1
|
Barakat N, Ismail E, Zahran F. The integrated effect of roflumilast and selenium nanoparticles on nephrotoxicity generated by cisplatin through the regulation of the antioxidant and apoptotic pathways. J Trace Elem Med Biol 2024; 86:127555. [PMID: 39442470 DOI: 10.1016/j.jtemb.2024.127555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
AIM The current investigations aimed to investigate the potential synergistic effect of Roflumilast (ROF) and Selenium nanoparticles (SeNPs) administration on Cisplatin (Cis) -induced nephrotoxicity. MATERIALS AND METHODS Fifty male rats were divided into five groups; Control group: animals were administered 0.9 % saline solution. Cis group: animals were injected with a single dose of 6 mg/kg. ROF group: Rats received a dosage of 1.2 mg/kg orally daily for 11 days. SeNPs group: animals orally received 0.5 mg/kg of ROF daily for 11 days. The ROF + SeNPs group was administered both after receiving a Cis injection for 11 days. Animals were sacrificed at 5 and 11 days, and the urine and blood samples were collected on day 5 and day 11 for chemical analysis, while kidney samples were obtained for molecular, histological, and immunohistochemical studies. RESULTS The levels of serum creatinine, Blood urea nitrogen (BUN), and total protein were elevated in the Cis group compared to the control group (p < 0.05). While the combination of ROF and SeNPs dramatically decreased these values after 5 and 11 days (p < 0.05). In addition, Cis caused renal oxidative stress by elevating MDA levels and suppressing the activities of SOD, GSH, and CAT. Similarly, these effects were modulated by ROF and SeNPs after 11 days (p < 0.05). Furthermore, the concurrent administration of ROF and SeNPs resulted in a significant increase in the expression of HO-1, Nrf2, and Bcl2, while decreasing the expression of BAX and IL-6 compared to the Cis group after 11 days (P < 0.05). CONCLUSION The study showed that both ROF and SeNPs had significant therapeutic potential in reducing the pathological alterations caused by Cis.
Collapse
Affiliation(s)
- Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - Ehab Ismail
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Faten Zahran
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
2
|
Ahmed MA, Kamel EO, Abd-Eldayem AM. Role of cAMP/pCREB and GSK-3β/NF-κB p65 signaling pathways in the renoprotective effect of mirabegron against renal ischemia-reperfusion injury in rats. Eur J Pharmacol 2024; 974:176617. [PMID: 38679120 DOI: 10.1016/j.ejphar.2024.176617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Acute kidney injury and other renal disorders are thought to be primarily caused by renal ischemia-reperfusion (RIR). Cyclic adenosine monophosphate (cAMP) has plenty of physiological pleiotropic effects and preserves tissue integrity and functions. This research aimed to examine the potential protective effects of the β3-adrenergic receptors agonist mirabegron in a rat model of RIR and its underlying mechanisms. Male rats enrolled in this work were given an oral dose of 30 mg/kg mirabegron for two days before surgical induction of RIR. Renal levels of kidney injury molecule-1 (KIM-1), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), Interleukin-10 (IL-10), cAMP, cAMP-responsive element binding protein (pCREB), and glycogen synthase kinase-3 beta (GSK-3β) were assessed along with blood urea nitrogen and serum creatinine. Additionally, caspase-3 and nuclear factor-kappa B (NF-κB) p65 were explored by immunohistochemical analysis. Renal specimens were inspected for histopathological changes. RIR led to renal tissue damage with elevated blood urea nitrogen and serum creatinine levels. The renal KIM-1, MCP-1, TNF-α, and GSK-3β were significantly increased, while IL-10, cAMP, and pCREB levels were reduced. Moreover, upregulation of caspase-3 and NF-κB p65 protein expression was seen in RIR rats. Mirabegron significantly reduced kidney dysfunction, histological abnormalities, inflammation, and apoptosis in the rat renal tissues. Mechanistically, mirabegron mediated these effects via modulation of cAMP/pCREB and GSK-3β/NF-κB p65 signaling pathways. Mirabegron administration could protect renal tissue and maintain renal function against RIR.
Collapse
Affiliation(s)
- Marwa A Ahmed
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Esam O Kamel
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Ahmed M Abd-Eldayem
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Pharmacology, Faculty of Medicine, Merit University, Sohag, Egypt.
| |
Collapse
|
3
|
Shokeir AA, Awadalla A, Hamam ET, Hussein AM, Mahdi MR, Abosteta AN, Shahin M, Barakat N, El-Adl M, El-Sherbiny M, Eldesoqui M, AlMadani M, Ali SK, El-Sherbini ES, Khirallah SM. Human Wharton's jelly-derived mesenchymal stromal stem cells preconditioned with valproic acid promote cell migration and reduce renal inflammation in ischemia/reperfusion injury by activating the AKT/P13K and SDF1/CXCR4 pathways. Arch Biochem Biophys 2024; 755:109985. [PMID: 38579957 DOI: 10.1016/j.abb.2024.109985] [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: 01/22/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/07/2024]
Abstract
OBJECTIVE To determine whether WJ-MSCs pretreated with VPA would enhance their migration to improve functional recovery of renal IRI in rats. METHODS 150 Sprague-Dawley rats were distributed into 5 groups; Sham, IRI, WJ-MSC, VPA, and WJ-MSCs + VPA. 10 rats were sacrificed after 3, 5, and 7 days. Role of WJ-MSCs pretreated with VPA was evaluated by assessment of renal function, antioxidant enzymes together with renal histopathological and immunohistopathological analyses and finally by molecular studies. RESULTS WJ-MSCs and VPA significantly improved renal function and increased antioxidants compared to IRI group. Regarding gene expression, WJ-MSCs and VPA decreased BAX and TGF-β1, up-regulated Akt, PI3K, BCL2, SDF1α, and CXCR4 related to IRI. Additionally, WJ-MSCs pretreated with VPA improved the measured parameters more than either treatment alone. CONCLUSION WJ-MSCs isolated from the umbilical cord and pretreated with VPA defended the kidney against IRI by more easily homing to the site of injury.
Collapse
Affiliation(s)
- Ahmed A Shokeir
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Amira Awadalla
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Eman T Hamam
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt; Nanomedicine Research Unit, Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Abdelaziz M Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed R Mahdi
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt; Department of Basic Medical Sciences, Faculty of Medicine, Galala University, Suez, Egypt
| | - Alyaa Naeem Abosteta
- Biochemistry Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mirna Shahin
- Mansoura Manchester Medical Program for Medical Education, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Adl
- Departement of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, 13713, Riyadh, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt; Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, 13713, Riyadh, Saudi Arabia
| | - Moneer AlMadani
- Department of Clinical Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, 13713, Riyadh, Saudi Arabia
| | - Sahar K Ali
- Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - El-Said El-Sherbini
- Biochemistry Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Salma M Khirallah
- Chemistry Department (Biochemistry Division), Faculty of Science, Port Said University, Port Said, 42526, Egypt.
| |
Collapse
|
4
|
Ba X, Ye T, Shang H, Tong Y, Huang Q, He Y, Wu J, Deng W, Zhong Z, Yang X, Wang K, Xie Y, Zhang Y, Guo X, Tang K. Recent Advances in Nanomaterials for the Treatment of Acute Kidney Injury. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12117-12148. [PMID: 38421602 DOI: 10.1021/acsami.3c19308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Acute kidney injury (AKI) is a serious clinical syndrome with high morbidity, elevated mortality, and poor prognosis, commonly considered a "sword of Damocles" for hospitalized patients, especially those in intensive care units. Oxidative stress, inflammation, and apoptosis, caused by the excessive production of reactive oxygen species (ROS), play a key role in AKI progression. Hence, the investigation of effective and safe antioxidants and inflammatory regulators to scavenge overexpressed ROS and regulate excessive inflammation has become a promising therapeutic option. However, the unique physiological structure and complex pathological alterations in the kidneys render traditional therapies ineffective, impeding the residence and efficacy of most antioxidant and anti-inflammatory small molecule drugs within the renal milieu. Recently, nanotherapeutic interventions have emerged as a promising and prospective strategy for AKI, overcoming traditional treatment dilemmas through alterations in size, shape, charge, and surface modifications. This Review succinctly summarizes the latest advancements in nanotherapeutic approaches for AKI, encompassing nanozymes, ROS scavenger nanomaterials, MSC-EVs, and nanomaterials loaded with antioxidants and inflammatory regulator. Following this, strategies aimed at enhancing biocompatibility and kidney targeting are introduced. Furthermore, a brief discussion on the current challenges and future prospects in this research field is presented, providing a comprehensive overview of the evolving landscape of nanotherapeutic interventions for AKI.
Collapse
Affiliation(s)
- Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wen Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zichen Zhong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kangyang Wang
- Department of Urology, Wenchang People's Hospital, Wenchang 571300, Hainan Province, China
| | - Yabin Xie
- Department of Urology, Wenchang People's Hospital, Wenchang 571300, Hainan Province, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang 550025, Guizhou Province, China
| | - Xiaolin Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
5
|
Mohammed ET, Safwat GM, Bahnasawy EA, Abdel-Razik ARH, Mohamed DS. Zinc Oxide Nanoparticles and Vitamin C Ameliorate Atrazine-Induced Hepatic Apoptosis in Rat via CYP450s/ROS Pathway and Immunomodulation. Biol Trace Elem Res 2023; 201:5257-5271. [PMID: 36790584 PMCID: PMC10509061 DOI: 10.1007/s12011-023-03587-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023]
Abstract
Atrazine, as an herbicide, is used widely worldwide. Because of its prolonged persistence in the environment and accumulation in the body, atrazine exposure is a potential threat to human health. The present study evaluated the possible protective effects of zinc oxide nanoparticles and vitamin C against atrazine-induced hepatotoxicity in rats. Atrazine administered to rats orally at a dose of 300 mg/kg for 21 days caused liver oxidative stress as it increased malondialdehyde (MDA) formation and decreased reduced glutathione (GSH) contents. Atrazine induced inflammation accompanied by apoptosis via upregulation of hepatic gene expression levels of NF-κB, TNF-α, BAX, and caspase-3 and downregulation of Bcl-2 gene expression levels. Additionally, it disturbed the metabolic activities of cytochrome P450 as it downregulated hepatic gene expression levels of CYP1A1, CYP1B1, CYP2E1. The liver function biomarkers were greatly affected upon atrazine administration, and the serum levels of AST and ALT were significantly increased, while BWG%, albumin, globulins, and total proteins levels were markedly decreased. As a result of the above-mentioned influences of atrazine, histopathological changes in liver tissue were recorded in our findings. The administration of zinc oxide nanoparticles or vitamin C orally at a dose of 10 mg/kg and 200 mg/kg, respectively, for 30 days prior and along with atrazine, could significantly ameliorate the oxidative stress, inflammation, and apoptosis induced by atrazine and regulated the hepatic cytochrome P450 activities. Furthermore, they improved liver function biomarkers and histopathology. In conclusion, our results revealed that zinc oxide nanoparticles and vitamin C supplementations could effectively protect against atrazine-induced hepatotoxicity.
Collapse
Affiliation(s)
- Eman T Mohammed
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, 62512, Egypt.
| | - Ghada M Safwat
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, 62512, Egypt
| | - Esraa A Bahnasawy
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, 62512, Egypt
| | - Abdel-Razik H Abdel-Razik
- Department of Histology, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, 62511, Egypt
| | - Doaa Sh Mohamed
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, 62512, Egypt
| |
Collapse
|
6
|
Dai XS, Wei QH, Guo X, Ding Y, Yang XQ, Zhang YX, Xu XY, Li C, Chen Y. Ferulic acid, ligustrazine, and tetrahydropalmatine display the anti-proliferative effect in endometriosis through regulating Notch pathway. Life Sci 2023; 328:121921. [PMID: 37429417 DOI: 10.1016/j.lfs.2023.121921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/12/2023]
Abstract
AIMS With an ambiguous anti-proliferative mechanism, the combination of ferulic acid, ligustrazine, and tetrahydropalmatine (FLT) shows good anti-endometriosis (EMS) activity. In EMS, the expression of Notch pathway and its role in proliferation are not yet unclear. In this study, we sought to uncover the role of Notch pathway's effect and FLT's anti-proliferative mechanism on EMS proliferation. MAIN METHODS In autograft and allograft EMS models, the proliferating markers (Ki67, PCNA), Notch pathway, and the effect of FLT on them were detected. Then, the anti-proliferative influence of FLT was measured in vitro. The proliferating ability of endometrial cells was investigated with a Notch pathway activator (Jagged 1 or VPA) or inhibitor (DAPT) alone, or in combination with FLT separately. KEY FINDINGS FLT presented the inhibitory effect on ectopic lesions in 2 EMS models. The proliferating markers and Notch pathway were promoted in ectopic endometrium, but FLT showed the counteraction. Meantime, FLT restrained the endometrial cell growth and clone formation along with a reduction in Ki67 and PCNA. Jagged 1 and VPA stimulated the proliferation. On the contrary, DAPT displayed the anti-proliferating effect. Furthermore, FLT exhibited an antagonistic effect on Jagged 1 and VPA by downregulating Notch pathway and restraining proliferation. FLT also displayed a synergistic effect on DAPT. SIGNIFICANCE This study indicated that the overexpressing Notch pathway induced EMS proliferation. FLT attenuated the proliferation by inhibiting Notch pathway.
Collapse
Affiliation(s)
- Xue-Shan Dai
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Qing-Hua Wei
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Xin Guo
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Yi Ding
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Xiao-Qian Yang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Yu-Xin Zhang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Xiao-Yu Xu
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China
| | - Cong Li
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yi Chen
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing, China; Engineering Research Center of Coptis Development and Utilization, Ministry of Education, College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China; National Demonstration Center for Experimental Pharmacy Education, Southwest University, Chongqing, China.
| |
Collapse
|
7
|
Wu J, Shang H, Zhang A, He Y, Tong Y, Huang Q, Liu X, Chen Z, Tang K. Antioxidant nanozymes in kidney injury: mechanism and application. NANOSCALE 2023; 15:13148-13171. [PMID: 37547960 DOI: 10.1039/d3nr01954c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Excessive production of reactive oxygen species (ROS) in the kidneys is involved in the pathogenesis of kidney diseases, such as acute kidney injury (AKI) and diabetic kidney disease (DKD), and is the main reason for the progression of kidney injury. ROS can easily lead to lipid peroxidation and damage the tubular epithelial cell membrane, proteins and DNA, and other molecules, which can trigger cellular oxidative stress. Effective scavenging of ROS can delay or halt the progression of kidney injury by reducing inflammation and oxidative stress. With the development of nanotechnology and an improved understanding of nanomaterials, more researchers are applying nanomaterials with antioxidant activity to treat kidney injury. This article reviews the detailed mechanism between ROS and kidney injury, as well as the applications of nanozymes with antioxidant effects based on different materials for various kidney injuries. To better guide the applications of antioxidant nanozymes in kidney injury and other inflammatory diseases, at the end of this review we also summarize the aspects of nanozymes that need to be improved. An in-depth understanding of the role played by ROS in the occurrence and progression of kidney injury and the mechanism by which antioxidant nanozymes reduce oxidative stress is conducive to improving the therapeutic effect in kidney injury and inflammation-related diseases.
Collapse
Affiliation(s)
- Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - An Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| |
Collapse
|
8
|
Awadalla A, Hamam ET, El-Senduny FF, Omar NM, Mahdi MR, Barakat N, Ammar OA, Hussein AM, Shokeir AA, Khirallah SM. Zinc oxide nanoparticles and spironolactone-enhanced Nrf2/HO-1 pathway and inhibited Wnt/β-catenin pathway in adenine-induced nephrotoxicity in rats. Redox Rep 2022; 27:249-258. [DOI: 10.1080/13510002.2022.2139947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Amira Awadalla
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Eman T. Hamam
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Fardous F. El-Senduny
- Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
- Department of Pathology & Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Nisreen Mansour Omar
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed R. Mahdi
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Omar A. Ammar
- Basic Science Department, Delta University for Science and Technology, Gamasa, Egypt
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed A. Shokeir
- Center of Excellence for Genome and Cancer Research, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Salma M. Khirallah
- Biochemistry Division, Chemistry Department, Faculty of Science, Port Said University, Port Said, Egypt
| |
Collapse
|
9
|
Guo W, Li Y, An D, Zhou M, Xiong J, Jiang Z, Ding Y, Huang R, Miao W. Sodium ferulate-functionalized silver nanopyramides with synergistic antithrombotic activity for thromboprophylaxis. Colloids Surf B Biointerfaces 2022; 220:112925. [DOI: 10.1016/j.colsurfb.2022.112925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/28/2022] [Accepted: 10/10/2022] [Indexed: 11/27/2022]
|
10
|
Barakat M, Hussein AM, Salama MF, Awadalla A, Barakat N, Serria M, El-Shafey M, El-Sherbiny M, El Adl MA. Possible Underlying Mechanisms for the Renoprotective Effect of Retinoic Acid-Pretreated Wharton's Jelly Mesenchymal Stem Cells against Renal Ischemia/Reperfusion Injury. Cells 2022; 11:cells11131997. [PMID: 35805083 PMCID: PMC9266019 DOI: 10.3390/cells11131997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives: The current work investigated the effect of Wharton jelly mesenchymal stem cells (WJ-MSCs) pretreated with all-trans-retinoic acid (ATRA) on renal ischemia in rats and the possible role of oxidative stress, apoptotic and Wnt/β-Catenin signaling pathways, and inflammatory cytokines in their effects. Methods: The study included 90 male Sprague Dawley rats that were allocated to five groups (n = 18 rats): (I) Sham-operated group (right nephrectomy was performed); (II) Ischemia/reperfusion injury (IRI) group, a sham group with 45-min renal ischemia on the left kidney; (III) ATRA group, an ischemic group with an intravenous (i.v.) administration of ATRA 10 µM, 10 min post-surgery); (IV) WJ-MSCs group, an IRI group with an i.v. administration of 150 µL containing 7 × 106 WJ-MSCs, 10 min post-surgery; (V) WJ-MSCs + ATRA group, an IRI group with an i.v. administration of 150 µL of 7 × 106 WJ-MSCs pretreated with 10 µM ATRA. At the end of the experiments, serum creatinine, BUN micro-albuminuria (MAU), urinary protein, markers of redox state in the left kidney (MDA, CAT, SOD, and GSH), and the expression of Bax, IL-6, HIF-1α, Wnt7B, and β-catenin genes at the level of mRNA as well as for immunohistochemistry for NFkB and β-Catenin markers were analyzed. Results: The current study found that 45-min of renal ischemia resulted in significant impairment of kidney function (evidenced by the increase in serum creatinine, BUN, and urinary proteins) and deterioration of the kidney morphology, which was associated with a significant increase in redox state (evidenced by an increase in MDA and a decrease in GSH, SOD, and CAT), and a significant increase in inflammatory and apoptotic processes (evidenced by an increase in Bax and IL-6, NFkB, Wnt7B, β-catenin and HIF-1α) in kidney tissues (p < 0.05). On the other hand, treatment with ATRA, WJ-MSCs, or a combination of both, caused significant improvement in kidney function and morphology, which was associated with significant attenuation of oxidative stress, apoptotic markers, and inflammatory cytokines (IL6 and NFkB) with the upregulation of HIF-1α and β-catenin in kidney tissues (p < 0.05). Moreover, the renoprotective effect of WJ-MSCs pretreated with ATRA was more potent than WJ-MSCs alone. Conclusions: It is concluded that preconditioning of WJ-MSCs with ATRA may enhance their renoprotective effect. This effect could be due to the upregulation of the beta-catenin/Wnt pathway and attenuation of apoptosis, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Mai Barakat
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
- Institute of Global Public Health and Human Ecology, School of Science and Engineering, American University, Cairo 11835, Egypt
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence: ; Tel.: +20-10-0242-1140
| | - Mohamed F. Salama
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
| | - Amira Awadalla
- Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt; (A.A.); (N.B.)
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt; (A.A.); (N.B.)
| | - Mohamed Serria
- Department of Biochemistry, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed El-Shafey
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
- Physiological Sciences Department, Fakeeh College for Medical Sciences, Jeddah 21461, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 71666, Saudi Arabia;
| | - Mohamed A. El Adl
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
| |
Collapse
|
11
|
Chen T, Niu L, Wang L, Zhou Q, Zhao X, Lai S, He X, He H, He M. Ferulic acid protects renal tubular epithelial cells against anoxia/reoxygenation injury mediated by AMPKα1. Free Radic Res 2022; 56:173-184. [PMID: 35382666 DOI: 10.1080/10715762.2022.2062339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Anoxia/reoxygenation (A/R) injury causes dysfunction of rat renal tubular epithelial cells (NRK-52E), which is associated with excess reactive oxygen species (ROS) generation and eventually leads to apoptosis. Ferulic acid (FA), a phenolic acid, which is abundant in fruits and vegetables. FA possesses the properties of scavenging free radicals and cytoprotection against oxygen stress. In the study, the protective effects of FA against NRK-52E cells damage induced by A/R were explored and confirmed the role of AMP-activated protein kinaseα1 (AMPKα1). We found that after NRK-52E cells suffered A/R damage, FA pretreatment increased the cell viability and decreased LDH activity in culture medium in a concentration-dependent manner, the activities of endogenous antioxidant enzymes such as glutathione peroxidase, superoxide dismutase and catalase improved, intracellular ROS generation and malondialdehyde contents mitigated. In addition, pretreatment of 75 μM FA ameliorated mitochondrial dysfunction by A/R-injury and ultimately decreased apoptosis (25.3 ± 0.61 vs 12.1 ± 0.60), which was evidenced by preventing the release of cytochrome c from mitochondria to the cytoplasm. 75 μM FA pretreatment also significantly upregulated AMPKα1 expression (3.16 ± 0.18 folds) and phosphorylation (2.56 ± 0.13 folds). However, compound C, a specific AMPK inhibitor, significantly attenuated FA pretreatment's effects, as mentionedabove. These results firstly clarified that FA pretreatment attenuated NRK-52E cell damage induced by A/R via upregulating AMPKα1 expression and phosphorylation.
Collapse
Affiliation(s)
- Tianpeng Chen
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Li Niu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Liang Wang
- Department of rehabilitation, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Songqing Lai
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xinlan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
12
|
Stompor-Gorący M, Machaczka M. Recent Advances in Biological Activity, New Formulations and Prodrugs of Ferulic Acid. Int J Mol Sci 2021; 22:ijms222312889. [PMID: 34884693 PMCID: PMC8657461 DOI: 10.3390/ijms222312889] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 01/18/2023] Open
Abstract
Trans-ferulic acid (FA) is a derivative of 4-hydroxycinnamic acid, which is found in many food products, fruits and beverages. It has scientifically proven antioxidant, anti-inflammatory and antibacterial properties. However, its low ability to permeate through biological barriers (e.g., the blood-brain barrier, BBB), its low bioavailability and its fast elimination from the gastrointestinal tract after oral administration limit its clinical use, e.g., for the treatment of neurodegenerative diseases, such as Alzheimer's disease. Therefore, new nanotechnological approaches are developed in order to regulate intracellular transport of ferulic acid. The objective of this review is to summarize the last decade's research on biological properties of ferulic acid and innovative ways of its delivery, supporting pharmacological therapy.
Collapse
Affiliation(s)
- Monika Stompor-Gorący
- Department of Human Pathophysiology, Institute of Medical Sciences, University of Rzeszow, Kopisto 2a, 35-959 Rzeszów, Poland;
- Correspondence:
| | - Maciej Machaczka
- Department of Human Pathophysiology, Institute of Medical Sciences, University of Rzeszow, Kopisto 2a, 35-959 Rzeszów, Poland;
- Department of Clinical Science and Education, Division of Internal Medicine, Södersjukhuset, Karolinska Institutet, 11883 Stockholm, Sweden
| |
Collapse
|
13
|
Makled MN, El-Awady MS, Abdel-Aziz RR, Shehab El-Din AB, Ammar EM, Gameil NM. Pomegranate extract ameliorates renal ischemia/reperfusion injury in rats via suppressing NF-κB pathway. Hum Exp Toxicol 2021; 40:S573-S582. [PMID: 34802289 DOI: 10.1177/09603271211041998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Inflammation and oxidative stress are the major pathways involved in ischemia-reperfusion (I/R)-induced renal injury. This study was designed to evaluate the potential effect of pomegranate against I/R-induced renal injury. I/R injury was induced in nephrectomized rats by unilateral occlusion of the left renal pedicle for 45 min followed by 24 h of perfusion. Pomegranate succeeded to decrease serum levels of creatinine, potassium, and urea nitrogen, along with increasing creatinine clearance. Pomegranate also decreased I/R-induced changes in histopathological examination. Pomegranate attenuated the renal inflammatory response reflected by the suppression of nuclear factor κB p65 DNA binding activity, the upregulation of inhibitory protein kappa B-alpha mRNA expression, the downregulation of mRNA and protein expression of tumor necrosis factor α, in addition to the reduced myeloperoxidase activity and mRNA expression. Additionally, pomegranate attenuated oxidative stress likely through the modulation of lipid peroxidation and antioxidant levels reflected by the decreased MDA content and the increased glutathione level and superoxide dismutase activity. Results confirm the potential protective effect of pomegranate against I/R-induced renal injury through its anti-inflammatory and anti-oxidant effects mediated through the upregulation of inhibitory protein kappa B-alpha, the inhibition of NF-κB activity, and the associated TNF-α release, neutrophil infiltration, and oxidative stress.
Collapse
Affiliation(s)
- Mirhan N Makled
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, 158395Mansoura University, Mansoura, Egypt
| | - Mohammed S El-Awady
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, 158395Mansoura University, Mansoura, Egypt
| | - Rania R Abdel-Aziz
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, 158395Mansoura University, Mansoura, Egypt
| | - Ahmed B Shehab El-Din
- Nephrology and Urology Center, Faculty of Medicine, 158395Mansoura University, Mansoura, Egypt
| | - Elsayed M Ammar
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, 158395Mansoura University, Mansoura, Egypt
| | - Nariman M Gameil
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, 158395Mansoura University, Mansoura, Egypt
| |
Collapse
|
14
|
Novel doxorubicin / folate-targeted trans-ferulic acid-loaded PLGA nanoparticles combination: In-vivo superiority over standard chemotherapeutic regimen for breast cancer treatment. Biomed Pharmacother 2021; 145:112376. [PMID: 34749055 DOI: 10.1016/j.biopha.2021.112376] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
AIM Doxorubicin/Cyclophosphamide (AC) is one of the standard adjuvant anthracycline-containing regimens that is still in use for breast cancer treatment. Cancer cell resistance and AC-induced side effects make treatment suboptimal and worsen patients' quality of life. This study aimed to improve trans-ferulic acid's (TFA) efficiency via loading into folate-receptor-targeted-poly lactic-co-glycolic acid nanoparticles (FA-PLGA-TFA NPs). Also, investigating both the antitumor efficacy of Doxorubicin (Dox)/FA-PLGA-TFA NPs combination against dimethylbenz[a]anthracene (DMBA)-induced breast cancer and its safety profile. METHODS FA-PLGA-TFA NPs were optimally fabricated and characterized. Levels of Notch1, Hes1, Wnt-3a, β-catenin, MMP-9, cyclin D1, Permeability-Glycoprotein (P-gp), ERα, PR, and HER2 were assessed as a measure of the antitumor efficacy of different treatment protocols. Histopathological examination of heart and bone, levels of ALT, AST, ALP, CK-MB, and WBCs count were evaluated to ensure the combination's safety profile. KEY FINDINGS Dox/FA-PLGA-TFA NPs not only inhibited Notch signaling but also suppressed Notch synergy with Wnt, estrogen, progesterone, and HER2 pathways. Interestingly, Dox/FA-PLGA-TFA NPs decreased P-gp level and preserved heart, bone, and liver health as well as WBCs count. SIGNIFICANCE Dox/FA-PLGA-TFA NPs reduced the side-effects of each single drug, and at the same time exerted excellent antitumor activity that surpass the AC regimen in evading cancer cell resistance and having a superior safety profile.
Collapse
|