1
|
Yang S, Guo J, Chen D, Sun Z, Pu L, Sun G, Yang M, Peng Y. The Cardioprotective Effect of Ginseng Derived Exosomes via Inhibition of Oxidative Stress and Apoptosis. ACS APPLIED BIO MATERIALS 2025; 8:814-824. [PMID: 39740230 DOI: 10.1021/acsabm.4c01644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Ginsenosides possess potential protective effects against cisplatin (CDDP)-induced toxicity, but the limited bioavailability of ginsenosides hampered their therapeutic application. Ginseng exosomes (G-Exo), which are active ingredients in ginseng, exhibit excellent biocompatibility and low immunogenicity. Here, G-Exo were isolated from ginseng roots through a combination of ultracentrifugation and sucrose gradient centrifugation techniques. Subsequently, the potential protective effect of G-Exo on CDDP induced cardiotoxicity, and its underlying mechanisms were explored. The findings demonstrated that G-Exo effectively mitigated CDDP-induced oxidative stress and apoptosis in vitro. Moreover, in vivo experiments revealed that G-Exo significantly inhibited the increases in serum cardiac troponin T (cTnT), creatine kinase (CK), and lactate dehydrogenase (LDH) levels in mice induced by CDDP. Histological assessment and tissue staining further corroborated that G-Exo alleviated the cardiac tissue damage and apoptosis caused by CDDP. Mechanistically, G-Exo were found to alleviate CDDP-induced apoptosis through blocking the MAPK signaling. Collectively, these results suggest that G-Exo hold the potential to mitigate cisplatin-induced cardiac injury by regulating the MAPK pathway, thereby highlighting the therapeutic potential of G-Exo as a protective agent against CDDP-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shuiyue Yang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Jia Guo
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
| | - Danyang Chen
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
| | - Zepeng Sun
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Li Pu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Guoying Sun
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Min Yang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
| | - Yinghua Peng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
| |
Collapse
|
2
|
El-Dessouki AM, Alzokaky AA, Raslan NA, Ibrahim S, Salama LA, Yousef EH. Piracetam mitigates nephrotoxicity induced by cisplatin via the AMPK-mediated PI3K/Akt and MAPK/JNK/ERK signaling pathways. Int Immunopharmacol 2024; 137:112511. [PMID: 38909496 DOI: 10.1016/j.intimp.2024.112511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/08/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024]
Abstract
AIMS Cisplatin (CDDP) is commonly employed as an antineoplastic agent, but its use is significantly limited by the occurrence of dose-dependent nephrotoxicity, the detailed mechanisms of which remain unclear. This research is aimed to explore the molecular mechanisms of Piracetam (PIR)'s protective effects on nephrotoxicity resulting from CDDP exposure and to elucidate the mechanisms responsible for these effects. MAIN METHODS PIR was given in dosages of 100 and 300 mg/kg body weight for a duration of 15 days; concurrently, on the last day, a single 10 mg/kg dose of CDDP was delivered via intraperitoneal injection. Forty-eight hours post-CDDP injection, the animals were sacrificed to assess nephrotoxicity. Blood samples and renal tissues were taken for biochemical and histopathological investigations. Serum creatinine and blood urea nitrogen (BUN) were measured. AMP-activated protein kinase (AMPK), caspase-9 and nuclear factor kappa b p65 (NF-κB p65) were assessed by immunohistochemistry method. Enzyme-linked immunosorbent assay (ELISA) analysis was employed to determine cytochrome c (Cyt. c), Bcl-2-associated X-protein (BAX), caspase-3, nuclear factor erythroid 2-related factor 2 (Nrf2), Heme oxygenase-1 (HO-1), superoxide dismutase (SOD), tumor necrosis factor alpha (TNF-α), myeloperoxidase (MPO), and interleukin-1β (IL-1β) levels in renal tissue homogenates. The mRNA levels of tumor protein P53 (TP53), phosphatidylinositol-3 kinase (PI3K), protein kinase B (Akt), p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinases (ERK), and c-Jun N-terminal kinases (JNK) were tested by quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, histopathological evaluations of the renal tissues and the binding affinity of PIR to AMPK by molecular docking were also performed. KEY FINDINGS Pre-treatment with PIR enhanced renal function markers such as urea and creatinine, mitigated histological damage, and diminished inflammatory cell presence in renal tubules. PIR demonstrated antioxidant effects by reestablishing the equilibrium between pro-oxidants and antioxidants such as MPO, HO-1, Nrf2, as well as SOD. Furthermore, PIR inhibited the inflammatory pathways through the MAPK/NF-κB pathway. Additionally, PIR counteracted the CDDP-induced decline in PI3K/Akt activity and hindered caspase-dependent apoptotic processes. SIGNIFICANCE In summary, PIR appears to be an effective therapeutic strategy for reducing CDDP-induced nephrotoxicity, attributed to its antioxidant, anti-inflammatory, and antiapoptotic mechanisms. Consequently, PIR may serve as a complementary treatment alongside CDDP to alleviate nephrotoxicity associated with CDDP.
Collapse
Affiliation(s)
- Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 12566, Giza, Egypt
| | - Amany A Alzokaky
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt; Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Nahed A Raslan
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt; Clinical Pharmacy Program, College of Health Sciences and Nursing, Al-Rayan Colleges, Madina, Saudi Arabia
| | - Samar Ibrahim
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Galala University, Ataka, Egypt
| | - Lamiaa A Salama
- Department of Microbiology and Immunology, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Eman H Yousef
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt.
| |
Collapse
|
3
|
Lyrio RMDC, Rocha BRA, Corrêa ALRM, Mascarenhas MGS, Santos FL, Maia RDH, Segundo LB, de Almeida PAA, Moreira CMO, Sassi RH. Chemotherapy-induced acute kidney injury: epidemiology, pathophysiology, and therapeutic approaches. FRONTIERS IN NEPHROLOGY 2024; 4:1436896. [PMID: 39185276 PMCID: PMC11341478 DOI: 10.3389/fneph.2024.1436896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024]
Abstract
Despite significant advancements in oncology, conventional chemotherapy remains the primary treatment for diverse malignancies. Acute kidney injury (AKI) stands out as one of the most prevalent and severe adverse effects associated with these cytotoxic agents. While platinum compounds are well-known for their nephrotoxic potential, other drugs including antimetabolites, alkylating agents, and antitumor antibiotics are also associated. The onset of AKI poses substantial risks, including heightened morbidity and mortality rates, prolonged hospital stays, treatment interruptions, and the need for renal replacement therapy, all of which impede optimal patient care. Various proactive measures, such as aggressive hydration and diuresis, have been identified as potential strategies to mitigate AKI; however, preventing its occurrence during chemotherapy remains challenging. Additionally, several factors, including intravascular volume depletion, sepsis, exposure to other nephrotoxic agents, tumor lysis syndrome, and direct damage from cancer's pathophysiology, frequently contribute to or exacerbate kidney injury. This article aims to comprehensively review the epidemiology, mechanisms of injury, diagnosis, treatment options, and prevention strategies for AKI induced by conventional chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Felipe Luz Santos
- Department of Medicine, Universidade Salvador (UNIFACS), Salvador, Brazil
| | | | | | | | | | - Rafael Hennemann Sassi
- Hematology Department, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
4
|
Mohamed B, Ghareib SA, Alsemeh AE, El-Sayed SS. Telmisartan ameliorates nephropathy and restores the hippo pathway in rats with metabolic syndrome. Eur J Pharmacol 2024; 973:176605. [PMID: 38653362 DOI: 10.1016/j.ejphar.2024.176605] [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: 11/05/2023] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
The main objective of this study was to determine if the telmisartan-ameliorative effects of metabolic syndrome (MetS)-evoked nephropathy are attributed to the Hippo pathway. A secondary objective was to investigate the potential of vitamin D3 to enhance telmisartan-favourable effects. A diet composed of 24% fat and 3% salt, along with drinking water containing 10% fructose, was administered for 12 weeks to induce MetS. MetS-rats were given telmisartan (5 mg/kg/day), vitamin D3 (10 μg/kg/day) or both by gavage, starting in the sixth week of experimental diet administration. Assessments performed at closure included renal function, histological examination, catalase, malondialdehyde (MDA), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6), peroxisome proliferator-activated receptor-γ (PPAR-γ), phosphatase and tensin homolog (PTEN), and transforming growth factor-β (TGF-β). Matrix metalloproteinase-9 (MMP-9) immunostaining was conducted. The expression of the Hippo pathway components, as well as that of angiotensin II type 1 and type 2 (AT1 and AT2), receptors was evaluated. Telmisartan attenuated MetS-evoked nephropathy, as demonstrated by improvement of renal function and histological features, enhancement of catalase, reduction of MDA, inflammation (NF-κB, IL-6), and renal fibrosis (increased PPAR-γ and PTEN and reduced MMP-9 and TGF-β). Telmisartan downregulated AT1-receptor, upregulated AT2-receptor and restored the Hippo pathway. Vitamin D3 replicated most of the telmisartan-elicited effects and enhanced the antifibrotic actions of telmisartan. The alleviative effects of telmisartan on MetS-evoked nephropathy may be related to the restoration of the Hippo pathway. The combination of vitamin D3 and telmisartan exerted more favourable effects on metabolic and nephropathic biomarkers compared with either one administered alone.
Collapse
Affiliation(s)
- Badria Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Salah A Ghareib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Amira Ebrahim Alsemeh
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Shaimaa S El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
5
|
McSweeney KR, Gadanec LK, Kubatka P, Caprnda M, Gaspar L, Prosecky R, Delev D, Kruzliak P, Apostolopoulos V, Zulli A. Cisplatin treatment reduces contraction to angiotensin II by altering expression of angiotensin II receptors: a pilot study. Mol Cell Biochem 2023; 478:2907-2916. [PMID: 37004639 DOI: 10.1007/s11010-023-04706-2] [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: 12/04/2022] [Accepted: 03/10/2023] [Indexed: 04/04/2023]
Abstract
The renin angiotensin system is a key regulator of blood pressure homeostasis. Angiotensin type 1 (AT1R) and 2 receptors (AT2R) have been investigated as targets for cisplatin-induced acute kidney injury; however, their therapeutic potential remains inconclusive. This pilot study aimed to determined the effect that acute cisplatin treatment had on angiotensin II (AngII)-induced contraction in blood vessels and expression profiles of AT1R and AT2R in mouse arteries and kidneys. Male C57BL/6 mice at 18 week of age (n = 8) were treated with vehicle or bolus dose of cisplatin (12.5 mg/kg). Thoracic aorta (TA), adnominal aorta (AA), brachiocephalic arteries (BC), iliac arteries (IL) and kidneys were collected for isometric tension and immunohistochemistry analysis. Cisplatin treatment reduced IL contraction to AngII at all doses (p < 0.01, p < 0.001, p < 0.0001); however, AngII did not induce contraction in TA, AA or BC in either treatment group. Following cisplatin treatment, AT1R expression was significantly upregulated in the media of TA (p < 0.0001) and AA (p < 0.0001), and in the endothelium (p < 0.05) media (p < 0.0001) and adventitia (p < 0.01) of IL. Cisplatin treatment significantly reduced AT2R expression in the endothelium (p < 0.05) and media (p < 0.05) of TA. In renal tubules, both AT1R (p < 0.01) and AT2R (p < 0.05) were increased following cisplatin treatment. Herein, we report that cisplatin reduces AngII-mediated contraction in IL and may be explained by an absence of normal counterregulatory expression of AT1R and AT2R, indicating other factors are involved.
Collapse
Affiliation(s)
| | - Laura Kate Gadanec
- Institute of Health and Sport, Victoria University, Melbourne, Vic, Australia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Ludovit Gaspar
- Faculty of Health Sciences, University of Ss. Cyril and Methodius in Trnava, Trnava, Slovakia
| | - Robert Prosecky
- 2nd Department of Internal Medicine, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Delian Delev
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Peter Kruzliak
- 2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic.
| | - Vasso Apostolopoulos
- Institute of Health and Sport, Victoria University, Melbourne, Vic, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Anthony Zulli
- Institute of Health and Sport, Victoria University, Melbourne, Vic, Australia.
| |
Collapse
|
6
|
Bekhit AA, Beshay ON, Fawzy MA, Abdel-Hafez SMN, Batiha GES, Ataya FS, Fathy M. Curative Effect of AD-MSCs against Cisplatin-Induced Hepatotoxicity in Rats is Potentiated by Azilsartan: Targeting Oxidative Stress, MAPK, and Apoptosis Signaling Pathways. Stem Cells Int 2023; 2023:6767735. [PMID: 37908315 PMCID: PMC10615573 DOI: 10.1155/2023/6767735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/02/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
Despite its clinical value, cisplatin (CISP) is complicated by marked hepatotoxicity via inducing oxidative stress, inflammatory, and apoptotic pathways. This study aims to explore the protective impact of azilsartan (AZIL), an antihypertensive drug, in addition to adipose tissue-derived mesenchymal stem cells (AD-MSCs) on CISP-induced hepatotoxicity. After characterization and labeling of AD-MSCs by PKH26 dye, 54 Wistar male albino rats were randomly divided into nine groups: I (CONT), II (AZIL.H), III (CISP), IV (CISP + AZIL.L), V (CISP + AZIL.H), VI (CISP + AD-MSCs), VII (CISP + AZIL.L + AD-MSCs), VIII (CISP + AZIL.H + AD-MSCs), and IX (CISP + VITA C). Serum alanine aminotransferase (ALT), alanine aminotransferase (AST), and albumin levels were determined. Assessment of reactive oxygen species, malondialdehyde, and glutathione contents, and superoxide dismutase activity and histopathological evaluations were done on hepatic tissue. Quantitative real-time PCR was utilized to estimate the expression of TNF-α and IL-6 genes. Cell homing of labeled AD-MSCs to the liver tissues was investigated. Hepatic expression of JNK1/2, ERK1/2, p38, Bax, Bcl-2, and cleaved caspase-3 proteins was investigated by western blot analysis. CISP elevated serum ALT and AST activities, reduced albumin level, and remarkably changed the hepatic architecture. It increased the expression TNF-α and IL-6 genes, raised the expression of JNK1/2, ERK1/2, p38, Bax, and cleaved caspase-3 proteins, and diminished the Bcl-2 protein. By contrast, treatment of animals with either AZIL or AD-MSCs dramatically reduced the effects of CISP injection. Moreover, treatment with combination therapy (AZIL.L or H + AD-MSCs) considerably mitigated all previously mentioned alterations superior to AZIL or AD-MSCs alone, which might be attributed to the AZIL-enhanced homing ability of AD-MSCs into the injured liver tissue. In conclusion, the present findings demonstrated that AZIL improves the hepatoprotective potential of AD-MSCs against CISP-induced hepatotoxicity by modulating oxidative stress, mitogen-activated protein kinase, and apoptotic pathways.
Collapse
Affiliation(s)
| | - Olivia N. Beshay
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Michael A. Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | | | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt
| | - Farid S. Ataya
- Department of Biochemistry, College of Science, King Saud University, P.O. Box, 2455, Riyadh 11451, Saudi Arabia
| | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
7
|
Hou YY, Qi SM, Leng J, Shen Q, Tang S, Zhang JT, Hu JN, Jiang S, Li W. Lobetyolin, a Q-marker isolated from Radix Platycodi, exerts protective effects on cisplatin-induced cytotoxicity in HEK293 cells. J Nat Med 2023; 77:721-734. [PMID: 37353674 DOI: 10.1007/s11418-023-01714-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: 10/12/2022] [Accepted: 05/28/2023] [Indexed: 06/25/2023]
Abstract
This study investigated the protective effect of lobetyolin (LBT), a Q-marker isolated from the roots of Platycodon grandiflorum (Radix Platycodi), against cisplatin-induced cytotoxicity in human embryonic kidney (HEK293) cells. Results showed that LBT at 20 μM significantly prevented cisplatin-induced cytotoxicity by improving the viability of HEK293 cells, decreasing levels of MDA, and decreasing GSH content triggered by cisplatin. It also suppressed reactive oxygen species (ROS) levels. Molecular docking analysis revealed a strong binding affinity between LBT and the NF-κB protein, with a docking fraction of - 6.5 kcal/mol. These results provide compelling evidence suggesting a potential link between the visualization analysis of LBT and its protective mechanism, specifically implicating the NF-κB signaling pathway. LBT also reduced the expression level of tumor necrosis factor-alpha (TNF-α), phosphorylation NF-κB and IκBα in HEK293 cells which were increased by cisplatin exposure, leading to inhibition of inflammation. Furthermore, western blotting showed that LBT antagonized the up-regulation of Bax, cleaved caspase 3, 8, and 9 expression and inhibited the MAPK signaling pathway by down-regulating phosphorylation JNK, ERK, and p38, partially ameliorating cisplatin-induced cytotoxicity in HEK293 cells. Therefore, these results indicate that LBT has potentially protected renal function by inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Yun-Yi Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Si-Min Qi
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jing Leng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Qiong Shen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shan Tang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
- National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| |
Collapse
|
8
|
Sharaf G, El Morsy EM, El-Sayed EK. Augmented nephroprotective effect of liraglutide and rabeprazole via inhibition of OCT2 transporter in cisplatin-induced nephrotoxicity in rats. Life Sci 2023; 321:121609. [PMID: 36958435 DOI: 10.1016/j.lfs.2023.121609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
AIMS Cisplatin, a widely used anticancer treatment, has a marked nephrotoxic effect. This nephrotoxic effect is linked to the triggering of oxidative stress, inflammation, activation of mitogen-activated protein kinase (MAPK) pathway as well as apoptosis. The purpose of the present research was to examine the possible ameliorative effect of liraglutide and/or rabeprazole on cisplatin-induced nephrotoxicity in rats and to underline the potential molecular pathways involved. MAIN METHODS Rats were divided into five groups: Control, cisplatin, liraglutide (200 μg/kg/day, i.p), rabeprazole (10 mg/kg/day, orally) and liraglutide + rabeprazole combination groups. All treatments were given for 7 days. Cisplatin was given as a single dose (7 mg/kg, i.p) at day 4 to induce nephrotoxicity in all groups except the control group. KEY FINDINGS Treatment with liraglutide and/or rabeprazole prior to cisplatin maintained the function and morphology of kidney via decreasing cisplatin renal uptake by significant inhibition of OCT2. Besides, they showed a significant increase in GLP-1 receptor expression. Liraglutide and/or rabeprazole significantly attenuated the levels of TNF-α. ICAM, NF-κB, and downregulated MAPK pathway proteins such as JNK, and ERK1/2. Moreover, they maintained oxidant antioxidant balance by decreasing MDA level and increasing GSH level and CAT activity. Additionally, liraglutide and/or rabeprazole exhibited antiapoptotic effect evidenced by the decreased caspase-3 level and Bax expression and the increased Bcl-2 expression. SIGNIFICANCE The current study showed that both liraglutide and rabeprazole exerted a nephroprotective effect against cisplatin-induced renal toxicity in rats. Interestingly, co-treatment with both drugs showed an augmented effect.
Collapse
Affiliation(s)
- Gehad Sharaf
- Nasr Hospital Health Insurance, Helwan, Cairo, Egypt.
| | - E M El Morsy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| | - Elsayed K El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| |
Collapse
|
9
|
Zhang S, Tang J, Sun C, Zhang N, Ning X, Li X, Wang J. Dexmedetomidine attenuates hepatic ischemia-reperfusion injury-induced apoptosis via reducing oxidative stress and endoplasmic reticulum stress. Int Immunopharmacol 2023; 117:109959. [PMID: 36881980 DOI: 10.1016/j.intimp.2023.109959] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Dexmedetomidine (DEX) affords a hepatoprotective effect during ischemia-reperfusion (IR) injury (IRI); however, the underlying mechanism remains elusive. In this work, using a rat liver IR model and a BRL-3A cell hypoxia-reoxygenation (HR) model, we explored whether DEX protects the liver against IRI by decreasing oxidative stress (OS), endoplasmic reticulum stress (ERS), and apoptotic pathways. We found that DEX significantly increased SOD and GSH activity while decreasing ROS and MDA levels in BRL-3A cells, successfully preventing HR-induced OS damage. DEX administration reduced JNK, ERK, and P38 phosphorylation and blocked HR-induced MAPK signaling pathway activation. Additionally, DEX administration reduced the expression of GRP78, IRE1α, XBP1, TRAF2, and CHOP, which reduced HR-induced ERS. NAC prevented the MAPK pathway from being activated and inhibited the ERS pathway. Further research showed that DEX significantly reduced HR-induced apoptosis by suppressing the expression of Bax/Bcl-2 and cleaved caspase-3. Similarly, animal studies demonstrated DEX exerted a protective effect of the liver by alleviating histopathological injury and enhancing liver function, mechanically DEX reduced cell apoptosis in liver tissue by reducing oxidative stress and ERS. In conclusion, DEX mitigates OS and ERS during IR, thereby suppressing cell apoptosis, thus providing protection to the liver.
Collapse
Affiliation(s)
- Shixia Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Jilang Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China.
| | - Chen Sun
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Nuannuan Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Xiaqing Ning
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Xueqin Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Jiaqi Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| |
Collapse
|
10
|
Oliveira BM, de Almeida LF, Deluque AL, Souza CS, Maciel ALD, Francescato HDC, Costa RS, Giovanini C, de Paula FJA, Coimbra TM. Calcitriol Reduces the Inflammation, Endothelial Damage and Oxidative Stress in AKI Caused by Cisplatin. Int J Mol Sci 2022; 23:ijms232415877. [PMID: 36555517 PMCID: PMC9783003 DOI: 10.3390/ijms232415877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/21/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cisplatin treatment is one of the most commonly used treatments for patients with cancer. However, thirty percent of patients treated with cisplatin develop acute kidney injury (AKI). Several studies have demonstrated the effect of bioactive vitamin D or calcitriol on the inflammatory process and endothelial injury, essential events that contribute to changes in renal function and structure caused by cisplatin (CP). This study explored the effects of calcitriol administration on proximal tubular injury, oxidative stress, inflammation and vascular injury observed in CP-induced AKI. Male Wistar Hannover rats were pretreated with calcitriol (6 ng/day) or vehicle (0.9% NaCl). The treatment started two weeks before i.p. administration of CP or saline and was maintained for another five days after the injections. On the fifth day after the injections, urine, plasma and renal tissue samples were collected to evaluate renal function and structure. The animals of the CP group had increased plasma levels of creatinine and of fractional sodium excretion and decreased glomerular filtration rates. These changes were associated with intense tubular injury, endothelial damage, reductions in antioxidant enzymes and an inflammatory process observed in the renal outer medulla of the animals from this group. These changes were attenuated by treatment with calcitriol, which reduced the inflammation and increased the expression of vascular regeneration markers and antioxidant enzymes.
Collapse
Affiliation(s)
- Beatriz M. Oliveira
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Lucas Ferreira de Almeida
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Amanda L. Deluque
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Claudia S. Souza
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Ana Lívia D. Maciel
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Heloísa D. C. Francescato
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Roberto S. Costa
- Department of Medical Clinic, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Cleonice Giovanini
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Francisco José A. de Paula
- Department of Medical Clinic, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
| | - Terezila M. Coimbra
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 140490-900, Sao Paulo, Brazil
- Correspondence: ; Tel.: +55-16-3315–3021
| |
Collapse
|
11
|
Expression of ER stress markers (GRP78 and PERK) in experimental nephrotoxicity induced by cisplatin and gentamicin: roles of inflammatory response and oxidative stress. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:789-801. [PMID: 36482225 DOI: 10.1007/s00210-022-02358-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
This study aimed to establish the relationship between two endoplasmic reticulum (ER) stress proteins, glucose-regulated protein 78 (GRP78/BiP) and PKR-like endoplasmic reticulum kinase (PERK), and oxidative stress markers in cisplatin (CIS)-induced and gentamicin (GEN)-induced nephrotoxicity.The study consisted of five groups: control (saline solution only), CIS D2 (2.5 mg/kg for 2 days), CIS D7 (2.5 mg/kg for 7 days), GEN D2 (160 mg/kg for 2 days), and GEN D7 (160 mg/kg for 7 days). All rats were sacrificed 24 h after the last injection for standard clinical chemistry, and ultrastructural and histological evaluation of the kidney.CIS and GEN increased blood urea nitrogen (BUN) and serum creatinine (Cr) levels, as well as total oxidant status (TOS), while decreasing total antioxidant status (TAS) level in CIS D7 and GEN D7 groups. Histopathological and ultrastructural findings were also consistent with renal tubular damage. In addition, expression of markers of renal inflammation (tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β)) and ER stress markers (GRP78 and PERK) was significantly increased in the kidney tissue of rats treated with CIS and GEN for 7 days.These findings suggest that CIS and GEN administration for 7 days aggravates nephrotoxicity through the enhancement of oxidative stress, inflammation, and ER stress-related markers. As a result, the recommended course of action is to utilize CIS and GEN as an immediate but brief induction therapy, stopping after 3 days and switching to other drugs instead.
Collapse
|
12
|
Chen JY, Tsai CL, Tseng CY, Yu PR, Chang YH, Wong YC, Lin HH, Chen JH. In Vitro and In Vivo Nephroprotective Effects of Nelumbo nucifera Seedpod Extract against Cisplatin-Induced Renal Injury. PLANTS (BASEL, SWITZERLAND) 2022; 11:3357. [PMID: 36501396 PMCID: PMC9737562 DOI: 10.3390/plants11233357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Cisplatin has been considered a chemotherapeutic drug for treating human tumors, and one of the noteworthy side effects of cisplatin is nephrotoxicity. Amelioration of cisplatin-induced nephrotoxicity is necessary. Lotus seedpod extract (LSE) mainly composed of quercetin-3-glucuronide has been revealed for antioxidant and anti-tumor effects. However, the effects of LSE on cisplatin-induced nephrotoxicity are still unknown. This study aims to explore the in vitro and in vivo protective effect and possible mechanism of LSE on cisplatin-induced nephrotoxicity. Results showed that co-treatment of LSE with cisplatin raised the viability of rat renal tubular epithelial NRK-52E cells and decreased oxidative stress and cell apoptosis when compared to the cells treated with cisplatin alone. The molecular mechanisms analyzed found that LSE could reduce the expressions of apoptotic factors, including Bax, Bad, t-Bid, and caspases. In the in vivo study, LSE improved the cisplatin-induced levels of serum markers of kidney function, glomerular atrophy, and the degree of apoptosis in the kidneys. This is the first study to display that LSE prevents cisplatin-induced nephrotoxicity by reducing oxidative stress and apoptosis. Thus, LSE could be a novel and natural chemoprotective agent for cisplatin chemotherapy in the future.
Collapse
Affiliation(s)
- Jui-Yi Chen
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan City 71004, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan City 71710, Taiwan
| | - Chia-Lin Tsai
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Chiao-Yun Tseng
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Pei-Rong Yu
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Yu-Hsuan Chang
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Yue-Ching Wong
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Hui-Hsuan Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Jing-Hsien Chen
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| |
Collapse
|
13
|
Duan YY, Mi XJ, Su WY, Tang S, Jiang S, Wang Z, Zhao LC, Li W. Trilobatin, an Active Dihydrochalcone from Lithocarpus polystachyus, Prevents Cisplatin-Induced Nephrotoxicity via Mitogen-Activated Protein Kinase Pathway-Mediated Apoptosis in Mice. ACS OMEGA 2022; 7:37401-37409. [PMID: 36312396 PMCID: PMC9607670 DOI: 10.1021/acsomega.2c04142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/31/2022] [Indexed: 06/16/2023]
Abstract
Although naturally occurring flavonoids have shown beneficial effects on the side effects caused by cisplatin, there are few reports on the protective effect of dihydrochalcone on the cisplatin-induced toxicity. Trilobatin (TLB), as the major sweetener and active ingredient in Lithocarpus polystachyus Rehd, is a dihydrochalcone-like compound that can be present in concentrations of up to 10% or more in tender leaves. Herein, a cisplatin-induced acute kidney injury (AKI) model was established to investigate the protective effect and mechanism of TLB against the cisplatin-induced nephrotoxicity in mice. The results showed that TLB significantly reversed the inhibition of CRE, BUN, and MDA levels compared with the cisplatin group. Furthermore, TLB treatment (50 and 100 mg/kg) for 10 days significantly alleviated cisplatin-induced renal pathological changes. TUNEL staining showed that TLB administration can effectively improve the occurrence of apoptosis of renal tissue cells caused by cisplatin exposure. Importantly, western blot analysis verified that TLB alleviated cisplatin-induced nephrotoxicity by regulating the AKT/MAPK signaling pathway and apoptosis. In summary, our findings showed clearly that TLB has a significant preventive effect on cisplatin-induced AKI.
Collapse
Affiliation(s)
- Yue-yang Duan
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
and Local Joint Engineering Research Center for Ginseng Breeding and
Development, Changchun 130118, China
| | - Xiao-jie Mi
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Wen-ya Su
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Shan Tang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Shuang Jiang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Zi Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Li-Chun Zhao
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530001, China
| | - Wei Li
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
and Local Joint Engineering Research Center for Ginseng Breeding and
Development, Changchun 130118, China
| |
Collapse
|
14
|
Alqahtani MJ, Mostafa SA, Hussein IA, Elhawary S, Mokhtar FA, Albogami S, Tomczyk M, Batiha GES, Negm WA. Metabolic Profiling of Jasminum grandiflorum L. Flowers and Protective Role against Cisplatin-Induced Nephrotoxicity: Network Pharmacology and In Vivo Validation. Metabolites 2022; 12:metabo12090792. [PMID: 36144196 PMCID: PMC9502427 DOI: 10.3390/metabo12090792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Cisplatin (CP) is a powerful chemotherapeutic agent; however, its therapeutic use is restricted due to its nephrotoxicity. In this work, we profiled the phytoconstituents of Jasminum grandiflorum flower extract (JGF) using LC-MS/MS and explored the possible molecular mechanisms against acute renal failure through pharmacological network analysis. Furthermore, the possible molecular mechanisms of JGF against acute renal failure were verified in an in vivo nephrotoxicity model caused by cisplatin. LC-MS analysis furnished 26 secondary metabolites. Altogether, there were 112 total hit targets for the identified metabolites, among which 55 were potential consensus targets related to nephrotoxicity based on the network pharmacology approach. Upon narrowing the scope to acute renal failure, using the DisGeNET database, only 30 potential targets were determined. The computational pathway analysis illustrated that JGF might inhibit renal failure through PI3K-Akt, MAPK signaling pathway, and EGFR tyrosine kinase inhibitor resistance. This study was confirmed by in vivo experiment in which kidneys were collected for histopathology and gene expression of mitogen-activated protein kinase 4 (MKK4), MKK7, I-CAM 1, IL-6, and TNF receptor-associated factor 2 (TRAF2). The animal-administered cisplatin exhibited a substantial rise in the expression levels of the MMK4, MKK7, I CAM 1, and TRFA2 genes compared to the control group. To summarize, J. grandiflorum could be a potential source for new reno-protective agents. Further experiments are needed to confirm the obtained activities and determine the therapeutic dose and time.
Collapse
Affiliation(s)
- Moneerah J. Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sally A. Mostafa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35511, Egypt
| | - Ismail A. Hussein
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Seham Elhawary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Fatma A. Mokhtar
- Department of Pharmacognosy, Faculty of Pharmacy, ALSalam University, Al Gharbiya, Kafr El Zayat 31616, Egypt
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Michał Tomczyk
- Department of Pharmacognosy, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
- Correspondence:
| |
Collapse
|
15
|
Liu Z, Xu Y, Bai X, Guo L, Li X, Gao J, Teng Y, Yu P. Prediction of the mechanisms of action of Zhibai Dihaung Granule in cisplatin-induced acute kidney injury: A network pharmacology study and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115241. [PMID: 35351575 DOI: 10.1016/j.jep.2022.115241] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/03/2022] [Accepted: 03/24/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhibai Dihuang Granule (ZDG) is known as traditional Chinese patent medicine with the functions of "Ziyin decrease internal heat" in Traditional Chinses medicine. In clinical, it is also used to treat various kidney diseases. AIM OF THE STUDY We aimed to provide a basis for the curative effect of ZDG on acute kidney injury induced by cisplatin (CIAKI). MATERIALS AND METHODS The active compounds and protein targets of ZDG, as well as the potential targets of the CIAKI were searched from the database. The protein-protein interaction (PPI) network diagram and the drug-compounds-targets-disease network were constructed. Enrichment analysis was performed by Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG). Subsequently, the effect of ZDG on the prevention and treatment of CIAKI was experimentally validated in vivo and in vitro. RESULTS From the database, we screened 22 active compounds of ZDG and 226 related targets. We obtained 498 gene targets related to CIAKI, among which 40 genes overlapped with ZDG-related targets. Go enrichment and KEGG analysis got 339 terms and 64 pathways, respectively. Based on the above study, we speculated that ZDG has the potential effect on treatment CIAKI, and the mechanism may be related to cell apoptosis and inflammation. The results in vitro experiments showed that ZDG reduced the cytotoxicity of cisplatin to HK-2 and 293T cells, but did not affect the antitumor effect of cisplatin. Moreover, in vivo experiments further proved that ZDG effectively controlled kidney damage caused by cisplatin in SD rats. The results showed that ZDG could regulate the expression of CASP3, p65 and MAPK pathway related proteins, suggesting that ZDG's prevention of CIAKI may be related to apoptosis and inflammatory response. CONCLUSIONS Our study showed that ZDG could prevent and treat CIAKI by inhibiting cell apoptosis and inflammation, which provided a new efficacy and clinical application for ZDG.
Collapse
Affiliation(s)
- Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Ye Xu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Xinming Bai
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Lvqian Guo
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Xinran Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Junling Gao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| |
Collapse
|
16
|
Wei X, Ma Y, Li Y, Zhang W, Zhong Y, Yu Y, Zhang LC, Wang Z, Tu Y. Anti-Apoptosis of Podocytes and Pro-Apoptosis of Mesangial Cells for Telmisartan in Alleviating Diabetic Kidney Injury. Front Pharmacol 2022; 13:876469. [PMID: 35517816 PMCID: PMC9061946 DOI: 10.3389/fphar.2022.876469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022] Open
Abstract
Podocytes damage and mesangial cells expansion are two important pathological manifestations of glomerular injury in early diabetes. Telmisartan, as an angiotensin type 1 (AT1) receptor inhibitor, could improve advanced glycation end (AGE) products or angiotensin Ⅱ (Ang Ⅱ)-induced podocytes injury including detachment or apoptosis. In this current paper, we first confirmed the protective effect of telmisartan on early diabetic kidney injury in type 1 diabetic rats. Telmisartan reduced the loss of podocin and inhibited the expression of α-SMA, reflecting its protective effect on podocyte injury and mesangial proliferation, respectively. More interestingly we observed an opposite effect of telmisartan on the cell viability and apoptosis of podocytes and mesangial cells in a high-glucose environment in vitro. The anti-apoptotic effect of telmisartan on podocytes might be related to its inhibition of swiprosin-1 (a protein can mediate high glucose-induced podocyte apoptosis) expression. While telmisartan induced a high expression of PPARγ in mesangial cells, and GW9662 (a PPARγ antagonist) partially inhibited telmisartan-induced apoptosis and reduced viability of mesangial cells. In addition, high glucose-induced PKCβ1/TGFβ1 expression in mesangial cells could be blocked by telmisartan. These data provide a more precise cellular mechanism for revealing the protective effect of telmisartan in diabetic kidney injury.
Collapse
Affiliation(s)
- Xin Wei
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yabin Ma
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ya Li
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Wenzhao Zhang
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Yuting Zhong
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yue Yu
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Zhibin Wang
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Ye Tu
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
17
|
Gowda BR, Prakash N, Santhosh CR, Pavithra BH, Rajashekaraiah R, Sathyanarayana ML, Rao S, Waghe P, Kumar KRA, Shivaprasad GR, Muralidhar Y. Effect of Telmisartan on Arsenic-Induced (Sub-chronic) Perturbations in Redox Homeostasis, Pro-inflammatory Cascade and Aortic Dysfunction in Wistar Rats. Biol Trace Elem Res 2022; 200:1776-1790. [PMID: 34339004 DOI: 10.1007/s12011-021-02804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/19/2021] [Indexed: 11/26/2022]
Abstract
An experimental study was conducted in male Wistar rats to explore the antioxidant potential of telmisartan (an AT1 receptor blocker) to overcome arsenic ('As')-induced perturbations in redox homeostasis pro-inflammatory cytokines, prostaglandin-E2 levels and aortic dysfunction in Wistar rats. Wistar rats were randomly divided into four groups of six each. Group-I served as untreated control, while group-II received sodium (meta) arsenite (NaAsO2) (10 mg/kg b.wt. p.o) for a period of 60 days. Experimental rats in group-III received treatment similar to group-II, but in addition received telmisartan (with 1% aqueous solution of Tween 80) @ 10 mg/kg b.wt. (p.o) for a similar duration, while rats in group-IV received telmisartan alone. Arsenic exposure resulted in significant (p < 0.05) elevation in the levels of superoxide anion ([Formula: see text]) radicals (control: 768.20 ± 126.77 vs group-II: 1232.75 ± 97.85 pmol of NBT reduced/min/mg protein). Telmisartan administration showed significant (p < 0.05) reduction in [Formula: see text] generation (815.34 ± 43.41 pmol of NBT reduced/min/mg protein). Sub-chronic exposure to 'As' significantly (p < 0.05) decreased the activities of SOD, CAT, GPx and GR activity and GSH levels in the aorta, thus induced lipid peroxidation (LPO) measured as measured in terms of thiobarbituric acid reactive substances (TBARS) called as malondialdehyde (MDA). However, the administration of telmisartan effectively countered the LPO (24.03 ± 1.18 nmol of MDA/g) on account of restoring the levels of aforesaid antioxidant defense system. Telmisartan administration effectively attenuated the 'As'-induced surge in pro-inflammatory cytokines (viz., IL-1β, IL-6 and TNF-α) levels, as well as countered the activity of cyclooxygenase (COX2) as indicated by a significant (p < 0.05) decrease in PGE2 level in the aorta. In addition to it, there was a significant (p < 0.05) decrease in plasma angiotensin II (Ang-II) levels in experimental rats receiving telmisartan. Quantitative RT-PCR studies revealed that sub-chronic exposure to 'As' upregulated the Nox2 mRNA expression, but there was a 1.2-fold reduction in expression level upon co-administration of telmisartan. Histopathological examination revealed marked recovery from 'As'-induced disruption of tunica adventitia and loss of connective tissue in experimental rats receiving telmisartan. The study concludes that telmisartan can overcome aortic dysfunction induced by sub-chronic exposure to arsenic through drinking water in experimental rats through restoration of redox balance, attenuation of pro-inflammatory cytokines and mediators and downregulation of Nox2 mRNA expression.
Collapse
Affiliation(s)
- B Rudresh Gowda
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - N Prakash
- Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Vinobanagar, Shivamogga, Karnataka, 577 204, India.
| | - C R Santhosh
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - B H Pavithra
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - Rashmi Rajashekaraiah
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - M L Sathyanarayana
- Department of Veterinary Pathology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - Suguna Rao
- Department of Veterinary Pathology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - Prashantkumar Waghe
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Nandinagar, Bidar, Karnataka, 585 226, India
| | - K R Anjan Kumar
- Department of Veterinary Pathology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - G R Shivaprasad
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| | - Y Muralidhar
- Department of Veterinary Pharmacology and Toxicology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Hebbal, Bengaluru, Karnataka, 560 024, India
| |
Collapse
|
18
|
Telmisartan restricts Chikungunya virus infection in vitro and in vivo through the AT1/PPAR-γ/MAPKs pathways. Antimicrob Agents Chemother 2021; 66:e0148921. [PMID: 34748384 DOI: 10.1128/aac.01489-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) has re-emerged as a global public health threat. The inflammatory pathways of RAS and PPAR-γ are usually involved in viral infections. Thus, Telmisartan (TM) with known capacity to block AT1 receptor and activate PPAR-γ, was investigated against CHIKV. The anti-CHIKV effect of TM was investigated in vitro (Vero, RAW 264.7 cells and hPBMCs) and in vivo (C57BL/6 mice). TM was found to abrogate CHIKV infection efficiently (IC50 of 15.34-20.89μM in the Vero and RAW 264.7 cells respectively). Viral RNA and proteins were reduced remarkably. Additionally, TM interfered in the early and late stages of CHIKV life cycle with efficacy in both pre and post-treatment assay. Moreover, the agonist of AT1 receptor and antagonist of PPAR-γ increased CHIKV infection suggesting TM's anti-viral potential by modulating host factors. Besides, reduced activation of all major MAPKs, NF-κB (p65) and cytokines by TM through the inflammatory axis supported the fact that the anti-CHIKV efficacy of TM is partly mediated through the AT1/PPAR-γ/MAPKs pathways. Interestingly, at the human equivalent dose, TM abrogated CHIKV infection and inflammation significantly leading to reduced clinical score and complete survival of C57BL/6 mice. Additionally, TM reduced infection in hPBMC derived monocyte-macrophage populations in vitro. Hence, TM was found to reduce CHIKV infection by targeting both viral and host factors. Considering its safety and in vivo efficacy, it can be a suitable candidate in future for repurposing against CHIKV.
Collapse
|
19
|
Yuan J, Liang X, Zhou W, Feng J, Wang Z, Shen S, Guan X, Zhao L, Deng F. TRPA1 promotes cisplatin-induced nephrotoxicity through inflammation mediated by the MAPK/NF-κB signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1578. [PMID: 34790784 PMCID: PMC8576655 DOI: 10.21037/atm-21-5125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND The nephrotoxicity induced by cisplatin (DDP) has been a severe obstacle for its clinical use in anticancer treatment. The apoptosis and inflammation induced by DDP are the main causes of the nephrotoxicity. Transient receptor potential ankyrin 1 (TRPA1) is a non-selective cation ligand-gated channel that is involved in the inflammation progress. METHODS The apoptosis, inflammation, MAPK/NF-κB signaling pathway, and TRPA1 expression were assessed after HEK293 cells had been induced by DDP, and the role of TRPA1 in apoptosis and inflammation of DDP-induced HEK293 cells treated with TRPA1 antagonist HC-030031 was also evaluated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), flow cytometry, and western blot assays. RESULTS The cell viability was reduced by DDP in both a time-dependent and dose-dependent manner with a minimal cytotoxic concentration of 10 μM. Moreover, DDP induced an enhancement of the apoptosis and inflammation in a dose-dependent manner, as indicated by the increase of the relative protein level of cleaved-caspase3 (cleaved-cas3), the cleavage product of caspase-3 substrate poly-ADP-ribose polymerase (cleaved-PARP) and inducible nitric oxide synthase (iNOS), and the messenger RNA (mRNA) expression level of interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), and interferon-γ (INF-γ). Additionally, DDP treatment increased the protein phosphorylation expression of IKKβ, JNK, ERK, and p38 in a dose-dependent manner, which was antagonized by the treatment of NF-κB-specific inhibitor BAY 11-7082 and pan-MAPK inhibitor U0126. It was also found that DDP upregulated the expression of TRPA1 at both the mRNA and protein levels in a dose-dependent manner. Besides, block of TRPA1 with HC-030031 relieved the apoptosis, diminished the level of IL-1β, IL-6, TNF-α, and INF-γ, reduced the level of cleaved-cas3, cleaved-PARP, and iNOS, decreased the p-IKKβ, p-JNK, p-ERK, and p-p38 expression, and enhanced the expression of IκBα. CONCLUSIONS Taken together, these results indicate that TRPA1 regulates DDP-induced nephrotoxicity via inflammation mediated by the MAPK/NF-κB signaling pathway in HEK293 cells.
Collapse
Affiliation(s)
- Jinyan Yuan
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Liang
- Department of Internal Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Zhou
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Feng
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenyang Wang
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shaoxian Shen
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| | - Xin Guan
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| | - Liangbin Zhao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Deng
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| |
Collapse
|
20
|
Docosahexaenoic Acid-Acylated Astaxanthin Esters Exhibit Superior Renal Protective Effect to Recombination of Astaxanthin with DHA via Alleviating Oxidative Stress Coupled with Apoptosis in Vancomycin-Treated Mice with Nephrotoxicity. Mar Drugs 2021; 19:md19090499. [PMID: 34564161 PMCID: PMC8467572 DOI: 10.3390/md19090499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/30/2022] Open
Abstract
Prevention of acute kidney injury caused by drugs is still a clinical problem to be solved urgently. Astaxanthin (AST) and docosahexaenoic acid (DHA) are important marine-derived active ingredients, and they are reported to exhibit renal protective activity. It is noteworthy that the existing forms of AST in nature are mainly fatty acid-acylated AST monoesters and diesters, as well as unesterified AST, in which DHA is an esterified fatty acid. However, no reports focus on the different bioactivities of unesterified AST, monoesters and diesters, as well as the recombination of DHA and unesterified AST on nephrotoxicity. In the present study, vancomycin-treated mice were used to evaluate the effects of DHA-acylated AST monoesters, DHA-acylated AST diesters, unesterified AST, and the recombination of AST and DHA in alleviating nephrotoxicity by determining serum biochemical index, histopathological changes, and the enzyme activity related to oxidative stress. Results found that the intervention of DHA-acylated AST diesters significantly ameliorated kidney dysfunction by decreasing the levels of urea nitrogen and creatinine, alleviating pathological damage and oxidative stress compared to AST monoester, unesterified AST, and the recombination of AST and DHA. Further studies revealed that dietary DHA-acylated AST esters could inhibit the activation of the caspase cascade and MAPKs signaling pathway, and reduce the levels of pro-inflammatory cytokines. These findings indicated that the administration of DHA-acylated AST esters could alleviate vancomycin-induced nephrotoxicity, which represented a potentially novel candidate or therapeutic adjuvant for alleviating acute kidney injury.
Collapse
|
21
|
Zhang JJ, Zhou YD, Liu YB, Wang JQ, Li KK, Gong XJ, Lin XH, Wang YP, Wang Z, Li W. Protective Effect of 20(R)-Ginsenoside Rg3 Against Cisplatin-Induced Renal Toxicity via PI3K/AKT and NF-[Formula: see text]B Signaling Pathways Based on the Premise of Ensuring Anticancer Effect. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1739-1756. [PMID: 34461812 DOI: 10.1142/s0192415x21500828] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although the protective effect of ginsenoside on cisplatin-induced renal injury has been extensively studied, whether ginsenoside interferes with the antitumor effect of cisplatin has not been confirmed. In this paper, we verified the main molecular mechanism of 20(R)-ginsenoside Rg3 (R-Rg3) antagonizing cisplatin-induced acute kidney injury (AKI) through the combination of in vivo and in vitro models. It is worth mentioning that the two cell models of HK-2 and HepG2 were used simultaneously for the first time to explore the effect of the activation site of tumor-associated protein p53 on apoptosis and tumor suppression. The results showed that a single injection of cisplatin (20 mg/kg) led to weight loss, the kidney index of the mice increased, and creatinine (CRE) and blood urea nitrogen (BUN) levels in mice sharply increased. Continuous administration of R-Rg3 at doses of 10 and 20 mg/kg for 10 days could significantly alleviate this symptom. Similarly, R-Rg3 treatment reduced oxidative stress damage caused by cisplatin. Moreover, R-Rg3 could observably reduce the apoptosis and inflammatory infiltration of renal tubular cells induced by cisplatin. We used western blotting analysis to demonstrate that R-Rg3 restored cisplatin-induced AKI might be related to PI3K/AKT and NF-[Formula: see text]B mediated apoptosis and inflammation pathways. In the meantime, we also verified that R-Rg3 could activate different sites of p53 to control renal cell apoptosis induced by cisplatin without affecting its antitumor effect.
Collapse
Affiliation(s)
- Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yan-Dan Zhou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yong-Bo Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jian-Qiang Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Ke-Ke Li
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiao-Jie Gong
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiang-Hui Lin
- Liaoning XIFENG Pharmaceutical Group Co., Ltd., Huanren 117200, P. R. China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China.,Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| |
Collapse
|
22
|
Shi HH, Wang CC, Ding L, Mao XZ, Xue CH, Yanagita T, Zhang TT, Wang YM. Comparative evaluation of phosphatidylcholine and phosphatidylserine with different fatty acids on nephrotoxicity in vancomycin-induced mice. Biosci Biotechnol Biochem 2021; 85:1873-1884. [PMID: 34196365 DOI: 10.1093/bbb/zbab105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/01/2021] [Indexed: 01/06/2023]
Abstract
Phospholipids reportedly alleviate drug-induced acute kidney injury. However, no study has compared the effect of phospholipids with different fatty acids and polar heads on drug-induced nephrotoxicity. In the present study, we aimed to compare the possible nephroprotection afforded by phosphatidylcholine and phosphatidylserine with different fatty acids in a mouse model of vancomycin-induced nephrotoxicity. Pretreatment with phospholipids rich in docosahexaenoic acid (DHA) or eicosapentaenoic acid (EPA) doubled the survival time when compared with the model group. Moreover, phospholipids rich in DHA/EPA significantly reduced the serum levels of renal function biomarkers and ameliorated kidney pathologies. In terms of alleviating renal damage, no significant differences were observed between different polar heads in DHA-enriched phospholipids, while phosphatidylserine from soybean was better than phosphatidylcholine in mitigating renal injury. Furthermore, DHA/EPA-enriched phospholipids inhibited vancomycin-induced nephrotoxicity mainly by inhibiting apoptosis and oxidative stress. These results provide a scientific basis for phospholipids as potential ingredients to prevent acute kidney injury.
Collapse
Affiliation(s)
- Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Xiang-Zhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, P. R. China
| | - Teruyoshi Yanagita
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, Japan
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, P. R. China
| |
Collapse
|
23
|
Ala M, Ghasemi M, Mohammad Jafari R, Dehpour AR. Beyond its anti-migraine properties, sumatriptan is an anti-inflammatory agent: A systematic review. Drug Dev Res 2021; 82:896-906. [PMID: 33792938 DOI: 10.1002/ddr.21819] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
Sumatriptan is the first available medication from triptans family that was approved by the U.S. Food and Drug Administration for migraine attacks and cluster headaches in 1991. Most of its action is mediated by selective 5-HT1B/1D receptor agonism. Recent investigations raised the possibility of repositioning of this drug to other indications beyond migraine, as increasing evidence suggests for an anti-inflammatory property of sumatriptan. We performed a literature search using PubMed, Web of Science, Scopus, and Google Scholar using "inflammation AND sumatriptan" or "inflammation AND 5HT1B/D" as the keywords. Then, articles were screened for their relevance and those directly discussing the correlation between inflammation and sumatriptan or 5HT1B/D were included. Total references reviewed or inclusion/exclusion were 340 retrieved full-text articles (n = 340), then based on critical assessment 66 of them were included in this systematic review. Our literature review indicates that at low doses, sumatriptan can reduce inflammatory markers (e.g., interleukin-1β, tumor necrosis factor-α, and nuclear factor-κB), affects caspases and changes cells lifespan. Additionally, nitric oxide synthase and nitric oxide signaling seem to be regulated by this drug. It also inhibits the release of calcitonin gene-related peptide. Sumatriptan protects against many inflammatory conditions including cardiac and mesenteric ischemia/reperfusion, skin flap, pruritus, peripheral, and central nervous system injuries such as spinal cord injury, testicular torsion-detorsion, oral mucositis, and other experimental models. Considering the safety and potency of low dose sumatriptan compared to corticosteroids and other immunosuppressive medications, it is worth to take advantage of sumatriptan in inflammatory conditions.
Collapse
Affiliation(s)
- Moein Ala
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Ibrahim MA, Albahlol IA, Wani FA, Abd-Eltawab Tammam A, Kelleni MT, Sayeed MU, Abd El-Fadeal NM, Mohamed AA. Resveratrol protects against cisplatin-induced ovarian and uterine toxicity in female rats by attenuating oxidative stress, inflammation and apoptosis. Chem Biol Interact 2021; 338:109402. [PMID: 33587916 DOI: 10.1016/j.cbi.2021.109402] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/02/2021] [Accepted: 01/31/2021] [Indexed: 12/20/2022]
Abstract
Cisplatin is an important antineoplastic drug used in multiple chemotherapeutic regimens but unfortunately causes serious toxic effects as ovarian and uterine toxicity. This study aimed to investigate the potential protective effect of resveratrol (RSV) against cisplatin-induced ovarian and uterine toxicity in female rats. Thirty-two female Wistar rats were divided randomly into four groups (n = 8 in each). Control group received oral normal saline for 28 days; RSV group received RSV (10 mg/kg; daily) via oral gavage; CIS group received a single dose of CIS (7 mg/kg; i.p.) on the 21st day; (CIS + RSV) group received both RSV and CIS by the same schedules and doses of RSV and CIS groups, respectively. Results demonstrated a significant decrease in MDA level and a significant increase in both glutathione content and activity of the antioxidant enzymes GPx, SOD, and CAT in the tissues of the ovary and uterus of CIS + RSV group in comparison to that of CIS group (P<0.05), also there are significantly decreased tissue levels of the proinflammatory cytokines and enzymes (NF-κB, IL-1β, IL-6, TNF-α, COX-2, and iNOS), increased estradiol, progesterone, prolactin and decreased FSH serum levels in CIS + RSV group compared to CIS group (P < 0.05). Moreover, there is downregulation of tissues Cleaved Caspase-3, NF-κB and Cox-2 proteins as shown in Western blot analysis, also apoptosis was significantly inhibited, evidenced by downregulation of Bax and upregulation of Bcl-2 proteins, and the ovarian and uterine histological architecture and integrity were maintained in CIS + RSV group compared to CIS group. In conclusion, these findings indicate that RSV has beneficial effects in ameliorating cisplatin-induced oxidative stress, inflammation, and apoptosis in the ovarian and uterine tissues of female rats.
Collapse
Affiliation(s)
- Mahrous Abdelbasset Ibrahim
- Forensic Medicine and Clinical Toxicology, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia, 41522, Egypt.
| | - Ibrahim Abdelkhalek Albahlol
- Obstetrics and Gynecology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Obstetrics and Gynecology Department, Faculty of Medicine, Mansoura University, Egypt.
| | - Farooq Ahmed Wani
- Pathology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia.
| | - Ahmed Abd-Eltawab Tammam
- Physiology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt.
| | - Mina Thabet Kelleni
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt.
| | | | - Noha M Abd El-Fadeal
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University (SCU), Ismailia, Egypt.
| | - Alaa Abdelhamid Mohamed
- Medical Biochemistry Division, Pathology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Medical Biochemistry Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
25
|
McSweeney KR, Gadanec LK, Qaradakhi T, Ali BA, Zulli A, Apostolopoulos V. Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers (Basel) 2021; 13:1572. [PMID: 33805488 PMCID: PMC8036620 DOI: 10.3390/cancers13071572] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Administration of the chemotherapeutic agent cisplatin leads to acute kidney injury (AKI). Cisplatin-induced AKI (CIAKI) has a complex pathophysiological map, which has been linked to cellular uptake and efflux, apoptosis, vascular injury, oxidative and endoplasmic reticulum stress, and inflammation. Despite research efforts, pharmaceutical interventions, and clinical trials spanning over several decades, a consistent and stable pharmacological treatment option to reduce AKI in patients receiving cisplatin remains unavailable. This has been predominately linked to the incomplete understanding of CIAKI pathophysiology and molecular mechanisms involved. Herein, we detail the extensively known pathophysiology of cisplatin-induced nephrotoxicity that manifests and the variety of pharmacological and genetic alteration studies that target them.
Collapse
|
26
|
Abd El Aziz AE, Sayed RH, Sallam NA, El Sayed NS. Neuroprotective Effects of Telmisartan and Nifedipine Against Cuprizone-Induced Demyelination and Behavioral Dysfunction in Mice: Roles of NF-κB and Nrf2. Inflammation 2021; 44:1629-1642. [PMID: 33709265 DOI: 10.1007/s10753-021-01447-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/01/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022]
Abstract
Multiple sclerosis is a chronic inflammatory neurodegenerative disease of the central nervous system which injures the myelin sheath. Telmisartan and nifedipine are antihypertensive drugs that recently showed neuroprotective properties against neurodegenerative diseases. This study evaluated the neuroprotective effect of telmisartan or nifedipine in cuprizone-induced demyelination in mice by examining the underlying mechanisms. C57BL/6 mice received a diet containing 0.7% (w/w) cuprizone for 7 days followed by 3 weeks on a 0.2% cuprizone diet. Telmisartan (5 mg/kg/day, p.o.) or nifedipine (5 mg/kg/day, p.o.) was administered for 3 weeks starting from the second week. Telmisartan or nifedipine improved locomotor activity and enhanced motor coordination as demonstrated by open field, rotarod, and grip strength tests. Furthermore, telmisartan or nifedipine restored myelin basic protein mRNA and protein expression and increased luxol fast blue-staining intensity. Telmisartan or nifedipine attenuated cuprizone-induced oxidative stress and apoptosis by decreasing brain malondialdehyde and caspase-3 along with restoring reduced glutathione and brain-derived neurotrophic factor levels. Telmisartan or nifedipine exerted an anti-inflammatory effect by reducing the expression of nuclear factor kappa B (NF-κB p65) as well as pro-inflammatory cytokines and elevating the expression of IκB-α. In parallel, telmisartan or nifedipine upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and the levels of heme oxygenase-1 and NADPH quinone oxidoreductase 1 enzymes. In conclusion, the current study provides evidence for the protective effect of telmisartan and nifedipine in cuprizone-induced demyelination and behavioral dysfunction in mice possibly by modulating NF-κB and Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Amira E Abd El Aziz
- Center of Excellence, Arab Academy for Science and Technology and Maritime Transport, Alexandria, Egypt
| | - Rabab Hamed Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt.
| | - Nada A Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt
| |
Collapse
|
27
|
Mao R, Shen J, Hu X. RETRACTED: BMSCs-derived exosomal microRNA-let-7a plays a protective role in diabetic nephropathy via inhibition of USP22 expression. Life Sci 2021; 268:118937. [PMID: 33347877 DOI: 10.1016/j.lfs.2020.118937] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 6B, and 2F, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0). The journal requested the corresponding author comment on these concerns and provide the raw data. However the authors were not able to satisfactorily fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Ruiyang Mao
- Department of Medicine, Hangzhou Mingzhou Hospital, Hangzhou 311200, Zhejiang, China
| | - Jiaoni Shen
- Department of Medicine, Hangzhou Mingzhou Hospital, Hangzhou 311200, Zhejiang, China.
| | - Xiaoli Hu
- Department of Medicine, Hangzhou Mingzhou Hospital, Hangzhou 311200, Zhejiang, China
| |
Collapse
|
28
|
Guo Q, Liu L, Nie P, Luo P. Telmisartan alleviates collagen type III glomerulopathy: A case report with literature review. Exp Ther Med 2020; 20:140. [PMID: 33093878 PMCID: PMC7571403 DOI: 10.3892/etm.2020.9269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 07/14/2020] [Indexed: 01/28/2023] Open
Abstract
Collagen type III is commonly detected in the renal interstitium and vasculature; however, it is absent in healthy glomeruli. Deposition of collagen type III in the glomerular mesangium and capillary basement membranes may arise in two rare diseases, namely collagen type III glomerulopathy (CG) and nail patella syndrome. CG is a rare glomerular disease with no specific treatment, although supportive measures for control of hypertension and edema may help to relieve symptoms. With progression to end-stage renal disease, patients with CG may come to require dialysis and/or renal transplantation. The present study reported on a 59-year-old male who was diagnosed with CG nephrotic syndrome by immunohistochemical and electron microscopic examination of biopsy material. To the best of our knowledge, this is the first case reported in northeastern China. The angiotensin II blocker telmisartan was successfully used to alleviate renal symptoms and a literature review was performed. The present case supports the use of telmisartan as a first choice of treatment for CG.
Collapse
Affiliation(s)
- Qiaoyan Guo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Lihua Liu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
29
|
Azouz AA, Abdel-Nassir Abdel-Razek E, Abo-Youssef AM. Amlodipine alleviates cisplatin-induced nephrotoxicity in rats through gamma-glutamyl transpeptidase (GGT) enzyme inhibition, associated with regulation of Nrf2/HO-1, MAPK/NF-κB, and Bax/Bcl-2 signaling. Saudi Pharm J 2020; 28:1317-1325. [PMID: 33250641 PMCID: PMC7679434 DOI: 10.1016/j.jsps.2020.08.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/27/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The therapeutic utility of the effective chemotherapeutic agent cisplatin is hampered by its nephrotoxic effect. We aimed from the current study to examine the possible protective effects of amlodipine through gamma-glutamyl transpeptidase (GGT) enzyme inhibition against cisplatin nephrotoxicity. METHODS Amlodipine (5 mg/kg, po) was administered to rats for 14 successive days. On the 10th day, nephrotoxicity was induced by a single dose of cisplatin (6.5 mg/kg, ip). On the last day, blood samples were collected for estimation of kidney function, while kidney samples were used for determination of GGT activity, oxidative stress, inflammatory, and apoptotic markers, along with histopathological evaluation. RESULTS Amlodipine alleviated renal injury that was manifested by significantly diminished serum creatinine and blood urea nitrogen levels, compared to cisplatin group. Amlodipine inhibited GGT enzyme, which participates in the metabolism of extracellular glutathione (GSH) and platinum-GSH-conjugates to a reactive toxic thiol. Besides, amlodipine diminished mRNA expression of NADPH oxidase in the kidney, while enhanced the anti-oxidant defense by activating Nrf2/HO-1 signaling. Additionally, it showed marked anti-inflammatory response by reducing expressions of p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor-kappa B (NF-κB), with subsequent down-regulation of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and vascular cell adhesion molecule-1 (VCAM-1). Moreover, amlodipine reduced Bax/Bcl-2 ratio and elevated hepatocyte growth factor (HGF), thus favoring renal cell survival. CONCLUSIONS Effective GGT inhibition by amlodipine associated with enhancement of anti-oxidant defense and suppression of inflammatory signaling and apoptosis support our suggestion that amlodipine could replace toxic GGT inhibitors in protection against cisplatin nephrotoxicity.
Collapse
Key Words
- Amlodipine
- Anti-inflammatory response
- Anti-oxidant defense
- BUN, Blood urea nitrogen
- Bax, Bcl-2-associated X protein
- Bcl-2, B-cell lymphoma 2
- CMC, Carboxymethyl cellulose
- Cisplatin nephrotoxicity
- GGT inhibition
- GGT, gamma-glutamyl transpeptidase
- GSH, Reduced glutathione
- H & E, Hematoxylin and eosin
- HGF, Hepatocyte growth factor
- HO-1, Heme oxygenase-1
- IL-6, Interleukin-6
- Keap1, Kelch-like ECH-associated protein 1
- MAPK, Mitogen-activated protein kinase
- MDA, Malondialdehyde
- NADPH, Nicotinamide adenine dinucleotide phosphate
- NF-κB, Nuclear factor-kappa B
- NO, Nitric oxide
- NOx, Total nitrate/nitrite
- Nrf2, Nuclear factor erythroid 2-related factor 2
- ROS, Reactive oxygen species
- Renal cell survival
- TNF-α, Tumor necrosis factor-alpha
- VCAM-1, vascular cell adhesion molecule-1
Collapse
Affiliation(s)
- Amany A. Azouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | | | - Amira M. Abo-Youssef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
30
|
Wang S, Tang S, Chen X, Li X, Jiang S, Li HP, Jia PH, Song MJ, Di P, Li W. Pulchinenoside B4 exerts the protective effects against cisplatin-induced nephrotoxicity through NF-κB and MAPK mediated apoptosis signaling pathways in mice. Chem Biol Interact 2020; 331:109233. [PMID: 32991863 DOI: 10.1016/j.cbi.2020.109233] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 01/17/2023]
Abstract
Cisplatin (cis-Dichlorodiammine platinum, CP), as the first-line chemotherapy drug of choice for many cancers such as urogenital system tumors and digestive tract tumors, also causes toxicity and side effects to the kidney. Previous studies have shown that Pulsatilla chinensis has significant anti-inflammatory and antioxidant activities, but the mechanism of cisplatin induced acute kidney injury (AKI) in vivo has not been thoroughly studied. The purpose of this study is to investigate the protective effect of pulchinenoside B4 (PB4), a representative and major component with a content of up to 10% in root of P. chinensis, on AKI induced by CP in mice. Our results indicated the significant protective effect of PB4 by evaluating renal function indicators, inflammatory factor levels and renal histopathological changes. In addition, PB4 may mainly act on NF-κB signaling pathway to reduce the levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) in the kidney after CP exposure, thus exerting anti-inflammatory activity. Furthermore, PB4 regulated MAPK signaling pathway and its downstream apoptotic factors to inhibit the occurrence of apoptosis, such as Bax, Bcl-2, caspase 3 and caspase 9. Notably, the activations of caspase 3 induced by cisplatin were strikingly reduced in PB4-treated mice. Therefore, the above evidence suggested that PB4 is a potential renal protectant with significant anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Shuang Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shan Tang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Xuan Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Hui-Ping Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Pin-Hui Jia
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Ming-Jie Song
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Peng Di
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| |
Collapse
|
31
|
Hu JN, Leng J, Shen Q, Liu Y, Li XD, Wang SH, Li HP, Wang Z, Wang YP, Li W. Platycodin D suppresses cisplatin-induced cytotoxicity by suppressing ROS-mediated oxidative damage, apoptosis, and inflammation in HEK-293 cells. J Biochem Mol Toxicol 2020; 35:e22624. [PMID: 32881195 DOI: 10.1002/jbt.22624] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/29/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022]
Abstract
Cisplatin, a proven effective chemotherapeutic agent, has been used clinically to treat malignant solid tumors, whereas its clinical use is limited by serious side effect including nephrotoxicity. Platycodin D (PD), the major and marked saponin isolated from Platycodon grandiflorum, possesses many pharmacological effects. In this study, we evaluated its protective effect against cisplatin-induced human embryonic kidney 293 (HEK-293) cells injury and elucidated the related mechanisms. Our results showed that PD (0.25, 0.5, and 1 μM) can dose-dependently alleviate oxidative stress by decreasing malondialdehyde and reactive oxygen species, while increasing the levels of glutathione, superoxide dismutase, and catalase. Moreover, the elevation of apoptosis including Bax, Bad, cleaved caspase-3,-9, and decreased protein levels of Bcl-2, Bcl-XL induced by cisplatin were reversed after PD treatment. Importantly, PD pretreatment can also regulate PI3K/Akt and ERK/JNK/p38 signaling pathways. Furthermore, PD was found to reduce NF-κB-mediated inflammatory relative proteins. Our finding indicated that PD exerted significant effects on cisplatin induced oxidative stress, apoptosis and inflammatory, which will provide evidence for the development of PD to attenuate cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jing Leng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Qiong Shen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Ying Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Xin-Dian Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Shi-Han Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Hui-Ping Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| |
Collapse
|
32
|
Hu JN, Xu XY, Jiang S, Liu Y, Liu Z, Wang YP, Gong XJ, Li KK, Ren S, Li W. Protective effect of ginsenoside Rk1, a major rare saponin from black ginseng, on cisplatin-induced nephrotoxicity in HEK-293 cells. Kaohsiung J Med Sci 2020; 36:732-740. [PMID: 32374939 DOI: 10.1002/kjm2.12220] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/16/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Cisplatin, as one of the most effective chemotherapeutic agents, its clinical use is limited by serious side effect of nephrotoxicity. Cisplatin-induced nephrotoxicity is closely related to apoptosis induction and activation of caspase. The present study aimed to explore the potential protective effect of ginsenoside Rk1 (Rk1), a rare ginsenoside generated during steaming ginseng, on cisplatin-induced nephrotoxicity and the underlying mechanisms in human embryonic kidney 293 (HEK-293) cells. Our results showed that the reduced cell viability induced by cisplatin could significantly recover by Rk1. Furthermore, glutathione (GSH) as an oxidative index, was elevated and the lipid peroxidation product malondialdehyde (MDA) was significantly decreased after Rk1 treatment compared to the cisplatin group. Additionally, Rk1 can also decrease the ROS fluorescence expression and increase the protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) compared to the cisplatin group, which suggested a suppression of oxidative response. More importantly, the cisplatin-induced elevated protein levels of Bax, cleaved caspase-3, cleaved caspase-9, and decreased protein level of Bcl-2 were reversed after treatment with Rk1. Our results elucidated the possible protective mechanism of Rk1 for the first time, which may involve in its anti-oxidation and anti-apoptosis effects.
Collapse
Affiliation(s)
- Jun-Nan Hu
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Xing-Yue Xu
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Shuang Jiang
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Ying Liu
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Zhi Liu
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Ying-Ping Wang
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Xiao-Jie Gong
- Department of Biological Engineering, College of Life Science, Dalian Minzu University, Dalian, China
| | - Ke-Ke Li
- Department of Biological Engineering, College of Life Science, Dalian Minzu University, Dalian, China
| | - Shen Ren
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Wei Li
- Department of Chinese Medicine, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| |
Collapse
|
33
|
Dexmedetomidine Protects against Ischemia and Reperfusion-Induced Kidney Injury in Rats. Mediators Inflamm 2020; 2020:2120971. [PMID: 32317860 PMCID: PMC7157761 DOI: 10.1155/2020/2120971] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/24/2020] [Indexed: 01/21/2023] Open
Abstract
Acute kidney injury (AKI), a clinical syndrome, is a sudden onset of kidney failure that severely affects the kidney tubules. One potential treatment is dexmedetomidine (DEX), a highly selective α 2-adrenoreceptor agonist that is used as an anesthetic adjuvant. It also has anti-inflammatory, neuroprotective, and sympatholytic qualities. The aim of this study was to establish whether DEX also offers protection against ischemia and reperfusion- (I/R-) induced AKI in rats. An intraperitoneal injection of DEX (25 μg/kg) was administered 30 min prior to the induction of I/R. The results indicate that in the I/R rats, DEX played a protective role by reducing the damage to the tubules and maintaining renal function. Furthermore, in response to I/R, the DEX treatment reduced the mRNA expression of TNF-α, IL-1β, IL-6, and MCP-1 in the kidney tissues and the serum levels of TNF-α, IL-1β, IL-6, and MCP-1. DEX also reduced the levels of oxidative stress and apoptosis in the tubular cells. These results indicate that in response to I/R kidney injury, DEX plays a protective role by inhibiting inflammation and tubular cell apoptosis, reducing the production of reactive oxygen species, and promoting renal function.
Collapse
|
34
|
Sarangi SC, Pattnaik SS, Katyal J, Kaleekal T, Dinda AK. An interaction study of Ocimum sanctum L. and levetiracetam in pentylenetetrazole kindling model of epilepsy. JOURNAL OF ETHNOPHARMACOLOGY 2020; 249:112389. [PMID: 31739106 DOI: 10.1016/j.jep.2019.112389] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/24/2019] [Accepted: 11/11/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ocimum sanctum L. commonly known as tulsi (synonym of Ocimum tenuiflorum L.) is widely used in Ayurveda medicine and is having multitude neuromodulatory effect including the anticonvulsant effect in acute seizure models as per previous studies. In India, it is used for the treatment of epilepsy as traditional medicine. However, its role in chronic seizure model and interaction with newer antiepileptic drugs has not been investigated, which will enhance its translational value. AIM OF THE STUDY Current study investigated the effect of Ocimum on chronic seizure model and its interaction with levetiracetam (LEV), a newer antiepileptic drug. MATERIALS AND METHODS The adjuvant role of Ocimum sanctum hydroalcoholic extracts (OSHE) 1000 mg/kg along with LEV 300 mg/kg was studied in adult male Wistar rats with mean weight of 227.84 ± 21.68 g using pentylenetetrazole (30 mg/kg, i.p.) kindling (K) (with maximum 24 injections on alternate days and challenge on 7th-day). Along with seizure score, neurobehavioral, brain tissue oxidative stress and histopathology status were assessed. Pharmacokinetic interaction was assessed between LEV and OSHE after 14 days of drug treatment. RESULTS K-LEV + OSHE had least seizure score during kindling and on the pentylenetetrazole-challenge test (p=0.031) than other kindling groups. Seizure protection was more in K-LEV + OSHE (85.72%) than others (K-LEV-42.86%, K-OSHE-42.86%, and K-Control-28.58%). Ocimum treated groups had better memory retention potential as evident from Morris water maze (MWM), passive avoidance test but not in an elevated plus maze test. Oxidative-stress was lower in Ocimum treated groups than K-Control group. As per histopathology, K-LEV + OSHE group had the least neuronal degeneration among kindling groups. There was no significant pharmacokinetic interaction between LEV and OSHE, except increased Tmax in LEV + OSHE group than LEV alone (p=0.009). CONCLUSIONS Ocimum per se and combination with levetiracetam treatment exerted better seizure control, memory retention, oxidative stress reduction, and neuronal structure preservation than kindling control group. There was a very minimal drug interaction between Ocimum and LEV. So, Ocimum as an adjuvant to LEV may be shelpful in enhancing the antiepileptic effect and also in minimizing the adverse effects.
Collapse
Affiliation(s)
| | - Soumya S Pattnaik
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Jatinder Katyal
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Thomas Kaleekal
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - A K Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
35
|
Astragalus Polysaccharide Attenuates Cisplatin-Induced Acute Kidney Injury by Suppressing Oxidative Damage and Mitochondrial Dysfunction. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2851349. [PMID: 31998784 PMCID: PMC6970487 DOI: 10.1155/2020/2851349] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/15/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022]
Abstract
Cisplatin is a widely used chemotherapeutic drug in the treatment of various solid tumors. However, the cisplatin-induced acute kidney injury remains a disturbing complication, which still lacks effective prevention. Cisplatin-induced oxidative damage and mitochondrial dysfunction are anticipated to be crucial in the occurrence of kidney injury. Astragalus polysaccharide (APS) has been reported to possess multiple biological activities including anti-inflammatory, antioxidant, and mitochondria protection. In this study, we investigated the potentially protective effect of APS against cisplatin-induced kidney injury both in vivo and in vitro. We found that APS pretreatment attenuated the cisplatin-induced renal dysfunction and histopathological damage in mice; in addition, it also protected the viability of HK-2 cells upon cisplatin exposure. APS attenuated the cisplatin-induced oxidative damage by reducing reactive oxygen species (ROS) generation and recovering the activities of total superoxide dismutase and glutathione peroxidase in mice kidney. In addition, electron microscope analysis indicated that cisplatin induced extensive mitochondrial vacuolization in mice kidney. However, APS administration reversed these mitochondrial morphology changes. In HK-2 cells, APS reduced the cisplatin-induced mitochondrial and intracellular ROS generation. Furthermore, APS protected the normal morphology of mitochondria, blocked the cisplatin-induced mitochondrial permeability transition pore opening, and reduced the cytochrome c leakage. Subsequently, APS reduced the cisplatin-induced apoptosis in mice renal and HK-2 cells. In conclusion, our data suggested that APS pretreatment might prevent cisplatin-induced kidney injury through attenuating oxidative damage, protecting mitochondria, and ameliorating mitochondrial-mediated apoptosis.
Collapse
|
36
|
Khairnar SI, Mahajan UB, Patil KR, Patel HM, Shinde SD, Goyal SN, Belemkar S, Ojha S, Patil CR. Disulfiram and Its Copper Chelate Attenuate Cisplatin-Induced Acute Nephrotoxicity in Rats Via Reduction of Oxidative Stress and Inflammation. Biol Trace Elem Res 2020; 193:174-184. [PMID: 30825159 DOI: 10.1007/s12011-019-01683-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 02/19/2019] [Indexed: 12/19/2022]
Abstract
The use of cisplatin (CP) in chemotherapy of resistant cancers is limited due to its dose-dependent nephrotoxicity. Disulfiram (DSF), the aversion therapy for alcoholism, has recently emerged as an anticancer and chemopreventive agent. Its anticancer activity is potentiated in the presence of copper. However, such use of copper leads to several adverse effects. In the present study, the protective effect of DSF and its copper chelate (Cu-DEDC) against CP-induced nephrotoxicity in rats was evaluated. Nephrotoxicity was induced by a single intraperitoneal injection of CP (5 mg/kg). The treatment groups included control (vehicle treated), CP (CP-treated), CP + DSF (CP followed by DSF), CP + DSF + Cu (CP followed by DSF and CuCl2), CP + Cu-DEDC (CP followed by Cu-DEDC), and CP + AMF (amifostine pre-treated and CP-treated). The DSF, Cu-DEDC, and CuCl2 were administered orally at 50 mM/kg/day dose for 5 days post CP injection. AMF served as a standard chemo protectant, administered intravenously 30 min prior to CP. The markers of oxidative stress, inflammation, and kidney function estimated on the 6th day revealed that both DSF and Cu-DEDC significantly attenuated the CP-induced rise in the serum/urine creatinine and blood urea nitrogen (BUN). The CP-induced rise in serum alkaline phosphatase (ALPase) was reversed by these drugs. Both drugs reduced the levels of malondialdehyde and nitric oxide (NO) in kidney tissues. These drugs reversed CP-induced depletion of SOD, catalase, and GSH in the kidneys. There was a significant reduction in the CP-induced TNF-α and IL-1β production along with prevention of histological alterations. Above observations indicate that DSF and Cu-DEDC may have significance as adjuvants to protect against CP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Shraddha I Khairnar
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India
| | - Umesh B Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India
| | - Kalpesh R Patil
- Department of Pharmacology, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India
| | - Harun M Patel
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India
| | - Sachin D Shinde
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India
| | - Sameer N Goyal
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Sateesh Belemkar
- Department of Pharmacology, School of Pharmacy & Technology Management, SVKM's NMIMS, Shirpur, India, Shirpur, Dist. Dhule, Maharashtra, 425405, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Chandragouda R Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, Maharashtra, 425405, India.
| |
Collapse
|
37
|
Tomar A, Kaushik S, Khan SI, Bisht K, Nag TC, Arya DS, Bhatia J. The dietary isoflavone daidzein mitigates oxidative stress, apoptosis, and inflammation in CDDP-induced kidney injury in rats: Impact of the MAPK signaling pathway. J Biochem Mol Toxicol 2019; 34:e22431. [PMID: 31833131 DOI: 10.1002/jbt.22431] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/04/2019] [Accepted: 12/02/2019] [Indexed: 01/14/2023]
Abstract
Cisplatin-induced nephrotoxicity persists as a clinical problem despite several supportive measures to alleviate renal damage. Daidzein (DZ), a dietary isoflavone having antioxidant and anti-inflammatory activity, is investigated in this study for protective effects against cisplatin-induced renal injury in rats. DZ (25, 50, or 100 mg/kg; intraperitoneally; 10 days) was administered along with Cisplatin, single dose, on the 7th day of the experiment. On the 11th day, the rats were euthanized, and different samples were collected for analysis. Biochemical, histopathological, and molecular parameters were assessed to evaluate the effect of daidzein. Cisplatin injection resulted in renal dysfunction, lipid peroxidation that led to consumption of antioxidants, exaggerated apoptosis, and inflammation. These changes were associated with increase in the signaling proteins. DZ attenuated the toxic effects of cisplatin on the kidney at 100 mg/kg dose. The study concludes with the finding that daidzein imparts protection against the nephrotoxic effect of Cisplatin and can be considered as a novel, potential therapy.
Collapse
Affiliation(s)
- Ameesha Tomar
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Kaushik
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sana Irfan Khan
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Khushboo Bisht
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
38
|
Ma N, Wei W, Fan X, Ci X. Farrerol Attenuates Cisplatin-Induced Nephrotoxicity by Inhibiting the Reactive Oxygen Species-Mediated Oxidation, Inflammation, and Apoptotic Signaling Pathways. Front Physiol 2019; 10:1419. [PMID: 31849693 PMCID: PMC6901966 DOI: 10.3389/fphys.2019.01419] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cisplatin is a chemotherapy drug that is often used in clinical practice, but its frequent use often leads to nephrotoxicity. Therefore, we urgently need a drug that reduces the nephrotoxicity induced by cisplatin. Farrerol reportedly has antioxidant potential, but its renal protective effects and potential mechanisms remain unclear. In this study, we used both cell and mouse models to determine the mechanism of farrerol in cisplatin-induced nephrotoxicity. The in vitro experiments revealed that farrerol improved cisplatin-induced nephrotoxicity and reactive oxygen species (ROS) production via nuclear factor erythrocyte 2-related factor 2 (Nrf2) activation. Moreover, farrerol effectively activated Nrf2 and subsequently increased the expression of Nrf2-targeted antioxidant enzymes, including heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase-1 (NQO1), but inhibited Kelch-like ECH-associated protein 1 (Keap1) and NADPH oxidase type 4 (NOX4). Furthermore, farrerol attenuated the phosphorylation of C-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and p38 mitogen-activated protein kinase (p38); the activation of phosphorylated nuclear factor-κB (p-NF-κB) and nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3); and the expression of phosphorylated p53 (p-p53), Bax, and cleaved caspase-3. In vivo, farrerol significantly improved cisplatin-induced renal damage, as demonstrated by the recovery of blood urea nitrogen (BUN), serum creatinine (SCr), kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and pathological damage. Moreover, farrerol inhibited inflammatory and apoptotic protein expression in vivo. Notably, farrerol exerted slight protection in Nrf2-knockout mice compared with wild-type mice. These findings indicate that farrerol can effectively activate Nrf2 and can serve as a therapeutic target in the treatment of acute kidney injury (AKI).
Collapse
Affiliation(s)
- Ning Ma
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China.,Department of Urology, The First Hospital, Jilin University, Changchun, China
| | - Wei Wei
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Xiaoye Fan
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
39
|
Zhao Q, Yang H, Liu F, Luo J, Zhao Q, Li X, Yang Y. Naringenin Exerts Cardiovascular Protective Effect in a Palmitate‐Induced Human Umbilical Vein Endothelial Cell Injury Model via Autophagy Flux Improvement. Mol Nutr Food Res 2019; 63:e1900601. [PMID: 31622021 DOI: 10.1002/mnfr.201900601] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/01/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Qiang Zhao
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical University Urumqi 830054 China
- Xinjiang Key Laboratory of Cardiovascular Disease Research Urumqi 830054 China
| | - Hongyan Yang
- School of Aerospace MedicineFourth Military Medical University Xi'an 710032 China
| | - Fen Liu
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical University Urumqi 830054 China
- Xinjiang Key Laboratory of Cardiovascular Disease Research Urumqi 830054 China
| | - Junyi Luo
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical University Urumqi 830054 China
- Xinjiang Key Laboratory of Cardiovascular Disease Research Urumqi 830054 China
| | - Qian Zhao
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical University Urumqi 830054 China
- Xinjiang Key Laboratory of Cardiovascular Disease Research Urumqi 830054 China
| | - Xiaomei Li
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical University Urumqi 830054 China
- Xinjiang Key Laboratory of Cardiovascular Disease Research Urumqi 830054 China
| | - Yining Yang
- Department of CardiologyFirst Affiliated Hospital of Xinjiang Medical University Urumqi 830054 China
- Xinjiang Key Laboratory of Cardiovascular Disease Research Urumqi 830054 China
| |
Collapse
|
40
|
Abdulla MH, Brennan N, Ryan E, Sweeney L, Manning J, Johns EJ. Tacrolimus restores the high- and low-pressure baroreflex control of renal sympathetic nerve activity in cisplatin-induced renal injury rats. Exp Physiol 2019; 104:1726-1736. [PMID: 31468631 DOI: 10.1113/ep087829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/22/2019] [Indexed: 01/04/2025]
Abstract
NEW FINDINGS What is the central question of this study? Does immunosuppression restore the baroreflex control of renal sympathetic nerve activity (RSNA) in an animal model of kidney injury? What is the main finding and its importance? Tacrolimus, a calcineurin inhibitor, restored the high- and low-pressure baroreflex control of RSNA following cisplatin-induced renal injury. ABSTRACT Cisplatin administration causes depression of renal haemodynamic and excretory function and is associated with renal sympatho-excitation and loss of baroreflex regulation of renal sympathetic nerve activity (RSNA). This study investigated whether administration of the immunosuppressant tacrolimus in this cisplatin-mediated renal injury model could restore, or the acute intra-renal infusion of tumour necrosis factor α (TNF-α) could blunt, the high- or low-pressure baroreflex control of RSNA. Groups of control and cisplatin-treated (5 mg kg-1 , i.p. on day 0) rats received either saline or tacrolimus (0.25 mg kg-1 day-1 , i.p.) for 7 days prior to study. Rats were anaesthetised and prepared for measurement of mean arterial pressure (MAP), heart rate (HR) and RSNA. Baroreflex gain curves were generated and the degree of renal sympatho-inhibition determined (area under the curve (AUC) reported as %RSNA min) during acute volume expansion. Intrarenal TNF-α infusion (0.3 µg kg-1 h-1 ) in control rats decreased baroreflex gain by 32% (P < 0.05) compared to intra-renal saline infusion. In the cisplatin group (MAP: 98 ± 14 mmHg; HR: 391 ± 24beats min-1 ), the baroreflex gain for RSNA was 39% (P < 0.05) lower than that for the control group (MAP: 91 ± 7 mmHg; HR: 382 ± 29 beats min-1 ). In cisplatin-treated rats given daily tacrolimus (MAP: 84 ± 12 mmHg; HR: 357 ± 30 beats min-1 ), the baroreflex gain and renal sympatho-inhibition (AUC, 2440 ± 1071 vs. 635 ± 498% min) were restored to normal values. These findings provide evidence for the view that cisplatin administration initiates an injury involving inflammation which may contribute to the deranged baroreflex regulation of RSNA. This phenomenon appears mediated in part via the renal innervation.
Collapse
Affiliation(s)
- Mohammed H Abdulla
- Department of Physiology, Western Gateway Building, University College Cork, College Road, Cork, Ireland
| | - Nicola Brennan
- Department of Physiology, Western Gateway Building, University College Cork, College Road, Cork, Ireland
| | - Eimear Ryan
- Department of Physiology, Western Gateway Building, University College Cork, College Road, Cork, Ireland
| | - Linda Sweeney
- Department of Physiology, Western Gateway Building, University College Cork, College Road, Cork, Ireland
| | - Jennifer Manning
- Department of Physiology, Western Gateway Building, University College Cork, College Road, Cork, Ireland
| | - Edward J Johns
- Department of Physiology, Western Gateway Building, University College Cork, College Road, Cork, Ireland
| |
Collapse
|
41
|
Raman spectroscopy: A novel experimental approach to evaluating cisplatin induced tissue damage. Talanta 2019; 207:120343. [PMID: 31594623 DOI: 10.1016/j.talanta.2019.120343] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 01/24/2023]
Abstract
The aim of this work is to clarify the effect of curcumin and beta-carotene on cisplatin-induced tissue damage and to demonstrate the potential of Raman spectroscopy to detect tissue changes consistent with liver and kidney histopathology as a potential diagnostic adjunct. İn the study, 56 Wistar albino female rats were used and randomly divided into 7 groups (n:8). Sham group received only sesame oil; Cisplatin group, received a single dose injection of cisplatin; Beta-carotene group, treated with beta-carotene orally; Cisplatin + Beta-carotene group, pretreated with beta-carotene 30 min prior to the cisplatin injection, then received cisplatin; Curcumin group, orally treated with curcumin; Cisplatin + Curcumin group, pretreated with curcumin 30min prior to the cisplatin injection, then received cisplatin. The second application was performed 1 week after the first application. One of the liver and kidney tissues was taken to 10% form for histopathological examinations and the others were taken to -80 °C for raman spectroscopy. Received sections were hematoxylin-eosin stained. The avidin-biotin peroxidase method was used for to investigate anti-TNF-α and IL1-β activities. TUNEL method was applied to determine apoptotic cells. According to our histopathological findings, beta-carotene and especially curcumin have been found to possess hepatorenal protective activities. These datas were supported by the microscopic damage scores. Although some of these findings were observed in both the cisplatin + curcumin and cisplatin + beta-carotene groups, the incidence and severity of histopathological lesions were less than the cisplatin group. Both immunohistochemical studies and Raman spectroscopy results consistent with histopathological examination of hematoxylen-eosin stained sections. Raman spectroscopy represents a suitable tool to provide insights into structural factors involved in the mechanisms underlying antitumor effects of platinum drug.
Collapse
|
42
|
Ali FF, Abdelzaher WY, Ibrahim RA, Elroby Ali DM. Amelioration of estrogen-induced endometrial hyperplasia in female rats by hemin via heme-oxygenase-1 expression, suppression of iNOS, p38 MAPK, and Ki67. Can J Physiol Pharmacol 2019; 97:1159-1168. [PMID: 31505119 DOI: 10.1139/cjpp-2019-0287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although heme oxygenase-1 (HO-1) is part of an endogenous defense system implicated in the homeostatic response, its role in cell proliferation and tumor progression is still controversial. Endometrial hyperplasia (EH) is associated with high risk of endometrial cancer (EC). Therefore, we aimed to evaluate the effect of hemin, a HO-1 inducer, against EH. Thirty-two female rats (60-70 days old) were divided into 4 groups treated for 1 week: vehicle control group, hemin group (25 mg/kg; i.p. 3 times/week), estradiol valerate (EV) group (2 mg/kg per day, p.o.), and hemin plus EV group. Sera were obtained for reduced glutathione level. Uterine malondialdehyde, superoxide dismutase, total nitrite/nitrate, and interleukin-1β levels were estimated. HO-1 and p38 mitogen-activated protein kinase expressions were obtained in uterine tissue. Uterine histological and immunohistochemical assessment of iNOS and Ki67 were also done. Results demonstrated that upregulation of HO-1 expression in hemin plus EV rats led to amelioration of EH which was confirmed with histological examination. This was associated with significant decrease in oxidative stress parameters, p38 mitogen-activated protein kinase expression, and interleukin-1β level. Also, uterine iNOS and Ki67 expressions were markedly suppressed. In conclusion, upregulation of HO-1 expression via hemin has ameliorative effect against EH through its antioxidant, anti-inflammatory, and antiproliferative actions.
Collapse
Affiliation(s)
- Fatma F Ali
- Department of Medical Physiology, Faculty of Medicine, Minia University, Egypt
| | | | - Randa Ahmed Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Egypt
| | | |
Collapse
|
43
|
|
44
|
Kandemir FM, Yildirim S, Caglayan C, Kucukler S, Eser G. Protective effects of zingerone on cisplatin-induced nephrotoxicity in female rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:22562-22574. [PMID: 31165450 DOI: 10.1007/s11356-019-05505-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
Zingerone (ZO), one of the active components of ginger (Zingiber officinale), is a phenolic alkanone with antioxidant, antiapoptotic, and anti-inflammatory properties. Cisplatin (CP) is a widely used chemotherapeutic drug for solid tumors, but its therapeutic use is limited due to dose-dependent nephrotoxicity. In the present study, we investigated the ameliorative effect of ZO against CP-induced nephrotoxicity. Intraperitoneal administration of single-dose CP (7 mg/kg body weight) on the first day enhanced kidney lipid peroxidation and reduced antioxidant enzyme activities such as catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione (GSH). CP increased serum urea and creatinine levels and disrupted histological integrity while causing a decrease aquaporin 1 (AQP1) level in the kidney tissues. CP induced inflammatory responses by elevating the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-33 (IL-33) and nuclear factor kappa B (NF-κB), and activities of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Moreover, it also caused oxidative DNA damage and activation of apoptotic pathway by increasing of 8-hydroxy-2'-deoxyguanosine (8-OHdG), p53, cysteine aspartate-specific protease-3 (caspase-3), and Bcl-2-associated x protein (bax) while decreasing B cell lymphoma-2 (Bcl-2). However, treatment with ZO at a dose of 25 and 50 mg/kg b.wt. for 7 days significantly decreased oxidative stress, apoptosis, inflammation, and histopathological alterations while increased AQP1 levels in the kidney tissue. The results of the current study suggested that ZO as an effective natural product attenuates CP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Cuneyt Caglayan
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingol University, 12000, Bingol, Turkey.
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Gizem Eser
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
45
|
Wang L, Wu D, Xu Z. USP10 protects against cerebral ischemia injury by suppressing inflammation and apoptosis through the inhibition of TAK1 signaling. Biochem Biophys Res Commun 2019; 516:1272-1278. [PMID: 31301769 DOI: 10.1016/j.bbrc.2019.06.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 06/08/2019] [Indexed: 12/11/2022]
Abstract
Cerebral ischemia is a leading cause of death and long-term disability in the world. Multiple signaling pathways play essential roles in the process. Therefore, identifying the unknown important modulators of these pathways may supply promising therapeutic targets for the treatment of cerebral ischemia. Ubiquitin-specific protease 10 (USP10) is a member of the ubiquitin-specific protease family of cysteine proteases with enzymatic activity to cleave ubiquitin from ubiquitin-conjugated protein substrates, and is involved in multiple pathologies. However, the effects of USP10 in cerebral ischemia-reperfusion (I/R) injury remain unclear. Here, we reported that USP10 expression was markedly decreased in wild type (WT) mice after cerebral I/R injury. USP10 knockout (KO) mice showed significantly elevated infarct size and the neurological deficit score after cerebral I/R operation. USP10 deletion also promoted inflammatory response in ischemic penumbra of cortical regions by further accelerating nuclear factor κB (NF-κB) signaling pathway. In addition, apoptosis was markedly induced in USP10-knockout mice after cerebral I/R injury compared to the WT mice. The c-Jun N-terminal kinase-mitogen-activated protein kinase (JNK-MAPK) signaling induced by cerebral I/R injury was further aggravated in USP10-KO mice. Finally, USP10 was found to display protective effects against cerebral I/R injury through direct interaction with transforming growth factor β-activated kinase 1 (TAK1). Thus, USP10 might be a protective factor in cerebral I/R injury. Modulation of USP10/TAK1 might be a promising strategy to prevent this pathological process.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, The Second People's Hospital of Dongying, Shandong Province, 257335, China
| | - Dongchuan Wu
- Department of Neurology, People's Hospital of Dongying City, Dongying, Shandong Province, 257091, China
| | - Zongrong Xu
- Department of Neurology, People's Hospital of Dongying City, Dongying, Shandong Province, 257091, China.
| |
Collapse
|
46
|
Michel HE, Menze ET. Tetramethylpyrazine guards against cisplatin-induced nephrotoxicity in rats through inhibiting HMGB1/TLR4/NF-κB and activating Nrf2 and PPAR-γ signaling pathways. Eur J Pharmacol 2019; 857:172422. [PMID: 31152701 DOI: 10.1016/j.ejphar.2019.172422] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022]
Abstract
Cisplatin-induced acute renal injury is the most common and serious side effect, sometimes requiring discontinuation of the treatment. Thus, the development of new protective strategies is essential. The present study aimed to investigate the potential nephroprotective effect of tetramethylpyrazine (TMP) against acute renal damage induced by cisplatin in rats. Rats were administered 50 and 100 mg/kg TMP intraperitoneally before cisplatin (7 mg/kg). Acute nephrotoxicity was evident in cisplatin-treated rats where relative kidney weight, BUN and serum creatinine were markedly elevated. Cisplatin administration resulted in enhanced oxidative stress, evidenced by depleted GSH level as well as catalase and superoxide dismutase activities. Also, lipid peroxidation was boosted in comparison to the control. This was associated with inhibition of Nrf2 defense pathway. Moreover, cisplatin increased the expression of pro-inflammatory mediators in the kidney tissues. Cisplatin-induced apoptosis was depicted by elevated Bax mRNA expression and caspase-3 activity, as well as decreased Bcl2 mRNA expression. In addition, high mobility group box 1/toll-like receptor 4/nuclear factor-kappa B (HMGB1/TLR4/NF-κB) signaling pathway was significantly upregulated, while peroxisome proliferator-activated receptor-gamma (PPAR-γ) expression was significantly diminished in cisplatin-treated rats. Cisplatin-induced nephrotoxicity, oxidative stress, inflammation, apoptosis and the effect on Nrf2 defense pathway and HMGB1/TLR4/NF-κB as well as PPAR-γ expression were markedly ameliorated by TMP administration. Given the major nephrotoxicity of cisplatin cancer chemotherapy, TMP might be a potential candidate for neoadjuvant chemotherapy due to its antioxidant, anti-inflammatory and anti-apoptotic effects, in addition to its effect on Nrf2, HMGB1/TLR4/NF-κB signaling pathway and PPAR-γ expression.
Collapse
Affiliation(s)
- Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
47
|
Yao S, Wei B, Yu M, Meng X, He M, Yao R. Design, synthesis and evaluation of PD176252 analogues for ameliorating cisplatin-induced nephrotoxicity. MEDCHEMCOMM 2019; 10:757-763. [PMID: 31191866 PMCID: PMC6533884 DOI: 10.1039/c8md00632f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/19/2019] [Indexed: 11/21/2022]
Abstract
Cisplatin is a clinical chemotherapy drug for cancers; however, its remarkably high kidney toxicity and other toxicities pose a danger to patients. As the small molecule inhibitor of GRPR, PD176252 can inhibit the growth and proliferation of various cancer cells, but the characteristics of high toxicity and poor water solubility has limited its use as a drug. When we studied PD176252 for the reduction of toxicity of cisplatin, we modified its structure to synthesize 16 analogues. Surprisingly, the analogues showed reduced cisplatin-induced renal toxicity, and unlike PD176252, the analogues 5d and 5m were almost non-toxic to the normal HK2 cells. Furthermore, the analogue 5d and PD176252 were subjected to cisplatin-induced inflammatory response in vitro. The results showed that 5d was able to better prevent this condition by effectively inhibiting its inflammatory response. Thus, this study will help in clinically reducing the side effects of cisplatin.
Collapse
Affiliation(s)
- Sen Yao
- School of Food and Biological Engineering , Hefei University of Technology , Hefei 230000 , China .
| | - Biao Wei
- School of Pharmacy , Anhui Medical University , Hefei , 230032 , China
| | - Mingjun Yu
- School of Food and Biological Engineering , Hefei University of Technology , Hefei 230000 , China .
| | - Xiaoming Meng
- School of Pharmacy , Anhui Medical University , Hefei , 230032 , China
| | - Meng He
- School of Food and Biological Engineering , Hefei University of Technology , Hefei 230000 , China .
| | - Risheng Yao
- School of Food and Biological Engineering , Hefei University of Technology , Hefei 230000 , China .
- Engineering Research Center of Bioprocess , Ministry of Education , PRC , Hefei University of Technology , Hefei 230009 , China
| |
Collapse
|
48
|
Sherif IO, Sarhan OM. Candesartan in a rat model of testicular toxicity: New insight on its protective mechanism. Exp Biol Med (Maywood) 2019; 244:593-601. [PMID: 31042053 DOI: 10.1177/1535370219842149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPACT STATEMENT Cisplatin is a commonly used drug in the treatment of solid tumors and its application is associated with testicular toxicity. The effect of candesartan in cisplatin-induced testicular toxicity and its fundamental mechanism of action were investigated. Candesartan had certainly repaired the testicular injury and ameliorated both biochemical and histopathological changes. Candesartan mitigated the gonadotoxicity induced by cisplatin via antioxidative, anti-inflammatory, and antiapoptotic actions.
Collapse
Affiliation(s)
- Iman O Sherif
- 1 Emergency Hospital, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Osama M Sarhan
- 2 Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
49
|
Kaushik S, Tomar A, Puthanmadhom Narayanan S, Nag TC, Arya DS, Bhatia J. Pitavastatin attenuates cisplatin-induced renal injury by targeting MAPK and apoptotic pathways. ACTA ACUST UNITED AC 2019; 71:1072-1081. [PMID: 30957246 DOI: 10.1111/jphp.13090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 03/09/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Anti-neoplastic drug cisplatin is prescribed widely for treatment of a variety of malignancies. Its use has been restricted lately due to severe renal toxicity. The purpose of current study was to investigate the effect of pitavastatin (a hypolipidaemic drug) in cisplatin-induced acute kidney injury in rats. METHOD Male Wistar rats (150-200 g) were treated with different doses of pitavastatin (0.16, 0.32 and 0.64 mg/kg per day p.o.; 10 days). On 7th day of the study, rats were administered cisplatin (8 mg/kg i.p.). Rats were euthanized (11th day), and blood and tissues were processed to evaluate biochemical, histopathological and ultrastructural parameters along with the analysis of immunohistochemistry and DNA-fragmentation studies. Protein expressions were analysed to demonstrate the underlying molecular mechanisms. KEY FINDINGS In the study group with cisplatin insult, KFT parameters were found to be elevated, concentration of apoptotic markers was found to be increased, histopathological and ultramicroscopical architecture was found to be distorted and the expression of MAPK proteins was also found to be elevated as compared to the normal group rats. Pitavastatin treatment alleviated all these anomalies. CONCLUSION Cisplatin-induced acute renal injury was improved on administration of pitavastatin via inhibition of MAPK and apoptotic pathway.
Collapse
Affiliation(s)
- Swati Kaushik
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Ameesha Tomar
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir Singh Arya
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
50
|
Wu J, Li DD, Li JY, Yin YC, Li PC, Qiu L, Chen LM. Identification of microRNA-mRNA networks involved in cisplatin-induced renal tubular epithelial cells injury. Eur J Pharmacol 2019; 851:1-12. [PMID: 30768982 DOI: 10.1016/j.ejphar.2019.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/01/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
Cisplatin is a widely used chemotherapeutic drug that often causes acute kidney injury (AKI) in cancer patients. The contribution of miRNAs to the cisplatin-induced renal tubular epithelial cell injury remains largely unknown. Here we performed an integrative network analysis of miRNA and mRNA expression profiles to shed light into the underlying mechanism of cisplatin-induced renal tubular epithelial cell injury. Microarray analysis identified 47 differentially expressed miRNAs, among them 26 were upregulated and 21 were downregulated. Moreover, integrating dysregulated miRNAs target prediction and altered mRNA expression enabled us to identify 1181 putative target genes for further bioinformatics analysis. Gene ontology (GO) analysis revealed that the putative target genes were involved in apoptosis process and regulation of transcription. Pathway analysis indicated that the top upregulated pathways included MAPK and p53 signaling pathway, while the top downregulated pathways were PI3K-Akt and Wnt signaling pathway. Further network analysis showed that MAPK signaling pathway and apoptosis with the highest degree were identified as core pathways, hsa-miR-9-3p and hsa-miR-371b-5p as the most critical miRNAs, and CASK, ASH1L, CDK6 etc. as hub target genes. In addition, the expression level change of selected five microRNAs (hsa-miR-4299, hsa-miR-297, hsa-miR-3135b, hsa-miR-9-3p, and hsa-miR-371b-5p) and two mRNAs( CASK and CDK6) were validated in cisplatin-induced HK-2 cells. Furthermore, a similar trend of expression level change was observed in NRK-52E cells by cisplatin treatment. Overall, our results provide the molecular basis and potential targets for the treatment of cisplatin-induced renal tubular cell injury.
Collapse
Affiliation(s)
- Jie Wu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Dan-Dan Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Jia-Yao Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Yi-Cong Yin
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Peng-Chang Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Ling Qiu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China.
| | - Li-Meng Chen
- Department of Nephrology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China.
| |
Collapse
|