1
|
Reum Kwon B, Jo AR, Lee I, Lee G, Joo Park Y, Pyo Lee J, Park NY, Kho Y, Kim S, Ji K, Choi K. Thyroid, neurodevelopmental, and kidney toxicities of common organic UV filters in embryo-larval zebrafish (Danio rerio), and their potential links. ENVIRONMENT INTERNATIONAL 2024; 192:109030. [PMID: 39341038 DOI: 10.1016/j.envint.2024.109030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Organic UV filters (OUVFs) have been commonly used in sunscreen and many consumer products. Following dermal application, these compounds can enter circulation and may cause systemic effects in humans. In the present study, we chose four OUVFs frequently detected in the environment, i.e., avobenzone (AVB), benzophenone-3 (BP-3), octocrylene (OC), and octyl methoxycinnamate (OMC), and evaluated their thyroid, neurodevelopmental, and kidney toxicities. For this purpose, zebrafish embryos (<4 h post fertilization, hpf) were exposed to sublethal concentrations of AVB, BP-3, OC, or OMC until 120 hpf. Exposure to all OUVFs decreased thyroid hormone (TH) levels, probably by enhanced metabolism and excretion of THs (ugt1ab and/or sult1 st5) in the larval fish. Exposure to the OUVFs also induced hypoactivities and/or anxiety-like behaviors: Regulatory changes of mbp, gfap, c-fos, syn2a, sty1a, and stxbp1b genes, support the changes in normal neurobehavior of the larval fish. Moreover, the OUVFs exposure caused increased proteinuria in the fish, along with transcriptional changes of wt1, nephrin, podocin, and cdh17 genes, which could explain the observed reduction in kidney functions. Principal component analysis (PCA) implied the potential interplay of THs with neurogenesis, or podocyte differentiation of the larval fish. Toxicological consequences of altered TH homeostasis, neurobehavior, and kidney function at the early life stage warrant further investigations not only in humans but also in aquatic ecosystems.
Collapse
Affiliation(s)
- Ba Reum Kwon
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Ah-Reum Jo
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Inae Lee
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Gowoon Lee
- Department of Safety Engineering, Korea National University of Transportation, Chungju, Chungbuk 27469, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea
| | - Na-Youn Park
- Department of Health, Environment & Safety, Eulji University, Seongnam, Gyeonggi 13135, Republic of Korea
| | - Younglim Kho
- Department of Health, Environment & Safety, Eulji University, Seongnam, Gyeonggi 13135, Republic of Korea
| | - Sungkyoon Kim
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyunghee Ji
- Department of Environmental Health, Yongin University, Yongin, Gyeonggi 17092, Republic of Korea; Department of Occupational and Environmental Health, Yongin University, Yongin, Gyeonggi 17092, Republic of Korea
| | - Kyungho Choi
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
2
|
Golmohammadi M, Ivraghi MS, Hasan EK, Huldani H, Zamanian MY, Rouzbahani S, Mustafa YF, Al-Hasnawi SS, Alazbjee AAA, Khalajimoqim F, Khalaj F. Protective effects of pioglitazone in renal ischemia-reperfusion injury (RIRI): focus on oxidative stress and inflammation. Clin Exp Nephrol 2024; 28:955-968. [PMID: 38935212 DOI: 10.1007/s10157-024-02525-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (RIRI) is a critical phenomenon that compromises renal function and is the most serious health concern related to acute kidney injury (AKI). Pioglitazone (Pio) is a known agonist of peroxisome proliferator-activated receptor-gamma (PPAR-γ). PPAR-γ is a nuclear receptor that regulates genes involved in inflammation, metabolism, and cellular differentiation. Activation of PPAR-γ is associated with antiinflammatory and antioxidant effects, which are relevant to the pathophysiology of RIRI. This study aimed to investigate the protective effects of Pio in RIRI, focusing on oxidative stress and inflammation. METHODS We conducted a comprehensive literature search using electronic databases, including PubMed, ScienceDirect, Web of Science, Scopus, and Google Scholar. RESULTS The results of this study demonstrated that Pio has antioxidant, anti-inflammatory, and anti-apoptotic activities that counteract the consequences of RIRI. The study also discussed the underlying mechanisms, including the modulation of various pathways such as TNF-α, NF-κB signaling systems, STAT3 pathway, KIM-1 and NGAL pathways, AMPK phosphorylation, and autophagy flux. Additionally, the study presented a summary of various animal studies that support the potential protective effects of Pio in RIRI. CONCLUSION Our findings suggest that Pio could protect the kidneys from RIRI by improving antioxidant capacity and decreasing inflammation. Therefore, these findings support the potential of Pio as a therapeutic strategy for preventing RIRI in different clinical conditions.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1988873554, Iran
| | | | | | - Huldani Huldani
- Department of Physiology, Faculty of Medicine Lambung, Mangkurat University, South Kalimantan, Banjarmasin, Indonesia
| | - Mohammad Yasin Zamanian
- Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Shiva Rouzbahani
- Miller School of Medicine, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
- Department of Community Medicine and Family Physician, School of Medicine, Isfahan University of Medical Sciences, Hezar Jarib Blvd, Isfahan, Iran
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | | | - Faranak Khalajimoqim
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Fattaneh Khalaj
- Digestive Diseases Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Salama RM, Darwish SF, Yehia R, Sallam AA, Elmongy NF, Abd-Elgalil MM, El Wakeel SA. Lactoferrin alleviates gentamicin-induced acute kidney injury in rats by suppressing ferroptosis: Highlight on ACSL4, SLC7A11, NCOA4, FSP1 pathways and miR-378a-3p, LINC00618 expression. Food Chem Toxicol 2024; 193:115027. [PMID: 39357596 DOI: 10.1016/j.fct.2024.115027] [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: 06/04/2024] [Revised: 07/23/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
The use of gentamicin (GNT) is associated with acute kidney injury (AKI). Ferroptosis is a newly recognized iron-dependent, non-apoptotic cell death that can lead to AKI. Lactoferrin (LF), an iron-binding glycoprotein, was previously reported to be renoprotective. Nonetheless, LF's impact on GNT-induced AKI and ferroptosis has not yet been investigated. Accordingly, we assessed the dose-dependent effect of LF on GNT-induced AKI and its influence on ferroptosis. Thirty-six male rats were allocated as control, LF, GNT (100 mg/kg/day, i.p.), and groups given LF (100, 200, and 300 mg/kg, p.o.) for 14 days prior concurrently with GNT (Day 8-14). The high dose of LF (300 mg/kg) showed better histopathological picture, higher creatinine clearance, reduced serum and urine levels of kidney injury markers when compared to the GNT group and the lower two doses. These nephroprotective effects of LF can be attributed to the observed reduction in renal ferrous iron, 4-HNE, and MDA, miR-378a-3p and ALOX15 expression, TFR1, NCOA4, and ACSL4 protein expression and the increased LINC00618 expression, GSH levels, GPX4, SLC7A11, and FSP1 protein expression. In conclusion, LF high dose was the most renoprotective against GNT-induced AKI, in which suppression of ferroptosis pathways was a likely contributor to its protective mechanism.
Collapse
Affiliation(s)
- Rania M Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| | - Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt.
| | - Rana Yehia
- Clinical Pharmacy, Faculty of Pharmacy, British University in Egypt (BUE), Cairo, Egypt.
| | - Al Aliaa Sallam
- Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Noura F Elmongy
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt.
| | - Mona M Abd-Elgalil
- Histology and Cell Biology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt.
| | - Sara A El Wakeel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| |
Collapse
|
4
|
Bolat I, Terim–Kapakin KA, Apaydin Yildirim B, Manavoğlu Kirman E. Protective effect of Helichrysum plicatum on head shock protein inflammation and apoptosis in Gentamicin induced nephrotoxicity. REVISTA CIENTÍFICA DE LA FACULTAD DE CIENCIAS VETERINARIAS 2024; XXXIV:1-9. [DOI: 10.52973/rcfcv-e34388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Gentamicin (GM) is an aminoglycoside antibiotic the most common used in the treatment of infectious diseases in humans and animals. However, GM causes damage to many tissues and organs in the body, especially the kidneys. Helichrysum plicatum (Hp), native to the Balkans and Anatolia, is a plant used in various diseases such as diabetes, liver and kidney damage. In this study, Male Spraque Dawley rats (n=36 and 200–250 g) were randomly divided into 6 experimental groups: Group 1: Control; received normal saline (intraperitoneally –i.p.–), Group 2: Hp (100 mg·kg–1 day i.p.), Group 3: Hp (200 mg·kg–1 day i.p.), Group 4: GM (80 mg·kg–1 day i.p.), Group 5: GM 80 + Hp 100 (mg·kg–1 day i.p.), and Group 6: GM 80 + Hp 200 (mg·kg–1 day i.p.). Then kidney tissue samples were collected for evaluations. All of our results showed that Hp (100 mg·kg–1 day) reduced the levels of pro–inflammatory cytokines such as IL–8, IL–6, and TNF– while increasing the level of anti–inflammatory cytokine IL–10. It was also observed that Hp reduced the expressions of the caspase3, NOS and Heat shock proteins such as Hsp27 and Hsp70. With this study, we have shown that Hp probably due to its chemical properties has a protective effect against GM induced nephrototoxicity by reducing the values stated above to normal values.
Collapse
Affiliation(s)
- Ismail Bolat
- Atatürk University, Faculty of Veterinary Medicine, Departments of Pathology. Erzurum, Türkiye
| | | | - Betul Apaydin Yildirim
- Atatürk University, Faculty of Veterinary Medicine, Departments of Biochemistry. Erzurum, Türkiye
| | - Esra Manavoğlu Kirman
- Atatürk University, Faculty of Veterinary Medicine, Departments of Pathology. Erzurum, Türkiye
| |
Collapse
|
5
|
Babaeenezhad E, Dezfoulian O, Moradi Sarabi M, Ahmadvand H. Monoterpene linalool restrains gentamicin-mediated acute kidney injury in rats by subsiding oxidative stress, apoptosis, and the NF-κB/iNOS/TNF-α/IL-1β pathway and regulating TGF-β. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5701-5714. [PMID: 38294506 DOI: 10.1007/s00210-024-02978-z] [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: 12/15/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024]
Abstract
The clinical use of gentamicin (GM) is restricted by its nephrotoxic effects. This study aimed for the first time to elucidate the ameliorative effects of the monoterpene linalool (Lin) against GM-mediated acute kidney injury in rats. A total of thirty-two rats were subdivided into four equal groups: control (saline), Lin (100 mg/kg/day), GM (100 mg/kg/day), and GM + Lin (100 and 100 mg/kg/day). Lin and GM were intraperitoneally administered for 12 days. Our results illustrated that Lin ameliorated GM-mediated renal histopathological abnormalities and reduced serum urea and creatinine levels in rats exposed to GM. Lin treatment mitigated oxidative stress in nephrotoxic animals as manifested by reducing serum and renal levels of malondialdehyde and increasing the activities of serum and renal glutathione peroxidase and renal catalase. Moreover, Lin markedly inhibited GM-triggered inflammation by downregulating NF-κB, iNOS, TNF-α, and IL-1β and reducing renal myeloperoxidase activity and nitric oxide levels. Interestingly, Lin repressed GM-induced apoptosis, as reflected by a marked downregulation of Bax and caspase-3 expression, concurrent with the upregulation of Bcl2 expression. Finally, Lin administration led to a significant downregulation of TGF-β expression in nephrotoxic animals. In summary, Lin ameliorated GM-mediated nephrotoxicity in rats, at least through its antioxidant, anti-inflammatory, and anti-apoptotic activities and by modulating TGF-β.
Collapse
Affiliation(s)
- Esmaeel Babaeenezhad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Omid Dezfoulian
- Department of Pathobiology, School of Veterinary Medicine, Lorestan University, Khorramabad, Iran.
| | - Mostafa Moradi Sarabi
- Razi Herbal Medicines Research Center, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Hassan Ahmadvand
- Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
6
|
Ye Z, Zhang J, Xu Z, Li Z, Huang G, Tong B, Xia P, Shen Y, Hu H, Yu P, Xi X. Pioglitazone ameliorates ischemia/reperfusion-induced acute kidney injury via oxidative stress attenuation and NLRP3 inflammasome. Hum Cell 2024; 37:959-971. [PMID: 38607518 DOI: 10.1007/s13577-024-01059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/27/2024] [Indexed: 04/13/2024]
Abstract
Acute kidney injury (AKI) induced by renal ischemia/reperfusion injury (IRI) is a severe clinical condition. ROS accumulation, antioxidant pathways deficiency, and inflammation are involved in IRI. Pioglitazone (Pio) exerts anti-inflammatory and antioxidant effects. The aim of this study was to explore the protective effects of pioglitazone against IRI-induced AKI. Pathogen-free Sprague-Dawley (SD) rats were arbitrarily divided into four groups: Sham operation group Control (CON) group, CON + Pio group, I/R + Saline group, and I/R + Pio group. In addition, HK-2 cells were subjected to hypoxia and reoxygenation to develop an H/R model for investigation of the protective mechanism of Pio. Pretreatment with pioglitazone in the model rats reduced urea nitrogen and creatinine levels, histopathological scores, and cytotoxicity after IRI. Pioglitazone treatment significantly attenuated renal cell apoptosis, decreased cytotoxicity, increased Bcl-2 expression, and downregulated Bax expression. Besides, the levels of ROS and inflammatory factors, including NLRP3, ASC, pro-IL-1β, pro-caspase-1, cleaved-caspase-1, TNF-α, IL-6, and IL-1β, in I/R rats and H/R cells were normalized by the pioglitazone treatment. Pioglitazone improved IRI-induced AKI by attenuating oxidative stress and NLRP3 inflammasome activation. Therefore, pioglitazone has the potential to serve as a novel agent for renal IRI treatment and prevention.
Collapse
Affiliation(s)
- Zhenfeng Ye
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China
| | - Zhou Xu
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Zhangwang Li
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Gaomin Huang
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China
| | - Bin Tong
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Jiangxi, 330006, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China.
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Jiangxi, 330006, Nanchang, China.
| | - Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China.
| |
Collapse
|
7
|
Rahimi Monfared S, Valibeik A, Tavakoli Dastjerd N, Jafaripour L, Jafarian A, Nabi Moradi M, Ahmadvand H. Protective role of citronellol on antioxidant enzymes and oxidative damage induced by gentamicin in experimental nephrotoxic rats. Mol Biol Rep 2024; 51:382. [PMID: 38430358 DOI: 10.1007/s11033-024-09212-2] [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: 08/04/2023] [Accepted: 01/04/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Gentamicin leads to nephrotoxicity with increasing oxidative stress. In the present research the role of citronellol on oxidative damage induced by gentamicin in nephrotoxic rats was evaluated. METHODS AND RESULTS Forty-twomale Wistar rats were randomly divided into seven equal groups; healthy control, gentamicin, DMSO, citronellol 50, citronellol 100, citronellol 200 and vitamin E. The animals were anesthetized after 12 days of treatment. Kidney and serum samples were received for biochemical, histological changes, and gene expression assessments. The levels of serum glutathione (GSH), serum and kidney glutathione peroxidase (GPX) and the expression of GPX gene against gentamicin group were increased in citronellol treatment groups. The levels of serum and kidney malondialdehyde (MDA), urine protein, serum creatinine and the gene expression of inflammatory factors including tumor necrosis factor-alpha (TNF-α) and Interleukin 6 (IL-6) against gentamicin group were decreased in these groups. Moreover, recuperation in histological alterations was shown in three groups receiving citronellol compared to the gentamicin group. CONCLUSIONS Citronellol with its antioxidant and anti-inflammatory properties can decrease kidney damage caused by nephrotoxicity induced by gentamicin.
Collapse
Affiliation(s)
- Sobhan Rahimi Monfared
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Clinical Biochemistry, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ali Valibeik
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Clinical Biochemistry, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Niloufar Tavakoli Dastjerd
- Department of Medical Biotechnology, School of Allied Medical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Leila Jafaripour
- Department of Anatomy, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Ashkan Jafarian
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Nabi Moradi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hassan Ahmadvand
- Medical Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
8
|
Dogan T, Yildirim BA, Kapakin KAT. Investigation of the effects of crocin on inflammation, oxidative stress, apoptosis, NF-κB, TLR-4 and Nrf-2/HO-1 pathways in gentamicin-induced nephrotoxicity in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 106:104374. [PMID: 38246228 DOI: 10.1016/j.etap.2024.104374] [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: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
The primary limitation of gentamicin (Gm) treatment is its potential to induce nephrotoxicity, which can restrict both its duration and efficacy. This study aims to investigate the protective effects of Crocin (Cr) against Gm-induced nephrotoxicity and its underlying mechanisms, including inflammation, apoptosis, TLR-4, Nrf-2/HO-1 pathways. 36 Sprague Dawley rats were divided into 6 groups for the study. Group I received only saline. Groups II and III were administered 25 and 50 mg/kg of crocin, respectively. Group IV was treated with 80 mg/kg of Gm. Groups V and VI received 25 and 50 mg/kg of crocin, respectively, in addition to Gm administration. Crocin demonstrated protective effects on kidney tissue. It down-regulated the genes NF-κB, COX-2, TLR-4, Bax, and Caspase-3, while up-regulating Bcl-2, Nrf-2, and HO-1. In conclusion, these findings hold promise for the prevention of Gm-induced nephrotoxicity through the modulation of the Nrf-2/HO-1 pathway.
Collapse
Affiliation(s)
- Tuba Dogan
- Ataturk University, Veterinary Faculty, Biochemistry Department, Erzurum 25100, Turkey.
| | | | | |
Collapse
|
9
|
Hanna DA, Messiha BAS, Abo-Saif AA, Ali FEM, Azouz AA. Lysosomal membrane stabilization by imipramine attenuates gentamicin-induced renal injury: Enhanced LAMP2 expression, down-regulation of cytoplasmic cathepsin D and tBid/cytochrome c/cleaved caspase-3 apoptotic signaling. Int Immunopharmacol 2024; 126:111179. [PMID: 37995569 DOI: 10.1016/j.intimp.2023.111179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023]
Abstract
Nephrotoxicity is a serious complication commonly encountered with gentamicin (GTM) treatment. Permeabilization of lysosomes with subsequent cytoplasmic release of GTM and cathepsins is considered a crucial issue in progression of GTM toxicity. This study was designed to evaluate the prospective defensive effect of lysosomal membrane stabilization by imipramine (IMP) against GTM nephrotoxicity in rats. GTM (30 mg/kg/h) was intraperitoneally administered over 4 h daily (120 mg/kg/day) for 7 days. IMP (30 mg/kg/day) was orally administered for 14 days; starting 7 days before and then concurrently with GTM. On 15th day, samples (urine, blood, kidney) were collected to estimate biomarkers of kidney function, lysosomal stability, apoptosis, and inflammation. IMP administration to GTM-treated rats ameliorated the disruption in lysosomal membrane stability induced by GTM. That was evidenced by enhanced renal protein expressions of LAMP2 and PI3K, but reduced cathepsin D cytoplasmic expression in kidney sections. Besides, IMP guarded against apoptosis in GTM-treated rats by down-regulation of the pro-apoptotic (tBid, Bax, cytochrome c) and the effector cleaved caspase-3 expressions, while the anti-apoptotic Bcl-2 expression was enhanced. Additionally, the inflammatory cascade p38 MAPK/NF-κB/TNF-α was attenuated in GTM + IMP group along with marked improvement in kidney function biomarkers, compared to GTM group. These findings were supported by the obvious improvement in histological architecture. Furthermore, in vitro enhancement of the antibacterial activity of GTM by IMP confers an additional benefit to their combination. Conclusively, lysosomal membrane stabilization by IMP with subsequent suppression of tBid/cytochrome c/cleaved caspase-3 apoptotic signaling could be a promising protective strategy against GTM nephrotoxicity.
Collapse
Affiliation(s)
- Dina A Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Basim A S Messiha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ali A Abo-Saif
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Fares E M Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Amany A Azouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
10
|
Matouk AI, Awad EM, Mousa AAK, Abdelhafez SMN, Fahmy UA, El-Moselhy MA, Abdel-Naim AB, Anter A. Dihydromyricetin protects against gentamicin-induced nephrotoxicity via upregulation of renal SIRT3 and PAX2. Life Sci 2024; 336:122318. [PMID: 38035992 DOI: 10.1016/j.lfs.2023.122318] [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: 08/24/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023]
Abstract
AIM Gentamicin-induced nephrotoxicity limits its widespread use as an effective antibacterial agent. Oxidative stress, inflammatory cytokines and apoptotic cell death are major participants in gentamicin-induced nephrotoxicity. We therefore, investigated whether dihydromyricetin (DHM), the antioxidant and anti-inflammatory flavonoid, could protect against the nephrotoxic effects of gentamicin. METHODS Male Wistar rats administrated gentamicin (100 mg/kg/day, i.p.) for 8 days. DHM (400 mg/kg, p.o.) was concurrently given with gentamicin for 8 days. Control group received the vehicle of DHM and gentamicin. Histopathological examinations, biochemical measurements and immunohistochemical analyses were done at the end of the study. KEY FINDINGS Treatment with DHM improved the gentamicin induced deterioration of renal functions; serum levels of urea, creatinine and cystatin-C as well as urinary levels of Kim-1 and NGAL, the sensitive indicators for early renal damage, were declined. Additionally, DHM abrogated gentamicin-induced changes in kidney morphology. These nephroprotective effects were possibly mediated via decreasing renal gentamicin buildup, activating the antioxidant enzymes GSH, SOD and CAT and decreasing lipid peroxidation and nitric oxide levels. Further, DHM suppressed renal inflammation and apoptotic cell death by decreasing the expression of nuclear factor-kappa B (NF-κB), TNF-alpha and caspase-3. These effects were correlated to the upregulation of renal SIRT3 expression. Also, DHM activated the regeneration and replacement of injured tubular cells with new ones via enhancing PAX2 expression. SIGNIFICANCE DHM is a promising therapeutic target that could prevent acute renal injury induced by gentamicin and help renal tubular cells to recover through its antioxidant, anti-inflammatory and antiapoptotic properties.
Collapse
Affiliation(s)
- Asmaa I Matouk
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Eman M Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Amr A K Mousa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Sara M N Abdelhafez
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Usama A Fahmy
- Center of Research Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A El-Moselhy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt; Clinical Pharmacy and Pharmacology Department, Ibn Sina National College for Medical Studies, Jeddah 21589, Saudi Arabia
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aliaa Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
11
|
Albalawi RS, Binmahfouz LS, Hareeri RH, Shaik RA, Bagher AM. Parthenolide Phytosomes Attenuated Gentamicin-Induced Nephrotoxicity in Rats via Activation of Sirt-1, Nrf2, OH-1, and NQO1 Axis. Molecules 2023; 28:2741. [PMID: 36985711 PMCID: PMC10053629 DOI: 10.3390/molecules28062741] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Nephrotoxicity is a serious complication that limits the clinical use of gentamicin (GEN). Parthenolide (PTL) is a sesquiterpene lactone derived from feverfew with various therapeutic benefits. However, PTL possesses low oral bioavailability. This study aimed to evaluate the therapeutic protective effects of PTL-phytosomes against GEN-induced nephrotoxicity in rats. The PTL was prepared as phytosomes to improve the pharmacological properties with a particle size of 407.4 nm, and surface morphology showed oval particles with multiple edges. Rats were divided into six groups: control, nano-formulation plain vehicle, PTL-phytosomes (10 mg/kg), GEN (100 mg/kg), GEN + PTL-phytosomes (5 mg/kg), and GEN + PTL-phytosomes (10 mg/kg). The administration of PTL-phytosomes alleviated GEN-induced impairment in kidney functions and histopathological damage, and decreased kidney injury molecule-1 (KIM-1). The anti-oxidative effect of PTL-phytosomes was demonstrated by the reduced malondialdehyde (MDA) concentration and increased superoxide dismutase (SOD) and catalase (CAT) activities. Furthermore, PTL-phytosomes treatment significantly enhanced sirtuin 1 (Sirt-1), nuclear factor erythroid-2-related factor-2 (Nrf2), NAD(P)H quinone dehydrogenase 1 (NQO1), and heme oxygenase-1 (HO-1). Additionally, PTL-phytosomes treatment exhibited anti-inflammatory and anti-apoptotic properties in the kidney tissue. These findings suggest that PTL-phytosomes attenuate renal dysfunction and structural damage by reducing oxidative stress, inflammation, and apoptosis in the kidney.
Collapse
Affiliation(s)
| | | | | | | | - Amina M. Bagher
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
12
|
Peroxisome proliferator-activated receptor ɣ agonist mediated inhibition of heparanase expression reduces proteinuria. EBioMedicine 2023; 90:104506. [PMID: 36889064 PMCID: PMC10043778 DOI: 10.1016/j.ebiom.2023.104506] [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: 09/05/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Proteinuria is associated with many glomerular diseases and a risk factor for the progression to renal failure. We previously showed that heparanase (HPSE) is essential for the development of proteinuria, whereas peroxisome proliferator-activated receptor ɣ (PPARɣ) agonists can ameliorate proteinuria. Since a recent study showed that PPARɣ regulates HPSE expression in liver cancer cells, we hypothesized that PPARɣ agonists exert their reno-protective effect by inhibiting glomerular HPSE expression. METHODS Regulation of HPSE by PPARɣ was assessed in the adriamycin nephropathy rat model, and cultured glomerular endothelial cells and podocytes. Analyses included immunofluorescence staining, real-time PCR, heparanase activity assay and transendothelial albumin passage assay. Direct binding of PPARɣ to the HPSE promoter was evaluated by the luciferase reporter assay and chromatin immunoprecipitation assay. Furthermore, HPSE activity was assessed in 38 type 2 diabetes mellitus (T2DM) patients before and after 16/24 weeks treatment with the PPARɣ agonist pioglitazone. FINDINGS Adriamycin-exposed rats developed proteinuria, an increased cortical HPSE and decreased heparan sulfate (HS) expression, which was ameliorated by treatment with pioglitazone. In line, the PPARɣ antagonist GW9662 increased cortical HPSE and decreased HS expression, accompanied with proteinuria in healthy rats, as previously shown. In vitro, GW9662 induced HPSE expression in both endothelial cells and podocytes, and increased transendothelial albumin passage in a HPSE-dependent manner. Pioglitazone normalized HPSE expression in adriamycin-injured human endothelial cells and mouse podocytes, and adriamycin-induced transendothelial albumin passage was reduced as well. Importantly, we demonstrated a regulatory effect of PPARɣ on HPSE promoter activity and direct PPARy binding to the HPSE promoter region. Plasma HPSE activity of T2DM patients treated with pioglitazone for 16/24 weeks was related to their hemoglobin A1c and showed a moderate, near significant correlation with plasma creatinine levels. INTERPRETATION PPARɣ-mediated regulation of HPSE expression appears an additional mechanism explaining the anti-proteinuric and renoprotective effects of thiazolidinediones in clinical practice. FUNDING This study was financially supported by the Dutch Kidney Foundation, by grants 15OI36, 13OKS023 and 15OP13. Consortium grant LSHM16058-SGF (GLYCOTREAT; a collaboration project financed by the PPP allowance made available by Top Sector Life Sciences & Health to the Dutch Kidney Foundation to stimulate public-private partnerships).
Collapse
|
13
|
Nadeem RI, Aboutaleb AS, Younis NS, Ahmed HI. Diosmin Mitigates Gentamicin-Induced Nephrotoxicity in Rats: Insights on miR-21 and -155 Expression, Nrf2/HO-1 and p38-MAPK/NF-κB Pathways. TOXICS 2023; 11:48. [PMID: 36668774 PMCID: PMC9865818 DOI: 10.3390/toxics11010048] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
Gentamicin (GNT) is the most frequently used aminoglycoside. However, its therapeutic efficacy is limited due to nephrotoxicity. Thus, the potential anticipatory effect of Diosmin (DIOS) against GNT-prompted kidney damage in rats together with the putative nephroprotective pathways were scrutinized. Four groups of rats were used: (1) control; (2) GNT only; (3) GNT plus DIOS; and (4) DIOS only. Nephrotoxicity was elucidated, and the microRNA-21 (miR-21) and microRNA-155 (miR-155) expression and Nrf2/HO-1 and p38-MAPK/NF-κB pathways were assessed. GNT provoked an upsurge in the relative kidney weight and serum level of urea, creatinine, and KIM-1. The MDA level was markedly boosted, with a decline in the level of TAC, SOD, HO-1, and Nrf2 expression in the renal tissue. Additionally, GNT exhibited a notable amplification in TNF-α, IL-1β, NF-κB p65, and p38-MAPK kidney levels. Moreover, caspase-3 and BAX expression were elevated, whereas the Bcl-2 level was reduced. Furthermore, GNT resulted in the down-regulation of miR-21 expression along with an up-regulation of the miR-155 expression. Histological examination revealed inflammation, degradation, and necrosis. GNT-provoked pathological abnormalities were reversed by DIOS treatment, which restored normal kidney architecture. Hence, regulating miR-21 and -155 expression and modulating Nrf2/HO-1 and p38-MAPK/NF-κB pathways could take a vital part in mediating the reno-protective effect of DIOS.
Collapse
Affiliation(s)
- Rania I. Nadeem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Amany S. Aboutaleb
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
| | - Nancy S. Younis
- Pharmaceutical Sciences Department, Faculty of Clinical Pharmacy, King Faisal University, Al-Ahsa, Al-Hofuf 31982, Saudi Arabia
| | - Hebatalla I. Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
| |
Collapse
|
14
|
Dose-dependent ameliorating effect of lipoxin A4 on gentamicin-induced nephrotoxicity in rats: The role of TNFα, TGF-β, ICAM-1, and JNK signaling. Chem Biol Interact 2022; 366:110139. [DOI: 10.1016/j.cbi.2022.110139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/03/2022]
|
15
|
The N-Methyl-D-Aspartate Receptor Blocker REL-1017 (Esmethadone) Reduces Calcium Influx Induced by Glutamate, Quinolinic Acid, and Gentamicin. Pharmaceuticals (Basel) 2022; 15:ph15070882. [PMID: 35890179 PMCID: PMC9319291 DOI: 10.3390/ph15070882] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
REL-1017 (esmethadone) is a novel N-methyl-D-aspartate receptor (NMDAR) antagonist and promising rapid antidepressant candidate. Using fluorometric imaging plate reader (FLIPR) assays, we studied the effects of quinolinic acid (QA) and gentamicin, with or without L-glutamate and REL-1017, on intracellular calcium ([Ca2+]in) in recombinant cell lines expressing human GluN1-GluN2A, GluN1-GluN2B, GluN1-GluN2C, and GluN1-GluN2D NMDAR subtypes. There were no effects of QA on [Ca2+]in in cells expressing GluN1-GluN2C subtypes. QA acted as a low-potency, subtype-selective, NMDAR partial agonist in GluN1-GluN2A, GluN1-GluN2B, and GluN1-GluN2D subtypes. REL-1017 reduced [Ca2+]in induced by QA. In cells expressing the GluN1-GluN2D subtype, QA acted as an agonist in the presence of 0.04 μM L-glutamate and as an antagonist in the presence of 0.2 μM L-glutamate. REL-1017 reduced [Ca2+]in induced by L-glutamate alone and with QA in all cell lines. In the absence of L-glutamate, gentamicin had no effect. Gentamicin was a positive modulator for GluN1-GluN2B subtypes at 10 μM L-glutamate, for GluN1-GluN2A at 0.2 μM L-glutamate, and for GluN1-GluN2A, GluN1-GluN2B, and GluN1-GluN2D at 0.04 μM L-glutamate. No significant changes were observed with GluN1-GluN2C NMDARs. REL-1017 reduced [Ca2+]in induced by the addition of L-glutamate in all NMDAR cell lines in the presence or absence of gentamicin. In conclusion, REL-1017 reduced [Ca2+]in induced by L-glutamate alone and when increased by QA and gentamicin. REL-1017 may protect cells from excessive calcium entry via NMDARs hyperactivated by endogenous and exogenous molecules.
Collapse
|
16
|
Brkić BM, Rovčanin B, Stojanović M, Srebro D, Vučković S, Savić Vujović K. Chloroquine Attenuates Oxidative Stress in Gentamicin-Induced Nephrotoxicity in Rats. Dose Response 2022; 20:15593258221119871. [PMID: 36003319 PMCID: PMC9393693 DOI: 10.1177/15593258221119871] [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] [Revised: 07/18/2022] [Accepted: 07/24/2022] [Indexed: 11/25/2022]
Abstract
The wider application of gentamicin is limited by potential adverse effects
(nephrotoxicity and ototoxicity). The goal of our study was to investigate the effects of
chloroquine on biochemical and oxidative stress parameters in gentamicin-induced
nephrotoxicity in rats. Animals were randomly divided into 1 of 5 groups. First was Sham
group (0.9% NaCl) (n = 8); second group received gentamicin (n = 8); while third (n = 8),
fourth (n = 8) and fifth group (n = 8) received gentamicin and chloroquine in a dose of
0.3, 1 and 3 mg/kg, respectively. The urea and creatinine levels were significantly lower
in chloroquine treated groups in doses of 0.3 mg/kg and 1 mg/kg (P <
0.001). Total oxidant status and the oxidative stress index showed significantly lower
values in all chloroquine treated groups (P < 0.001;
P < 0.005). Malondialdehyde was lower in chloroquine treatment in
doses of 0.3 mg/kg (P < 0.005) and 3 mg/kg (P <
0.05). Chloroquine treatment markedly reduced the level of superoxide dismutase in doses
of 1 mg/kg (P < 0.01) and 3 mg/kg (P < 0.05). Our
study showed that chloroquine attenuates gentamicin-induced nephrotoxicity in rats
regarding biochemical and oxidative stress parameters.
Collapse
Affiliation(s)
- Branislava Medić Brkić
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branislav Rovčanin
- Centre for Endocrine Surgery, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marko Stojanović
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragana Srebro
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Sonja Vučković
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Katarina Savić Vujović
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Althunibat OY, Abukhalil MH, Aladaileh SH, Qaralleh H, Al-Amarat W, Alfwuaires MA, Algefare AI, Namazi NI, Melebary SJ, Babalghith AO, Conte-Junior CA. Formononetin Ameliorates Renal Dysfunction, Oxidative Stress, Inflammation, and Apoptosis and Upregulates Nrf2/HO-1 Signaling in a Rat Model of Gentamicin-Induced Nephrotoxicity. Front Pharmacol 2022; 13:916732. [PMID: 35712704 PMCID: PMC9197219 DOI: 10.3389/fphar.2022.916732] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022] Open
Abstract
Gentamicin (GEN) is a bactericidal aminoglycoside known to cause nephrotoxicity. Formononetin (FN) is a potent flavonoid that exhibits numerous promising pharmacological activities. In this study, we have assessed the nephroprotective efficacy of FN against GEN-induced renal injury in rats. Rats were orally administered with FN (60 mg/kg/day, for 2 weeks) and were co-treated with intraperitoneal (i.p.) injection of GEN (100 mg/kg/day) during the days 8–14. GEN-treated rats demonstrated increased urea and creatinine levels in serum associated with marked histopathological changes in the kidney. Malondialdehyde (MDA) and protein carbonyl contents were elevated, whereas glutathione concentration and catalase and superoxide dismutase activities were lowered in GEN-administered rats. The FN largely prevented tissue damage, attenuated renal function, reduced MDA and protein carbonyl, and enhanced antioxidant capacity in the kidney of GEN-administrated animals. The kidney of GEN-treated rats demonstrated elevated Bax and caspase-3 protein expression, accompanied by lowered Bcl-2 protein expression, an effect that FN attenuated. Moreover, FN treatment caused upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) expression in renal tissue of GEN-intoxicated animals. Collectively, FN protects against GEN-caused renal damage via exhibiting antioxidant, anti-inflammatory, and antiapoptotic activities and augmenting Nrf2 signaling, suggesting FN as a promising agent for preventing drug-induced organ damage.
Collapse
Affiliation(s)
- Osama Y. Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an, Jordan
- *Correspondence: Osama Y. Althunibat, ; Mohammad H. Abukhalil,
| | - Mohammad H. Abukhalil
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an, Jordan
- Department of Biology, College of Science, Al-Hussein Bin Talal University, Ma’an, Jordan
- *Correspondence: Osama Y. Althunibat, ; Mohammad H. Abukhalil,
| | - Saleem H. Aladaileh
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an, Jordan
| | - Haitham Qaralleh
- Department of Medical Laboratory Sciences, Mutah University, Karak, Jordan
| | - Wesam Al-Amarat
- Department of Medical Support, Al-karak University College, Al-Balqa’ Applied University, As-Salt, Jordan
| | - Manal A. Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abdulmohsen I. Algefare
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Nader Ibrahim Namazi
- Pharmaceutics and Pharmaceutical Technology Department, College of Pharmacy, Taibah University, Al Madinah Al Munawarah, Saudi Arabia
| | - Sahar J. Melebary
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahmad O. Babalghith
- Medical Genetics Department, College of Medicine, Umm al-qura University, Makkah, Saudi Arabia
| | - Carlos Adam Conte-Junior
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Gao J, Gu Z. The Role of Peroxisome Proliferator-Activated Receptors in Kidney Diseases. Front Pharmacol 2022; 13:832732. [PMID: 35308207 PMCID: PMC8931476 DOI: 10.3389/fphar.2022.832732] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/14/2022] [Indexed: 12/20/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors. Accumulating evidence suggests that PPARs may play an important role in the pathogenesis of kidney disease. All three members of the PPAR subfamily, PPARα, PPARβ/δ, and PPARγ, have been implicated in many renal pathophysiological conditions, including acute kidney injury, diabetic nephropathy, and chronic kidney disease, among others. Emerging data suggest that PPARs may be potential therapeutic targets for renal disease. This article reviews the physiological roles of PPARs in the kidney and discusses the therapeutic utility of PPAR agonists in the treatment of kidney disease.
Collapse
Affiliation(s)
- Jianjun Gao
- Department of Nephrology, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Zhaoyan Gu
- Department of Endocrinology, Second Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhaoyan Gu,
| |
Collapse
|
19
|
Ungur RA, Borda IM, Codea RA, Ciortea VM, Năsui BA, Muste S, Sarpataky O, Filip M, Irsay L, Crăciun EC, Căinap S, Jivănescu DB, Pop AL, Singurean VE, Crișan M, Groza OB, Martiș (Petruț) GS. A Flavonoid-Rich Extract of Sambucus nigra L. Reduced Lipid Peroxidation in a Rat Experimental Model of Gentamicin Nephrotoxicity. MATERIALS (BASEL, SWITZERLAND) 2022; 15:772. [PMID: 35160718 PMCID: PMC8837157 DOI: 10.3390/ma15030772] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 12/04/2022]
Abstract
The use of gentamicin (GM) is limited due to its nephrotoxicity mediated by oxidative stress. This study aimed to evaluate the capacity of a flavonoid-rich extract of Sambucus nigra L. elderflower (SN) to inhibit lipoperoxidation in GM-induced nephrotoxicity. The HPLC analysis of the SN extract recorded high contents of rutin (463.2 ± 0.0 mg mL-1), epicatechin (9.0 ± 1.1 µg mL-1), and ferulic (1.5 ± 0.3 µg mL-1) and caffeic acid (3.6 ± 0.1 µg mL-1). Thirty-two Wistar male rats were randomized into four groups: a control group (C) (no treatment), GM group (100 mg kg-1 bw day-1 GM), GM+SN group (100 mg kg-1 bw day-1 GM and 1 mL SN extract day-1), and SN group (1 mL SN extract day-1). Lipid peroxidation, evaluated by malondialdehyde (MDA), and antioxidant enzymes activity-superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX)-were recorded in renal tissue after ten days of experimental treatment. The MDA level was significantly higher in the GM group compared to the control group (p < 0.0001), and was significantly reduced by SN in the GM+SN group compared to the GM group (p = 0.021). SN extract failed to improve SOD, CAT, and GPX activity in the GM+SN group compared to the GM group (p > 0.05), and its action was most probably due to the ability of flavonoids (rutin, epicatechin) and ferulic and caffeic acids to inhibit synthesis and neutralize reactive species, to reduce the redox-active iron pool, and to inhibit lipid peroxidation. In this study, we propose an innovative method for counteracting GM nephrotoxicity with a high efficiency and low cost, but with the disadvantage of the multifactorial environmental variability of the content of SN extracts.
Collapse
Affiliation(s)
- Rodica Ana Ungur
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Ileana Monica Borda
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Răzvan Andrei Codea
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania;
| | - Viorela Mihaela Ciortea
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Bogdana Adriana Năsui
- Department of Community Health, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Sevastița Muste
- Food Engineering Department, University of Agricultural Sciences and Veterinary Medicine, 64 Calea Floresti, 400509 Cluj-Napoca, Romania; (S.M.); (G.S.M.)
| | - Orsolya Sarpataky
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania;
| | - Miuța Filip
- Raluca Ripan Institute for Research in Chemistry, Babeş-Bolyai University, 30 Fântânele Street, 400294 Cluj-Napoca, Romania;
| | - Laszlo Irsay
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Elena Cristina Crăciun
- Department of Pharmaceutical Biochemistry and Clinical Laboratory, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - Simona Căinap
- Department of Mother and Child, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Delia Bunea Jivănescu
- Department of Internal Medicine, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Anca Lucia Pop
- Department of Clinical Laboratory, Food Safety, Nutrition, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania;
| | - Victoria Emilia Singurean
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Maria Crișan
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Oana Bianca Groza
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Georgiana Smaranda Martiș (Petruț)
- Food Engineering Department, University of Agricultural Sciences and Veterinary Medicine, 64 Calea Floresti, 400509 Cluj-Napoca, Romania; (S.M.); (G.S.M.)
| |
Collapse
|
20
|
El-Sayed K, Ali DA, Maher SA, Ghareeb D, Selim S, Albogami S, Fayad E, Kolieb E. Prophylactic and Ameliorative Effects of PPAR-γ Agonist Pioglitazone in Improving Oxidative Stress, Germ Cell Apoptosis and Inflammation in Gentamycin-Induced Testicular Damage in Adult Male Albino Rats. Antioxidants (Basel) 2022; 11:antiox11020191. [PMID: 35204074 PMCID: PMC8868260 DOI: 10.3390/antiox11020191] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPAR-γ) is ubiquitously expressed in testicular tissue and plays a crucial role in regulating various physiological processes. Pioglitazone (PIO) is one of the PPAR-γ agonists, having anti-oxidant and anti-inflammatory effects. Patients on gentamycin treatment may undergo serious side effects such as testicular damage. To the best of our knowledge, this was the first study to investigate the possible protective anti-inflammatory and anti-apoptotic effects of PIO on gentamycin-induced testicular damage. Fifty adult male Wistar albino rats included in the study as the control group (CTL) received normal saline; a gentamycin-induced testicular damage group (GM) received gentamycin (100 mg/kg); PIO5, PIO10, PIO20 groups received PIO at a dose of 5, 10, and 20 mg/ kg, respectively, for 21 days, and gentamycin was started at day 15 of the experiment for 6 days. The parameters of spermatozoa and histopathological alterations in the testes were significantly improved in the PIO20 group. Moreover, MDA levels, inflammatory mediators, and apoptotic Bax expression were decreased. The activity of glutathione peroxidase, catalase, total antioxidant capacity, and anti-apoptotic Bcl-2 genes expression were increased. It was concluded that PIO20 could protect against gentamycin-induced testicular damage in Wistar rats through its anti-oxidant, anti-inflammatory, and antiapoptotic effects.
Collapse
Affiliation(s)
- Karima El-Sayed
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Dina A. Ali
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Shymaa Ahmed Maher
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Dalia Ghareeb
- Clinical Pathology Department, Faculty of Medicine, Suez University, Suez 41522, Egypt;
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Sarah Albogami
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia; (S.A.); (E.F.)
| | - Eman Fayad
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia; (S.A.); (E.F.)
| | - Eman Kolieb
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Correspondence: ; Tel.: +20-1006738513
| |
Collapse
|
21
|
Renoprotective Effect of Fucoidan from Seaweed Sargassum angustifolium C. Agardh 1820 on Gentamicin-Induced Nephrotoxicity: From Marine Resources to Therapeutic Uses. Jundishapur J Nat Pharm Prod 2021. [DOI: 10.5812/jjnpp.119081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Nephrotoxicity is a major side effect of aminoglycoside antibiotics, caused by oxidative damage and inflammation. Fucoidan, a group of sulfated polysaccharides derived from different species of brown algae, are well recognized for their antioxidant and anti-inflammatory activities. Objectives: In the present study, we aimed to investigate, for the first time, the efficacy of fucoidan extracted from Sargassum angustifolium C. Agardh 1820 against gentamicin-induced nephrotoxicity in rats. Methods: Twenty-eight male Wistar rats were divided into 4 groups of control, gentamicin (100 mg/kg), and gentamicin plus 50- and 100-mg/kg/day fucoidan pretreatment. In the end, all rats were killed, and then urine, blood, and tissue samples were prepared. Kidney weight (KW), body weight (BW), and 24-hour urine volume, as well as serum creatinine (Cr), blood urea nitrogen (BUN), Cr clearance, and malondialdehyde (MDA) levels and superoxide dismutase (SOD) activity, were measured. Kidney samples were also evaluated for histopathological changes. Results: Gentamicin significantly increased KW, KW/BW ratio, 24-hour urine volume, serum Cr, MDA, and BUN levels; however, fucoidan pretreatment, especially at a dose of 50 mg/kg, significantly returned these variables near to the control group values. Gentamicin also decreased BW gain, Cr clearance, SOD activity, and the degree of renal tissue damage compared to the control group, while treatment with fucoidan significantly reversed these alterations. Conclusions: The results show that fucoidan from S. angustifolium C. Agardh 1820 ameliorates gentamicin-induced nephrotoxicity by alleviating oxidative stress and augmenting antioxidant enzymes activity in renal tissue, suggesting the potential use of this fucoidan in a clinical setting.
Collapse
|
22
|
Tomşa AM, Răchişan AL, Pandrea SL, Benea A, Uifălean A, Parvu AE, Junie LM. Accelerated lipid peroxidation in a rat model of gentamicin nephrotoxicity. Exp Ther Med 2021; 22:1218. [PMID: 34584563 DOI: 10.3892/etm.2021.10652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
Kidney disease represents a burden for the health care system worldwide. As the prevalence continues to rise, discovering new biomarkers of early kidney damage has become crucial. Oxidative stress (OS) represents one of the main factors involved in the early stages of many syndromes leading to kidney damage. Therefore, it must be studied in detail. To date, many studies have focused on OS in advanced stages of acute kidney injury (AKI), with great success. The aim of the present study was to ascertain whether even mild renal function impairment can be linked to specific systemic markers of OS and systemic antioxidants in order to pinpoint certain biomarkers for early kidney damage. We used male rats (Rattus norvegicus) in which we induced kidney damage by injecting gentamicin for 7 days. Blood was collected 24 h after the last dose of gentamicin. Urea, creatinine, 3-nitrotyrosine (3-NT), nitric oxide (NO), malondialdehyde (MDA), thiols (TS), total oxidative stress (TOS), and interferon-γ (IFN-γ) were determined. In addition, for the antioxidant status we measured total antioxidant capacity (TAC) and interleukin-10 (IL-10). Our results demonstrated that the rats had mild renal impairment consistent with a pre-AKI stage due to the nephrotoxic effect of gentamicin. However, TOS, MDA and NO were significantly higher in the gentamicin group compared to the control group. In addition, TAC was higher in the control group. Hence, OS markers reach higher levels and may potentially be used as markers of kidney damage even in cases of mild renal function impairment.
Collapse
Affiliation(s)
- Anamaria Magdalena Tomşa
- Department 9-Mother and Child, Second Clinic of Pediatrics, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400177 Cluj-Napoca, Romania.,Department of Microbiology, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andreea Liana Răchişan
- Department 9-Mother and Child, Second Clinic of Pediatrics, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400177 Cluj-Napoca, Romania
| | - Stanca Lucia Pandrea
- Department of Microbiology, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania.,Laboratory Department, 'Prof. Dr. Octavian Fodor' Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Andreea Benea
- Laboratory Department, 'Prof. Dr. Octavian Fodor' Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Ana Uifălean
- Department of Pathophysiology, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alina Elena Parvu
- Department of Pathophysiology, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Lia Monica Junie
- Department of Microbiology, 'Iuliu Haţieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
23
|
Kaur T, Singh D, Pathak D, Singh AP, Singh B. Umbelliferone attenuates glycerol-induced myoglobinuric acute kidney injury through peroxisome proliferator-activated receptor-γ agonism in rats. J Biochem Mol Toxicol 2021; 35:e22892. [PMID: 34409680 DOI: 10.1002/jbt.22892] [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: 06/17/2021] [Accepted: 08/11/2021] [Indexed: 11/07/2022]
Abstract
Rhabdomyolysis is a clinical syndrome caused by damage to skeletal muscle, which consequently releases breakdown products into circulation and causes acute kidney injury (AKI) in humans. Intramuscular injection of glycerol mimics rhabdomyolysis and associated AKI. In this study, we explored the role of umbelliferone against glycerol-induced AKI in rats. Kidney function was assessed by measuring serum creatinine, urea, electrolytes, and microproteinuria. Renal oxidative stress was quantified using thiobarbituric acid reactive substances, superoxide anion generation, and reduced glutathione assay. Renal histological changes were determined using periodic acid Schiff and hematoxylin-eosin staining, and immunohistology of apoptotic markers (Bax, Bcl-2) was done. Serum creatine kinase was quantified to assess glycerol-induced muscle damage. Umbelliferone attenuated glycerol-induced change in biochemical parameters, oxidative stress, histological alterations, and renal apoptosis. Pretreatment with bisphenol A diglycidyl ether, a peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist, attenuated umbelliferone-mediated protection. It is concluded that umbelliferone attenuates glycerol-induced AKI possibly through PPAR-γ agonism in rats.
Collapse
Affiliation(s)
- Tajpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Devendra Pathak
- Department of Veterinary Anatomy, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Amrit P Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
24
|
Alzoubi K, Khabour O, Alfaqih M, Tashtoush M, Al-Azzam S, Mhaidat N, Alrabadi N. The protective effects of pioglitazone against cognitive impairment caused by L-Methionine administration in a rat model. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:77-84. [PMID: 34370649 DOI: 10.2174/1871527320666210809122523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/02/2021] [Accepted: 04/30/2021] [Indexed: 11/22/2022]
Abstract
PURPOSE Accumulating evidence indicates that elevated levels of methionine are associated with cognitive decline including loss of memory. The exact mechanisms behind this observation are not completely understood but could be related to an increase in oxidative stress markers in hippocampal tissues. The above increase in oxidative stress could be directly caused by an increase in the blood levels of methionine (hypermethioninemia) or one of its metabolites, such as homocysteine. Pioglitazone is a drug primarily used for the treatment of type 2 diabetes mellitus. Several reports showed that using pioglitazone protects against cognitive decline observed in Alzheimer's disease. Pioglitazone has antioxidant properties independent of its hypoglycemic effects. Taken together, we hypothesized that pioglitazone protects against memory loss triggered by elevated levels of methionine through lowering of oxidative stress in the hippocampus. METHOD To test this hypothesis, we used chronic administration of L-methionine in a rat model. Spatial learning and memory were evaluated in the model using a radial arm water maze (RAWM). The levels of several markers related to oxidative stress were measured in hippocampal tissues recovered from experimental rats. RESULTS Current results showed that administration of L-methionine was associated with a significant loss of short- and long-term memory and an increase in blood homocysteine levels. The above memory changes were associated with an increase in lipid peroxidation and a decrease in the activity of catalase and glutathione peroxidase antioxidant enzymes in the hippocampus. The combined treatment of pioglitazone with L-methionine protected rat model from memory loss. It also prevented changes observed in lipid peroxidation and changes in the activity of catalase and glutathione peroxidase enzymes. CONCLUSION Current findings indicate that pioglitazone is a viable therapeutic option that protects against cognitive changes observed upon administration of L-methionine.
Collapse
Affiliation(s)
- Karem Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Omar Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid. Jordan
| | - Mahmoud Alfaqih
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Murad Tashtoush
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Sayer Al-Azzam
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Nizar Mhaidat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid-22110. Jordan
| | - Nasr Alrabadi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid-22110. Jordan
| |
Collapse
|
25
|
Babaeenezhad E, Nouryazdan N, Nasri M, Ahmadvand H, Moradi Sarabi M. Cinnamic acid ameliorate gentamicin-induced liver dysfunctions and nephrotoxicity in rats through induction of antioxidant activities. Heliyon 2021; 7:e07465. [PMID: 34278037 PMCID: PMC8264605 DOI: 10.1016/j.heliyon.2021.e07465] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/29/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022] Open
Abstract
This study was the first to evaluate the possible protective effects of cinnamic acid (CA) against Gentamicin (GM) induced liver and kidney dysfunctions in rats. Adult male Wistar rats were randomly assigned to 4 equal groups (n = 8): Control group (saline, 0.5 ml/day), CA group (CA, 50 mg/kg/day), GM group (GM, 100 mg/kg/day), and GM + CA group (100 & 50 mg/kg/day). Following 12 days of treatments, blood and 24 h urine samples were collected and kidneys were taken out for biochemical, histopathological, and molecular studies. Following CA treatment, renal function markers and transaminases activities including serum urea (59.92%) and creatinine (50.41%), protein excretion rate (43.67%), and serum activities of aspartate aminotransferase (AST) (54.34%) and alanine aminotransferase (ALT) (47.26%) significantly reduced in the treated group as compared with the GM group (P < 0.05). Also, CA could significantly ameliorate the levels of triglyceride (29.70%), cholesterol (13.02%), very low-density lipoprotein (29.69%) and high-density lipoprotein-cholesterol (7.28%). CA could also attenuate oxidative stress through a decrease of serum malondialdehyde (MDA) (50.86%) and nitric oxide (NO) (0.85%) and an increase of renal catalase (CAT) (196.14%) and glutathione peroxidase (GPX) activities (45.88%) as well as GPX mRNA expression (44.42-fold) as compared with the GM group (P < 0.05). Moreover, histopathological evaluations revealed attenuated tubular damages and reduced inflammatory cellular infiltration in CA treated animals. Overall, CA alleviates GM-induced nephrotoxicity and alterations in transaminases activities in rats through its antioxidant activities.
Collapse
Affiliation(s)
- Esmaeel Babaeenezhad
- Department of Clinical Biochemistry, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Nouryazdan
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Maryam Nasri
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hassan Ahmadvand
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mostafa Moradi Sarabi
- Department of Clinical Laboratory Sciences, School of Allied Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
26
|
Telek V, Erlitz L, Caleb I, Nagy T, Vecsernyés M, Balogh B, Sétáló G, Hardi P, Jancsó G, Takács I. Effect of Pioglitazone on endoplasmic reticulum stress regarding in situ perfusion rat model. Clin Hemorheol Microcirc 2021; 79:311-325. [PMID: 33867357 DOI: 10.3233/ch-211163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) can cause insufficient microcirculation of the transplanted organ and results in a diminished and inferior graft survival rate. OBJECTIVE This study aimed to investigate the effect of different doses of an anti-diabetic drug, Pioglitazone (Pio), on endoplasmic reticulum stress and histopathological changes, using an in situ perfusion rat model. METHODS Sixty male Wistar rats were used and were divided into six groups, consisting of the control group, vehicle-treated group and four Pio-treated groups (10, 20, 30 and 40 mg/kg Pio was administered). The rats were perfused through vena cava and an outflow on the abdominal aorta occurred. Following the experiment, kidneys and livers were collected. The level of the endoplasmic reticulum stress markers (XBP1 and Caspase 12) was analyzed using Western blot and histopathological changes were evaluated. RESULTS Histopathological findings were correlated with the Western blot results and depict a protective effect corresponding to the elevated dosage of Pioglitazone regarding in situ perfusion rat model. CONCLUSIONS In our study, Pioglitazone can reduce the endoplasmic reticulum stress, and the most effective dosage proved to be the 40 mg/kg Pio referencing the kidney and liver samples.
Collapse
Affiliation(s)
- Vivien Telek
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Luca Erlitz
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Ibitamuno Caleb
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Tibor Nagy
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Mónika Vecsernyés
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Bálint Balogh
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - György Sétáló
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary.,Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, Hungary
| | - Péter Hardi
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Gábor Jancsó
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Ildikó Takács
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
27
|
Ali FEM, Sayed AM, El-Bahrawy AH, Omar ZMM, Hassanein EHM. Targeting KEAP1/Nrf2, AKT, and PPAR-γ signals as a potential protective mechanism of diosmin against gentamicin-induced nephrotoxicity. Life Sci 2021; 275:119349. [PMID: 33744325 DOI: 10.1016/j.lfs.2021.119349] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 03/01/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022]
Abstract
AIM Gentamicin (GM) is an aminoglycoside antibiotic effectively used for severe/life-threatening infections. However, the clinical application of GM is limited by nephrotoxic side effects. Diosmin (DS) is a flavonoid with a wide range of bioactivities. However, its therapeutic potential in GM-induced nephrotoxicity remains unclear. METHODS Rats received GM (100 mg/kg, i.p.) for 7 days either separately or in combination with oral DS (50 mg/kg). RESULTS GM injection disrupted kidney function along with oxidant/antioxidant imbalance. Also, GM significantly decreased renal nuclear factor erythroid 2-related factor 2 (Nrf2), glutamyl cysteine synthetase (GCLC), heme oxygenase-1 (HO-1), superoxide dismutase3 (SOD-3), protein kinase B (AKT), and p-AKT expressions along with Kelch-like ECH-associated protein 1 (KEAP1) up-regulation. On the contrary, DS administration significantly attenuated GM-induced kidney dysfunction and restored kidney oxidant/antioxidant status. In addition, co-treatment with DS plus GM significantly enhanced Nrf2, GCLC, HO-1, SOD3, AKT, and p-AKT expressions along with KEAP1 down-regulation. Additionally, GM-treated rats exhibited a significant decrease in the expressions of renal peroxisome-proliferator activated receptor-gamma (PPAR-γ) and this reduction was alleviated by DS treatment. Furthermore, histopathological findings demonstrated that DS significantly reduced the GM-induced histological abrasions. Besides, an in-silico study was conducted to confirm our biochemical results. Interestingly, in-silico results strongly supported our biochemical investigation by studying the binding affinity of DS to KEAP1, AKT, and PPAR-γ proteins. SIGNIFICANCE DS could be a promising protective agent against GM-induced nephrotoxicity through targeting of KEAP1/Nrf2/ARE, AKT, and PPAR-γ signaling pathways.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Egypt
| | - Ali H El-Bahrawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Zainab M M Omar
- Department of Pharmacology, College of Medicine, Al-Azhar University, Assiut 71524, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
28
|
Chandekar KV, Shkir M, Alshahrani T, Ibrahim EH, Kilany M, Ahmad Z, Manthrammel MA, AlFaify S, Kateb B, Kaushik A. One-spot fabrication and in-vivo toxicity evaluation of core-shell magnetic nanoparticles. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111898. [PMID: 33641901 DOI: 10.1016/j.msec.2021.111898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/01/2020] [Accepted: 01/17/2021] [Indexed: 12/17/2022]
Abstract
This research, for the first time, report the synthesis of core-shell magnetic nanoparticles (NPs) consisting poly acrylic acid (PAA) coated cobalt ferrite (CF) using a simple co-precipitation route. Nanocrystalline PAA@CF-NPs, particle size of 9.2 nm, exhibited saturation magnetization as 28.9 emu/g, remnant magnetization as 8.37 emu/g, and coercivity as 543 Oe. Keeping biomedical applications into consideration, PAA@CF-NPs were further analysed to evaluate antimicrobial performance against Gram positive (Staphylococcus aureus and Bacillus subtilis) and Gram negative (Pseudomonas aeruginosa and Escherichia coli) bacteria, and biocompatibility with reference to activated splenic cells. The PAA@CF-NPs were viable to the normal splenic cells (up to 1000 μg/ml) and do not affect the ability of fast dividing ability of the cells (activated splenic cells). An optimized dose of PAA@CF-NPs was intramuscularly administrated (100 μg/ml) into Albino mice to evaluate acute toxicity. The results of these studies suggest that injected PAA@CF-NPs do not affect vital organs mainly including liver and kidneys that confirmed the heptic/renal biocompatibility. The outcomes of this research project such developed nano-system for biomedical applications, mainly for magnetically guided drug delivery and image guided therapies development. However, to support the proposed claims, extended in-vivo studies are required to explore bio-distribution, chronic toxicity, and homeostatic conditions.
Collapse
Affiliation(s)
- Kamlesh V Chandekar
- Department of Physics, Rayat Shikshan Sanstha's, Karmaveer Bhaurao Patil College, Vashi, Navi Mumbai 400703, Maharashtra, India.
| | - Mohd Shkir
- Advanced Functional Materials and Optoelectronics Laboratory (AFMOL), Department of Physics, College of Science, King Khalid University, Abha 61413, Saudi Arabia.
| | - Thamraa Alshahrani
- Department of Physics, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia.
| | - Essam H Ibrahim
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Research Centre for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo 12611, Egypt
| | - Mona Kilany
- Research Centre for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Department of Microbiology, National Organization for Drug Control and Research (NODCAR), Cairo 12611, Egypt
| | - Zubair Ahmad
- Research Centre for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia; Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Saudi Arabia
| | - M Aslam Manthrammel
- Advanced Functional Materials and Optoelectronics Laboratory (AFMOL), Department of Physics, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - S AlFaify
- Advanced Functional Materials and Optoelectronics Laboratory (AFMOL), Department of Physics, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Babak Kateb
- National Center for NanoBioElectronics, West Hollywood, CA, USA; California Neurosurgical Institute, Los Angeles, CA, USA; Brain Mapping Foundation, West Hollywood, CA, USA; Society for Brain Mapping and Therapeutics, West Hollywood, CA, USA
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Natural Sciences, Division of Sciences, Art and Mathematics, Florida Polytechnic University, Lakeland, FL 33805, USA.
| |
Collapse
|
29
|
Ibrahim EH, Ghramh HA, Alshehri A, Kilany M, Khalofah A, El-Mekkawy HI, Sayed MA, Alothaid H, Taha R. Lepidium sativum and Its Biogenic Silver Nanoparticles Activate Immune Cells and Induce Apoptosis and Cell Cycle Arrest in HT-29 Colon Cancer Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2551] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
There is an increased demand for plants with antioxidants and anticancer properties. Lepidium sativum L. is an edible plant with medical importance. In this study, we aimed to investigate the anticancer activity; antioxidant capacity and antibacterial impact of Lepidium sativum
L. seed acetone extract (LSSAExt), alone and with its biogenic silver nanoparticles (AgNPs). LSSAExt-produced AgNPs were characterized using SEM, XRD and Vis/UV analysis. Biomolecules in LSSAExt and LSSAExt + AgNPs were explored utilizing FTIR. The ability of LSSAExt and LSSAExt + AgNPs to
induce apoptosis and mitotic cell arrest in the HT-29 colon cancer cells, compared to normal and repeated cell division activated splenic cells was determined by florescent stains and flow cytometry. Antibacterial power was tested using well diffusion technique. LSSAExt and LSSAExt + AgNPs
showed a good antibacterial impact. LSSAExt contains ROS, which could help in cancer cells apoptosis. LSSAExt and LSSAExt+AgNPs were not toxic to splenic cells and increased the rate of their cell division. LSSAExt and LSSAExt+AgNPs increased p53 expression and could arrest cell division of
HT-29 colon cancer cells but not of normal fast dividing cells. LSSAExt and LSSAExt+AgNPs caused apoptosis in cancer cells rather than necrosis. In conclusion, acetone preparation of the edible plant L. sativum is a good antibacterial agent, good anticancer preparation at least against
colon cancer as it is shown to be targeted, effective and can boost immune cells.
Collapse
Affiliation(s)
- Essam H. Ibrahim
- Biology Department, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Hamed A. Ghramh
- Biology Department, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Ali Alshehri
- Biology Department, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Mona Kilany
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
| | - Ahlam Khalofah
- Biology Department, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Haitham I. El-Mekkawy
- Biology Department, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Mahmoud A. Sayed
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, 4781, Saudi Arabia
| | - Ramadan Taha
- Biology Department, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
| |
Collapse
|
30
|
Resveratrol reduces gentamicin-induced EMT in the kidney via inhibition of reactive oxygen species and involving TGF-β/Smad pathway. Life Sci 2020; 258:118178. [PMID: 32739468 DOI: 10.1016/j.lfs.2020.118178] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
AIMS Gentamicin (GEN) is one of the most valuable aminoglycoside antibiotics utilized against life-threatening bacterial infections. Unfortunately, GEN-induced nephrotoxicity limited its clinical utility. The pathologic process of nephrotoxicity caused by GEN may involve epithelial to mesenchymal transition (EMT). Resveratrol (RES) is a natural compound was revealed to inhibit EMT in kidney. The present work was conducted to explore the potential renoprotective role of RES on GEN-induced EMT. Moreover, the underlying signaling pathway of this inhibition was investigated. MAIN METHODS Mice were treated with GEN by intraperitoneal (i.p.) route daily for 15 days to identify EMT onset with regard to GEN-induced nephrotoxicity. To assess the ameliorative role of RES against GEN-induced EMT, RES was i.p. administrated in high and low doses before and concurrently with GEN treatment. KEY FINDINGS GEN administration significantly deteriorated kidney functions. In addition, reduced glutathione (GSH) content and catalase (CAT) activity were significantly decreased with a concomitant increase in the content of kidney malondialdehyde (MDA) after GEN treatment. Histological changes and deposition of collagen were extensive in renal corpuscles and tubules. Increased expression of alpha smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1) and phosphorylated (p)-Smad2 were observed after GEN administration, while E-cadherin expression was decreased. On the contrary, pretreatment with both doses of RES reversed the modifications caused by GEN administration. SIGNIFICANCE We concluded that EMT contributes to pathogenesis of GEN-induced nephrotoxicity. RES has a protective effect on GEN-induced EMT via suppressing oxidative stress and a possible involvement of TGF-β/Smad signaling pathway.
Collapse
|
31
|
Dual soluble epoxide hydrolase inhibitor/PPAR-γ agonist attenuates renal fibrosis. Prostaglandins Other Lipid Mediat 2020; 150:106472. [PMID: 32569747 DOI: 10.1016/j.prostaglandins.2020.106472] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is a contributor to chronic kidney disease and an important predictor of long-term prognosis. We developed a dual soluble epoxide hydrolase inhibitor-PPAR-γ agonist (sEHi/PPAR-γ), RB394, and investigated its ability to attenuate renal fibrosis in a mouse unilateral ureteral obstruction (UUO) model. RB394 efficacy was compared to an sEH inhibitor (sEHi), a PPAR-γ agonist rosiglitazone (Rosi), or their combination (sEHi + Rosi). All interventional treatments were administrated in drinking water 3 days after UUO induction surgery and continued for 7 days. UUO mice developed renal fibrosis with higher collagen formation and RB394 significantly attenuated fibrosis (P < 0.05). Renal expression of α-smooth muscle actin (α-SMA) was elevated in UUO mice and all treatments except sEHi significantly attenuated renal α-SMA expression. Renal mRNA expression fibrotic and fibrosis regulators were higher in UUO mice and RB394 and sEHi + Rosi treatments attenuated their expression. Renal inflammation was evident in UUO mice with increased infiltration of CD45 and F4/80 positive cells. RB394 and sEHi + Rosi treatments attenuated renal inflammation in UUO mice. UUO mice had renal tubular and vascular injury. Renal tubular and vascular injuries were attenuated to a greater extent by RB394 and sEHi + Rosi than sEHi or Rosi treatment alone. Renal mRNA expression of oxidative stress markers were significantly higher in UUO mice (P < 0.05). RB394 and sEHi + Rosi attenuated expression of oxidative stress markers to a greater extent than other interventional treatments (P < 0.05). These findings demonstrate that RB394 can attenuate renal fibrosis by reducing renal inflammation, oxidative stress, tubular injury, and vascular injury. In conclusion, RB394 demonstrates exciting potential as a therapeutic for renal fibrosis and chronic kidney disease.
Collapse
|