1
|
Azmat F, Safdar M, Ahmad H, Khan MRJ, Abid J, Naseer MS, Aggarwal S, Imran A, Khalid U, Zahra SM, Islam F, Cheema SA, Shehzadi U, Ali R, Kinki AB, Ali YA, Suleria HAR. Phytochemical profile, nutritional composition of pomegranate peel and peel extract as a potential source of nutraceutical: A comprehensive review. Food Sci Nutr 2024; 12:661-674. [PMID: 38370077 PMCID: PMC10867480 DOI: 10.1002/fsn3.3777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/30/2023] [Accepted: 10/07/2023] [Indexed: 02/20/2024] Open
Abstract
The current study focuses on Punica granatum L. (pomegranate) peel and peel extract and their use as functional foods, food additives, or physiologically active constituents in nutraceutical formulations. The pomegranate peel extract is a good source of bioactive substances needed for the biological activity of the fruit, including phenolic acids, minerals, flavonoids (anthocyanins), and hydrolyzable tannins (gallic acid). The macromolecules found in pomegranate peel and peel extract have been recommended as substitutes for synthetic nutraceuticals, food additives, and chemo-preventive agents because of their well-known ethno-medical significance and chemical properties. Moreover, considering the promises for both their health-promoting activities and chemical properties, the dietary and nutraceutical significance of pomegranate peel and pomegranate peel extract appears to be underestimated. The present review article details their nutritional composition, phytochemical profile, food applications, nutraceutical action, and health benefits.
Collapse
Affiliation(s)
- Faiza Azmat
- Department of Nutritional Sciences and Environmental DesignAllama Iqbal Open UniversityIslamabadPakistan
| | - Mahpara Safdar
- Department of Nutritional Sciences and Environmental DesignAllama Iqbal Open UniversityIslamabadPakistan
| | - Hajra Ahmad
- Department of Nutritional Sciences and Environmental DesignAllama Iqbal Open UniversityIslamabadPakistan
| | | | - Junaid Abid
- Department of Food Science and TechnologyUniversity of HaripurHaripurPakistan
| | | | - Saurabh Aggarwal
- Department of Mechanical Engineering Uttaranchal Institute of TechnologyUttaranchal UniversityDehradunIndia
| | - Ali Imran
- Department of Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Urma Khalid
- Department of Nutritional Sciences and Environmental DesignAllama Iqbal Open UniversityIslamabadPakistan
| | - Syeda Mahvish Zahra
- Department of Nutritional Sciences and Environmental DesignAllama Iqbal Open UniversityIslamabadPakistan
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
| | - Fakhar Islam
- Department of Clinical NutritionNUR International UniversityLahorePakistan
- Department of Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Sadia Arif Cheema
- Department of Nutritional Sciences and Environmental DesignAllama Iqbal Open UniversityIslamabadPakistan
| | - Umber Shehzadi
- Department of Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Rehman Ali
- Department of Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Abdela Befa Kinki
- Food Science and NutritionEthiopian Institute of Agricultural ResearchAddis AbabaEthiopia
| | - Yuosra Amer Ali
- Department of Food Sciences, College of Agriculture and ForestryUniversity of MosulMosulIraq
| | | |
Collapse
|
2
|
Li X, Shi J, Teng Y, Liu Z. The preventative effect of Baihe Gujin Pill on cisplatin-induced acute kidney injury by activating the PI3K/AKT and suppressing the NF-κB/MAPK pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117071. [PMID: 37619855 DOI: 10.1016/j.jep.2023.117071] [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: 06/13/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baihe Gujin Pill (BHGJP) is a traditional Chinese medicine (TCM) derived from the "Collection of Medical Formulas". BHGJP is applied to treat lung and kidney deficiency by nourishing yin and clearing heat. However, the role and preventative mechanism of BHGJP in cisplatin induced acute kidney injury (CIAKI) are poorly understood. AIM OF THE STUDY The preventative effect of BHGJP on CIAKI by the in vitro and in vivo experiments based on network pharmacology was investigated. METHODS Network pharmacology was used to predict the protective effect of BHGJP on CIAKI. The effect and mechanism of BHGJP against CIAKI were detected and verified by the in vitro kidney cells 293T and HK-2 as well as the in vivo mice model established by a single injection of cisplatin. RESULTS Network pharmacology predicted that BHGJP prevented CIAKI by regulating PI3K/AKT and NF-κB/MAPK signaling pathways. BHGJP could reverse the reduced cell viability of HK-2 and 293T cells caused by cisplatin without decreasing its cytotoxic effects on H460, H1299, and A549 cells. Meanwhile, BHGJP effectively controlled kidney injury in the CIAKI model. Moreover, cisplatin induced cell apoptosis and accumulation of reactive oxygen species (ROS) were downregulated after treatment with BHGJP. The changes of oxidative stress indexes of GSH, MDA, and SOD as well as the inflammatory factors of TNF-α, IL-6, and IL-1β in the CIAKI model were recovered to normal state when BHGJP treatment. Furthermore, BHGJP activated PI3K/AKT pathway and suppressed the NF-κB/MAPK pathway in the CIAKI model. CONCLUSION The study found that BHGJP prevented CIAKI by inhibiting apoptosis, oxidative stress, and inflammation via regulating PI3K/AKT and NF-κB/MAPK pathways, providing new efficacy and clinical applications for BHGJP.
Collapse
Affiliation(s)
- Xinran Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Jieya Shi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| |
Collapse
|
3
|
Yuan W, Kou S, Ma Y, Qian Y, Li X, Chai Y, Jiang Z, Zhang L, Sun L, Huang X. Hyperoside ameliorates cisplatin-induced acute kidney injury by regulating the expression and function of Oat1. Xenobiotica 2023; 53:559-571. [PMID: 37885225 DOI: 10.1080/00498254.2023.2270046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
Cisplatin is a widely used chemotherapeutic agent to treat solid tumours in clinics. However, cisplatin-induced acute kidney injury (AKI) limits its clinical application. This study investigated the effect of hyperoside (a flavonol glycoside compound) on regulating AKI.The model of cisplatin-induced AKI was established, and hyperoside was preadministered to investigate its effect on improving kidney injury.Hyperoside ameliorated renal pathological damage, reduced the accumulation of SCr, BUN, Kim-1 and indoxyl sulphate in vivo, increased the excretion of indoxyl sulphate into the urine, and upregulated the expression of renal organic anion transporter 1 (Oat1). Moreover, evaluation of rat kidney slices demonstrated that hyperoside promoted the uptake of PAH (p-aminohippurate, the Oat1 substrate), which was confirmed by transient over-expression of OAT1 in HEK-293T cells. Additionally, hyperoside upregulated the mRNA expression of Oat1 upstream regulators hepatocyte nuclear factor-1α (HNF-1α) and pregnane X receptor (PXR).These findings indicated hyperoside could protect against cisplatin-induced AKI by promoting indoxyl sulphate excretion through regulating the expression and function of Oat1, suggesting hyperoside may offer a potential tactic for cisplatin-induced AKI treatment.
Collapse
Affiliation(s)
- Wenjing Yuan
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Shanshan Kou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Ying Ma
- Foreign Language Teaching Department, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Yusi Qian
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Xinyu Li
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Yuanyuan Chai
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Zhenzhou Jiang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Lixin Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Xin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| |
Collapse
|
4
|
ALRashdi B, Mohamed R, Mohamed A, Samoul F, Mohamed M, Moussa M, Alrashidi S, Dawod B, Habotta O, Abdel Moneim A, Ramadan S. Therapeutic activity of green synthesized selenium nanoparticles from turmeric against cisplatin-induced oxido-inflammatory stress and cell death in mice kidney. Biosci Rep 2023; 43:BSR20231130. [PMID: 37902021 PMCID: PMC10643052 DOI: 10.1042/bsr20231130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/09/2023] [Accepted: 10/27/2023] [Indexed: 10/31/2023] Open
Abstract
Cisplatin (CDDP) is a commonly prescribed chemotherapeutic agent; however, its associated nephrotoxicity limits its clinical efficacy and sometimes requires discontinuation of its use. The existing study was designed to explore the reno-therapeutic efficacy of turmeric (Tur) alone or conjugated with selenium nanoparticles (Tur-SeNPs) against CDDP-mediated renal impairment in mice and the mechanisms underlying this effect. Mice were orally treated with Tur extract (200 mg/kg) or Tur-SeNPs (0.5 mg/kg) for 7 days after administration of a single dose of CDDP (5 mg/kg, i.p.). N-acetyl cysteine NAC (100 mg/kg) was used as a standard antioxidant compound. The results revealed that Tur-SeNPs counteracted CDDP-mediated serious renal effects in treated mice. Compared with the controls, Tur or Tur-SeNPs therapy remarkably decreased the kidney index along with the serum levels of urea, creatinine, Kim-1, and NGAL of the CDDP-injected mice. Furthermore, Tur-SeNPs ameliorated the renal oxidant status of CDDP group demonstrated by decreased MDA and NO levels along with elevated levels of SOD, CAT, GPx, GR, GSH, and gene expression levels of HO-1. Noteworthy, lessening of renal inflammation was exerted by Tur-SeNPs via lessening of IL-6 and TNF-α besides down-regulation of NF-κB gene expression in mouse kidneys. Tur-SeNPs treatment also restored the renal histological features attained by CDDP challenge and hindered renal apoptosis through decreasing the Bax levels and increasing Bcl-2 levels. Altogether, these outcomes suggest that the administration of Tur conjugated with SeNPs is effective neoadjuvant chemotherapy to guard against the renal adverse effects that are associated with CDDP therapy.
Collapse
Affiliation(s)
- Barakat M. ALRashdi
- Department of Biology, College of Science, Jouf University, Sakaka 72388, Saudi Arabia
| | - Roaya A. Mohamed
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Amal H. Mohamed
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Feryal A. Samoul
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mazen I. Mohamed
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mohsen M. Moussa
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Saad M. Alrashidi
- Consultant Radiation Oncology, Comprehensive Cancer Centre, King Fahad Medical City and College of medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Bassel Dawod
- McMaster Children’s Hospital, Faculty of Health Sciences, Hamilton, Ontario, Canada
- Department of Biology, College of Science, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
- Department of Zoology, Faculty of Science, Fayoum University, Fayoum, Egypt
| | - Ola A. Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed E. Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Shimaa S. Ramadan
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
5
|
Revankar AG, Bagewadi ZK, Shaikh IA, Mannasaheb BA, Ghoneim MM, Khan AA, Asdaq SMB. In-vitro and computational analysis of Urolithin-A for anti-inflammatory activity on Cyclooxygenase 2 (COX-2). Saudi J Biol Sci 2023; 30:103804. [PMID: 37727526 PMCID: PMC10505678 DOI: 10.1016/j.sjbs.2023.103804] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
Cyclooxygenase 2 (COX-2) participates in the inflammation process by converting arachidonic acid into prostaglandin G2 which increases inflammation, pain and fever. COX-2 has an active site and a heme pocket and blocking these sites stops the inflammation. Urolithin A is metabolite of ellagitannin produced from humans and animals gut microbes. In the current study, Urolithin A showed good pharmacokinetic properties. Molecular docking of the complex of Urolithin A and COX-2 revealed the ligand affinity of -7.97 kcal/mol with the ligand binding sites at TYR355, PHE518, ILE517 and GLN192 with the 4-H bonds at a distance of 2.8 Å, 2.3 Å, 2.5 Å and 1.9 Å. The RMSD plot for Urolithin A and COX-2 complex was observed to be constant throughout the duration of dynamics. A total of 3 pair of hydrogen bonds was largely observed on average of 3 simulation positions for dynamics duration of 500 ns. The MMPBSA analysis showed that active site amino acids had a binding energy of -22.0368 kJ/mol indicating that throughout the simulation the protein of target was bounded by Urolithin A. In-silico results were validated by biological assays. Urolithin A strongly revealed to exhibit anti-inflammatory effect on COX-2 with an IC50 value of 44.04 µg/mL. The anti-inflammatory capability was also depicted through reduction of protein denaturation that showed 37.6 ± 0.1 % and 43.2 ± 0.07 % reduction of protein denaturation for BSA and egg albumin respectively at 500 µg/mL. The present study, suggests Urolithin A to be an effective anti-inflammatory compound for therapeutic use.
Collapse
Affiliation(s)
- Archana G. Revankar
- Department of Biotechnology, KLE Technological University, Hubballi, Karnataka 580031, India
| | - Zabin K. Bagewadi
- Department of Biotechnology, KLE Technological University, Hubballi, Karnataka 580031, India
| | - Ibrahim Ahmed Shaikh
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia
| | | | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia
| | - Aejaz Abdullatif Khan
- Department of General Science, Ibn Sina National College for Medical Studies, Jeddah 21418, Saudi Arabia
| | | |
Collapse
|
6
|
Wojciechowska O, Kujawska M. Urolithin A in Health and Diseases: Prospects for Parkinson's Disease Management. Antioxidants (Basel) 2023; 12:1479. [PMID: 37508017 PMCID: PMC10376282 DOI: 10.3390/antiox12071479] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disorder characterized by a complex pathophysiology and a range of symptoms. The prevalence increases with age, putting the ageing population at risk. Disease management includes the improvement of symptoms, the comfort of the patient's life, and palliative care. As there is currently no cure, growing evidence points towards the beneficial role of polyphenols on neurodegeneration. Numerous studies indicate the health benefits of the family of urolithins, especially urolithin A (UA). UA is a bacterial metabolite produced by dietary ellagitannins and ellagic acid. An expanding body of literature explores the involvement of the compound in mitochondrial health, and its anti-inflammatory, anti-oxidant, and anti-apoptotic properties. The review organizes the existing knowledge on the role of UA in health and diseases, emphasizing neurodegenerative diseases, especially PD. We gathered data on the potential neuroprotective effect in in vivo and in vitro models. We discussed the possible mechanisms of action of the compound and related health benefits to give a broader perspective of potential applications of UA in neuroprotective strategies. Moreover, we projected the future directions of applying UA in PD management.
Collapse
Affiliation(s)
- Olga Wojciechowska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland
| |
Collapse
|
7
|
Alkhalaf M, Mohamed NA, El-Toukhy SE. Prophylactic consequences of sodium salicylate nanoparticles in cisplatin-mediated hepatotoxicity. Sci Rep 2023; 13:10045. [PMID: 37344526 DOI: 10.1038/s41598-023-35916-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/25/2023] [Indexed: 06/23/2023] Open
Abstract
Unintended side effects linked to the antineoplastic drug cisplatin are a major drawback in its clinical application. The underlying source of these side effects include the generation of reactive oxygen species which are toxic and damaging to tissues and organs. In the present study the anti-inflammatory and antioxidant potential of sodium salicylate was assessed against cisplatin-induced hepatotoxicity in albino rats. Sodium salicylate was used as a model drug and loading into hollow structured porous silica using ultrasound-assisted sol-gel method to produce a nanoemulsion. Transmission Electron Microscopy and Dynamic Light scattering analysis were employed to assess the structural properties and stability of this model. Liver function was assessed by measuring biomarkers including ALT, AST & GGT and oxidant/antioxidant markers including MDA, NO, PON, GSH, MCP1 & AVP in serum or liver tissue. Additionally, blood leukocyte DNA damage was evaluated. Cisplatin significantly altered the normal levels of all biomarkers confirming its hepatotoxic effects. In contrast, treatment with sodium salicylate-loaded silica nanoemulsion significantly restored the levels of these markers. The finding suggests the protective effects of this model drug in preventing cisplatin-induced hepatotoxicity, and therefore may have implications in attenuating cisplatin-induced hepatotoxicity.
Collapse
Affiliation(s)
- Maha Alkhalaf
- Biochemistry Department, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia.
| | - Nadia A Mohamed
- Medical Biochemistry Department, National Research Centre, Cairo, Egypt
| | | |
Collapse
|
8
|
Jakobek L, Blesso C. Beneficial effects of phenolic compounds: native phenolic compounds vs metabolites and catabolites. Crit Rev Food Sci Nutr 2023; 64:9113-9131. [PMID: 37140183 DOI: 10.1080/10408398.2023.2208218] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In the human body, the positive effects of phenolic compounds are increasingly observed through their presence in tissues and organs in their native form or in the form of metabolites or catabolites formed during digestion, microbial metabolism, and host biotransformation. The full extent of these effects is still unclear. The aim of this paper is to review the current knowledge of beneficial effects of native phenolic compounds or their metabolites and catabolites focusing on their role in the health of the digestive system, including disorders of the gastrointestinal and urinary tracts and liver. Studies are mostly connecting beneficial effects in the gastrointestinal and urinary tract to the whole food rich in phenolics, or to the amount of phenolic compounds/antioxidants in food. Indeed, the bioactivity of parent phenolic compounds should not be ignored due to their presence in the digestive tract, and the impact on the gut microbiota. However, the influence of their metabolites and catabolites might be more important for the liver and urinary tract. Distinguishing between the effects of parent phenolics vs metabolites and catabolites at the site of action are important for novel areas of food industry, nutrition and medicine.
Collapse
Affiliation(s)
- Lidija Jakobek
- Faculty of Food Technology Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Department of Statistics and Data Science, Yale University, New Haven, Connecticut, USA
| | - Christopher Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
9
|
Jin XH, Fang JQ, Wang JG, Xu B, Wang X, Liu SH, Chen F, Liu JJ. PCL NGCs integrated with urolithin-A-loaded hydrogels for nerve regeneration. J Mater Chem B 2022; 10:8771-8784. [PMID: 36196763 DOI: 10.1039/d2tb01624a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammation and oxidative stress are among the leading causes of poor prognosis after peripheral nerve injury (PNI). Urolithin-A (UA), an intermediate product produced by the catabolism of ellagitannins in the gastrointestinal tract, has anti-inflammatory, antioxidant, and immunomodulatory properties for inflammation, oxidative damage, and aging-related diseases. Hence, we prepared UA-loaded hydrogels and embedded them in the lumen of PCL nerve guide conduits (NGCs). The hydrogels continuously released appropriate doses of UA into the microenvironment. Based on in vitro studies, UA facilitates cell proliferation and reduces oxidative damage. Besides, the experimental evaluation revealed good biocompatibility of the materials involved. We implanted NGCs into rat models to bridge the sciatic nerve defects in an in vivo study. The sciatic functional index of the PCL/collagen/UA group was comparable to that of the autograft group. Additionally, the consequences of electrophysiological, gastrocnemius muscle and nerve histology assessment of the PCL/collagen/UA group were better than those in the PCL and PCL/collagen groups and close to those in the autograft group. In this study, UA sustained release via the PCL/collagen/UA NGC was found to be an effective alternative treatment for PNI, validating our hypothesis that UA could promote regeneration of nerve tissue.
Collapse
Affiliation(s)
- Xue-Han Jin
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| | - Jia-Qi Fang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| | - Jian-Guang Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| | - Bo Xu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| | - Xu Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Shu-Hao Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| | - Feng Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| | - Jun-Jian Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai, 200072, P. R. China.
| |
Collapse
|
10
|
Chafik SG, Michel HE, El-Demerdash E. The Cannabinoid-2 receptor agonist, 1-phenylisatin, protects against cisplatin-induced nephrotoxicity in mice. Life Sci 2022; 308:120928. [PMID: 36058263 DOI: 10.1016/j.lfs.2022.120928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/15/2022]
Abstract
AIM The present study investigated the potential protective effect of a selective Cannabinoid-2 (CB2) receptor agonist, 1-phenylisatin, in acute nephrotoxicity induced by cisplatin. MATERIALS AND METHODS Animals were arranged into 5 groups. Group I; normal saline, group II; 1-phenylisatin for 7 days, group III: received a single injection of cisplatin (20 mg/kg, i.p.) on day 5, group IV: 1-phenylisatin for 7 days and cisplatin on day 5 and group V: AM630, CB2 antagonist, 15 min before 1-phenylisatin for 7 days and a single injection of cisplatin on day 5. Mice were sacrificed 72 h after cisplatin injection. Kidneys were isolated for histopathological and biochemical analyses. Nephrotoxicity parameters including serum creatinine and urea were assessed as well as histopathological examination was done. Also, Oxidative stress markers; MDA and GSH, inflammatory markers; TNF-α, NF-κB (p65), MCP-1, MIP-2, and ICAM-1, along with apoptotic markers, Bax, Bcl2, and caspase-3 were studied. Further, CB2 receptor expression was investigated. KEY FINDINGS Cisplatin injection increased serum creatinine and urea levels, and increased lipid peroxidation, decreased glutathione level and increased the renal expression of pro-inflammatory markers, TNF-α, NF-κB, MCP-1, MIP-2, and ICAM-1, along with increased apoptotic markers and significantly reduced the expression of the anti-apoptotic Bcl2. Pretreatment with 1-phenylisatin significantly counteracted these effects. The CB2 receptor antagonist; AM630, increased the renal expression of caspase-3 and Bax whereas Bcl2 expression decreased. SIGNIFICANCE 1-Phenylisatin protected against cisplatin-induced nephrotoxicity owing to its anti-apoptotic, anti-inflammatory, and antioxidant effects. These actions were mostly mediated through CB2 receptor.
Collapse
Affiliation(s)
| | - Haidy E Michel
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
11
|
García‐Villalba R, Giménez‐Bastida JA, Cortés‐Martín A, Ávila‐Gálvez MÁ, Tomás‐Barberán FA, Selma MV, Espín JC, González‐Sarrías A. Urolithins: a Comprehensive Update on their Metabolism, Bioactivity, and Associated Gut Microbiota. Mol Nutr Food Res 2022; 66:e2101019. [PMID: 35118817 PMCID: PMC9787965 DOI: 10.1002/mnfr.202101019] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/22/2022] [Indexed: 12/30/2022]
Abstract
Urolithins, metabolites produced by the gut microbiota from the polyphenols ellagitannins and ellagic acid, are discovered by the research group in humans almost 20 years ago. Pioneering research suggests urolithins as pleiotropic bioactive contributors to explain the health benefits after consuming ellagitannin-rich sources (pomegranates, walnuts, strawberries, etc.). Here, this study comprehensively updates the knowledge on urolithins, emphasizing the review of the literature published during the last 5 years. To date, 13 urolithins and their corresponding conjugated metabolites (glucuronides, sulfates, etc.) have been described and, depending on the urolithin, detected in different human fluids and tissues (urine, blood, feces, breastmilk, prostate, colon, and breast tissues). There has been a substantial advance in the research on microorganisms involved in urolithin production, along with the compositional and functional characterization of the gut microbiota associated with urolithins metabolism that gives rise to the so-called urolithin metabotypes (UM-A, UM-B, and UM-0), relevant in human health. The design of in vitro studies using physiologically relevant assay conditions (molecular forms and concentrations) is still a pending subject, making some reported urolithin activities questionable. In contrast, remarkable progress has been made in the research on the safety, bioactivity, and associated mechanisms of urolithin A, including the first human interventions.
Collapse
Affiliation(s)
- Rocío García‐Villalba
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - Juan Antonio Giménez‐Bastida
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - Adrián Cortés‐Martín
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - María Ángeles Ávila‐Gálvez
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - Francisco A. Tomás‐Barberán
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - María Victoria Selma
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - Juan Carlos Espín
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| | - Antonio González‐Sarrías
- Laboratory of Food & HealthResearch Group on QualitySafety and Bioactivity of Plant FoodsCEBAS‐CSICMurciaCampus de EspinardoSpain
| |
Collapse
|
12
|
Xu Z, Li S, Li K, Wang X, Li X, An M, Yu X, Long X, Zhong R, Liu Q, Wang X, Yang Y, Tian N. Urolithin A ameliorates diabetic retinopathy via activation of the Nrf2/HO-1 pathway. Endocr J 2022; 69:971-982. [PMID: 35321989 DOI: 10.1507/endocrj.ej21-0490] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy (DR) is a progressive microvascular complication of diabetes mellitus and is characterised by excessive inflammation and oxidative stress. Urolithin A (UA), a major metabolite of ellagic acid, exerts anti-inflammatory and antioxidant functions in various human diseases. This study, for the first time, uncovered the role of UA in DR pathogenesis. Streptozotocin-induced diabetic rats were used to determine the effects of UA on blood glucose levels, retinal structures, inflammation, and oxidative stress. High glucose (HG)-induced human retinal endothelial cells (HRECs) were used to elucidate the anti-inflammatory and antioxidant mechanisms of UA in DR in vitro. The in vivo experiments demonstrated that UA injection reduced blood glucose levels, decreased albumin and vascular endothelial growth factor concentrations, and ameliorated the injured retinal structures caused by DR. UA administration also inhibited inflammation and oxidative damage in the retinal tissues of diabetic rats. Similar anti-inflammatory and antioxidant effects of UA were observed in HRECs induced by HG. Furthermore, we found that UA elevated the levels of nuclear Nrf2 and HO-1 both in vivo and in vitro. Nrf2 silencing reversed the inhibitory effects of UA on inflammation and oxidative stress during DR progression. Together, our findings indicate that UA can ameliorate DR by repressing inflammation and oxidative stress via the Nrf2/HO-1 pathway, which suggests that UA could be an effective drug for clinical DR treatment.
Collapse
Affiliation(s)
- Zepeng Xu
- Department of Ophthalmology, Wuyi Hospital of Traditional Chinese Medicine, Guangdong Province, 529000, China
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Songtao Li
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Kunmeng Li
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Xiaoyu Wang
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Xiaojie Li
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Meixia An
- Department of Ophthalmology, The Third Affiliated Hospital of Southern Medical University, Guangdong Province, 510630, China
| | - Xiaoyi Yu
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Xinguang Long
- Department of Ophthalmology, Wuyi Hospital of Traditional Chinese Medicine, Guangdong Province, 529000, China
| | - Ruiying Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Qiuhong Liu
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Xiaochuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Yan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| | - Ni Tian
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province, 510504, China
| |
Collapse
|
13
|
The Therapeutic Relevance of Urolithins, Intestinal Metabolites of Ellagitannin-Rich Food: A Systematic Review of In Vivo Studies. Nutrients 2022; 14:nu14173494. [PMID: 36079752 PMCID: PMC9460125 DOI: 10.3390/nu14173494] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 12/09/2022] Open
Abstract
The therapeutic effects of food rich in ellagitannins have been established to stem from its microbial metabolite, urolithin. Over the past decade, there has been a growing trend in urolithin research pertaining to its pharmacological properties. The purpose of this systematic review is to collate and synthesise all available data on urolithin’s therapeutic ability, to highlight its potential as a pharmaceutical agent, and prospective direction on future research. Methods: This systematic review was written based on the PRISMA guideline and was conducted across Ovid via Embase, Ovid MEDLINE, Cochrane Central Register for Controlled Trials, and Web of Science Core Collection. Results: A total of 41 animal studies were included in this systematic review based on the appropriate keyword. The included studies highlighted the neuroprotective, anti-metabolic disorder activity, nephroprotective, myocardial protective, anti-inflammatory, and musculoskeletal protection of urolithin A, B, and its synthetic analogue methylated urolithin A. The Sirt1, AMPK, and PI3K/AKT/mTOR signalling pathways were reported to be involved in the initiation of autophagy and mitochondrial biogenesis by urolithin A. Conclusions: This review methodically discusses the therapeutic prospects of urolithins and provides scientific justification for the potential development of urolithin A as a potent natural mitophagy inducer for anti-ageing purposes.
Collapse
|
14
|
Gandhi GR, Antony PJ, Ceasar SA, Vasconcelos ABS, Montalvão MM, Farias de Franca MN, Resende ADS, Sharanya CS, Liu Y, Hariharan G, Gan RY. Health functions and related molecular mechanisms of ellagitannin-derived urolithins. Crit Rev Food Sci Nutr 2022; 64:280-310. [PMID: 35959701 DOI: 10.1080/10408398.2022.2106179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ellagitannins are vital bioactive polyphenols that are widely distributed in a variety of plant-based foods. The main metabolites of ellagitannins are urolithins, and current research suggests that urolithins provide a variety of health benefits. This review focused on the role of the gut bacteria in the conversion of ellagitannins to urolithins. Based on the results of in vitro and in vivo studies, the health benefits of urolithins, including antioxidant, anti-inflammatory, anti-cancer, anti-obesity, anti-diabetic, anti-aging, cardiovascular protective, neuroprotective, kidney protective, and muscle mass protective effects, were thoroughly outlined, with a focus on their associated molecular mechanisms. Finally, we briefly commented on urolithins' safety. Overall, urolithins' diverse health benefits indicate the potential utilization of ellagitannins and urolithins in the creation of functional foods and nutraceuticals to treat and prevent some chronic diseases.
Collapse
Affiliation(s)
- Gopalsamy Rajiv Gandhi
- Department of Biosciences, Rajagiri College of Social Sciences, Kalamaserry, Kochi, India
| | | | | | - Alan Bruno Silva Vasconcelos
- Postgraduate Program of Physiological Sciences (PROCFIS), Federal University of Sergipe (UFS), São Cristóvão, Sergipe, Brazil
| | - Monalisa Martins Montalvão
- Postgraduate Program of Physiological Sciences (PROCFIS), Federal University of Sergipe (UFS), São Cristóvão, Sergipe, Brazil
| | - Mariana Nobre Farias de Franca
- Postgraduate Program of Health Sciences (PPGCS), Federal University of Sergipe (UFS), Campus Prof. João Cardoso Nascimento, Aracaju, CEP, Sergipe, Brazil
| | - Ayane de Sá Resende
- Postgraduate Program of Health Sciences (PPGCS), Federal University of Sergipe (UFS), Campus Prof. João Cardoso Nascimento, Aracaju, CEP, Sergipe, Brazil
| | | | - Yi Liu
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences (CAAS), Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Govindasamy Hariharan
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous) affiliated to the Bharathidasan University, Tiruchirapalli, India
| | - Ren-You Gan
- Nepal Jesuit Society, St. Xavier's College, Jawalakhel, Lalitpur Dt. Kathmandu, Nepal
| |
Collapse
|
15
|
Ameliorative Effects of Gut Microbial Metabolite Urolithin A on Pancreatic Diseases. Nutrients 2022; 14:nu14122549. [PMID: 35745279 PMCID: PMC9229509 DOI: 10.3390/nu14122549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 12/11/2022] Open
Abstract
Urolithin A (Uro A) is a dietary metabolite of the intestinal microbiota following the ingestion of plant-based food ingredients ellagitannins and ellagic acid in mammals. Accumulating studies have reported its multiple potential health benefits in a broad range of diseases, including cardiovascular disease, cancer, cognitive impairment, and diabetes. In particular, Uro A is safe via direct oral administration and is non-genotoxic. The pancreas plays a central role in regulating energy consumption and metabolism by secreting digestive enzymes and hormones. Numerous pathophysiological factors, such as inflammation, deficits of mitophagy, and endoplasmic reticulum stress, can negatively affect the pancreas, leading to pancreatic diseases, including pancreatitis, pancreatic cancer, and diabetes mellitus. Recent studies showed that Uro A activates autophagy and inhibits endoplasmic reticulum stress in the pancreas, thus decreasing oxidative stress, inflammation, and apoptosis. In this review, we summarize the knowledge of Uro A metabolism and biological activity in the gut, as well as the pathological features and mechanisms of common pancreatic diseases. Importantly, we focus on the potential activities of Uro A and the underlying mechanisms in ameliorating various pancreatic diseases via inhibiting inflammatory signaling pathways, activating autophagy, maintaining the mitochondrial function, and improving the immune microenvironment. It might present a novel nutritional strategy for the intervention and prevention of pancreatic diseases.
Collapse
|
16
|
Mo Y, Ma J, Gao W, Zhang L, Li J, Li J, Zang J. Pomegranate Peel as a Source of Bioactive Compounds: A Mini Review on Their Physiological Functions. Front Nutr 2022; 9:887113. [PMID: 35757262 PMCID: PMC9218663 DOI: 10.3389/fnut.2022.887113] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
The production and consumption of pomegranates have always been increasing owing to their taste and nutrition. However, during fruit processing, a large number of by-products are produced, such as peels and seeds, which can lead to environmental pollution problems if not handled properly. The pomegranate peel takes up about 26-30% of the total weight, while it contains abundant bioactive substances. This paper carries out a mini review of the characterization and physiological functions of key bioactive compounds in pomegranate peel, comprehensively assessing their effects on human health. The overview summarizes the main phenolic substances in pomegranate peel, including tannins, flavonoids, and phenolic acids. Dietary fiber and other bioactive substances such as alkaloids, minerals, and vitamins are also mentioned. These components act as antioxidants by improving oxidative biomarkers and scavenging or neutralizing reactive oxygen species, further contributing to their extensive functions like anti-inflammatory, anti-cancer, antibacterial, and cardiovascular protection. Overall, it is envisaged that through the deeper understanding of bioactive compounds in pomegranate peel, the waste sources can be better reused for physiological applications.
Collapse
Affiliation(s)
- Yaxian Mo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jiaqi Ma
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Wentao Gao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Lei Zhang
- College of Forestry and Landscape Architecture, Xinjiang Agricultural University, Ürümqi, China
| | - Jiangui Li
- College of Forestry and Landscape Architecture, Xinjiang Agricultural University, Ürümqi, China
| | - Jingming Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jiachen Zang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
17
|
Wang X, Jiang L, Liu Q. miR-18a-5p derived from mesenchymal stem cells-extracellular vesicles inhibits ovarian cancer cell proliferation, migration, invasion, and chemotherapy resistance. J Transl Med 2022; 20:258. [PMID: 35672774 PMCID: PMC9172103 DOI: 10.1186/s12967-022-03422-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/02/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Ovarian cancer (OC) is a major threat to women's health. Mesenchymal stem cells (MSCs) are key regulators in cellular communication by secreting extracellular vesicles (EVs) that are involved in OC. This study probed into the mechanism of human MSCs derived-EVs (hMSC-EVs) in regulating OC cell growth and chemotherapy resistance. METHODS hMSCs and EVs were isolated and identified. After adding EVs, the uptake of EVs by OC CAOV3/ES2 cells (for in vitro studies), and cell proliferation, migration, and invasion were detected. Downregulated miRNAs in hMSC-EVs were screened and miR-18a-5p expression in OC patients was detected. The prognosis of OC patients was analyzed. Binding sites of miR-18a-5p and NACC1 were predicted and validated. NACC1 expression in OC tissues was measured by RT-qPCR, and its correlation with miR-18a-5p was analyzed by Pearson method. AKT/mTOR pathway activation was assessed by WB. The cisplatin sensitivity of EVs-treated CAOV3 cells was evaluated via MTT assay and tested by tumor formation assay in nude mice. RESULTS hMSC-EVs suppressed OC cell proliferation, migration, and invasion. miR-18a-5p was downregulated in OC and miR-18a-5p low expression was associated with a poor prognosis. EV-encapsulated miR-18a-5p targeted NACC1. NACC1 was upregulated in OC tissues. miR-18a-5p knockdown and NACC1 overexpression both annulled the inhibition of hMSC-EVs on OC cell growth. AKT and mTOR were elevated in OC and NACC1 activated the AKT/mTOR pathway in OC cells. hMSC-EVs promoted cisplatin sensitivity of OC cells by carrying miR-18a-5p. CONCLUSION hMSC-EVs-derived miR-18a-5p inhibits OC cell proliferation, migration, invasion, and chemotherapy resistance.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Lili Jiang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Qifang Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
18
|
Singh A, D'Amico D, Andreux PA, Fouassier AM, Blanco-Bose W, Evans M, Aebischer P, Auwerx J, Rinsch C. Urolithin A improves muscle strength, exercise performance, and biomarkers of mitochondrial health in a randomized trial in middle-aged adults. Cell Rep Med 2022; 3:100633. [PMID: 35584623 PMCID: PMC9133463 DOI: 10.1016/j.xcrm.2022.100633] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 02/24/2022] [Accepted: 04/21/2022] [Indexed: 12/14/2022]
Abstract
Targeting mitophagy to activate the recycling of faulty mitochondria during aging is a strategy to mitigate muscle decline. We present results from a randomized, placebo-controlled trial in middle-aged adults where we administer a postbiotic compound Urolithin A (Mitopure), a known mitophagy activator, at two doses for 4 months (NCT03464500). The data show significant improvements in muscle strength (∼12%) with intake of Urolithin A. We observe clinically meaningful improvements with Urolithin A on aerobic endurance (peak oxygen oxygen consumption [VO2]) and physical performance (6 min walk test) but do not notice a significant improvement on peak power output (primary endpoint). Levels of plasma acylcarnitines and C-reactive proteins are significantly lower with Urolithin A, indicating higher mitochondrial efficiency and reduced inflammation. We also examine expression of proteins linked to mitophagy and mitochondrial metabolism in skeletal muscle and find a significant increase with Urolithin A administration. This study highlights the benefit of Urolithin A to improve muscle performance. Oral supplementation with Urolithin A increases muscle strength High dose of Urolithin A positively impacts exercise-performance measures An increase in mitophagy proteins in human skeletal muscle observed in parallel Supplementation is safe and increases circulating levels of Urolithin A
Collapse
Affiliation(s)
- Anurag Singh
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland.
| | - Davide D'Amico
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland
| | - Pénélope A Andreux
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland
| | | | | | - Mal Evans
- KGK Science, 255 Queens Avenue #1440, London, ON N6A 5R8, Canada
| | - Patrick Aebischer
- Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland
| |
Collapse
|
19
|
Wang T, Zhang C, Li H, Zhou R, Ye X, Yang Y, He K. The underlying rationality of Chinese medicine herb pair Coptis chinensis and Dolomiaea souliei: From the perspective of metabolomics and intestinal function. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115065. [PMID: 35122977 DOI: 10.1016/j.jep.2022.115065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The combination of Coptis chinensis (RC) and Dolomiaea souliei (VR) has long been used as a classic herb pair for the treatment of gastrointestinal diseases, but the underlying mechanisms remain unknown. MATERIALS AND METHODS In this study, the rationality of evidence-based RC and VR combination was explored from the perspective of metabolism, gut microbiota and gastrointestinal function. RESULTS After 5 weeks treatment, VR extracts (700 mg/kg) and RC alkaloids (800 mg/kg) showed no toxic effect on mice. However, RC administration significantly decreased the body weight of mice. Gastric emptying, gastrointestinal motility function and the absorption of FITC dextran were retarded in the mice of RC group, taking RC along with low dose VR (RC-VRL) and high dose VR (RC-VRH) reversed the impaired gastrointestinal function caused by RC. RC administration significantly increased villus height/crypt depth value. Notably, VR administration increased the number of crypts in mice ileum and reduced villus height/crypt depth value in VR and RC combination group. RC treatment significantly increased the expression of occludin compared to NC group; RC-VRL treatment reversed this tendency. While, VR administration increased ZO1 expression by 99.4% compared to NC mice. As for gut microbiota, RC gavage decreased the gut microbiota diversity, but gut microbiota in VR group was similar to NC group, and VR and RC combination increased gut microbiota diversity. RC administration obviously increased the proportion of Akkermansia muciniphila, Bacteroides thetaiotaomicron, Parabacteroides distasonis, and Escherichia coli, compared to NC mice. VR treatment increased the richness of Bacteroides thetaiotaomicron, Parabacteroides distasonis. RC-VRL and RC-VRH treatment dose-dependently increased the richness of Rikenellaceae RC9, Lactobacillus, and decreased the abundance of Psychrobacter, Bacteroides and Ruminococcus in mice. Serum metabolomic analysis revealed that RC gavage significantly down regulated 76 metabolites and up regulated 31 metabolites. VR treatment significantly down regulated 30 metabolites and up regulated 12 metabolites. Weight loss caused by RC may attribute to the elevated methylxanthine level in mice. The potential adverse effects caused by high dose RC intake may partially alleviate by high serum contents of adenosine, inosine and urolithin A resulted from VR coadministration. CONCLUSION VR may alleviate RC caused "fluid retention" via normalizing gastrointestinal function, gut microbiota and modulating the perturbed metabolism.
Collapse
Affiliation(s)
- Ting Wang
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, 418000, Hunan, China
| | - Chongyang Zhang
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, 418000, Hunan, China
| | - Hui Li
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, 418000, Hunan, China
| | - Rui Zhou
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, 418000, Hunan, China
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Yong Yang
- School of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China
| | - Kai He
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, 418000, Hunan, China.
| |
Collapse
|
20
|
Wei W, Peng C, Gu R, Yan X, Ye J, Xu Z, Sheng X, Huang G, Guo Y. Urolithin A attenuates RANKL-induced osteoclastogenesis by co-regulating the p38 MAPK and Nrf2 signaling pathway. Eur J Pharmacol 2022; 921:174865. [PMID: 35231470 DOI: 10.1016/j.ejphar.2022.174865] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/17/2022] [Accepted: 02/23/2022] [Indexed: 12/30/2022]
Abstract
As a critical regulator of bone resorption. osteoclastogenesis is closely associated with osteoporosis (OP) and commonly induced by receptor activator of nuclear factor-κB ligand (RANKL), suggesting that suppression of inflammation may improve OP. Urolithin A (UroA), an active metabolite of ellagic acid, is known to exert anti-inflammatory and antioxidative effects. However, whether UroA attenuates osteoclastogenesis remains unclear. Using a lipopolysaccharide (LPS)-induced bone loss model, we evaluated the effects of UroA on inflammatory osteoclastogenesis in mice and explored the potential mechanism from RANKL-related signaling pathway. UroA significantly improved LPS-induced bone loss and rescued the imbalance in bone microarchitecture parameters. Hematoxylin&eosin (H&E) and tartrate resistant acid phosphatase (TRAP) staining of femurs showed that UroA suppressed LPS-induced osteoclastogenesis accompanied by the activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling. In RANKL-triggered mouse bone marrow-derived macrophages (BMDMs), UroA inhibited the formation of osteoclasts and Fibrous actin rings (F-actin rings), and decreased TRAP activity. Moreover, UroA significantly decreased mRNA and protein expression of major inflammatory cytokines in LPS-challenged RAW264.7 cells by decreasing the phosphorylation of NF-κB p65, c-Jun N-terminal kinase (JNK), extracellular signal regulated kinase1/2 (Erk1/2), and p38. Furthermore, UroA may activate the Nrf2 signaling pathway by increasing mRNA and protein expression of antioxidant proteins. We conclude that UroA attenuated RANKL-induced osteoclastogenesis by suppressing the p38 mitogen-activated protein kinase (MAPK) pathway and inducing Nrf2 nuclear translocation. Thus, supplementation with UroA may help alleviate inflammation-induced bone loss and bone resorption.
Collapse
Affiliation(s)
- Wei Wei
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chenjian Peng
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China
| | - Renjun Gu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiwu Yan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiapeng Ye
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhuicheng Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xianjie Sheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guicheng Huang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yang Guo
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
21
|
Zhang M, Cui S, Mao B, Zhang Q, Zhao J, Zhang H, Tang X, Chen W. Ellagic acid and intestinal microflora metabolite urolithin A: A review on its sources, metabolic distribution, health benefits, and biotransformation. Crit Rev Food Sci Nutr 2022; 63:6900-6922. [PMID: 35142569 DOI: 10.1080/10408398.2022.2036693] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Foods rich in ellagic tannins are first hydrolyzed into ellagic acid in the stomach and small intestine, and then converted into urolithins with high bioavailability by the intestinal flora. Urolithin has beneficially biological effects, it can induce adipocyte browning, improve cholesterol metabolism, inhibit graft tumor growth, relieve inflammation, and downregulate neuronal amyloid protein formation via the β3-AR/PKA/p38MAPK, ERK/AMPKα/SREBP1, PI3K/AKT/mTOR signaling pathways, and TLR4, AHR receptors. But differences have been reported in urolithin production capacity among different individuals. Thus, it is of great significance to explore the biological functions of urolithin, screen the strains responsible for biotransformation of urolithin, and explore the corresponding functional genes. Tannin acyl hydrolase can hydrolyze tannins into ellagic acid, and the genera Gordonibacter and Ellagibacter can metabolize ellagic acid into urolithins. Therefore, application of "single bacterium", "single bacterium + enzyme", and "microflora" can achieve biotransformation of urolithin A. In this review, the source and metabolic pathway of ellagic tannins, and the mechanisms of the biological function of a metabolite, urolithin A, are discussed. The current strategies of biotransformation to obtain urolithin A are expounded to provide ideas for further studies on the relationship between urolithin and human health.
Collapse
Affiliation(s)
- Mengwei Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R China
- Wuxi Translational Medicine Research Center, Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, P. R China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R China
| |
Collapse
|
22
|
Vini R, Azeez JM, Remadevi V, Susmi TR, Ayswarya RS, Sujatha AS, Muraleedharan P, Lathika LM, Sreeharshan S. Urolithins: The Colon Microbiota Metabolites as Endocrine Modulators: Prospects and Perspectives. Front Nutr 2022; 8:800990. [PMID: 35187021 PMCID: PMC8849129 DOI: 10.3389/fnut.2021.800990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/10/2021] [Indexed: 12/19/2022] Open
Abstract
Selective estrogen receptor modulators (SERMs) have been used in hormone related disorders, and their role in clinical medicine is evolving. Tamoxifen and raloxifen are the most commonly used synthetic SERMs, and their long-term use are known to create side effects. Hence, efforts have been directed to identify molecules which could retain the beneficial effects of estrogen, at the same time produce minimal side effects. Urolithins, the products of colon microbiota from ellagitannin rich foodstuff, have immense health benefits and have been demonstrated to bind to estrogen receptors. This class of compounds holds promise as therapeutic and nutritional supplement in cardiovascular disorders, osteoporosis, muscle health, neurological disorders, and cancers of breast, endometrium, and prostate, or, in essence, most of the hormone/endocrine-dependent diseases. One of our findings from the past decade of research on SERMs and estrogen modulators, showed that pomegranate, one of the indirect but major sources of urolithins, can act as SERM. The prospect of urolithins to act as agonist, antagonist, or SERM will depend on its structure; the estrogen receptor conformational change, availability and abundance of co-activators/co-repressors in the target tissues, and also the presence of other estrogen receptor ligands. Given that, urolithins need to be carefully studied for its SERM activity considering the pleotropic action of estrogen receptors and its numerous roles in physiological systems. In this review, we unveil the possibility of urolithins as a potent SERM, which we are currently investigating, in the hormone dependent tissues.
Collapse
Affiliation(s)
- Ravindran Vini
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Juberiya M. Azeez
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Viji Remadevi
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - T. R. Susmi
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - R. S. Ayswarya
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | | | - Lakshmi Mohan Lathika
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sreeja Sreeharshan
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: Sreeja Sreeharshan
| |
Collapse
|
23
|
Zhang Y, Liu M, Zhang Y, Tian M, Chen P, Lan Y, Zhou B. Urolithin A alleviates acute kidney injury induced by renal ischemia reperfusion through the p62-Keap1-Nrf2 signaling pathway. Phytother Res 2022; 36:984-995. [PMID: 35040204 DOI: 10.1002/ptr.7370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) induced by renal ischemia reperfusion (RIR) is typically observed in renal surgeries and is a leading cause of renal failure. However, there is still an unmet medical need currently in terms of clinical treatments. Herein, we report the effect of Urolithin A (UA) in a mouse RIR model, wherein we demonstrated its underlying mechanism both in vitro and in vivo. The expression levels of p62 and Keap1 significantly decreased, while that of nuclear Nrf2 increased in vitro in a hypoxia cell model after UA treatment. Furthermore, the apoptosis of tubular cells was attenuated and the reactive oxygen species (ROS) levels were reduced in the kidneys in a mouse RIR model after UA administration. In this study, we demonstrated that UA can alleviate oxidative stress and promote autophagy by activating the p62-Keap1-Nrf2 signaling pathway, which could protect the kidneys from ischemia reperfusion injury.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China.,School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Mengmeng Liu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Yaoyuan Zhang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mi Tian
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Lan
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China.,School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Gwon MG, Gu H, Leem J, Park KK. Protective Effects of 6-Shogaol, an Active Compound of Ginger, in a Murine Model of Cisplatin-Induced Acute Kidney Injury. Molecules 2021; 26:5931. [PMID: 34641472 PMCID: PMC8512008 DOI: 10.3390/molecules26195931] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 12/27/2022] Open
Abstract
Acute kidney injury (AKI) is a dose-limiting side effect of cisplatin therapy in cancer patients. However, effective therapies for cisplatin-induced AKI are not available. Oxidative stress, tubular cell death, and inflammation are known to be the major pathological processes of the disease. 6-Shogaol is a major component of ginger and exhibits anti-oxidative and anti-inflammatory effects. Accumulating evidence suggest that 6-shogaol may serve as a potential therapeutic agent for various inflammatory diseases. However, whether 6-shogaol exerts a protective effect on cisplatin-induced renal side effect has not yet been determined. The aim of this study was to evaluate the effect of 6-shogaol on cisplatin-induced AKI and to investigate its underlying mechanisms. An administration of 6-shogaol after cisplatin treatment ameliorated renal dysfunction and tubular injury, as shown by a reduction in serum levels of creatinine and blood urea nitrogen and an improvement in histological abnormalities. Mechanistically, 6-shogaol attenuated cisplatin-induced oxidative stress and modulated the renal expression of prooxidant and antioxidant enzymes. Apoptosis and necroptosis induced by cisplatin were also suppressed by 6-shogaol. Moreover, 6-shogaol inhibited cisplatin-induced cytokine production and immune cell infiltration. These results suggest that 6-shogaol exhibits therapeutic effects against cisplatin-induced AKI via the suppression of oxidative stress, tubular cell death, and inflammation.
Collapse
Affiliation(s)
- Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea; (M.-G.G.); (H.G.); (K.-K.P.)
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea; (M.-G.G.); (H.G.); (K.-K.P.)
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea; (M.-G.G.); (H.G.); (K.-K.P.)
| |
Collapse
|
25
|
Hasheminezhad SH, Boozari M, Iranshahi M, Yazarlu O, Sahebkar A, Hasanpour M, Iranshahy M. A mechanistic insight into the biological activities of urolithins as gut microbial metabolites of ellagitannins. Phytother Res 2021; 36:112-146. [PMID: 34542202 DOI: 10.1002/ptr.7290] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/06/2021] [Accepted: 09/04/2021] [Indexed: 12/26/2022]
Abstract
Urolithins are the gut metabolites produced from ellagitannin-rich foods such as pomegranates, tea, walnuts, as well as strawberries, raspberries, blackberries, and cloudberries. Urolithins are of growing interest due to their various biological activities including cardiovascular protection, anti-inflammatory activity, anticancer properties, antidiabetic activity, and antiaging properties. Several studies mostly based on in vitro and in vivo experiments have investigated the potential mechanisms of urolithins which support the beneficial effects of urolithins in the treatment of several diseases such as Alzheimer's disease, type 2 diabetes mellitus, liver disease, cardiovascular disease, and various cancers. It is now obvious that urolithins can involve several cellular mechanisms including inhibition of MDM2-p53 interaction, modulation of mitogen-activated protein kinase pathway, and suppressing nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activity. Antiaging activity is the most appealing and probably the most important property of urolithin A that has been investigated in depth in recent studies, owing to its unique effects on activation of mitophagy and mitochondrial biogenesis. A recent clinical trial showed that urolithin A is safe up to 2,500 mg/day and can improve mitochondrial biomarkers in elderly patients. Regarding the importance of mitochondria in the pathophysiology of many diseases, urolithins merit further research especially in clinical trials to unravel more aspects of their clinical significance. Besides the nutritional value of urolithins, recent studies proved that urolithins can be used as pharmacological agents to prevent or cure several diseases. Here, we comprehensively review the potential role of urolithins as new therapeutic agents with a special focus on the molecular pathways that have been involved in their biological effects. The pharmacokinetics of urolithins is also included.
Collapse
Affiliation(s)
| | - Motahareh Boozari
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Omid Yazarlu
- Department of General Surgery, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maede Hasanpour
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Iranshahy
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Pavitrakar V, Mody R, Ravindran S. Amelioration of Cisplatin-induced renal inflammation by Recombinant Human Golimumab in Mice. Curr Pharm Biotechnol 2021; 23:970-977. [PMID: 35135447 DOI: 10.2174/1389201022666210810141139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/31/2021] [Accepted: 06/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND One of the most commonly used anti-cancer agents, Cisplatin (CDDP) often causes nephrotoxicity by eliciting inflammation and oxidative stress. Golimumab, an anti-TNF biologic, is prescribed for the management of numerous inflammatory ailments like psoriatic and rheumatoid arthritis ulcerative colitis, and ankylosing spondylitis. OBJECTIVE Current study has explored the effects of anti-TNF biologics golimumab on mice due to cisplatin-induced nephrotoxicity. METHOD Renal toxicity was caused by administration of single cisplatin injection at 25 mg/kg by intraperitoneal (i/p) route. Golimumab (24 mg/kg, s.c.) was administered consecutively for 7 days. The parameters such as renal functions, oxidative stress, inflammation, and renal damage were evaluated on the 7th day of experiments. RESULTS Cisplatin administration caused nephrotoxicity as shown by a significant elevation of various parameters viz; serum creatinine, neutrophil gelatinase-associated lipocalin (NGAL), urea nitrogen (BUN), and cystatin C. There was a significant rise in urinary clusterin, kidney injury molecule 1 (KIM-1), and β-N-acetylglucosaminidase (NAG) concentrations in the animals treated with cisplatin-. The markers of oxidative stress (malondialdehyde, reduced glutathione, and catalase), inflammation (IL-6, TNF-α, IL-10, IL-1β, MCP-1, ICAM-1, and TGF-β1), and apoptosis (caspase-3) were also altered in serum and/or kidneys of cisplatin animals. Further, cisplatin-caused histopathological changes in proximal tubular cells as observed in the H&E staining of renal tissue. Golimumab treatment reduced all markers of kidney injury and attenuated cell death. Golimumab significantly reduced inflammatory cytokines TNFα, IL- 6, MCP-1, IL- 1β, ICAM-1, and TGF-β1 and increased anti-inflammatory cytokine IL-10 in cisplatin-intoxicated mice. CONCLUSION The study results suggest that golimumab prevented nephrotoxicity induced by cisplatin- through inhibition of oxidative stress, apoptotic cell death inflammatory response, thus improving renal function.
Collapse
Affiliation(s)
- Vishal Pavitrakar
- Biotechnology division, Vishal N. Pavitrakar, Lupin Limited, Pune. India
| | - Rustom Mody
- Biotechnology division, Rustom Mody, Lupin Limited, Pune. India
| | - Selvan Ravindran
- Faculty of health Sciences, Symbiosis School of Biological Sciences, Selvan Ravindran, Symbiosis International (Deemed) University, Pune. India
| |
Collapse
|
27
|
Giménez-Bastida JA, Ávila-Gálvez MÁ, Espín JC, González-Sarrías A. Evidence for health properties of pomegranate juices and extracts beyond nutrition: A critical systematic review of human studies. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.06.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Hong Y, Mu X, Ji X, Chen X, Geng Y, Zhang Y, Liu Q, Li F, Wang Y, He J. In-utero exposure to HT-2 toxin affects meiotic progression and early oogenesis in foetal oocytes by increasing oxidative stress. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 279:116917. [PMID: 33744629 DOI: 10.1016/j.envpol.2021.116917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 06/12/2023]
Abstract
HT-2 toxin (HT-2), a mycotoxin produced by Fusarium species, is detected in a variety of cereal grain-based human food and animal feed. Apart from its well-established immunotoxicity and haematotoxicity, it also causes reproductive disorders. In the present study, we revealed the adverse effects of HT-2 on early oogenesis at the foetal stage. Pregnant mice were orally administered with HT-2 for 3 days at mid-gestation. Oocytes from female foetuses exposed to HT-2 displayed defects in meiotic prophase, including unrepaired DNA damage, elevated recombination levels, and reduced expression of meiotic-related genes. Subsequently, increased oxidative stress was observed in the foetal ovaries exposed to HT-2, along with the elevated levels of reactive oxygen species, malondialdehyde, catalase, and superoxide dismutase 1/2, thereby resulting in impaired mitochondrial membrane potential and cell apoptosis. Furthermore, pre-treatment with urolithin A, a natural compound with antioxidant activities, partially reversed the delayed meiotic process by alleviating oxidative stress. Since early oogenesis is essential to determine female fertility in adult life, this study indicated that brief maternal exposure to HT-2 toxin may compromise the fertility of a developing female foetus.
Collapse
Affiliation(s)
- Yi Hong
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xinyi Mu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xingduo Ji
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; Chenghua District Center for Disease Control and Prevention, Chengdu, 610057, PR China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yan Zhang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qiqi Liu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Fangfang Li
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
29
|
D'Amico D, Andreux PA, Valdés P, Singh A, Rinsch C, Auwerx J. Impact of the Natural Compound Urolithin A on Health, Disease, and Aging. Trends Mol Med 2021; 27:687-699. [PMID: 34030963 DOI: 10.1016/j.molmed.2021.04.009] [Citation(s) in RCA: 181] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Urolithin A (UA) is a natural compound produced by gut bacteria from ingested ellagitannins (ETs) and ellagic acid (EA), complex polyphenols abundant in foods such as pomegranate, berries, and nuts. UA was discovered 40 years ago, but only recently has its impact on aging and disease been explored. UA enhances cellular health by increasing mitophagy and mitochondrial function and reducing detrimental inflammation. Several preclinical studies show how UA protects against aging and age-related conditions affecting muscle, brain, joints, and other organs. In humans, benefits of UA supplementation in the muscle are supported by recent clinical trials in elderly people. Here, we review the state of the art of UA's biology and its translational potential as a nutritional intervention in humans.
Collapse
Affiliation(s)
- Davide D'Amico
- Amazentis SA, EPFL Innovation Park, Bâtiment C, CH-1015 Lausanne, Switzerland.
| | - Pénélope A Andreux
- Amazentis SA, EPFL Innovation Park, Bâtiment C, CH-1015 Lausanne, Switzerland
| | - Pamela Valdés
- Amazentis SA, EPFL Innovation Park, Bâtiment C, CH-1015 Lausanne, Switzerland
| | - Anurag Singh
- Amazentis SA, EPFL Innovation Park, Bâtiment C, CH-1015 Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis SA, EPFL Innovation Park, Bâtiment C, CH-1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
30
|
Fang Q, Liu N, Zheng B, Guo F, Zeng X, Huang X, Ouyang D. Roles of Gut Microbial Metabolites in Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2021; 12:636175. [PMID: 34093430 PMCID: PMC8173181 DOI: 10.3389/fendo.2021.636175] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a highly prevalent metabolic disease that has emerged as a global challenge due to its increasing prevalence and lack of sustainable treatment. Diabetic kidney disease (DKD), which is one of the most frequent and severe microvascular complications of diabetes, is difficult to treat with contemporary glucose-lowering medications. The gut microbiota plays an important role in human health and disease, and its metabolites have both beneficial and harmful effects on vital physiological processes. In this review, we summarize the current findings regarding the role of gut microbial metabolites in the development and progression of DKD, which will help us better understand the possible mechanisms of DKD and explore potential therapeutic approaches for DKD.
Collapse
Affiliation(s)
- Qing Fang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Na Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Binjie Zheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Fei Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Xiangchang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Xinyi Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| |
Collapse
|
31
|
Immunomodulatory Role of Urolithin A on Metabolic Diseases. Biomedicines 2021; 9:biomedicines9020192. [PMID: 33671880 PMCID: PMC7918969 DOI: 10.3390/biomedicines9020192] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022] Open
Abstract
Urolithin A (UroA) is a gut metabolite produced from ellagic acid-containing foods such as pomegranates, berries, and walnuts. UroA is of growing interest due to its therapeutic potential for various metabolic diseases based on immunomodulatory properties. Recent advances in UroA research suggest that UroA administration attenuates inflammation in various tissues, including the brain, adipose, heart, and liver tissues, leading to the potential delay or prevention of the onset of Alzheimer’s disease, type 2 diabetes mellitus, and non-alcoholic fatty liver disease. In this review, we focus on recent updates of the anti-inflammatory function of UroA and summarize the potential mechanisms by which UroA may help attenuate the onset of diseases in a tissue-specific manner. Therefore, this review aims to shed new insights into UroA as a potent anti-inflammatory molecule to prevent immunometabolic diseases, either by dietary intervention with ellagic acid-rich food or by UroA administration as a new pharmaceutical drug.
Collapse
|
32
|
Catalpol-Induced AMPK Activation Alleviates Cisplatin-Induced Nephrotoxicity through the Mitochondrial-Dependent Pathway without Compromising Its Anticancer Properties. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7467156. [PMID: 33510841 PMCID: PMC7826214 DOI: 10.1155/2021/7467156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 11/15/2020] [Accepted: 12/24/2020] [Indexed: 01/19/2023]
Abstract
Nephrotoxicity is a common complication of cisplatin chemotherapy and, thus, limits the clinical application of cisplatin. In this work, the effects of catalpol (CAT), a bioactive ingredient extracted from Rehmannia glutinosa, on cisplatin-induced nephrotoxicity and antitumor efficacy were comprehensively investigated. Specifically, the protective effect of CAT on cisplatin-induced injury was explored in mice and HK-2 cells. In vivo, CAT administration strikingly suppressed cisplatin-induced renal dysfunction, morphology damage, apoptosis, and inflammation. In vitro, CAT induced activation of adenosine 5′-monophosphate- (AMP-) activated protein kinase (AMPK), improved mitochondrial function, and decreased generation of cellular reactive oxygen species (ROS), leading to a reduction in inflammation and apoptosis, which ultimately protected from cisplatin-induced injury. However, the beneficial effects of CAT were mostly blocked by coincubation with compound C. Furthermore, molecular docking results indicated that CAT had a higher affinity for AMPK than other AMPK activators such as danthron, phenformin, and metformin. Importantly, CAT possessed the ability to reverse drug resistance without compromising the antitumor properties of cisplatin. These findings suggest that CAT exerts positive effects against cisplatin-induced renal injury through reversing drug resistance via the mitochondrial-dependent pathway without affecting the anticancer activity of cisplatin.
Collapse
|
33
|
Cheng F, Dou J, Zhang Y, Wang X, Wei H, Zhang Z, Cao Y, Wu Z. Urolithin A Inhibits Epithelial-Mesenchymal Transition in Lung Cancer Cells via P53-Mdm2-Snail Pathway. Onco Targets Ther 2021; 14:3199-3208. [PMID: 34040386 PMCID: PMC8139733 DOI: 10.2147/ott.s305595] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The epithelial-to-mesenchymal transition (EMT) is a fundamental process in tumor progression that endows cancer cells with migratory and invasive potential. Snail, a zinc finger transcriptional repressor, plays an important role in the induction of EMT by directly repressing the key epithelial marker E-cadherin. Here, we assessed the effect of urolithin A, a major metabolite from pomegranate ellagitannins, on Snail expression and EMT process. METHODS The role of Snail in urolithin A-induced EMT inhibition in lung cancer cells was explored by wound healing assay and cell invasion assay. The qRT-PCR and CHX assay were performed to investigate how urolithin A regulates Snail expression. Immunoprecipitation assays were established to determine the effects of urolithin A in mdm2-Snail interaction. In addition, the expression of p53 was manipulated to explore its effect on the expression of mdm2 and Snail. RESULTS The urolithin A dose-dependently upregulated epithelial marker and decreased mesenchymal markers in lung cancer cells. In addition, exposure to urolithin A decreased cell migratory and invasive capacity. We have further demonstrated that urolithin A inhibits lung cancer cell EMT by decreasing Snail protein expression and activity. Mechanistically, urolithin A disrupts the interaction of p53 and mdm2 which leads Snail ubiquitination and degradation. CONCLUSION We conclude that urolithin A could inhibit EMT process by controlling mainly Snail expression. These results highlighted the role of pomegranate in regulation of EMT program in lung cancer.
Collapse
Affiliation(s)
- Feng Cheng
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Jintao Dou
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- School of Anesthesiology, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Yong Zhang
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- School of Clinical Medicine, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Xiang Wang
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- School of Laboratory Medicine, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Huijun Wei
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Zhijian Zhang
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Yuxiang Cao
- School of Laboratory Medicine, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- Provincial Engineering Laboratory for Screening and Re-Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, 241001, People’s Republic of China
| | - Zhihao Wu
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- Anhui Province Key Laboratory of Active Biological Macro-Molecules Research, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, People’s Republic of China
- Correspondence: Zhihao Wu Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, 241001, People’s Republic of China Email
| |
Collapse
|
34
|
Lee J, Nguyen QN, Park JY, Lee S, Hwang GS, Yamabe N, Choi S, Kang KS. Protective Effect of Shikimic Acid against Cisplatin-Induced Renal Injury: In Vitro and In Vivo Studies. PLANTS (BASEL, SWITZERLAND) 2020; 9:E1681. [PMID: 33271750 PMCID: PMC7759863 DOI: 10.3390/plants9121681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 12/15/2022]
Abstract
Nephrotoxicity is a serious side effect of cisplatin, which is one of the most frequently used drugs for cancer treatment. This study aimed to assess the renoprotective effect of Artemisia absinthium extract and its bioactive compound (shikimic acid) against cisplatin-induced renal injury. An in vitro assay was performed in kidney tubular epithelial cells (LLC-PK1) with 50, 100, and 200 µg/mL A. absinthium extract and 25 and 50 µM shikimic acid, and cytotoxicity was induced by 25 µM cisplatin. BALB/c mice (6 weeks old) were injected with 16 mg/kg cisplatin once and orally administered 25 and 50 mg/kg shikimic acid daily for 4 days. The results showed that the A. absinthium extract reversed the decrease in renal cell viability induced by cisplatin, whereas it decreased the reactive oxidative stress accumulation and apoptosis in LLC-PK1 cells. Shikimic acid also reversed the effect on cell viability but decreased oxidative stress and apoptosis in renal cells compared with the levels in the cisplatin-treated group. Furthermore, shikimic acid protected against kidney injury in cisplatin-treated mice by reducing serum creatinine levels. The protective effect of shikimic acid against cisplatin-mediated kidney injury was confirmed by the recovery of histological kidney injury in cisplatin-treated mice. To the best of our knowledge, this study is the first report on the nephroprotective effect of A. absinthium extract and its mechanism of action against cisplatin-induced renal injury.
Collapse
Affiliation(s)
- Jinkyung Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (J.L.), (Q.N.N.); (G.S.H.); (N.Y.)
| | - Quynh Nhu Nguyen
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (J.L.), (Q.N.N.); (G.S.H.); (N.Y.)
| | - Jun Yeon Park
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Korea;
| | - Sullim Lee
- College of Bio-Nano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Korea;
| | - Gwi Seo Hwang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (J.L.), (Q.N.N.); (G.S.H.); (N.Y.)
| | - Noriko Yamabe
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (J.L.), (Q.N.N.); (G.S.H.); (N.Y.)
| | - Sungyoul Choi
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (J.L.), (Q.N.N.); (G.S.H.); (N.Y.)
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (J.L.), (Q.N.N.); (G.S.H.); (N.Y.)
| |
Collapse
|
35
|
Casanova AG, Hernández-Sánchez MT, Martínez-Salgado C, Morales AI, Vicente-Vicente L, López-Hernández FJ. A meta-analysis of preclinical studies using antioxidants for the prevention of cisplatin nephrotoxicity: implications for clinical application. Crit Rev Toxicol 2020; 50:780-800. [PMID: 33170047 DOI: 10.1080/10408444.2020.1837070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cisplatin is an effective chemotherapeutic drug whose clinical use and efficacy are limited by its nephrotoxicity, which affects mainly the renal tubules and vasculature. It accumulates in proximal and distal epithelial tubule cells and causes oxidative stress-mediated cell death and malfunction. Consequently, many antioxidants have been tested for their capacity to prevent cisplatin nephrotoxicity. In this study, we made a systematic review of the literature and meta-analyzed 152 articles, which tested the nephroprotective effect of isolated compounds or mixtures of natural origin on cisplatin nephrotoxicity in preclinical models. This meta-analysis identified the most effective candidates and examined the efficacy obtained by antioxidants administered by the oral and intraperitoneal routes. By comparing with a recent, similar meta-analysis performed on clinical studies, this article identifies a disconnection between preclinical and clinical research, and contextualizes, discusses, and integrates the existing preclinical information toward the optimized selection of candidates to be further explored (clinical level). Despite proved efficacy, this article discusses the barriers limiting the clinical development of natural mixtures, such as those in extracts from Calendula officinalis flowers and Heliotropium eichwaldii roots. On the contrary, isolated compounds are more straightforward candidates, among which arjunolic acid and quercetin stand out in this meta-analysis.
Collapse
Affiliation(s)
- Alfredo G Casanova
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - M Teresa Hernández-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martínez-Salgado
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I Morales
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| | - Laura Vicente-Vicente
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J López-Hernández
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| |
Collapse
|
36
|
Song Y, Hu T, Gao H, Zhai J, Gong J, Zhang Y, Tao L, Sun J, Li Z, Qu X. Altered metabolic profiles and biomarkers associated with astragaloside IV-mediated protection against cisplatin-induced acute kidney injury in rats: An HPLC-TOF/MS-based untargeted metabolomics study. Biochem Pharmacol 2020; 183:114299. [PMID: 33148504 DOI: 10.1016/j.bcp.2020.114299] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/18/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CDDP)-induced acute kidney injury (AKI) limits the therapeutic use of CDDP, which urgently needs to be addressed. Our previous study demonstrated that astragaloside IV (AS IV), an active compound of the traditional Chinese herb Astragalus membranaceus, alleviated CDDP-induced AKI. To explore the mechanism, we performed a metabolomics study to explore the altered metabolic pathways and screen for sensitive biomarkers. Twenty-four rats were randomly divided into three groups, which were treated with vehicle solutions (Control), intraperitoneally injected CDDP, and intraperitoneally injected CDDP plus oral AS IV, respectively. Metabolic profiles of serum, urine, and kidney samples were analyzed by high-performance liquid chromatography-time of flight mass spectrometry. There were 38 key metabolites in the urine samples, 20 in the serum samples, and 16 in the kidney samples that were significantly altered due to AS IV-mediated protection against CDDP-induced AKI relative to CDDP-only treatment. CDDP + AS IV co-treatment significantly altered two pathways in the blood (biosynthesis of unsaturated fatty acids and alanine, aspartate, and glutamate metabolism), five pathways in the urine (phenylalanine metabolism; phenylalanine, tyrosine, and tryptophan biosynthesis; arginine biosynthesis; arginine and proline metabolism; and histidine metabolism), and five pathways in the kidneys (glutathione metabolism; alanine, aspartate, and glutamate metabolism; glyoxylate and dicarboxylate metabolism; arginine and proline metabolism; and D-glutamine and D-glutamate metabolism). The metabolic pathways were mainly associated with improvements in inflammatory responses, oxidative stress, and energy metabolism. Adrenic acid in serum and L-histidine and L-methionine in urine were identified as sensitive biomarkers. This study provides new insights to understand the mechanism of AS IV-mediated protection against CDDP-induced AKI and has identified three candidate biomarkers to evaluate preventative treatment and assess therapeutic effectiveness.
Collapse
Affiliation(s)
- Yanqing Song
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Tingting Hu
- Department of Technical Center, Changchun Customs District, Changchun 130062, China
| | - Huan Gao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Jinghui Zhai
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Jiawei Gong
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Yueming Zhang
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Lina Tao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Jingmeng Sun
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhiyuan Li
- AB Sciex Analytical Instrument Trading Co., Ltd, Beijing 100015, China
| | - Xiaoyu Qu
- Department of Pharmacy, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
37
|
Chen D, Liang M, Jin C, Sun Y, Xu D, Lin Y. Expression of inflammatory factors and oxidative stress markers in serum of patients with coronary heart disease and correlation with coronary artery calcium score. Exp Ther Med 2020; 20:2127-2133. [PMID: 32765687 PMCID: PMC7401708 DOI: 10.3892/etm.2020.8958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/05/2020] [Indexed: 12/21/2022] Open
Abstract
Expression characteristics of inflammatory factors interleukin-23 and interleukin-35; oxidative stress markers of malondialdehyde, which is a final product of lipid peroxidation; superoxide dismutase; microRNA-126 and microRNA-146a in serum of patients with coronary heart disease were investigated. Correlation between these biomarkers and CACS (calcification score), as well as the underlying clinical significance were evaluated. A total of 192 patients diagnosed with coronary heart disease were recruited as the observation group, and 69 healthy adults who provided their blood samples were selected as the control group. Enzyme linked immunosorbent assay was carried out to measure the levels of inflammatory factors interleukin-23 and interleukin-35, and the levels of oxidative stress markers of malondialdehyde and superoxide dismutase in serum of the patients and healthy subjects. Real-time fluorescence-based quantitative PCR was performed to measure the expression levels of microRNA-126 and microRNA-146a in serum. The differences in expression of these biomarkers were analyzed, and correlation between these biomarkers and coronary artery calcium score were assessed. The differences in expression levels of interleukin-23, interleukin-35, malondialdehyde, superoxide dismutase, microRNA-126 and microRNA-146a were statistically significant in both groups. The expression levels of interleukin-23, interleukin-35, malondialdehyde, superoxide dismutase, microRNA-126 and microRNA-146a in the observation group were closely associated with severity of the disease. There were positive correlations between coronary artery calcium score and interleukin-23, interleukin-35, malondialdehyde, microRNA-126 and microRNA-146a, respectively; while a negative correlation existed between coronary artery calcium score and superoxide dismutase in the observation group. In conclusion, biomarkers interleukin-23, interleukin-35, malondialdehyde, superoxide dismutase, microRNA-126 and microRNA-146a were abnormally expressed in serum of patients with coronary heart disease, implicating their association with onset and progression of the disease. The biomarkers were found to be correlated with coronary artery calcium score. Detection of changes of related biomarkers in serum may have certain value in diagnosis of disease formation, as well as assessment of disease severity.
Collapse
Affiliation(s)
- Datong Chen
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Minghui Liang
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Cheng Jin
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yue Sun
- CT Room, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Dongbin Xu
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yueming Lin
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
38
|
Peiffer DS. Modulation of the host microbiome by black raspberries or their components and the therapeutic implications in cancer. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.40] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Daniel S Peiffer
- Health Sciences Division Loyola University Chicago Maywood Illinois
| |
Collapse
|
39
|
Taghizadeh F, Hosseinimehr SJ, Zargari M, Karimpour Malekshah A, Talebpour Amiri FB. Gliclazide attenuates cisplatin-induced nephrotoxicity through inhibiting NF-κB and caspase-3 activity. IUBMB Life 2020; 72:2024-2033. [PMID: 32687680 DOI: 10.1002/iub.2342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022]
Abstract
Cisplatin (CP), as a chemotherapeutic drug, causes nephrotoxicity that has limited the clinical utility of CP. Gliclazide (GLZ), as an antihyperglycemic drug, at low dose has antioxidant property. In this study, we aimed to investigate the protective effect of GLZ against CP-induced acute renal injury. Sixty-four BALB/c mice were randomly divided into eight groups. The groups were included as control, GLZ (5, 10, and 25 mg/kg), CP, and GLZ (5, 10, and 25 mg/kg) + CP. Renal function markers (serum creatinine and blood urea nitrogen), oxidative stress markers (malondialdehyde and glutathione), apoptotic marker (caspase-3), and NF-κB were histopathologically evaluated. The results of our study showed that increased urea and creatinine were evidence of CP-induced nephrotoxicity. Histopathological examination revealed tubular epithelial and Bowman degeneration, edema, and cytoplasmic vacuolation in renal tissue structure. Administration of GLZ reduced oxidative stress, caspase-3, and NF-κB activity, and improved kidney function markers in CP-treated mice compared with CP alone group. Also, we observed that the histological tissue structure of the kidney was maintained. GLZ at dose of 25 mg/kg had higher protective effect as compared with other doses. Overall, our study suggests that GLZ with antioxidant, antiapoptotic, and anti-inflammatory properties may be a promising new therapeutic agent to prevent CP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Fatemeh Taghizadeh
- Department of Anatomy, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Department of Biochemistry, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | | |
Collapse
|
40
|
Yin M, Li N, Makinde EA, Olatunji OJ, Ni Z. N6-2-hydroxyethyl-adenosine ameliorate cisplatin induced acute kidney injury in mice. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1760149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Min Yin
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Na Li
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | | | | | - Ziyuan Ni
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
41
|
Lin J, Zhuge J, Zheng X, Wu Y, Zhang Z, Xu T, Meftah Z, Xu H, Wu Y, Tian N, Gao W, Zhou Y, Zhang X, Wang X. Urolithin A-induced mitophagy suppresses apoptosis and attenuates intervertebral disc degeneration via the AMPK signaling pathway. Free Radic Biol Med 2020; 150:109-119. [PMID: 32105828 DOI: 10.1016/j.freeradbiomed.2020.02.024] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022]
Abstract
Intervertebral disc degeneration (IDD) is a major cause of low back pain (LBP), and effective therapies are still lacking. Previous studies reported that mitochondrial dysfunction contributes to apoptosis, and urolithin A (UA) specifically induces mitophagy. Herein, we aimed to investigate the protective effect of UA-induced mitophagy on tert-butyl hydroperoxide (TBHP)-induced apoptosis in nucleus pulposus (NP) cells in vitro and a rat model of IDD in vivo. Mitochondrial function, apoptosis, and mitophagy were measured in UA-treated NP cells by western blotting and immunofluorescence; the therapeutic effects of UA on IDD were assessed in rats with puncture-induced IDD. The results showed that UA could activate mitophagy in primary NP cells, and UA treatment inhibited TBHP-induced mitochondrial dysfunction and the intrinsic apoptosis pathway. Mechanistically, we revealed that UA promoted mitophagy by activating AMPK signaling in TBHP-induced NP cells. In vivo, UA was shown to effectively alleviate the progression of puncture-induced IDD in rats. Taken together, our results suggest that UA could be a novel and effective therapeutic strategy for IDD.
Collapse
Affiliation(s)
- Jialiang Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jinru Zhuge
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xuanqi Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuhao Wu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zengjie Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tianzhen Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zaher Meftah
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hongming Xu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital of Wenzhou Medical University, Ningbo, Zhejiang Province, China
| | - Yaosen Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, Zhejiang Province, China.
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
42
|
Cásedas G, Les F, Choya-Foces C, Hugo M, López V. The Metabolite Urolithin-A Ameliorates Oxidative Stress in Neuro-2a Cells, Becoming a Potential Neuroprotective Agent. Antioxidants (Basel) 2020; 9:antiox9020177. [PMID: 32098107 PMCID: PMC7070385 DOI: 10.3390/antiox9020177] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Urolithin A is a metabolite generated from ellagic acid and ellagitannins by the intestinal microbiota after consumption of fruits such as pomegranates or strawberries. The objective of this study was to determine the cytoprotective capacity of this polyphenol in Neuro-2a cells subjected to oxidative stress, as well as its direct radical scavenging activity and properties as an inhibitor of oxidases. Cells treated with this compound and H2O2 showed a greater response to oxidative stress than cells only treated with H2O2, as mitochondrial activity (MTT assay), redox state (ROS formation, lipid peroxidation), and the activity of antioxidant enzymes (CAT: catalase, SOD: superoxide dismutase, GR: glutathione reductase, GPx: glutathione peroxidase) were significantly ameliorated; additionally, urolithin A enhanced the expression of cytoprotective peroxiredoxins 1 and 3. Urolithin A also acted as a direct radical scavenger, showing values of 13.2 μM Trolox Equivalents for Oxygen Radical Absorbance Capacity (ORAC) and 5.01 µM and 152.66 µM IC50 values for superoxide and 2,2-diphenyss1-picrylhydrazyl (DPPH) radicals, respectively. Finally, inhibition of oxidizing enzymes, such as monoamine oxidase A and tyrosinase, was also detected in a dose-dependent manner. The cytoprotective effects of urolithin A could be attributed to the improvement of the cellular antioxidant battery, but also to its role as a direct radical scavenger and enzyme inhibitor of oxidases.
Collapse
Affiliation(s)
- Guillermo Cásedas
- Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain; (G.C.); (F.L.)
| | - Francisco Les
- Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain; (G.C.); (F.L.)
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), 50059 Zaragoza, Spain
| | - Carmen Choya-Foces
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28009 Madrid, Spain; (C.C.-F.); (M.H.)
| | - Martín Hugo
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28009 Madrid, Spain; (C.C.-F.); (M.H.)
| | - Víctor López
- Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain; (G.C.); (F.L.)
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), 50059 Zaragoza, Spain
- Correspondence:
| |
Collapse
|