
SIRT1 is an NAD+-dependent class III histone deacetylase that is abundantly expressed in the kidney, where it modulates gene expression, apoptosis, energy homeostasis, autophagy, acute stress responses, and mitochondrial biogenesis. Alterations in SIRT1 activity and NAD+ metabolism are frequently observed in acute and chronic kidney diseases of diverse origins, including obesity and diabetes. Nevertheless, in vitro and in vivo studies and clinical trials with humans show that the SIRT1-activating compounds derived from natural sources, such as polyphenols found in fruits, vegetables, and plants, including resveratrol, quercetin, and isoflavones, can prevent disease and be part of treatments for a wide variety of diseases. Here, we summarize the roles of SIRT1 and NAD+ metabolism in renal pathophysiology and provide an overview of polyphenols that have the potential to restore SIRT1 and NAD+ metabolism in renal diseases.
Collapse
|
34
|
Zhao J, Duan Q, Dong C, Cui J. Cul4a attenuates LPS-induced acute kidney injury via blocking NF-kB signaling pathway in sepsis. J Med Biochem 2022; 41:62-70. [PMID: 35611245 PMCID: PMC9069243 DOI: 10.5937/jomb0-33096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 11/24/2022] Open
Abstract
Background Acute kidney injury (AKI) is a common disease that can develop into end-stage kidney disease. Sepsis is one of the main causes of AKI. Currently, there is no satisfactory way to treat septic AKI. Therefore, we have shown the protective function of Cul4a in septic AKI and its molecular mechanism. Methods The cellular and animal models of septic AKI were established by using lipopolysaccharide (LPS). Western blot (WB) was employed to analyze Cul4a expression. RT-qPCR was employed to test the expression of Cul4a, SOD1, SOD2, GPX1, CAT, IL-6, TNF-a, Bcl-2, IL1b, Bax and KIM-1 mRNA. ELISA was performed to detect the contents of inflammatory factors and LDH. CCK-8 was utilized to detect cell viability. Flow cytometry was utilized to analyze the apoptosis. DHE-ROS kit was used to detect the content of ROS. Results Cul4a was down-regulated in cellular and animal models of septic AKI. Oxidative stress is obviously induced by LPS, as well as apoptosis and inflammation. However, these can be significantly inhibited by up-regulating Cul4a. Moreover, LPS induced the activation of the NF-kB pathway, which could also be inhibited by overexpression of Cul4a. Conclusions Cul4awas found to be a protective factor in septic AKI, which could inhibit LPS-induced oxidative stress, apoptosis and inflammation of HK-2 cells by inhibiting the NF-kB pathway.
Collapse
Affiliation(s)
- Jing Zhao
- Yantaishan Hospital, Department of Critical Care Medicine, Yantai, China
| | - Qiuxia Duan
- The Third People's Hospital of Qingdao, Department of Critical Care Medicine, Qingdao, China
| | - Cuihong Dong
- Shandong College of Traditional Chinese Medicine, Yantai, China
| | - Jing Cui
- The Third People's Hospital of Qingdao, Department of Emergency, Qingdao, China
| |
Collapse
|
35
|
Ungur RA, Borda IM, Codea RA, Ciortea VM, Năsui BA, Muste S, Sarpataky O, Filip M, Irsay L, Crăciun EC, Căinap S, Jivănescu DB, Pop AL, Singurean VE, Crișan M, Groza OB, Martiș (Petruț) GS. A Flavonoid-Rich Extract of Sambucus nigra L. Reduced Lipid Peroxidation in a Rat Experimental Model of Gentamicin Nephrotoxicity. MATERIALS (BASEL, SWITZERLAND) 2022; 15:772. [PMID: 35160718 PMCID: PMC8837157 DOI: 10.3390/ma15030772] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 12/04/2022]
Abstract
The use of gentamicin (GM) is limited due to its nephrotoxicity mediated by oxidative stress. This study aimed to evaluate the capacity of a flavonoid-rich extract of Sambucus nigra L. elderflower (SN) to inhibit lipoperoxidation in GM-induced nephrotoxicity. The HPLC analysis of the SN extract recorded high contents of rutin (463.2 ± 0.0 mg mL-1), epicatechin (9.0 ± 1.1 µg mL-1), and ferulic (1.5 ± 0.3 µg mL-1) and caffeic acid (3.6 ± 0.1 µg mL-1). Thirty-two Wistar male rats were randomized into four groups: a control group (C) (no treatment), GM group (100 mg kg-1 bw day-1 GM), GM+SN group (100 mg kg-1 bw day-1 GM and 1 mL SN extract day-1), and SN group (1 mL SN extract day-1). Lipid peroxidation, evaluated by malondialdehyde (MDA), and antioxidant enzymes activity-superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX)-were recorded in renal tissue after ten days of experimental treatment. The MDA level was significantly higher in the GM group compared to the control group (p < 0.0001), and was significantly reduced by SN in the GM+SN group compared to the GM group (p = 0.021). SN extract failed to improve SOD, CAT, and GPX activity in the GM+SN group compared to the GM group (p > 0.05), and its action was most probably due to the ability of flavonoids (rutin, epicatechin) and ferulic and caffeic acids to inhibit synthesis and neutralize reactive species, to reduce the redox-active iron pool, and to inhibit lipid peroxidation. In this study, we propose an innovative method for counteracting GM nephrotoxicity with a high efficiency and low cost, but with the disadvantage of the multifactorial environmental variability of the content of SN extracts.
Collapse
Affiliation(s)
- Rodica Ana Ungur
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Ileana Monica Borda
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Răzvan Andrei Codea
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania;
| | - Viorela Mihaela Ciortea
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Bogdana Adriana Năsui
- Department of Community Health, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Sevastița Muste
- Food Engineering Department, University of Agricultural Sciences and Veterinary Medicine, 64 Calea Floresti, 400509 Cluj-Napoca, Romania; (S.M.); (G.S.M.)
| | - Orsolya Sarpataky
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania;
| | - Miuța Filip
- Raluca Ripan Institute for Research in Chemistry, Babeş-Bolyai University, 30 Fântânele Street, 400294 Cluj-Napoca, Romania;
| | - Laszlo Irsay
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Elena Cristina Crăciun
- Department of Pharmaceutical Biochemistry and Clinical Laboratory, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - Simona Căinap
- Department of Mother and Child, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Delia Bunea Jivănescu
- Department of Internal Medicine, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Anca Lucia Pop
- Department of Clinical Laboratory, Food Safety, Nutrition, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania;
| | - Victoria Emilia Singurean
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Maria Crișan
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Oana Bianca Groza
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Georgiana Smaranda Martiș (Petruț)
- Food Engineering Department, University of Agricultural Sciences and Veterinary Medicine, 64 Calea Floresti, 400509 Cluj-Napoca, Romania; (S.M.); (G.S.M.)
| |
Collapse
|
36
|
CHEN C, WEN D, DU J, XIAO H, ZHONG S, WU Z, PENG J, LIU D, TANG H. Activation of SIRT1 signaling pathway by clove improves cognitive dysfunction in septic mice. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.82622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
| | | | - Juan DU
- Army Medical University, China
| | | | | | | | - Ji PENG
- Army Medical University, China
| | | | | |
Collapse
|
37
|
Gravina AG, Pellegrino R, Facchiano A, Palladino G, Loguercio C, Federico A. Evaluation of the Efficacy and Safety of a Compound of Micronized Flavonoids in Combination With Vitamin C and Extracts of Centella asiatica, Vaccinium myrtillus, and Vitis vinifera for the Reduction of Hemorrhoidal Symptoms in Patients With Grade II and III Hemorrhoidal Disease: A Retrospective Real-Life Study. Front Pharmacol 2021; 12:773320. [PMID: 34970145 PMCID: PMC8712720 DOI: 10.3389/fphar.2021.773320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/15/2021] [Indexed: 12/18/2022] Open
Abstract
Background and Aim: Several evidences have shown how, in hemorrhoidal disease, phlebotonic flavonoid agents such as quercetin reduce capillary permeability by increasing vascular walls resistance, how rutin and vitamin C have antioxidant properties, and that Centella asiatica has reparative properties towards the connective tissue. A retrospective study was designed in order to evaluate the efficacy and safety of a compound consisting of micronized flavonoids in combination with vitamin C and extracts of C. asiatica, Vaccinium myrtillus, and Vitis vinifera for grade II and III hemorrhoidal disease. Patients and Methods: Data of 49 patients, over 18, who were following a free diet regimen, not on therapy with other anti-hemorrhoid agents, treated with a compound consisting of 450 mg of micronized diosmin, 300 mg of C. asiatica, 270 mg of micronized hesperidin, 200 mg of V. vinifera, 160 mg of vitamin C, 160 mg of V. myrtillus, 140 mg of micronized quercetin, and 130 mg of micronized rutin (1 sachet or 2 tablets a day) for 7 days were collected. Hemorrhoid grade according to Goligher's scale together with anorectal symptoms (edema, prolapse, itching, thrombosis, burning, pain, tenesmus, and bleeding) both before treatment (T0) and after 7 days of therapy (T7) were collected. Primary outcomes were the reduction of at least one degree of hemorrhoids according to Goligher's scale assessed by proctological examination and compound safety. The secondary outcome was the reduction of anorectal symptoms assessed by questionnaires administered to patients. Results: Forty-four patients (89.8%) presented a reduction in hemorrhoidal grade of at least one grade (p < 0.001). No adverse events with the use of the compound were noted. A significant reduction was observed in all anorectal symptoms evaluated (p < 0.05). No predictors of response to the compound were identified among the clinical and demographic variables collected. Conclusion: The compound analyzed was effective and safe for patients with grade II and III hemorrhoidal disease according to Goligher's scale.
Collapse
Affiliation(s)
- Antonietta G. Gravina
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Network Pharmacology-Based Identification of Potential Targets of Lonicerae japonicae Flos Acting on Anti-Inflammatory Effects. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5507003. [PMID: 34595237 PMCID: PMC8478540 DOI: 10.1155/2021/5507003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/10/2021] [Accepted: 08/25/2021] [Indexed: 12/31/2022]
Abstract
Lonicerae japonicae flos (LJF) is widely used for the treatment of inflammation-related diseases in traditional Chinese medicine (TCM). To clarify the anti-inflammatory mechanism of LJF, 29 compounds with high content in LJF were selected for network pharmacology. Then, a comprehensive network pharmacology strategy was implemented, which involved compound-inflammation-target construction, protein-protein interaction (PPI) network analysis, and enrichment analysis. Finally, molecular docking and in vitro experiments were performed to verify the anti-inflammatory activity and targets of the key compound. As a result, 279 inflammation-associated proteins were identified, which are mainly involved in the AGE/RAGE signaling pathway in diabetic complications, the HIF-1 signaling pathway, the PI3K-AKT signaling pathway, and EGFR tyrosine kinase inhibitor resistance. A total of 12 compounds were linked to more than 35 targets, including apigenin, kaempferol, quercetin, luteolin, and ferulic acid. The results of molecular docking showed that AKT has the most binding activity, exhibiting certain binding activity with 10 compounds, including vanillic acid, protocatechuic acid, secologanic acid, quercetin, and luteolin; the results of qRT-PCR and WB confirmed that two key compounds, secologanic acid and luteolin, could significantly decrease the secretion of TNF-α and the AKT expression of RAW264.7 murine macrophages stimulated by LPS (lipopolysaccharide). These results demonstrate that the comprehensive strategy can serve as a universal method to illustrate the anti-inflammatory mechanisms of traditional Chinese medicine by identifying the pathways or targets.
Collapse
|
39
|
Han X, Ding C, Sang X, Peng M, Yang Q, Ning Y, Lv Q, Shan Q, Hao M, Wang K, Wu X, Zhang H, Cao G. Targeting Sirtuin1 to treat aging-related tissue fibrosis: From prevention to therapy. Pharmacol Ther 2021; 229:107983. [PMID: 34480962 DOI: 10.1016/j.pharmthera.2021.107983] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Fibrosis, which is characterized by excessive extracellular matrix (ECM) deposition, is a wound-healing response to organ injury and may promote cancer and failure in various organs, such as the heart, liver, lung, and kidney. Aging associated with oxidative stress and inflammation exacerbates cellular dysfunction, tissue failure, and body function disorders, all of which are closely related to fibrosis. Sirtuin-1 (SIRT1) is a class III histone deacetylase that regulates growth, transcription, aging, and metabolism in various organs. This protein is downregulated in organ injury and fibrosis associated with aging. Its expression and distribution change with age in different organs and play critical roles in tissue oxidative stress and inflammation. This review first described the background on fibrosis and regulatory functions of SIRT1. Second, we summarized the relationships of SIRT1 with other proteins and its protective action during fibrosis in the heart, liver, lung and kidney. Third, the activation of SIRT1 in therapies of tissue fibrosis, especially in liver fibrosis and aging-related tissue injury, was analyzed. In conclusion, SIRT1 targeting may be a new therapeutic strategy in fibrosis.
Collapse
Affiliation(s)
- Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - XiaNan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - MengYun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Lv
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - QiYuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - KuiLong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyan Zhang
- Cancer Hospital of The University of Chinese Academy of Sciences, Hangzhou, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
40
|
Karimi A, Naeini F, Asghari Azar V, Hasanzadeh M, Ostadrahimi A, Niazkar HR, Mobasseri M, Tutunchi H. A comprehensive systematic review of the therapeutic effects and mechanisms of action of quercetin in sepsis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 86:153567. [PMID: 33940332 DOI: 10.1016/j.phymed.2021.153567] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Sepsis is a life-threatening condition caused by a dysregulated host response to infection. Several studies have indicated that flavonoids exhibit a wide variety of biological actions including free radical scavenging and antioxidant activities. Quercetin, one of the most extensively distributed flavonoids in the vegetables and fruits, presents various biological activities including modulation of oxidative stress, anti-infectious, anti-inflammatory, and neuroprotective activities. METHODS The present systematic review was conducted according to the guidelines of the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) statements. We searched Web of Sciences, Google Scholar, PubMed, Scopus, and Embase databases up to February 2021 by using the relevant keywords. RESULTS Out of 672 records screened, 35 articles met the study criteria. The evidence reviewed here indicates that quercetin supplementation may exert beneficial effects on sepsis by attenuating inflammation and oxidative stress, downregulating the mRNA expression of toll-like receptors (TLRs), modulating the immune response, and alleviating sepsis-related organ dysfunctions. CONCLUSION Due to the promising therapeutic effects of quercetin on sepsis complications and the lack of clinical trials in this regard, future human randomized clinical trials are warranted.
Collapse
Affiliation(s)
- Arash Karimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
| | - Vahid Asghari Azar
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Malihe Hasanzadeh
- Department of Biology, Ardabil Branch Islamic Azad University, Ardabil, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Niazkar
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Majid Mobasseri
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helda Tutunchi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Combining edible coatings technology and nanoencapsulation for food application: A brief review with an emphasis on nanoliposomes. Food Res Int 2021; 145:110402. [PMID: 34112405 DOI: 10.1016/j.foodres.2021.110402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/28/2021] [Accepted: 05/06/2021] [Indexed: 01/06/2023]
Abstract
The use of bioactive compounds within the biopolymer-based Edible Coatings (EC) matrices has certain limitations for their application at the food industry level. Encapsulation has been considered as a strategy that enables protecting and improving the physical and chemical characteristics of the compounds; as a result, it extends the shelf life of coated foods. This review discusses recent progress in combining edible coatings with nanoencapsulation technology. We also described and discussed various works, in which nanoliposomes are used as encapsulation systems to prepare, and subsequently apply the edible coatings in plant products and meat products. The use of nanoliposomes for the encapsulation of phenolic compounds and essential oils provides an improvement in the antioxidant and antimicrobial properties of coatings by extending the shelf life of food matrices. However, when liposomes are stored for a long period of time, they may present some degree of instability manifested by an increase in size, polydispersity index, and zeta potential. This is reflected in an aggregation, fusion, and rupture of the vesicles. This investigation can help researchers and industries to select an appropriate and efficient biopolymer to form EC containing nanoencapsulated active compounds. This work also addresses the use of nanoliposomes to create EC extending markedly the shelf life of fruit, reducing the weight loss, and deterioration due to the action of microorganisms.
Collapse
|
42
|
Activators of SIRT1 in the kidney and protective effects of SIRT1 during acute kidney injury (AKI) (effect of SIRT1 activators on acute kidney injury). Clin Exp Nephrol 2021; 25:807-821. [PMID: 33779856 DOI: 10.1007/s10157-021-02057-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is a complex disorder and a clinical condition characterized by acute reduction in renal function. If AKI is not treated, it can lead to chronic kidney disease, which is associated with a high risk of death. SIRT1 (silent information regulator 1) is an NAD-dependent deacetylase. This enzyme is responsible for the processes of DNA repair or recombination, chromosomal stability, and gene transcription. This enzyme also plays a protective role in many diseases, including AKI. In this study, we review the mechanisms that mediate the protective effects of SIRT1 on AKI, including SIRT1 activators.
Collapse
|
43
|
Integrating metabolomics and network pharmacology to explore Rhizoma Coptidis extracts against sepsis-associated acute kidney injury. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1164:122525. [PMID: 33454441 DOI: 10.1016/j.jchromb.2021.122525] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/01/2020] [Accepted: 01/01/2021] [Indexed: 11/20/2022]
Abstract
Sepsis remains the most common cause of acute kidney injury (AKI) in critically ill patients, increasing the risk of in-hospital and long-term death. Rhizoma Coptidis (RC), a classical traditional Chinese herb, exhibits anti-inflammatory and antioxidant properties in various diseases including sepsis. This study aimed to investigate the protective effects of RC extracts (RCE) against sepsis-associated acute kidney injury (SA-AKI) and explore the underlying mechanisms with metabolomics-based network pharmacology. The results showed that RCE improved renal function and histological injury and decreased reactive oxygen species (ROS) production in SA-AKI. Using ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-Q-TOF/MS), 25 differential metabolites were identified that had a close connection with the pathological processes of SA-AKI and the effects of RCE. Afterward, a compound-metabolite-target-disease network was constructed and 17 overlapping target proteins of the components of RCE, the differential metabolites, and the disease-related genes were discovered. Among these overlapping target proteins, RCE increased the nuclear translocation of nuclear factor-erythroid 2-related factor-2 (Nrf2), the protein expression of heme oxygenase-1 (HO-1), the mRNA expression of peroxisome proliferator activated receptor α (PPARα) and reduced nitric oxide synthase 2 (NOS2) activity. In addition, molecular docking revealed that both berberine and quercetin could bond with NOS2 and PPARα, respectively. Therefore, RCE demonstrated protective effects for SA-AKI through the regulation of metabolism and different signaling pathways.
Collapse
|
44
|
Huang FM, Chang YC, Su CH, Wu SW, Lee SS, Lee MW, Yeh KL, Chiang CY, Tu DG, Lu YC, Kuan YH. Rutin-protected BisGMA-induced cytotoxicity, genotoxicity, and apoptosis in macrophages through the reduction of the mitochondrial apoptotic pathway and induction of antioxidant enzymes. ENVIRONMENTAL TOXICOLOGY 2021; 36:45-54. [PMID: 32830914 DOI: 10.1002/tox.23009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/03/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
Bisphenol-A-glycidyldimethacrylate (BisGMA) is a resin monomer frequently used in dentin restorative treatments. The leakage of BisGMA monomer from BisGMA-based polymeric resins can lead to cytotoxicity in macrophages. Rutin has various beneficial bioeffects, including antioxidation and antiinflammation. In this study, we found that pretreatment of RAW264.7 macrophages with rutin-inhibited cytotoxicity induced by BisGMA in a concentration-dependent manner. BisGMA-induced apoptosis, which was detected by levels of phosphatidylserine from the internal to the external membrane and formation of sub-G1, and genotoxicity, which was detected by cytokinesis-blocked micronucleus and single-cell gel electrophoresis assays, were inhibited by rutin in a concentration-dependent manner. Rutin suppressed the BisGMA-induced activation of caspase-3 and -9 rather than caspase-8. Rutin inhibited the activation of the mitochondrial apoptotic pathway, including cytochrome C release and mitochondria disruption, after macrophages were treated with BisGMA. Finally, BisGMA-induced reactive oxygen species (ROS) generation and antioxidant enzyme (AOE) deactivation could be reversed by rutin. Parallel trends were observed in the elevation of AOE activation and inhibition of ROS generation, caspase-3 activity, mitochondrial apoptotic pathway activation, and genotoxicity. These results suggested that rutin suppressed BisGMA-induced cytotoxicity through genotoxicity, the mitochondrial apoptotic pathway, and relatively upstream factors, including reduction of ROS generation and induction of AOE.
Collapse
Affiliation(s)
- Fu-Mei Huang
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chao Chang
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Hung Su
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Sheng-Wen Wu
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Nephrology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shiuan-Shinn Lee
- School of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Min-Wei Lee
- A Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| | - Kun-Lin Yeh
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chen-Yu Chiang
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Dom-Gene Tu
- Department of Nuclear Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Biomedical Science, National Chung Cheng University, Chiayi, Taiwan
| | - Yin-Che Lu
- Min-Hwei Junior College of Health Care Management, Tainan, Taiwan
- Division of Hematology-Oncology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
45
|
Rahbar Saadat Y, Hosseiniyan Khatibi SM, Ardalan M, Barzegari A, Zununi Vahed S. Molecular pathophysiology of acute kidney injury: The role of sirtuins and their interactions with other macromolecular players. J Cell Physiol 2020; 236:3257-3274. [PMID: 32989772 DOI: 10.1002/jcp.30084] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Acute kidney injury (AKI), a rapid drop in kidney function, displays high mortality and morbidity, and its repeated or severe status can shift into chronic kidney disease or even end-stage renal disease. How and which events cause AKI still is controversial. In addition, no specific therapies have emerged that can attenuate AKI or expedite recovery. Some central mechanisms including tubular epithelial cells injury, endothelial injury, renal cell apoptosis, and necrosis signaling cascades, and inflammation have been reported in the pathophysiology of AKI. However, the timing of the activation of each pathway, their interactions, and the hierarchy of these pathways remain unknown. The main molecular mechanisms that might be complicated in this process are the mitochondrial impairment and alteration/shifting of cellular metabolites (e.g., acetyl-CoA and NAD+ /NADH) acting as cofactors to alter the activities of many enzymes, for instance, sirtuins. Moreover, alteration of mitochondrial structure over the fusion and fission mechanisms can regulate cellular signaling pathways by modifying the rate of reactive oxygen species generation and metabolic activities. The aim of this review is to better understand the underlying pathophysiological and molecular mechanisms of AKI. In addition, we predicted the main other molecular players in interaction with sirtuins as energy/stresses monitoring proteins for the development of future approaches in the treatment or prevention of ischemic AKI.
Collapse
Affiliation(s)
- Yalda Rahbar Saadat
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Villetaneuse, France
| | | |
Collapse
|
46
|
MBD2 Mediates Septic AKI through Activation of PKCη/p38MAPK and the ERK1/2 Axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:76-88. [PMID: 33335794 PMCID: PMC7723772 DOI: 10.1016/j.omtn.2020.09.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022]
Abstract
Our previous study demonstrated that the methyl-CpG-binding domain protein 2 (MBD2) mediates vancomycin (VAN)-induced acute kidney injury (AKI). However, the role and regulation of MBD2 in septic AKI are unknown. Herein, MBD2 was induced by lipopolysaccharide (LPS) in Boston University mouse proximal tubules (BUMPTs) and mice. For both in vitro and in vivo experiments, we showed that inhibition of MBD2 by MBD2 small interfering RNA (siRNA) and MBD2-knockout (KO) substantially improved the survival rate and attenuated both LPS and cecal ligation and puncture (CLP)-induced AKI, renal cell apoptosis, and inflammatory factor production. Global genetic expression analyses and in vitro experiments suggest that the expression of protein kinase C eta (PKCη), caused by LPS, is markedly suppressed in MBD2-KO mice and MBD2 siRNA, respectively. Mechanistically, chromatin immunoprecipitation (ChIP) analysis indicates that MBD2 directly binds to promoter region CpG islands of PKCη via suppression of promoter methylation. Furthermore, PKCη siRNA improves the survival rate and attenuates LPS-induced BUMPT cell apoptosis and inflammatory factor production via inactivation of p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK)1/2, which were further verified by PKCη siRNA treatment in CLP-induced AKI. Finally, MBD2-KO mice exhibited CLP-induced renal cell apoptosis and inflammatory factor production by inactivation of PKCη/p38MAPK and ERK1/2 signaling. Taken together, the data indicate that MBD2 mediates septic-induced AKI through the activation of PKCη/p38MAPK and the ERK1/2 axis. MBD2 represents a potential target for treatment of septic AKI.
Collapse
|
47
|
Silva B, Biluca FC, Mohr ETB, Caon T, Gonzaga LV, Fett R, Dalmarco EM, Costa ACO. Effect of Mimosa scabrella Bentham honeydew honey on inflammatory mediators. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
48
|
Jiang N, Jingwei L, Wang H, Huang H, Wang Q, Zeng G, Li S, Liu X. Ginsenoside 20(S)-protopanaxadiol attenuates depressive-like behaviour and neuroinflammation in chronic unpredictable mild stress-induced depressive rats. Behav Brain Res 2020; 393:112710. [DOI: 10.1016/j.bbr.2020.112710] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/27/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022]
|
49
|
Nouri Z, Fakhri S, Nouri K, Wallace CE, Farzaei MH, Bishayee A. Targeting Multiple Signaling Pathways in Cancer: The Rutin Therapeutic Approach. Cancers (Basel) 2020; 12:E2276. [PMID: 32823876 PMCID: PMC7463935 DOI: 10.3390/cancers12082276] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple dysregulated signaling pathways are implicated in the pathogenesis of cancer. The conventional therapies used in cancer prevention/treatment suffer from low efficacy, considerable toxicity, and high cost. Hence, the discovery and development of novel multi-targeted agents to attenuate the dysregulated signaling in cancer is of great importance. In recent decades, phytochemicals from dietary and medicinal plants have been successfully introduced as alternative anticancer agents due to their ability to modulate numerous oncogenic and oncosuppressive signaling pathways. Rutin (also known as rutoside, quercetin-3-O-rutinoside and sophorin) is an active plant-derived flavonoid that is widely distributed in various vegetables, fruits, and medicinal plants, including asparagus, buckwheat, apricots, apples, cherries, grapes, grapefruit, plums, oranges, and tea. Rutin has been shown to target various inflammatory, apoptotic, autophagic, and angiogenic signaling mediators, including nuclear factor-κB, tumor necrosis factor-α, interleukins, light chain 3/Beclin, B cell lymphoma 2 (Bcl-2), Bcl-2 associated X protein, caspases, and vascular endothelial growth factor. A comprehensive and critical analysis of the anticancer potential of rutin and associated molecular targets amongst various cancer types has not been performed previously. Accordingly, the purpose of this review is to present an up-to-date and critical evaluation of multiple cellular and molecular mechanisms through which the anticancer effects of rutin are known to be exerted. The current challenges and limitations as well as future directions of research are also discussed.
Collapse
Affiliation(s)
- Zeinab Nouri
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Keyvan Nouri
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Carly E. Wallace
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| |
Collapse
|
50
|
Lu S, Dong L, Jing X, Gen-Yang C, Zhan-Zheng Z. Abnormal lncRNA CCAT1/microRNA-155/SIRT1 axis promoted inflammatory response and apoptosis of tubular epithelial cells in LPS caused acute kidney injury. Mitochondrion 2020; 53:76-90. [DOI: 10.1016/j.mito.2020.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
|