201
|
Hino A, Muto S, Shimada Y, Hori S, Isotani S, Nagata M, Horie S. Impact of cisplatin-induced acute kidney injury on long-term renal function in patients with solid tumors. Clin Exp Nephrol 2023; 27:506-518. [PMID: 36941500 DOI: 10.1007/s10157-023-02324-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/26/2023] [Indexed: 03/22/2023]
Abstract
BACKGROUND The reality of cisplatin-induced acute kidney injury (CIA) and its effects on long-term renal function remain unclear. The aim of this study was to investigate the incidence and risk factors for CIA development, and if CIA is a useful predictor of long-term renal dysfunction after cisplatin treatment. METHODS This was a retrospective, single-center, observational, longitudinal follow-up, large cohort study in adult patients with solid tumors treated with cisplatin-based systematic chemotherapy. Electronic medical records were used for all demographic and medical data. AKI was defined by Kidney Disease Improving Global Outcomes (KDIGO) criteria. We assessed long-term renal dysfunction using %ΔeGFR/Y; (the last eGFR value during follow-up)-(the baseline eGFR)/(the baseline eGFR)/year of follow-up × 100. RESULTS A total of 2191 patients received 8,482 cycles of cisplatin. CIA was observed 359 times (4.2%). Significant risk factors for developing CIA, using multiple linear regression analysis, included: cisplatin administration immediately before the onset of CIA (p < 0.01), liver cancer (p = 0.02), colon cancer (p = 0.04), hypertension (p = 0.03), high estimated glomerular filtration rate (eGFR) (p < 0.01), and high C-reactive protein (CRP) (p = 0.04). Significant risk factors for %ΔeGFR/Y, using multivariate linear regression analysis, included: esophageal cancer (p < 0.01), lung cancer (p < 0.01), pharyngeal cancer (p = 0.02), Head and neck cancer (p < 0.01), liver cancer (p = 0.02), potassium (p < 0.01), and CIA (p < 0.01). CONCLUSIONS To our knowledge, this is the first study to show that CIA is a significant predictive risk factor for long-term renal dysfunction after cisplatin administration. Effective strategies are needed to prevent CIA in cancer patients.
Collapse
Affiliation(s)
- Amiko Hino
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Satoru Muto
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Department of Advanced Informatics for Genetic Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yosuke Shimada
- Intelligent Systems Laboratory, SECOM CO., LTD, Mitaka, Tokyo, Japan
- Department of Medical Electronic Intelligence Management, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Hori
- Department of Medical Electronic Intelligence Management, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Infection Control Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shuji Isotani
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Masayoshi Nagata
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Department of Advanced Informatics for Genetic Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
202
|
Li Z, Qiao X, Liu XM, Shi SH, Qiao X, Xu JY. Blocking xCT and PI3K/Akt pathway synergized with DNA damage of Riluzole-Pt(IV) prodrugs for cancer treatment. Eur J Med Chem 2023; 250:115233. [PMID: 36863224 DOI: 10.1016/j.ejmech.2023.115233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Cancer treatment requires the participation of multiple targets/pathways, and single approach is hard to effectively curb the proliferation and metastasis of carcinoma cells. In this work, we conjugated FDA-approved riluzole and platinum(II) drugs into a series of unreported riluzole-Pt(IV) compounds, which were designed to simultaneously target DNA, the solute carrier family 7 member 11 (SLC7A11, xCT), and the human ether a go-go related gene 1 (hERG1), to exert synergistic anticancer effect. Among them, c,c,t-[PtCl2(NH3)2(OH)(glutarylriluzole)] (compound 2) displayed excellent antiproliferative activity with IC50 value of 300-times lower than that of cisplatin in HCT-116, and optimal selectivity index between carcinoma and human normal liver cells (LO2). Mechanism studies indicated that compound 2 released riluzole and active Pt(II) species after entering cells to exhibit a prodrug behavior against cancer, which obviously increased DNA-damage and cell apoptosis, as well as suppressed metastasis in HCT-116. Compound 2 persisted in the xCT-target of riluzole and blocked the biosynthesis of glutathione (GSH) to trigger oxidative stress, which could boost the killing to cancer cells and reduce Pt-drug resistance. Meanwhile, compound 2 significantly inhibited invasion and metastasis of HCT-116 cells by targeting hERG1 to interrupt the phosphorylation of phosphatidylinositide 3-kinases/proteinserine-threonine kinase (PI3K/Akt), and reverse epithelial-mesenchymal transformation (EMT). Based on our results, the riluzole-Pt(IV) prodrugs studied in this work could be regarded as a new class of very promising candidates for cancer treatment compared to traditional platinum drugs.
Collapse
Affiliation(s)
- Zhe Li
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Shu-Hao Shi
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
203
|
Yuan XX, Duan YF, Luo C, Li L, Yang MJ, Liu TY, Cao ZR, Huang W, Bu X, Yue X, Liu RY. Disulfiram enhances cisplatin cytotoxicity by forming a novel platinum chelate Pt(DDTC) 3. Biochem Pharmacol 2023; 211:115498. [PMID: 36913990 DOI: 10.1016/j.bcp.2023.115498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/15/2023]
Abstract
Despite the use of targeted therapy in non-small cell lung cancer (NSCLC) patients, cisplatin (DDP)-based chemotherapy is still the main option. However, DDP resistance is the major factor contributing to the failure of chemotherapy. In this study, we tried to screen DDP sensitizers from an FDA-approved drug library containing 1374 small-molecule drugs to overcome DDP resistance in NSCLC. As a result, disulfiram (DSF) was identified as a DDP sensitizer: DSF and DDP had synergistic anti-NSCLC effects, which are mainly reflected in inhibiting tumor cell proliferation, plate colony formation and 3D spheroidogenesis and inducing apoptosis in vitro, as well as the growth of NSCLC xenografts in mice. Although DSF has recently been reported to promote the antitumor effect of DDP by inhibiting ALDH activity or modulating some important factors or pathways, unexpectedly, we found that DSF reacted with DDP to form a new platinum chelate, Pt(DDTC)3+, which might be one of the important mechanisms for their synergistic effect. Moreover, Pt(DDTC)3+ has a stronger anti-NSCLC effect than DDP, and its antitumor activity is broad-spectrum. These findings reveal a novel mechanism underlying the synergistic antitumor effect of DDP and DSF, and provide a drug candidate or a lead compound for the development of a new antitumor drug.
Collapse
Affiliation(s)
- Xue-Xia Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - You-Fa Duan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chunxiang Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Lu Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Meng-Jie Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Ting-Yu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhi-Rui Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Tumor Targeted Drugs & Guangzhou Enterprise Key Laboratory of Gene Medicine, Guangzhou DoublleBioproduct Co., Ltd., Guangzhou 510535, China
| | - Xianzhang Bu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xin Yue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Institute of Precision Medicine, The First Affiliated Hospital, SunYat-sen University, Guangzhou 510080, China.
| | - Ran-Yi Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
204
|
Hirsutidin Prevents Cisplatin-Evoked Renal Toxicity by Reducing Oxidative Stress/Inflammation and Restoring the Endogenous Enzymatic and Non-Enzymatic Level. Biomedicines 2023; 11:biomedicines11030804. [PMID: 36979784 PMCID: PMC10045162 DOI: 10.3390/biomedicines11030804] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Recent research has shown that phytocomponents may be useful in the treatment of renal toxicity. This study was conducted to evaluate the renal disease hirsutidin in the paradigm of renal toxicity induced by cisplatin. Male Wistar rats were given cisplatin (3 mg/kg body weight/day, for 25 days, i.p.) to induce renal toxicity. Experimental rats were randomly allocated to four different groups: group I received saline, group II received cisplatin, group III received cisplatin + hirsutidin (10 mg/kg)and group IV (per se)received hirsutidin (10 m/kg)for 25 days. Various biochemical parameters were assessed, oxidative stress (superoxide dismutase (SOD), glutathione transferase (GSH), malonaldehyde (MDA) and catalase (CAT)), blood-chemistry parameters (blood urea nitrogen (BUN) and cholesterol), non-protein-nitrogenous components (uric acid, urea, and creatinine), and anti-inflammatory-tumor necrosis factor-α (TNF-α), interleukin-1β(IL-1β). IL-6 and nuclear factor-kB (NFκB) were evaluated and histopathology was conducted. Hirsutidin alleviated renal injury which was manifested by significantly diminished uric acid, urea, urine volume, creatinine, and BUN, compared to the cisplatin group. Hirsutidin restored the activities of several antioxidant enzyme parameters—MDA, CAT, GSH, and SOD. Additionally, there was a decline in the levels of inflammatory markers—TNF-α, IL-1β, IL-6, and NFκB—compared to the cisplatin group. The current research study shows that hirsutidin may act as a therapeutic agent for the treatment of nephrotoxicity induced by cisplatin.
Collapse
|
205
|
Susa K, Kobayashi K, Galichon P, Matsumoto T, Tamura A, Hiratsuka K, Gupta NR, Yazdi IK, Bonventre JV, Morizane R. ATP/ADP biosensor organoids for drug nephrotoxicity assessment. Front Cell Dev Biol 2023; 11:1138504. [PMID: 36936695 PMCID: PMC10017499 DOI: 10.3389/fcell.2023.1138504] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Drug nephrotoxicity is a common healthcare problem in hospitalized patients and a major limitation during drug development. Multi-segmented kidney organoids derived from human pluripotent stem cells may complement traditional cell culture and animal experiments for nephrotoxicity assessment. Here we evaluate the capability of kidney organoids to investigate drug toxicity in vitro. Kidney organoids express renal drug transporters, OAT1, OAT3, and OCT2, while a human proximal tubular cell line shows the absence of OAT1 and OAT3. Tenofovir and aristolochic acid (AA) induce proximal tubular injury in organoids which is ameliorated by an OAT inhibitor, probenecid, without damage to podocytes. Similarly, cisplatin causes proximal tubular damage that can be relieved by an OCT inhibitor, cimetidine, collectively suggesting the presence of functional OATs and OCTs in organoid proximal tubules. Puromycin aminonucleoside (PAN) induced segment-specific injury in glomerular podocytes in kidney organoids in the absence of tubular injury. Reporter organoids were generated with an ATP/ADP biosensor, which may be applicable to high-throughput screening in the future. In conclusion, the kidney organoid is a useful tool for toxicity assessment in the multicellular context and may contribute to nephrotoxicity assessment during drug development.
Collapse
Affiliation(s)
- Koichiro Susa
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Nephrology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenichi Kobayashi
- Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital, Boston, MA, United States
| | - Pierre Galichon
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Takuya Matsumoto
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Akitoshi Tamura
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Ken Hiratsuka
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital, Boston, MA, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Navin R. Gupta
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital, Boston, MA, United States
| | - Iman K. Yazdi
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard-MIT Division of Health Sciences &Technology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joseph V. Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard-MIT Division of Health Sciences &Technology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ryuji Morizane
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital, Boston, MA, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| |
Collapse
|
206
|
Wen L, Wei Q, Livingston MJ, Dong G, Li S, Hu X, Li Y, Huo Y, Dong Z. PFKFB3 mediates tubular cell death in cisplatin nephrotoxicity by activating CDK4. Transl Res 2023; 253:31-40. [PMID: 36243313 PMCID: PMC10416729 DOI: 10.1016/j.trsl.2022.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Nephrotoxicity is a major side effect of cisplatin, a widely used cancer therapy drug. However, the mechanism of cisplatin nephrotoxicity remains unclear and no effective kidney protective strategies are available. Here, we report the induction of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) in both in vitro cell culture and in vivo mouse models of cisplatin nephrotoxicity. Notably, PFKFB3 was mainly induced in the nucleus of kidney tubular cells, suggesting a novel function other than its canonical role in glycolysis. Both pharmacological inhibition and genetic silencing of PFKFB3 led to the suppression of cisplatin-induced apoptosis in cultured renal proximal tubular cells (RPTCs). Moreover, cisplatin-induced kidney injury or nephrotoxicity was ameliorated in renal proximal tubule-specific PFKFB3 knockout mice. Mechanistically, we demonstrated the interaction of PFKFB3 with cyclin-dependent kinase 4 (CDK4) during cisplatin treatment, resulting in CDK4 activation and consequent phosphorylation and inactivation of retinoblastoma tumor suppressor (Rb). Inhibition of CDK4 reduced cisplatin-induced apoptosis in RPTCs and kidney injury in mice. Collectively, this study unveils a novel pathological role of PFKFB3 in cisplatin nephrotoxicity through the activation of the CDK4/Rb pathway, suggesting a new kidney protective strategy for cancer patients by blocking PFKFB3.
Collapse
Affiliation(s)
- Lu Wen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Siyao Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Xiaoru Hu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ying Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
207
|
Huang L, Lou K, Wang K, Liang L, Chen Y, Zhang J. Let-7c-5p Represses Cisplatin Resistance of Lung Adenocarcinoma Cells by Targeting CDC25A. Appl Biochem Biotechnol 2023; 195:1644-1655. [PMID: 36355336 DOI: 10.1007/s12010-022-04219-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/12/2022]
Abstract
Cisplatin broadly functions as a routine treatment for lung adenocarcinoma (LUAD) patients. However, primary and acquired cisplatin resistances frequently occur in the treatment of LUAD patients, seriously affecting the therapeutic effect of cisplatin in patients. We intended to illustrate the impact of let-7c-5p/cell division cycle 25A (CDC25A) axis on cisplatin resistance in LUAD. Expression of let-7c-5p and CDC25A was analyzed via quantitative real-time polymerase chain reaction. The interaction between the two was verified by dual-luciferase reporter detection. For detecting half-maximal inhibitory concentration value of cisplatin in LUAD cells and cell proliferation, we separately applied Cell Counting Kit-8 and colony formation assays. Furthermore, we measured cell apoptosis and cell cycle distribution via flow cytometry, as well as cell cycle-related protein expression via Western blot. Let-7c-5p was evidently downregulated in LUAD, while CDC25A was remarkably upregulated. Let-7c-5p upregulation arrested LUAD cells to proliferate, stimulated cell apoptosis, and arrested cell cycle in G0/G1 phase, thus enhancing sensitivity of LUAD cells to cisplatin. In terms of mechanism, CDC25A was directly targeted by let-7c-5p, and the influence of let-7c-5p overexpression on LUAD proliferation, apoptosis, cell cycle, and cisplatin resistance could be reversed by CDC25A upregulation. Let-7c-5p improved sensitivity of LUAD cells to cisplatin by modulating CDC25A, and let-7c-5p/CDC25A axis was an underlying target for the intervention of LUAD cisplatin resistance.
Collapse
Affiliation(s)
- Liang Huang
- Department of General Surgery, Taizhou First People's Hospital, Taizhou, 318020, China
| | - Kai Lou
- Emergency Department, Taizhou First People's Hospital, Taizhou, 318020, China
| | - Kunyu Wang
- Department of Thoracic Surgery, Taizhou First People's Hospital, Huangyan District, No.218 Hengjie Road, Taizhou, 318020, Zhejiang, China
| | - Lingxin Liang
- Department of Thoracic Surgery, Taizhou First People's Hospital, Huangyan District, No.218 Hengjie Road, Taizhou, 318020, Zhejiang, China
| | - Yi Chen
- Department of Thoracic Surgery, Taizhou First People's Hospital, Huangyan District, No.218 Hengjie Road, Taizhou, 318020, Zhejiang, China
| | - Jichen Zhang
- Department of Thoracic Surgery, Taizhou First People's Hospital, Huangyan District, No.218 Hengjie Road, Taizhou, 318020, Zhejiang, China.
| |
Collapse
|
208
|
Hu X, Ma Z, Li S, Wen L, Huo Y, Wu G, Manicassamy S, Dong Z. Fibroblast Growth Factor 2 Is Produced By Renal Tubular Cells to Act as a Paracrine Factor in Maladaptive Kidney Repair After Cisplatin Nephrotoxicity. J Transl Med 2023; 103:100009. [PMID: 36925200 PMCID: PMC10394613 DOI: 10.1016/j.labinv.2022.100009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 01/11/2023] Open
Abstract
Kidney repair after injury involves the cross-talk of injured kidney tubules with interstitial fibroblasts and immune cells. Although tubular cells produce multiple cytokines, the role and regulation of specific cytokines in kidney repair are largely undefined. In this study, we detected the induction of fibroblast growth factor 2 (FGF2) in mouse kidneys after repeated low-dose cisplatin (RLDC) treatment and in RLDC-treated renal proximal tubule cells in vitro. We further detected FGF2 in the culture medium of RLDC-treated renal tubular cells but not in the medium of control cells, indicating that RLDC induces FGF2 expression and secretion. Compared with the medium of control cells, the medium of RLDC-treated renal tubular cells was twice as effective in promoting fibroblast proliferation. Remarkably, the proliferative effect of the RLDC-treated cell medium was diminished by FGF2-neutralizing antibodies. In addition, the RLDC-treated cell medium induced the expression of fibrosis-related proteins, which was partially suppressed by FGF2-neutralizing antibodies. In mice, FGF2 deficiency partially prevented RLDC-induced decline in kidney function, loss of kidney weight, renal fibrosis, and inflammation. Together, these results indicate that FGF2 is produced by renal tubular cells after kidney injury and acts as an important paracrine factor in maladaptive kidney repair and disease progression.
Collapse
Affiliation(s)
- Xiaoru Hu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Zhengwei Ma
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia.
| | - Siyao Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Lu Wen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Yuqing Huo
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Georgia
| | | | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia.
| |
Collapse
|
209
|
Orwick A, Sears SM, Sharp CN, Doll MA, Shah PP, Beverly LJ, Siskind LJ. Lung cancer-kidney cross talk induces kidney injury, interstitial fibrosis, and enhances cisplatin-induced nephrotoxicity. Am J Physiol Renal Physiol 2023; 324:F287-F300. [PMID: 36727944 PMCID: PMC9988526 DOI: 10.1152/ajprenal.00317.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Patients with cancer represent a unique patient population with increased susceptibility to kidney disease. Drug-induced acute kidney injury (AKI) in patients with cancer is a common problem. Cisplatin is a highly effective treatment used in many solid-organ cancers and causes AKI in 30% of patients, increasing the risk of chronic kidney disease development. Most preclinical cisplatin toxicity studies have been completed in mice without cancer. We believe that the physiology of patients with cancer is not adequately represented in preclinical models, and the objective of this study was to determine how lung cancer will alter the nephrotoxicity of cisplatin. A genetically engineered mouse model and a syngeneic xenograft model of lung cancer were used. Mice were divided into the following four groups: 1) noncancer/vehicle, 2) noncancer/cisplatin, 3) cancer/vehicle, and 4) cancer/cisplatin. Mice were administered cisplatin via intraperitoneal injection once a week for 4 wk. Animals were euthanized 72 h following their final cisplatin injection. Mice with lung cancer had increased renal toxicity, injury, and fibrosis following repeated low doses of cisplatin. In addition, lung cancer alone induced kidney injury and fibrosis in the kidney before cisplatin treatment. In conclusion, this is the first study that we are aware of that assesses the impact of cancer on the kidney in conjunction with the nephrotoxicity of cisplatin. We believe that cancer is providing the first hit to the kidney and the subsequent damage from repeated doses of cisplatin becomes unsurmountable, leading to AKI and progression to chronic kidney disease.NEW & NOTEWORTHY Patients with cancer have impaired kidney function and increased susceptibility to nephrotoxic agents. Cisplatin is a commonly used chemotherapeutic with nephrotoxicity as the dose-limiting side effect. Cisplatin nephrotoxicity is almost exclusively studied in mice without cancer. Our current preclinical models do not adequately represent the complexity of patients with cancer. This study demonstrates increased renal toxicity, injury, and fibrosis in mice with lung cancer, which is exacerbated with cisplatin treatment. These results highlight the necessity of using preclinical models that more accurately capture the altered physiology of patients with cancer treated with cisplatin.
Collapse
Affiliation(s)
- Andrew Orwick
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Cierra N Sharp
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| | - Levi J Beverly
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| |
Collapse
|
210
|
Mancini M, Cerny MEV, Cardoso NS, Verissimo G, Maluf SW. Grape Seed Components as Protectors of Inflammation, DNA Damage, and Cancer. Curr Nutr Rep 2023; 12:141-150. [PMID: 36692807 DOI: 10.1007/s13668-023-00460-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE OF REVIEW Oxidative stress is related to the pathogenesis of several chronic diseases, including inflammatory processes. Free radicals excess increase not only oxidative stress but also genomic instability. Polyphenols are non-enzymatic antioxidants that act as a defense barrier against free radicals and non-radical oxidants. The purpose of this article was to review published articles relating dietary polyphenols contained in grape seed proanthocyanidin extracts with its potential for reversing DNA damage. RECENT FINDINGS Proanthocyanidin components exert pleiotropic actions having several biological, biochemical, and significant pharmacological effects and showed the ability to reduce cytotoxicity and genotoxicity. Grape seed proanthocyanidin extracts showed the ability to reduce cytotoxicity and genotoxicity through the comet assay and the micronucleus technique.
Collapse
Affiliation(s)
- Melissa Mancini
- Cytogenetics and Genome Stability Laboratory, University Hospital and Pharmacy Postgraduate Program, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Maria Eduarda Vieira Cerny
- Cytogenetics and Genome Stability Laboratory, University Hospital and Pharmacy Postgraduate Program, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Natali Silva Cardoso
- Cytogenetics and Genome Stability Laboratory, University Hospital and Pharmacy Postgraduate Program, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Sharbel Weidner Maluf
- Cytogenetics and Genome Stability Laboratory, University Hospital and Pharmacy Postgraduate Program, Federal University of Santa Catarina, Florianópolis, Brazil.
| |
Collapse
|
211
|
Flavonoids of Haloxylon salicornicum (Rimth) prevent cisplatin-induced acute kidney injury by modulating oxidative stress, inflammation, Nrf2, and SIRT1. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:49197-49214. [PMID: 36773264 DOI: 10.1007/s11356-023-25694-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 01/30/2023] [Indexed: 02/12/2023]
Abstract
Cisplatin (CIS) is an effective chemotherapeutic drug used for the treatment of many types of cancers, but its use is associated with adverse effects. Nephrotoxicity is a serious side effect of CIS and limits its therapeutic utility. Haloxylon salicornicum is a desert shrub used traditionally in the treatment of inflammatory disorders, but neither its flavonoid content nor its protective efficacy against CIS nephrotoxicity has been investigated. In this study, seven flavonoids were isolated from H. salicornicum methanolic extract (HSE) and showed in silico binding affinity with NF-κB, Keap1, and SIRT1. The protective effect of HSE against CIS nephrotoxicity was investigated. Rats received HSE (100, 200, and 400 mg/kg) for 14 days followed by a single injection of CIS. The drug increased Kim-1, BUN, and creatinine and caused multiple histopathological changes. CIS-administered rats showed an increase in renal ROS, MDA, NO, TNF-α, IL-1β, and NF-κB p65. HSE prevented tissue injury, and diminished ROS, NF-κB, and inflammatory mediators. HSE enhanced antioxidants and Bcl-2 and downregulated pro-apoptosis markers. These effects were associated with downregulation of Keap1 and microRNA-34a, and upregulation of SIRT1 and Nrf2/HO-1 signaling. In conclusion, H. salicornicum is rich in flavonoids, and its extract prevented oxidative stress, inflammation, and kidney injury, and modulated Nrf2/HO-1 and SIRT1 signaling in CIS-treated rats.
Collapse
|
212
|
Wang S, Chen Y, Wu H, Li X, Xiao H, Pan Q, Liu HF. Role of Transcription Factor EB in Mitochondrial Dysfunction of Cisplatin-Induced Acute Kidney Injury. Int J Mol Sci 2023; 24:ijms24033028. [PMID: 36769347 PMCID: PMC9917568 DOI: 10.3390/ijms24033028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Cisplatin, a widely used anticancer agent, can cause nephrotoxicity, including both acute kidney injury (AKI) and chronic kidney diseases, by accumulating in renal tubular epithelial cells (TECs). Mitochondrial pathology plays an important role in the pathogenesis of AKI. Based on the regulatory role of transcription factor EB (TFEB) in mitochondria, we investigated whether TFEB is involved in cisplatin-induced TEC damage. The results show that the expression of TFEB decreased in a concentration-dependent manner in both mouse kidney tissue and HK-2 cells when treated with cisplatin. A knockdown of TFEB aggravated cisplatin-induced renal TEC injury, which was partially reversed by TFEB overexpression in HK-2 cells. It was further observed that the TFEB knockdown also exacerbated cisplatin-induced mitochondrial damage in vitro, and included the depolarization of membrane potential, mitochondrial fragmentation and swelling, and the production of reactive oxygen species. In contrast, TFEB overexpression alleviated cisplatin-induced mitochondrial damage in TECs. These findings suggest that decreased TFEB expression may be a key mechanism of mitochondrial dysfunction in cisplatin-induced AKI, and that upregulation of TFEB has the potential to act as a therapeutic target to alleviate mitochondrial dysfunction and cisplatin-induced TEC injury. This study is important for developing therapeutic strategies to manipulate mitochondria through TFEB to delay AKI progression.
Collapse
Affiliation(s)
- Shujun Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yanse Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Hongluan Wu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xiaoyu Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
- Correspondence: (Q.P.); (H.-F.L.); Tel.: +86-759-2387164 (H.-F.L.)
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
- Correspondence: (Q.P.); (H.-F.L.); Tel.: +86-759-2387164 (H.-F.L.)
| |
Collapse
|
213
|
Li H, Ge H, Song X, Tan X, Xiong Q, Gong Y, Zhang L, He Y, Zhang W, Zhu P, Lin W, Xiao X. Neferine mitigates cisplatin-induced acute kidney injury in mice by regulating autophagy and apoptosis. Clin Exp Nephrol 2023; 27:122-131. [PMID: 36326941 DOI: 10.1007/s10157-022-02292-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE The nephrotoxicity caused by cisplatin severely limits the application and affects related platinum-based therapeutics. Neferine is a dibenzylisoquinoline alkaloid extracted from a Chinese medicinal herb (Nelumbo nucifera Gaertn), which can decrease cisplatin-induced apoptosis of NRK-52E cells by activating autophagy in vitro in our previous study. In this article, we aimed to further investigate the protective effect of neferine, against to the cispltain-induced kidney damage in mice. METHODS Six groups were designed in our study. Renal index, mice serum creatinine and blood urea nitrogen levels were detected after the mice were killed. HE staining was used to observe the pathological changes of each group. The apoptosis of mouse kidney tissue was detected by TUNEL. Immunofluorescence and Western blot were used to detect the expression of cleaved-caspase3 and LC3. The transmission electron microscope was used to reveal the changes of apoptosis and autophagy of renal tubular epithelial cells in different groups. RESULTS In our findings, the pathological changes of acute kidney injury were easily observed in cisplatin-treated mice while those in the neferine-pretreated groups were significantly alleviated. The apoptosis induced by cisplatin in mice increased evidently compared with the control group, which was decreased in the mice with neferine pretreatment. What' more, we found that autophagy increased obviously in mice pretreated by neferine contrast to the cisplatin-treated mice. CONCLUSION In our study, neferine can effectively alleviate cisplatin-induced renal injury in mice, as well act as an autophagy-regulator in kidney protection.
Collapse
Affiliation(s)
- Hui Li
- Department of Respiratory Medicine, The First Hospital of Changsha, Changsha, 410008, Hunan, China
| | - Huipeng Ge
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.,National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun Song
- Department of Science and Education, The First Hospital of Changsha, Changsha, 410008, Hunan, China
| | - Xin Tan
- Department of Pediatrics, The First Hospital of Changsha, Changsha, 410008, Hunan, China
| | - Qi Xiong
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yizi Gong
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Linlin Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yikai He
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Weiwei Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Peng Zhu
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Wei Lin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China. .,National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
214
|
Demirtas L, Gürbüzel M, Akbas EM, Tahirler H, Karatas O, Kemal Arslan Y. The Effects of Sunitinib in Healthy and Cisplatin-Induced Rats. Chem Biodivers 2023; 20:e202200704. [PMID: 36703598 DOI: 10.1002/cbdv.202200704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/04/2023] [Indexed: 01/28/2023]
Abstract
Sunitinib is a multitargeted kinase inhibitor that inhibits many receptor tyrosine kinases and has been used in the treatment of gastrointestinal stromal tumors, metastatic renal cell carcinoma, and pancreatic neuroendocrine tumors. In this study, the effects of sunitinib given to rats, both alone and after stress with cisplatin, were investigated. The animals were divided into four groups - (1) control group (C) administered interperitoneally with a single dose 0.9 % saline, (2) Cis group administered a single dose (7 mg/kg) of cisplatin, (3) Sun group administered 10 mg/kg sunitinib for seven days, and (4) Cis+Sun group administered 10 mg/kg sunitinib for seven days after a single dose (7 mg/kg) of cisplatin. After these applications, the rats were sacrificed, and blood and tissue samples were taken for biochemical and histopathological evaluations. Sunitinib did not show any effect on urea, creatine, and kidney IL1β and TGF-β3 expression levels when administered alone; it increased ALT, AST, and IL-38 levels. When sunitinib was given to the cisplatin-induced rats, it was observed that the increase in ALT, AST, and IL-38 levels increased more than the rats that was given only sunitinib. According to the data obtained, sunitinib does not cause a significant change in kidney tissue under both normal and stress conditions, while it creates stress in liver tissue. In addition, its toxicity in the liver becomes more certain as a result of its combination with cisplatin.
Collapse
Affiliation(s)
- Levent Demirtas
- Department of Internal Medicine, Faculty of Medicine, Erzincan Binali Yıldırım University, 24100, Erzincan, Türkiye
| | - Mehmet Gürbüzel
- Department of Medical Biology, Faculty of Medicine, Erzincan Binali Yıldırım University, 24100, Erzincan, Türkiye
| | - Emin Murat Akbas
- Department of Endocrinology, Faculty of Medicine, Erzincan Binali Yıldırım University, 24100, Erzincan, Türkiye
| | - Hilal Tahirler
- Department of Internal Medicine, Gülhane Training and Research Hospital, Health Sciences University, 06010, Ankara, Türkiye
| | - Ozhan Karatas
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, 58140, Sivas, Türkiye
| | - Yusuf Kemal Arslan
- Department of Biostatistics, Medical Faculty, Çukurova University, 01330, Adana, Türkiye
| |
Collapse
|
215
|
Shahbazi M, Zhang X, Dinh PC, Sanchez VA, Trendowski MR, Shuey MM, Nguyen T, Regeneron Genetics Center, Feldman DR, Vaughn DJ, Fung C, Kollmannsberger C, Martin NE, Einhorn LH, Cox NJ, Frisina RD, Travis LB, Dolan ME. Comprehensive association analysis of speech recognition thresholds after cisplatin-based chemotherapy in survivors of adult-onset cancer. Cancer Med 2023; 12:2999-3012. [PMID: 36097363 PMCID: PMC9939144 DOI: 10.1002/cam4.5218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/05/2022] Open
Abstract
PURPOSE Deficits in speech understanding constitute one of the most severe consequences of hearing loss. Here we investigate the clinical and genetic risk factors for symmetric deterioration of speech recognition thresholds (SRT) among cancer survivors treated with cisplatin. METHODS SRT was measured using spondaic words and calculating the mean of measurements for both ears with symmetric SRT values. For clinical associations, SRT-based hearing disability (SHD) was defined as SRT≥15 dB hearing loss and clinical variables were derived from the study dataset. Genotyped blood samples were used for GWAS with rank-based inverse normal transformed SRT values as the response variable. Age was used as a covariate in association analyses. RESULTS SHD was inversely associated with self-reported health (p = 0.004). Current smoking (p = 0.002), years of smoking (p = 0.02), BMI (p < 0.001), and peripheral motor neuropathy (p = 0.003) were positively associated with SHD, while physical activity was inversely associated with SHD (p = 0.005). In contrast, cumulative cisplatin dose, peripheral sensory neuropathy, hypertension, and hypercholesterolemia were not associated with SHD. Although no genetic variants had an association p value < 5 × 10-8 , 22 genetic variants were suggestively associated (p < 10-5 ) with SRT deterioration. Three of the top variants in 10 respective linkage disequilibrium regions were either positioned within the coding sequence or were eQTLs for genes involved in neuronal development (ATE1, ENAH, and ZFHX3). CONCLUSION Current results improve our understanding of risk factors for SRT deterioration in cancer survivors. Higher BMI, lower physical activity, and smoking are associated with SHD. Larger samples would allow for expansion of the current findings on the genetic architecture of SRT.
Collapse
Affiliation(s)
| | - Xindi Zhang
- Department of MedicineUniversity of ChicagoChicagoIllinoisUSA
| | - Paul C. Dinh
- Department of Medical OncologyIndiana UniversityIndianapolisIndianaUSA
| | - Victoria A. Sanchez
- Department of Otolaryngology—Head and Neck SurgeryUniversity of South FloridaTampaFloridaUSA
| | | | - Megan M. Shuey
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tessa Nguyen
- Center for Audiology, Speech, Language and LearningNorthwesthern UniversityChicagoIllinoisUSA
| | | | - Darren R. Feldman
- Department of Medical Oncology, Memorial Sloan‐Kettering Cancer CenterNew YorkNew YorkUSA
| | - David J. Vaughn
- Department of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical CenterRochesterNew YorkUSA
| | | | - Neil E. Martin
- Department of Radiation OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | | | - Nancy J. Cox
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Robert D. Frisina
- Departments of Medical Engineering and Communication Sciences and Disorders, Global Center for Hearing and Speech ResearchUniversity of South FloridaTampaFloridaUSA
| | - Lois B. Travis
- Department of Medical OncologyIndiana UniversityIndianapolisIndianaUSA
| | | |
Collapse
|
216
|
Ghassemi-Barghi N, Ehsanfar Z, Mohammadrezakhani O, Ashari S, Ghiabi S, Bayrami Z. Mechanistic Approach for Protective Effect of ARA290, a Specific Ligand for the Erythropoietin/CD131 Heteroreceptor, against Cisplatin-Induced Nephrotoxicity, the Involvement of Apoptosis and Inflammation Pathways. Inflammation 2023; 46:342-358. [PMID: 36085231 DOI: 10.1007/s10753-022-01737-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 12/01/2022]
Abstract
ARA 290, an 11-amino acid linear nonhematopoietic peptide derived from the three-dimensional structure of helix B of the erythropoietin (EPO), interacts selectively with the innate repair receptor (IRR) that arbitrates tissue protection. The aim of this study was to investigate the protective effects of ARA290 against cisplatin-induced nephrotoxicity. For this purpose, HEK-293 and ACHN cells were treated with ARA290 (50-400 nM) and cisplatin (2.5 μM) in pretreatment condition. Then, cytotoxicity, genotoxicity, oxidative stress parameters (ROS, GPx, SOD, and MDA), and inflammatory markers (TNFα, IL6, and IL1β) were evaluated. Furthermore, apoptotic cell death was assessed via caspase-3 activity and tunnel assay. To determine the molecular mechanisms of the possible nephroprotective effects of ARA290, gene and protein expressions of TNFα, IL1β, IL6, Caspase-3, Bax, and Bcl2 were evaluated by real-time PCR and western blot assay, respectively. The findings indicated that ARA290 significantly reduced the DNA damage parameters of comet assay and the frequency of micronuclei induced by cisplatin. Besides, ARA290 improved cisplatin-induced oxidative stress by reducing MDA/ROS levels and enhancing antioxidant enzyme levels. In addition, reduced levels of pro-inflammatory cytokines indicated that cisplatin-induced renal inflammation was mitigated upon the treatment with ARA290. Besides, ARA290 ameliorates cisplatin-induced cell injury by antagonizing apoptosis. Furthermore, the molecular findings indicated that gene and protein levels of TNFα, IL1β, IL6, Caspase-3, and Bax were significantly decreased and gene and protein levels of Bcl2 significantly increased in the ARA290 plus cisplatin group compared with the cisplatin group. These findings revealed that ARA290 as a potent chemo-preventive agent exerted a protective effect on cisplatin-induced nephrotoxicity mostly through its anti-apoptotic, anti-inflammatory, and antioxidant potentials and also suggested that ARA290 might be a new therapeutic approach for patients with acute kidney injury.
Collapse
Affiliation(s)
- Nasrin Ghassemi-Barghi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | | | - Omid Mohammadrezakhani
- Student Research Committee, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Sorour Ashari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shamim Ghiabi
- Department of Medical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Bayrami
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
217
|
Nephroprotective Effect of Diosmin against Cisplatin-Induced Kidney Damage by Modulating IL-1β, IL-6, TNFα and Renal Oxidative Damage. Molecules 2023; 28:molecules28031302. [PMID: 36770968 PMCID: PMC9920922 DOI: 10.3390/molecules28031302] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
Cisplatin (CP) is a platinum compound of the alkylating agent class that is used for the treatment of various types of cancer. However, CP treatments in cancer patients are accountable for nephrotoxicity, as it is a major adverse effect. Hence, this research study was proposed to investigate the nephroprotective effect of diosmin, a flavonoid glycoside of hesperidin derivatives against cisplatin-induced kidney damage. Wistar rats received a single intraperitoneal (i.p) injection of CP (7.5 mg/kg, i.p) to induce nephrotoxicity. The administration of CP significantly (p < 0.001) increased the markers of kidney function test (creatinine, blood urea nitrogen, and uric acid) and demonstrated histopathological changes in the kidney of the CP-treated nephrotoxic group. In addition, the CP-treated nephrotoxic group demonstrated a significant (p < 0.001) increase in lipid peroxidation (LPO) levels and depleted activities of reduced glutathione (GSH), glutathione peroxidase (GPx), glutathione reductase (GR), superoxide dismutase (SOD) and catalase (CAT).However, diosmin (100 and 200 mg/kg) treatments significantly reduced the elevated levels of kidney function test parameters and restored structural changes in the kidney (p < 0.001). The administration of diosmin (100 and 200 mg/kg) significantly (p < 0.001) reduced LPO levels, increased GSH content and showed improvements in the activities of GPx, GR, SOD and CAT. The markers of inflammatory cytokines such as IL-1β, IL-6 and TNFα significantly (p < 0.001) increased in the CP-treated nephrotoxic group, whereas diosmin (100 and 200 mg/kg) treatments significantly (p < 0.001) reduced the elevated levels of these cytokines. The findings of this research demonstrate the nephroprotective effect of diosmin against CP-induced kidney damage. Therefore, we conclude that diosmin may be used as a supplement in the management of nephrotoxicity associated with CP treatments in cancer patients.
Collapse
|
218
|
Su J, He T, You J, Cao J, Wang Q, Cao S, Mei Q, Zeng J, Liu L. Therapeutic effect and underlying mechanism of Shenkang injection against cisplatin-induced acute kidney injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115805. [PMID: 36216195 DOI: 10.1016/j.jep.2022.115805] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenkang injection (SKI), a Chinese patent medicine injection, has been approved for the treatment of chronic kidney disease (CKD) due to its definite clinical therapeutic efficacy. However, the effect and associated underlying mechanism of Shenkang injection against cisplatin (CDDP)-induced acute kidney injury (AKI) has not yet been well elucidated. AIM OF THE STUDY This study aims to investigate the therapeutic effect and associated underlying mechanism of Shenkang injection against CDDP-induced AKI. MATERIALS AND METHODS We established a CDDP-induced AKI mouse model to evaluate renal function by biochemical markers measurement and to observe histopathological alterations by haemotoxylin and eosin (HE)-staining sections of renal. In addition, the distribution of representative components of SKI in the kidneys of mice was evaluated by liquid chromatography tandem mass spectrometry (LC-MS/MS). Furthermore, the degree of oxidative stress and inflammation were assessed by detecting the levels of inflammatory cytokines and oxidants, while the related mechanisms were elucidated by network pharmacology. RESULTS CDDP could induce excessive inflammation and severe injury to the kidneys of mice. However, SKI significantly ameliorated the kidney damages and improved the renal function by reducing the levels of renal function markers (SCr, BUN and urine protein), and inhibiting the production of inflammatory cytokines IL-34, IL-6 and TNF-α. SKI repaired oxidative balance through up-regulation of antioxidants SOD and GSH and down-regulated oxidants MDA. Moreover, 4 components from SKI were detected in the kidney by LC-MS/MS quantification. In addition, pharmacology network indicated the PI3K/AKT, TNF, MAPK, and p53 were the possible signaling pathways for the therapeutic effect of SKI against CDDP-induced AKI, which were related to inflammation, oxidative stress and apoptosis. CONCLUSION In the present study, we for the first time demonstrated that SKI alleviates CDDP-induced nephrotoxicity by antioxidant and anti-inflammation via regulating PI3K/AKT, MAPK, TNF, and p53 signaling pathways. The study may provide a scientific rationale for the clinical indication of SKI.
Collapse
Affiliation(s)
- Jiahan Su
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Luzhou New Drug Evaluation and Research Center, Luzhou, Sichuan, 646000, China
| | - Tingting He
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Luzhou New Drug Evaluation and Research Center, Luzhou, Sichuan, 646000, China
| | - Jing You
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; The People's Hospital of DaZhu, Dazhou, Sichuan, 635000, China
| | - Jingjie Cao
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qianru Wang
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shousong Cao
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qibing Mei
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Luzhou New Drug Evaluation and Research Center, Luzhou, Sichuan, 646000, China
| | - Jing Zeng
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Li Liu
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
219
|
Alassaf N, Attia H. Autophagy and necroptosis in cisplatin-induced acute kidney injury: Recent advances regarding their role and therapeutic potential. Front Pharmacol 2023; 14:1103062. [PMID: 36794281 PMCID: PMC9922871 DOI: 10.3389/fphar.2023.1103062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Cisplatin (CP) is a broad-spectrum antineoplastic agent, used to treat many different types of malignancies due to its high efficacy and low cost. However, its use is largely limited by acute kidney injury (AKI), which, if left untreated, may progress to cause irreversible chronic renal dysfunction. Despite substantial research, the exact mechanisms of CP-induced AKI are still so far unclear and effective therapies are lacking and desperately needed. In recent years, necroptosis, a novel subtype of regulated necrosis, and autophagy, a form of homeostatic housekeeping mechanism have witnessed a burgeoning interest owing to their potential to regulate and alleviate CP-induced AKI. In this review, we elucidate in detail the molecular mechanisms and potential roles of both autophagy and necroptosis in CP-induced AKI. We also explore the potential of targeting these pathways to overcome CP-induced AKI according to recent advances.
Collapse
Affiliation(s)
- Noha Alassaf
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,*Correspondence: Noha Alassaf,
| | - Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,Department of Biochemistry, College of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
220
|
Dobrek L. A Synopsis of Current Theories on Drug-Induced Nephrotoxicity. Life (Basel) 2023; 13:life13020325. [PMID: 36836682 PMCID: PMC9960203 DOI: 10.3390/life13020325] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
The overriding goal of the treatment of patients is its effectiveness and safety. However, all medications currently being used also exert some adverse pharmaceutical reactions, which may be regarded as an unintended but inevitable cost of pharmacotherapy. The kidney, as the main organ that eliminates xenobiotics, is an organ especially predisposed and vulnerable to the toxic effects of drugs and their metabolites during their excretion from the body. Moreover, some drugs (e.g., aminoglycosides, cyclosporin A, cisplatin, amphotericin B, and others) have a "preferential" nephrotoxicity potential, and their use is associated with an increased risk of kidney damage. Drug nephrotoxicity is, therefore, both a significant problem and a complication of pharmacotherapy. It should be noted that, currently, there is no generally recognized definition of drug-induced nephrotoxicity and no clear criteria for its diagnosis. This review briefly describes the epidemiology and diagnosis of drug-induced nephrotoxicity and characterizes its pathomechanisms, including immunological and inflammatory disturbances, altered kidney blood flow, tubulointerstitial injury, increased lithogenesis-crystal nephropathy, rhabdomyolysis, and thrombotic microangiopathy. The study also lists the basic drugs with nephrotoxicity potential and provides a short overview of the preventive methods for reducing the risk of drug-related kidney damage developing.
Collapse
Affiliation(s)
- Lukasz Dobrek
- Department of Clinical Pharmacology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
221
|
Xiang Y, Fu Y, Wu W, Tang C, Dong Z. Autophagy in acute kidney injury and maladaptive kidney repair. BURNS & TRAUMA 2023; 11:tkac059. [PMID: 36694860 PMCID: PMC9867874 DOI: 10.1093/burnst/tkac059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/30/2022] [Accepted: 12/20/2022] [Indexed: 01/23/2023]
Abstract
Acute kidney injury (AKI) is a major renal disease characterized by a sudden decrease in kidney function. After AKI, the kidney has the ability to repair, but if the initial injury is severe the repair may be incomplete or maladaptive and result in chronic kidney problems. Autophagy is a highly conserved pathway to deliver intracellular contents to lysosomes for degradation. Autophagy plays an important role in maintaining renal function and is involved in the pathogenesis of renal diseases. Autophagy is activated in various forms of AKI and acts as a defense mechanism against kidney cell injury and death. After AKI, autophagy is maintained at a relatively high level in kidney tubule cells during maladaptive kidney repair but the role of autophagy in maladaptive kidney repair has been controversial. Nonetheless, recent studies have demonstrated that autophagy may contribute to maladaptive kidney repair after AKI by inducing tubular degeneration and promoting a profibrotic phenotype in renal tubule cells. In this review, we analyze the role and regulation of autophagy in kidney injury and repair and discuss the therapeutic strategies by targeting autophagy.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Wenwen Wu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | | |
Collapse
|
222
|
Selim MS, Kassem AB, El-Bassiouny NA, Salahuddin A, Abu El-Ela RY, Hamza MS. Polymorphic renal transporters and cisplatin's toxicity in urinary bladder cancer patients: current perspectives and future directions. Med Oncol 2023; 40:80. [PMID: 36650399 PMCID: PMC9845168 DOI: 10.1007/s12032-022-01928-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/10/2022] [Indexed: 01/19/2023]
Abstract
Urinary bladder cancer (UBC) holds a potentially profound social burden and affects over 573,278 new cases annually. The disease's primary risk factors include occupational tobacco smoke exposure and inherited genetic susceptibility. Over the past 30 years, a number of treatment modalities have emerged, including cisplatin, a platinum molecule that has demonstrated effectiveness against UBC. Nevertheless, it has severe dose-limiting side effects, such as nephrotoxicity, among others. Since intracellular accumulation of platinum anticancer drugs is necessary for cytotoxicity, decreased uptake or enhanced efflux are the root causes of platinum resistance and response failure. Evidence suggests that genetic variations in any transporter involved in the entry or efflux of platinum drugs alter their kinetics and, to a significant extent, determine patients' responses to them. This review aims to consolidate and describe the major transporters and their polymorphic variants in relation to cisplatin-induced toxicities and resistance in UBC patients. We concluded that the efflux transporters ABCB1, ABCC2, SLC25A21, ATP7A, and the uptake transporter OCT2, as well as the organic anion uptake transporters OAT1 and OAT2, are linked to cisplatin accumulation, toxicity, and resistance in urinary bladder cancer patients. While suppressing the CTR1 gene's expression reduced cisplatin-induced nephrotoxicity and ototoxicity, inhibiting the expression of the MATE1 and MATE2-K genes has been shown to increase cisplatin's nephrotoxicity and resistance. The roles of ABCC5, ABCA8, ABCC10, ABCB10, ABCG1, ATP7B, ABCG2, and mitochondrial SLC25A10 in platinum-receiving urinary bladder cancer patients should be the subject of further investigation.
Collapse
Affiliation(s)
- Mohamed S Selim
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| | - Amira B Kassem
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Noha A El-Bassiouny
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Ahmad Salahuddin
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
- Biochemistry Department, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Raghda Y Abu El-Ela
- Medical Oncology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Marwa Samir Hamza
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
223
|
Abstract
A library of 12 palladium(II) complexes of the type [PdBr(iPr2-bimy)(L∧X)] comprising 10 dithiocarbamato (R2NCS2-) and two xanthato (ROCS2-) ligands have been prepared and fully characterized. With these complexes in hand, the electronic and steric properties of the bidentate, monoanionic ligands were evaluated using the HEP2 and %Vbur methodologies. Moreover, the construction of the first stereoelectronic map for dithiocarbamates enabled the in-principle identification of optimal ligand parameters for enhanced cytotoxic activities of their gold(III) complexes. This application of the stereoelectronic map showcases its viability as a useful tool to establish structure-activity relationships for rational ligand design.
Collapse
Affiliation(s)
- Jia Nuo Leung
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, Singapore 117453, Singapore
| | - Han Vinh Huynh
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, Singapore 117453, Singapore
| |
Collapse
|
224
|
Wei M, Gao Y, Cheng D, Zhang H, Zhang W, Shen Y, Huang Q, An X, Wang B, Yu Z, Wang N, Chen H, Xu Y, Gui D. Notoginsenoside Fc ameliorates renal tubular injury and mitochondrial damage in acetaminophen-induced acute kidney injury partly by regulating SIRT3/SOD2 pathway. Front Med (Lausanne) 2023; 9:1055252. [PMID: 36714147 PMCID: PMC9875593 DOI: 10.3389/fmed.2022.1055252] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/28/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Mitochondria dysfunction is one of the primary causes of tubular injury in acute kidney injury (AKI). Notoginsenoside Fc (Fc), a new saponin isolated from Panax notoginseng, exhibited numerous pharmacological actions. However, the beneficial effects of Fc on renal tubular impairment and mitochondrial dysfunction in AKI have not been fully studied. Methods In this study, we established acetaminophen (APAP)-induced AKI model in mice to examine the therapeutic impacts of Fc on AKI. Results Our results showed that Fc could decrease the levels of the serum creatinine (Scr), blood urea nitrogen (BUN) and Cystatin C in mice with AKI. Fc also ameliorated renal histopathology, renal tubular cells apoptosis and restored expression of apoptosis-related proteins such as Bax, Bcl-2 and caspase3 (C-caspase3). Additionally, Fc increased the protein expression of SIRT3 and SOD2 in kidneys from mice with AKI. In vitro studies further showed Fc reduced the apoptosis of HK-2 cells exposure to APAP, attenuated the loss of mitochondrial membrane potential and decreased the formation of mitochondrial superoxide. Fc also partly restored the protein expression of Bax, Bcl-2, C-Caspase3, SIRT3, and SOD2 in HK-2 cells exposure to APAP. Conclusion In summary, Fc might reduce renal tubular injury and mitochondrial dysfunction in AKI partly through the regulation of SIRT3/SOD2 pathway.
Collapse
Affiliation(s)
- Miaomiao Wei
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China,Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuancheng Gao
- The Third Affiliated Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dongsheng Cheng
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiying Zhang
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Department of Nephrology, Shanghai Yangpu Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yilan Shen
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qunwei Huang
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoning An
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Wang
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhonghai Yu
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongbo Chen
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China,*Correspondence: Hongbo Chen
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China,Youhua Xu
| | - Dingkun Gui
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Central Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Dingkun Gui
| |
Collapse
|
225
|
Mobasher MA, Ahmed EI, Hakami NY, Germoush MO, Awad NS, Khodeer DM. The Combined Effect of Licorice Extract and Bone Marrow Mesenchymal Stem Cells on Cisplatin-Induced Hepatocellular Damage in Rats. Metabolites 2023; 13:94. [PMID: 36677019 PMCID: PMC9861302 DOI: 10.3390/metabo13010094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Drug-induced liver damage is a life-threatening disorder, and one major form of it is the hepatotoxicity induced by the drug cisplatin. In folk medicine, Licorice (Glycyrrhiza glabra (is used for detoxification and is believed to be a potent antioxidant. Currently, the magically self-renewable potential of bone marrow mesenchymal stem cells (BM-MSCs) has prompted us to explore their hepatoregenerative capability. The impact of G. glabra extract (GGE) and BM-MSCs alone and, in combination, on protecting against hepatotoxicity was tested on cisplatin-induced liver injury in rats. Hepatic damage, as revealed by liver histopathology and increased levels of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), and malondialdehyde (MDA), was elevated in rats by received 7 mg/kg of cisplatin intraperitoneally. The combination of GGE and BM-MSCs returned the enzyme levels to near the normal range. It also improved levels of liver superoxide dismutase (SOD) and glutathione (GSH) and reduced MDA levels. Additionally, it was found that when GGE and BM-MSCs were used together, they significantly downregulated caspase9 (Casp9), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), and interleukin-1β (IL-1β), which are involved in severe proinflammatory and apoptotic signaling cascades in the liver. Moreover, combining GGE and BM-MSCs led to the normal result of hepatocytes in several examined liver histological sections. Therefore, our findings suggest that GGE may have protective effects against oxidative liver damage and the promising regenerative potential of BM-MSCs.
Collapse
Affiliation(s)
- Maysa A. Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Eman Ibrahim Ahmed
- Pharmacology and Therapeutics Department, College of Medicine, Jouf University, Sakaka 72346, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt
| | - Nora Y. Hakami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
| | - Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nabil S Awad
- Department of Genetics, Faculty of Agriculture and Natural Resources, Aswan University, Aswan 81528, Egypt
- College of Biotechnology, Misr University for Science and Technology, Giza 12563, Egypt
| | - Dina M. Khodeer
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
226
|
Corridon PR. Still finding ways to augment the existing management of acute and chronic kidney diseases with targeted gene and cell therapies: Opportunities and hurdles. Front Med (Lausanne) 2023; 10:1143028. [PMID: 36960337 PMCID: PMC10028138 DOI: 10.3389/fmed.2023.1143028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
The rising global incidence of acute and chronic kidney diseases has increased the demand for renal replacement therapy. This issue, compounded with the limited availability of viable kidneys for transplantation, has propelled the search for alternative strategies to address the growing health and economic burdens associated with these conditions. In the search for such alternatives, significant efforts have been devised to augment the current and primarily supportive management of renal injury with novel regenerative strategies. For example, gene- and cell-based approaches that utilize recombinant peptides/proteins, gene, cell, organoid, and RNAi technologies have shown promising outcomes primarily in experimental models. Supporting research has also been conducted to improve our understanding of the critical aspects that facilitate the development of efficient gene- and cell-based techniques that the complex structure of the kidney has traditionally limited. This manuscript is intended to communicate efforts that have driven the development of such therapies by identifying the vectors and delivery routes needed to drive exogenous transgene incorporation that may support the treatment of acute and chronic kidney diseases.
Collapse
Affiliation(s)
- Peter R. Corridon
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biomedical Engineering, Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- *Correspondence: Peter R. Corridon,
| |
Collapse
|
227
|
Altındağ F, Ergen H. Sinapic acid alleviates cisplatin-induced acute kidney injury by mitigating oxidative stress and apoptosis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:12402-12411. [PMID: 36107295 DOI: 10.1007/s11356-022-22940-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
Cisplatin is an anticancer agent with many side effects such as nephrotoxicity, as well as being widely used in the treatment of many tumor types. Sinapic acid has antioxidant, anti-inflammatory, antihyperglycemic, and antiapoptotic effects. This study aimed to investigate the possible beneficial effects of sinapic acid against cisplatin-induced nephrotoxicity. Twenty-eight Wistar albino male rats were used. The groups are as follows: control, cisplatin, cisplatin + sinapic acid, and sinapic acid groups (n = 7). The control group received 1 ml of single-dose intraperitoneal saline on the first day of the study. The cisplatin group was given a single dose of 7 mg/kg cisplatin intraperitoneal. Animals in the cisplatin + sinapic acid group were given sinapic acid for 7 days 25 mg/kg, 3 days after oral gavage administration of 7 mg/kg cisplatin intraperitoneal. The sinapic acid group was given 25 mg/kg/day of sinapic acid by oral gavage for 7 days after the 3rd day of the study. The kidney of the rats was examined by stereological, immunohistochemical, histopathological, and biochemical methods. According to the stereological findings of the study, while the volume of the glomerulus cortex and filtration gap increased, the volume of the medulla decreased, and there was no significant difference in tubular volume in the CP group compared to the control group. The volume of the glomerulus, cortex, and filtration gap of the cisplatin + sinapic acid group was significantly reduced compared to the cisplatin group (p˂0.05). Histopathologically, it was observed the enlargement of the filtration gap, tubular dilatation, atrophy, renal fibrosis, deterioration of the microvilli, and necrosis in the tubular epithelial cells in the cisplatin group. In the cisplatin + sinapic acid group, these pathologies decreased compared to the cisplatin group. Compared to the control group, caspase-3 expression, urea, creatine, and malondialdehyde increased, while Bcl-2 and catalase decreased in the cisplatin group. However, caspase-3 expression, urea, creatine, and malondialdehyde were decreased, while Bcl-2 and catalase increased in the cisplatin + sinapic acid group compared to the cisplatin group. The results of our study showed that sinapic acid reduced the nephrotoxicity induced by cisplatin.
Collapse
Affiliation(s)
- Fikret Altındağ
- Department of Histology and Embryology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey.
| | - Hidayet Ergen
- Department of Histology and Embryology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| |
Collapse
|
228
|
Fan J, Xu X, Li Y, Zhang L, Miao M, Niu Y, Zhang Y, Zhang A, Jia Z, Wu M. A novel 3-phenylglutaric acid derivative (84-B10) alleviates cisplatin-induced acute kidney injury by inhibiting mitochondrial oxidative stress-mediated ferroptosis. Free Radic Biol Med 2023; 194:84-98. [PMID: 36403736 DOI: 10.1016/j.freeradbiomed.2022.11.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
Cisplatin is one of the most effective chemotherapy drugs and is widely used for cancer treatment. However, its clinical use is limited by nephrotoxicity. Emerging findings suggested that both ferroptosis and mitochondrial dysfunction mediate cisplatin-induced nephrotoxicity. In the current study, a novel 3-phenylglutaric acid derivative 5-[[2-(4-methoxyphenoxy)-5-(trifluoromethyl)phenyl]amino]-5-oxo-3-phenylpentanoic acid (referred to as 84-B10) was found to play a protective role in cisplatin-induced acute kidney injury with no tumor promoting effects. A genome-wide transcriptome analysis indicated that the protective effect of 84-B10 might be dependent on antagonizing ferroptosis. In accordance, lipid peroxide accumulation and downregulation of key ferroptosis suppressors were reversed using 84-B10 treatment both in vivo and in vitro. In addition, 84-B10 inhibited cisplatin-induced mitochondrial damage and mitochondrial reactive oxygen species (mtROS) production and restored superoxide dismutases (SODs). Furthermore, 84-B10 showed similar therapeutic effects to MnTBAP (a cell-permeable SOD mimetic) in eliminating mtROS, restoring mitochondrial homeostasis, and inhibiting ferroptosis under cisplatin challenge. Comparable effects of 84-B10 and liproxstatin-1 in ameliorating cisplatin-induced ferroptosis were observed. However, liproxstatin-1 failed to prevent mitochondrial dysfunction. These data indicated that mtROS might act upstream of cisplatin-induced tubular ferroptosis. Taken together, the novel 3-phenylglutaric acid derivative 84-B10 showed therapeutic potential against cisplatin-induced nephrotoxicity possibly by restoring mitochondria homeostasis and inhibiting mtROS-induced ferroptosis, which suggests the potential use of 84-B10 in preventing and treating cisplatin-nephrotoxicity.
Collapse
Affiliation(s)
- Jiaojiao Fan
- School of Medicine, Southeast University, Nanjing, 210009, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Xinyue Xu
- School of Medicine, Southeast University, Nanjing, 210009, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yuting Li
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Lingge Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Mengqiu Miao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yujia Niu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Aihua Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| | - Mengqiu Wu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
229
|
Topcu A, Saral S, Mercantepe T, Akyildiz K, Tumkaya L, Yilmaz A. The effects of apelin-13 against cisplatin-induced nephrotoxicity in rats. Drug Chem Toxicol 2023; 46:77-87. [PMID: 34894944 DOI: 10.1080/01480545.2021.2011309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Acute kidney injury (AKI) is observed in nearly 60% of patients undergoing cisplatin (CP) therapy. The aim of this study was to reveal the potential effects of apelin-13 (AP-13) in the prevention of CP-induced renal toxicity, together with its antioxidant and anti-inflammatory effect mechanisms. Four experimental groups were established. Group 1, the control group, received 0.9% saline solution alone intraperitoneally (IP). Group 2, the CP group, received CP IP at 5 mg/kg once weekly for four weeks for induction of nephrotoxicity. In Group 3, the CP + Apelin-13 (AP-13) group, AP-13 was prepared at 20 nmol kg/d in sterile pyrogen-free saline before injection every day for four weeks and administered IP. CP was administered IP at 5 mg/kg once weekly for four weeks for induction of nephrotoxicity. In Group 4, the AP-13 group, AP-13 was prepared at 20 nmol kg/d in sterile pyrogen-free 0.9% saline before injection every day for four weeks and administered IP. Thiobarbituric acid reactive substances (TBARS), thiol (-SH), interleukin-1 beta, cleaved caspase-3, 8-hydroxy 2-deoxyguanosine (8-OHdG), and nuclear factor kappa B (NF-κβ/p65) levels were then measured. Increased oxidative stress, inflammation, and apoptosis as a result of CP application activated the cascade. However, AP-13 administration reduced the oxidative stress increased by CIS with the determined antioxidant effect and reduced the damage by increasing total -SH levels. 8-OHdG and NF-κβ/p65, which were up-regulated by triggering oxidative stress and inflammation, were down-regulated through the antioxidant and anti-inflammatory effects of AP-13.
Collapse
Affiliation(s)
- Atilla Topcu
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Sinan Saral
- Department of Physiology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerimali Akyildiz
- Department of Medical Services and Techniques, Health Care Services Vocational School, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
230
|
Marotta C, Giorgi E, Binacchi F, Cirri D, Gabbiani C, Pratesi A. An overview of recent advancements in anticancer Pt(IV) prodrugs: New smart drug combinations, activation and delivery strategies. Inorganica Chim Acta 2023. [DOI: 10.1016/j.ica.2023.121388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
231
|
Cisplatin nephrotoxicity: new insights and therapeutic implications. Nat Rev Nephrol 2023; 19:53-72. [PMID: 36229672 DOI: 10.1038/s41581-022-00631-7] [Citation(s) in RCA: 211] [Impact Index Per Article: 105.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 11/08/2022]
Abstract
Cisplatin is an effective chemotherapeutic agent for various solid tumours, but its use is limited by adverse effects in normal tissues. In particular, cisplatin is nephrotoxic and can cause acute kidney injury and chronic kidney disease. Preclinical studies have provided insights into the cellular and molecular mechanisms of cisplatin nephrotoxicity, which involve intracellular stresses including DNA damage, mitochondrial pathology, oxidative stress and endoplasmic reticulum stress. Stress responses, including autophagy, cell-cycle arrest, senescence, apoptosis, programmed necrosis and inflammation have key roles in the pathogenesis of cisplatin nephrotoxicity. In addition, emerging evidence suggests a contribution of epigenetic changes to cisplatin-induced acute kidney injury and chronic kidney disease. Further research is needed to determine how these pathways are integrated and to identify the cell type-specific roles of critical molecules involved in regulated necrosis, inflammation and epigenetic modifications in cisplatin nephrotoxicity. A number of potential therapeutic targets for cisplatin nephrotoxicity have been identified. However, the effects of renoprotective strategies on the efficacy of cisplatin chemotherapy needs to be thoroughly evaluated. Further research using tumour-bearing animals, multi-omics and genome-wide association studies will enable a comprehensive understanding of the complex cellular and molecular mechanisms of cisplatin nephrotoxicity and potentially lead to the identification of specific targets to protect the kidney without compromising the chemotherapeutic efficacy of cisplatin.
Collapse
|
232
|
Yu X, Ji X, Fan Y, Yu B, Wang X, Feng C, Zhang L, Song C, Zhang X. Static Magnetic Fields Protect against Cisplatin-Induced Kidney Toxicity. Antioxidants (Basel) 2022; 12:73. [PMID: 36670933 PMCID: PMC9854588 DOI: 10.3390/antiox12010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Cisplatin is one of the most widely used anti-cancer drugs that can effectively inhibit the growth of multiple types of cancer. However, its clinical application is limited by its severe side effects, especially kidney toxicity, caused by cisplatin-induced oxidative stress, inflammation and kidney cell apoptosis. Here, we found that moderate (a few hundred mT) quasi-uniform static magnetic fields (SMFs) could inhibit cisplatin-induced renal proximal tubular cell death, especially the vertically downward direction SMF. RNA-seq experiments demonstrate that SMFs induced differential gene expressions that are closely associated with oxidative stress, apoptosis, cytokine production, transmembrane transport and DNA repair. In vivo experiments show that SMFs can reduce cisplatin-induced kidney injury in cisplatin-administrated tumor-bearing mice by reducing oxidative stress, inflammation and cell apoptosis. Furthermore, high-dose cisplatin-induced acute nephrotoxicity can be effectively alleviated by SMF treatment of as little as one day, which significantly reduced the reactive oxygen species levels in kidneys and prolonged the mice's survival. Moreover, the concentration of cisplatin in the kidney was significantly attenuated in SMF-treated mice. Therefore, our study demonstrates the effects of moderate SMFs as a novel physical method to reduce oxidative stress, and revealed their future potential to be used against cisplatin-induced kidney toxicity in cancer treatment.
Collapse
Affiliation(s)
- Xin Yu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230036, China
| | - Xinmiao Ji
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yixiang Fan
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230036, China
| | - Biao Yu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xinyu Wang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Chuanlin Feng
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230036, China
| | - Lei Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Chao Song
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xin Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230036, China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
- International Magnetobiology Frontier Research Center, Science Island, Hefei 230036, China
| |
Collapse
|
233
|
Pharmacodynamic Modeling to Evaluate the Impact of Cimetidine, an OCT2 Inhibitor, on the Anticancer Effects of Cisplatin. Cells 2022; 12:cells12010057. [PMID: 36611850 PMCID: PMC9818342 DOI: 10.3390/cells12010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Despite potent anticancer activity, the clinical utilization of cisplatin is limited due to nephrotoxicity. As Organic Cation Transporter 2 (OCT2) has been shown to be one of the key transporters involved in the uptake of cisplatin into renal proximal tubules, OCT2 inhibitors such as cimetidine have been explored to suppress cisplatin-induced nephrotoxicity. Nonetheless, the impact of OCT2 inhibition or cimetidine on the anti-cancer effects of cisplatin has not been extensively examined. The main objective of the present study was to quantitatively characterize the anticancer effects of cisplatin and cimetidine and determine their nature of interactions in two cancer cell lines, OCT2-negative hepatocellular carcinoma (HCC) cell line, Huh7, and OCT2-positive breast cancer cell line, MDA-MB-468. First, we determined the static concentration-response curves of cisplatin and cimetidine as single agents. Next, with the help of three-dimensional (3D) response surface analyses and a competitive interaction model, we determined their nature of interactions at static concentrations to be modestly synergistic or additive in Huh7 and antagonistic in MDA-MB-468. These results were consistent with the cell-level pharmacodynamic (PD) modeling analysis which leveraged the time-course effects of drugs as single agents and drug combinations. Our developed PD model can be further used to design future preclinical studies to further investigate the cisplatin and cimetidine combinations in different in vitro and in vivo cancer models.
Collapse
|
234
|
de Godoy Torso N, Visacri MB, Quintanilha JCF, Cursino MA, Pincinato EDC, Moriel P. Assessment of Renal Function in Head and Neck Cancer Patients Treated with Cisplatin: Different Biomarkers and Acute Kidney Injury Classifications. Int J Mol Sci 2022; 24:ijms24010141. [PMID: 36613585 PMCID: PMC9820437 DOI: 10.3390/ijms24010141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Cisplatin is associated with dose-limiting nephrotoxicity, and the timely detection of acute kidney injury (AKI) can affect morbimortality. Therefore, this study aimed to investigate the tools for monitoring renal function in AKI. This was a retrospective, cohort study. Cisplatin-treated patients with head and neck cancer were included. Nephrotoxicity was assessed using serum creatinine, estimated creatinine clearance, serum electrolytic alterations, and plasma kidney injury molecule-1 (KIM-1). The toxicity severity was classified according to Common Terminology Criteria for Adverse Events (CTCAE), and AKI was classified by Risk, Injury, Failure, Loss, and End-stage kidney disease (RIFLE) and Acute Kidney Injury Network (AKIN). A total of 81 participants were included, of whom only 32 did not have AKI. Almost 90% of participants had a decreased estimated glomerular filtration rate five (D5) days after chemotherapy. The AKI estimate differs between AKIN and RIFLE; more participants were diagnosed by the RIFLE at D5, 19.5% versus 2.4% by AKIN, and fifteen had a discordance between these classifications. All laboratory markers showed significant changes on D5. KIM-1 appeared a possible biomarker when considering CTCAE or AKIN classifications (p < 0.05 on D5), but not when RIFLE classification was used (p = 0.0780). Further studies may seek to understand the profiles of different biomarkers together.
Collapse
Affiliation(s)
| | | | | | | | | | - Patricia Moriel
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083-970, Brazil
- Correspondence: ; Tel.: +55-19-35218153
| |
Collapse
|
235
|
Kantauskaite M, Hucke A, Snieder B, Ciarimboli G. Exacerbation of Cisplatin Cellular Toxicity by Regulation of the Human Organic Cation Transporter 2 through Angiotensin II. Int J Mol Sci 2022; 23:ijms232415866. [PMID: 36555515 PMCID: PMC9779897 DOI: 10.3390/ijms232415866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cisplatin (CDDP) is an efficient chemotherapeutic drug, whose use is associated with the development of serious undesired toxicities, such as nephrotoxicity. The human organic cation transporter 2 (hOCT2), which is highly expressed in the basolateral membrane domain of renal proximal tubules seems to play an important role in the development of CDDP nephrotoxicity. The role of angiotensin II (AII) signaling by binding to the AII receptor type 1 (AT1R) in the development and/or progression of CDDP nephrotoxicity is debated. Therefore, in this work, the regulation of hOCT2 activity by AII and its role in the development of CDDP cellular toxicity was investigated. To do this, hOCT2 was overexpressed by viral transduction in Madin-Darby Canine Kidney (MDCK) cells which were cultivated on a filter. This approach allows the separation of an apical and a basolateral membrane domain, which are easily accessible for experimentation. In this system, hOCT2 was mainly localized on the basolateral plasma membrane domain of the cells. The transporter was functional since a specific uptake of the fluorescent organic cation 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+) with an affinity (Km) of 35 µM was only detectable by the addition of ASP+ to the basolateral compartment of hOCT2 expressing MDCK (hOCT2-MDCK) cells. Similarly, CDDP toxicity was evident mainly by CDDP addition to the basolateral compartment of hOCT2-MDCK cells cultivated on a filter. The addition of 1 nM AII stimulated hOCT2 function via PKC activation and worsened CDDP cytotoxicity via binding to AT1R. Therefore, the AII signaling pathway may be implicated in the development and/or progression of CDDP nephrotoxicity. This signaling pathway may be a target for protective interventions for example by blocking AT1R in the kidneys. However, it should be further investigated whether these findings obtained in a cell culture system may have translational relevance for the clinical situation. For toxicity experiments, a 100 µM CDDP concentration was used, which is high but allows us to identify clearly toxic effects due to hOCT2. In summary, down-regulation of hOCT2 activity by the inhibition of the AII signaling pathway may protect against CDDP nephrotoxicity.
Collapse
Affiliation(s)
- Marta Kantauskaite
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Anna Hucke
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Beatrice Snieder
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-83-56981
| |
Collapse
|
236
|
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
|
237
|
Hałka J, Spaleniak S, Kade G, Antosiewicz S, Sigorski D. The Nephrotoxicity of Drugs Used in Causal Oncological Therapies. Curr Oncol 2022; 29:9681-9694. [PMID: 36547174 PMCID: PMC9776938 DOI: 10.3390/curroncol29120760] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
In recent years, a dynamic development of oncology has been observed, resulting from the increasingly frequent occurrence of neoplasms and therefore, increasing population of patients. The most effective form of therapy for cancer patients is complex multidisciplinary specialized disease management, including nephro-oncology care. Different forms of renal function impairment are frequently diagnosed in cancer patients. They are caused by different co-morbidities existing before starting the oncologic treatment as well as the direct undesirable effects of this therapy which may cause temporary or irreversible damage of the urinary system-especially kidneys. According to different therapeutic programs, in such cases the degree of renal damage is often crucial for the possibility of further anti-cancer treatment. Medical personnel responsible for delivering care to oncology patients should be properly educated on current methods of prevention and treatment of renal complications resulting from anti-cancer therapy. The development of oncologic medicines design, including especially immuno-oncological agents, obliges us to learn new patomechanisms determining potential adverse effects, including renal complications. This publication is focused on the most important undesirable nephrotoxic effects of the frequently used anti-cancer drugs.
Collapse
Affiliation(s)
- Janusz Hałka
- Department of Clinical Hematology, Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration’s Hospital, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| | - Sebastian Spaleniak
- Department of Internal Diseases and Nephrodiabetology, Medical University of Lodz, Żeromskiego 113, 90-549 Lodz, Poland
- Correspondence:
| | - Grzegorz Kade
- Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration’s Hospital, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| | - Stefan Antosiewicz
- Military Institute of Aviation Medicine, Center of Aeromedical Examination and Occupational Medicine, Zygmunta Krasińskiego 54/56, 01-755 Warsaw, Poland
| | - Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Aleja Warszawska 30, 11-082 Olsztyn, Poland
- Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of the Ministry of the Interior and Administration’s Hospital, Wojska Polskiego 37, 10-228 Olsztyn, Poland
| |
Collapse
|
238
|
Shin Y, Kim B, Kim W. Cisplatin-Induced Nausea and Vomiting: Effect of Herbal Medicines. PLANTS (BASEL, SWITZERLAND) 2022; 11:3395. [PMID: 36501434 PMCID: PMC9736559 DOI: 10.3390/plants11233395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Cisplatin is a chemotherapeutic agent that is widely used to treat various types of cancers. However, its side effects, most commonly nausea and vomiting, limit its widespread use. Although various drugs, such as ondansetron and aprepitant, are used to alleviate these side effects, their efficacy is still debated. This review aims to summarize the results of 14 studies on the effects of seven single herbal extracts, one multiple herbal extract, and one ginger sub-component (i.e., [6]-gingerol) on cisplatin-induced nausea and vomiting. The results of the included studies were subdivided into four categories: kaolin consumption, retching and vomiting, food intake, and weight loss. Most studies used rodents, whereas four studies used minks or pigeons. The doses of cisplatin used in the studies varied from 3 mg/kg to 7.5 mg/kg, and only a single injection was used. Nine studies analyzed the mechanisms of action of herbal medicines and assessed the involvement of neurotransmitters, cytokines, enzymes, and various hematological parameters. Although further research is needed, this review suggests herbal medicine as a viable treatment option for cisplatin-induced neuropathic pain.
Collapse
Affiliation(s)
- Yuchan Shin
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woojin Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
239
|
Hashimoto T, Shibata K, Hasumi K, Honda K, Nobe K. Effect of SMTP-7 on Cisplatin-Induced Nephrotoxicity in Mice. Biol Pharm Bull 2022; 45:1832-1838. [DOI: 10.1248/bpb.b22-00620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Terumasa Hashimoto
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University
| | | | - Keiji Hasumi
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology
| | - Kazuo Honda
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University
| | - Koji Nobe
- Pharmacological Research Center, Showa University
| |
Collapse
|
240
|
7-hydroxycoumarin modulates Nrf2/HO-1 and microRNA-34a/SIRT1 signaling and prevents cisplatin-induced oxidative stress, inflammation, and kidney injury in rats. Life Sci 2022; 310:121104. [PMID: 36270424 DOI: 10.1016/j.lfs.2022.121104] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
The kidneys are vulnerable to toxicity and acute kidney injury (AKI) is the main adverse effect associated with the clinical use of the chemotherapeutic agent cisplatin (CIS). Oxidative stress and inflammation are implicated in CIS nephrotoxicity. In this study, the effect of the antioxidant 7-hydroxycoumarin (7-HC) against CIS-induced renal intoxication was evaluated. Rats received 7-HC (25, 50, and 100 mg/kg) orally for 14 days and CIS (7 mg/kg) at day 15, and samples were collected 3 days after CIS administration. CIS increased serum urea, creatinine and kidney injury molecule (Kim)-1, caused multiple histopathological changes and increased renal reactive oxygen species (ROS), malondialdehyde (MDA), nitric oxide (NO), NF-κB p65, iNOS, and pro-inflammatory cytokines. 7-HC dose-dependently prevented kidney dysfunction and tissue injury and suppressed ROS and inflammatory mediators. 7-HC boosted renal antioxidants and Bcl-2 while decreased Bax and caspase-3 expression in CIS-administered rats. In addition, 7-HC downregulated Keap-1 and microRNA-34a and upregulated Nrf2, NQO-1, HO-1, and SIRT1. Molecular docking revealed the binding affinity of 7-HC towards NF-κB, Keap-1, and SIRT1. In Conclusion, 7-HC prevented CIS nephrotoxicity by attenuating tissue injury, oxidative stress, inflammation, and apoptotic cell death. The protective efficacy of 7-HC was associated with inhibiting NF-κB and Keap-1, and modulating Nrf2/HO-1 and microRNA34a/Sirt1 signaling.
Collapse
|
241
|
Alotaibi B, El-Masry TA, Elekhnawy E, El-Kadem AH, Saleh A, Negm WA, Abdelkader DH. Aqueous core epigallocatechin gallate PLGA nanocapsules: characterization, antibacterial activity against uropathogens, and in vivo reno-protective effect in cisplatin induced nephrotoxicity. Drug Deliv 2022; 29:1848-1862. [PMID: 35708451 PMCID: PMC9225707 DOI: 10.1080/10717544.2022.2083725] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 12/17/2022] Open
Abstract
Epigallocatechin-3-gallate (EGCG) was isolated from Cycas thouarsii leaves for the first time and encapsulated in aqueous core poly(lactide-co-glycolide) (PLGA) nanocapsules (NCs). This work investigates antimicrobial activity and in vivo reno-protective effects of EGCG-PLGA NCs in cisplatin-induced nephrotoxicity. A double emulsion solvent evaporation process was adopted to prepare PLGA NCs loaded with EGCG. Particle size, polydispersity index (PDI), zeta potential, percent entrapment efficiency (%EE), structural morphology, and in vitro release platform were all studied in vitro. The optimum formula (F2) with particle size (61.37 ± 5.90 nm), PDI (0.125 ± 0.027), zeta potential (-11.83 ± 3.22 mV), %EE (85.79 ± 5.89%w/w), initial burst (36.85 ± 4.79), and percent cumulative release (87.79 ± 9.84) was selected for further in vitro/in vivo studies. F2 exhibited an enhanced antimicrobial activity against uropathogens as it had lower minimum inhibitory concentration (MIC) values and a more significant impact on bacterial growth than free EGCG. Forty male adult mice were randomly allocated into five groups: control vehicle, untreated methotrexate, MTX groups treated with a daily oral dose of free EGCG, placebo PLGA NCs, and EGCG PLGA NCs (F2) for 10 days. Results showed that EGCG PLGA NCs (F2) exerted promising renoprotective effects compared to free EGCG. EGCG PLGA NCs group induced a significant decrease in kidney index, serum creatinine, kidney injury molecule-1 (KIM-1), NGAL serum levels, and pronounced inhibition of NLPR-3/caspase-1/IL/1β inflammasome pathway. It also significantly ameliorated oxidative stress and decreased NFκB, Bax expression levels. Aqueous core PLGA NCs are a promising formulation strategy that provides high polymeric protection and sustained release pattern for hydrophilic therapeutic agents.
Collapse
Affiliation(s)
- Badriyah Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Thanaa A. El-Masry
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Aya H. El-Kadem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt
| | - Walaa A. Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Dalia H. Abdelkader
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
242
|
Davoudi M, Jadidi Y, Moayedi K, Farrokhi V, Afrisham R. Ameliorative impacts of polymeric and metallic nanoparticles on cisplatin-induced nephrotoxicity: a 2011-2022 review. J Nanobiotechnology 2022; 20:504. [PMID: 36457031 PMCID: PMC9714065 DOI: 10.1186/s12951-022-01718-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Cisplatin (CDDP) is a well-known platinum-based drug used in the treatment of various malignancies. However, the widespread side effects that this drug leaves on normal tissues make its use limited. Since cisplatin is mainly eliminated from the kidneys, CDDP-induced nephrotoxicity is the most significant dose-limiting complication attributed to cisplatin, which often leads to dose withdrawal. Considering the high efficiency of cisplatin in chemotherapy, finding renoprotective drug delivery systems for this drug is a necessity. In this regard, we can take advantages of different nanoparticle-based approaches to deliver cisplatin into tumors either using passive targeting or using specific receptors. In an effort to find more effective cisplatin-based nano-drugs with less nephrotoxic effect, the current 2011-2022 review study was conducted to investigate some of the nanotechnology-based methods that have successfully been able to mitigate CDDP-induced nephrotoxicity. Accordingly, although cisplatin can cause renal failures through inducing mitochondria dysfunction, oxidative stress, lipid peroxidation and endoplasmic reticulum stress, some CDDP-based nano-carriers have been able to reverse a wide range of these advert effects. Based on the obtained results, it was found that the use of different metallic and polymeric nanoparticles can help renal cells to strengthen their antioxidant systems and stay alive through reducing CDDP-induced ROS generation, inhibiting apoptosis-related pathways and maintaining the integrity of the mitochondrial membrane. For example, nanocurcumin could inhibit oxidative stress and acting as a ROS scavenger. CONPs could reduce lipid peroxidation and pro-inflammatory cytokines. CDDP-loaded silver nanoparticles (AgNPs) could inhibit mitochondria-mediated apoptosis. In addition, tea polyphenol-functionalized SeNPs (Se@TE) NPs could mitigate the increased level of dephosphorylated AKT, phosphorylated p38 MAPK and phosphorylated c-Jun N-terminal kinase (JNK) induced by cisplatin. Moreover, exosomes mitigated cisplatin-induced renal damage through inhibiting Bcl2 and increasing Bim, Bid, Bax, cleaved caspase-9, and cleaved caspase-3. Hence, nanoparticle-based techniques are promising drug delivery systems for cisplatin so that some of them, such as lipoplatins and nanocurcumins, have even reached phases 1-3 trials.
Collapse
Affiliation(s)
- Maryam Davoudi
- grid.411705.60000 0001 0166 0922Department of Clinical Laboratory Sciences, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Jadidi
- grid.411705.60000 0001 0166 0922Department of Clinical Laboratory Sciences, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiana Moayedi
- grid.411705.60000 0001 0166 0922Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vida Farrokhi
- grid.411705.60000 0001 0166 0922Department of Hematology, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- grid.411705.60000 0001 0166 0922Department of Clinical Laboratory Sciences, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
243
|
FFAR4 improves the senescence of tubular epithelial cells by AMPK/SirT3 signaling in acute kidney injury. Signal Transduct Target Ther 2022; 7:384. [PMID: 36450712 PMCID: PMC9712544 DOI: 10.1038/s41392-022-01254-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 12/02/2022] Open
Abstract
Acute kidney injury (AKI) is a serious clinical complication with high morbidity and mortality rates. Despite substantial progress in understanding the mechanism of AKI, no effective therapy is available for treatment or prevention. We previously found that G protein-coupled receptor (GPCR) family member free fatty acid receptor 4 (FFAR4) agonist TUG891 alleviated kidney dysfunction and tubular injury in AKI mice. However, the versatile role of FFAR4 in kidney has not been well characterized. In the study, the expression of FFAR4 was abnormally decreased in tubular epithelial cells (TECs) of cisplatin, cecal ligation/perforation and ischemia/reperfusion injury-induced AKI mice, respectively. Systemic and conditional TEC-specific knockout of FFAR4 aggravated renal function and pathological damage, whereas FFAR4 activation by TUG-891 alleviated the severity of disease in cisplatin-induced AKI mice. Notably, FFAR4, as a key determinant, was firstly explored to regulate cellular senescence both in injured kidneys of AKI mice and TECs, which was indicated by senescence-associated β-galactosidase (SA-β-gal) activity, marker protein p53, p21, Lamin B1, phospho-histone H2A.X, phospho-Rb expression, and secretory phenotype IL-6 level. Mechanistically, pharmacological activation and overexpression of FFAR4 reversed the decrease of aging-related SirT3 protein, where FFAR4 regulated SirT3 expression to exhibit anti-senescent effect via Gq subunit-mediated CaMKKβ/AMPK signaling in cisplatin-induced mice and TECs. These findings highlight the original role of tubular FFAR4 in cellular senescence via AMPK/SirT3 signaling and identify FFAR4 as a potential drug target against AKI.
Collapse
|
244
|
A Hydrodistillate of Gynostemma pentaphyllum and Damulin B Prevent Cisplatin-Induced Nephrotoxicity In Vitro and In Vivo via Regulation of AMPKα1 Transcription. Nutrients 2022; 14:nu14234997. [PMID: 36501027 PMCID: PMC9737728 DOI: 10.3390/nu14234997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
The clinical application of cisplatin, one of the most effective chemotherapeutic agents used to treat various cancers, has been limited by the risk of adverse effects, notably nephrotoxicity. Despite intensive research for decades, there are no effective approaches for alleviating cisplatin nephrotoxicity. This study aimed to investigate the protective effects and potential mechanisms of a Gynostemma pentaphyllum leaves hydrodistillate (GPHD) and its major component, damulin B, against cisplatin-induced nephrotoxicity in vitro and in vivo. A hydro-distillation method can extract large amounts of components within a short period of time using non-toxic, environmentally friendly solvent. We found that the levels of AMP-activated protein kinase α1 (AMPKα1), reactive oxygen species (ROS), and apoptosis were tightly associated with each other in HEK293 cells treated with cisplatin. We demonstrated that AMPKα1 acted as an anti-oxidant factor and that ROS generated by cisplatin suppressed the expression of AMPKα1 at the transcriptional level, thereby resulting in induction of apoptosis. Treatment with GPHD or damulin B effectively prevented cisplatin-induced apoptosis of HEK293 cells and cisplatin-induced acute kidney injury in mice by suppressing oxidative stress and maintaining AMPKα1 levels. Therefore, our study suggests that GPHD and damulin B may serve as prospective adjuvant agents against cisplatin-induced nephrotoxicity.
Collapse
|
245
|
Ozkul O, Ozkul B, Erdogan MA, Erbas O. Ameliorating Effect of Propofol on Cisplatin-Induced Liver and Kidney Damage in Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.1623.1635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
246
|
Dewaeles E, Carvalho K, Fellah S, Sim J, Boukrout N, Caillierez R, Ramakrishnan H, Van der Hauwaert C, Vijaya Shankara J, Martin N, Massri N, Launay A, Folger JK, de Schutter C, Larrue R, Loison I, Goujon M, Jung M, Le Gras S, Gomez-Murcia V, Faivre E, Lemaire J, Garat A, Beauval N, Maboudou P, Gnemmi V, Gibier JB, Buée L, Abbadie C, Glowacki F, Pottier N, Perrais M, Cunha RA, Annicotte JS, Laumet G, Blum D, Cauffiez C. Istradefylline protects from cisplatin-induced nephrotoxicity and peripheral neuropathy while preserving cisplatin antitumor effects. J Clin Invest 2022; 132:152924. [PMID: 36377661 PMCID: PMC9663157 DOI: 10.1172/jci152924] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is a potent chemotherapeutic drug that is widely used in the treatment of various solid cancers. However, its clinical effectiveness is strongly limited by frequent severe adverse effects, in particular nephrotoxicity and chemotherapy-induced peripheral neuropathy. Thus, there is an urgent medical need to identify novel strategies that limit cisplatin-induced toxicity. In the present study, we show that the FDA-approved adenosine A2A receptor antagonist istradefylline (KW6002) protected from cisplatin-induced nephrotoxicity and neuropathic pain in mice with or without tumors. Moreover, we also demonstrate that the antitumoral properties of cisplatin were not altered by istradefylline in tumor-bearing mice and could even be potentiated. Altogether, our results support the use of istradefylline as a valuable preventive approach for the clinical management of patients undergoing cisplatin treatment.
Collapse
Affiliation(s)
- Edmone Dewaeles
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France
| | - Kévin Carvalho
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Sandy Fellah
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Jaewon Sim
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA.,Cell and Molecular Biology Graduate program, Michigan State University, East Lansing, Michigan, USA
| | - Nihad Boukrout
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Raphaelle Caillierez
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | | | - Cynthia Van der Hauwaert
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Département de la Recherche en Santé, Lille, France
| | - Jhenkruthi Vijaya Shankara
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Nathalie Martin
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Noura Massri
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA.,Cell and Molecular Biology Graduate program, Michigan State University, East Lansing, Michigan, USA
| | - Agathe Launay
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Joseph K. Folger
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Clémentine de Schutter
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Romain Larrue
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service de Toxicologie et Génopathies, Lille, France
| | - Ingrid Loison
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Marine Goujon
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Matthieu Jung
- University of Strasbourg, CNRS UMR 7104, INSERM U1258 – GenomEast Platform – IGBMC – Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Stéphanie Le Gras
- University of Strasbourg, CNRS UMR 7104, INSERM U1258 – GenomEast Platform – IGBMC – Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Victoria Gomez-Murcia
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Faivre
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Julie Lemaire
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Anne Garat
- CHU Lille, Service de Toxicologie et Génopathies, Lille, France.,University of Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483, IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Lille, France
| | - Nicolas Beauval
- CHU Lille, Service de Toxicologie et Génopathies, Lille, France.,University of Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483, IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Lille, France
| | - Patrice Maboudou
- CHU Lille, Service de Biochimie Automatisée, Protéines et Biologie Prédictive, Lille, France
| | - Viviane Gnemmi
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service d’Anatomopathologie, Lille, France
| | - Jean-Baptiste Gibier
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service d’Anatomopathologie, Lille, France
| | - Luc Buée
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Corinne Abbadie
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Francois Glowacki
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service de Néphrologie, Lille, France
| | - Nicolas Pottier
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service de Toxicologie et Génopathies, Lille, France
| | - Michael Perrais
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Rodrigo A. Cunha
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Jean-Sébastien Annicotte
- University of Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, INSERM U1283-UMR8199 – EGID, Lille, France.,University of Lille, INSERM, CHU Lille, Institut Pasteur de Lille, RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - David Blum
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Christelle Cauffiez
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
247
|
Protective Effect of Natural Antioxidants on Reducing Cisplatin-Induced Nephrotoxicity. DISEASE MARKERS 2022; 2022:1612348. [PMID: 36419843 PMCID: PMC9678481 DOI: 10.1155/2022/1612348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 09/24/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022]
Abstract
The clinical application of cisplatin is limited by its adverse events, of which nephrotoxicity is the most commonly observed. In a cisplatin-induced pathological response, oxidative stress is one of the upstream reactions which inflicts different degrees of damages to the intracellular material components. Reactive oxygen species (ROS) are also one of the early signaling molecules that subsequently undergo a series of pathological reactions, such as apoptosis and necrosis. This review summarizes the mechanism of intracellular ROS generation induced by cisplatin, mainly from the consumption of endogenous antioxidants, destruction of antioxidant enzymes, induction of mitochondrial crosstalk between the endoplasmic reticulum by ROS and Ca2+, and destruction of the cytochrome P450 (CYP) system in the endoplasmic reticulum, all of which result in excessive accumulation of intracellular ROS and oxidative stress. In addition, studies demonstrated that natural antioxidants can protect against the cisplatin-induced nephrotoxicity, by reducing or even eliminating excess free radicals and also affecting other nonredox pathways. Therefore, this review on the one hand provides theoretical support for the research and clinical application of natural antioxidants and on the other hand provides a new entry point for the detailed mechanism of cisplatin nephrotoxicity, which may lay a solid foundation for the future clinical use of cisplatin.
Collapse
|
248
|
Sugimoto K, Taguchi S, Kishitani K, Kawai T, Masuda K, Nakamura Y, Kinjo M, Tambo M, Miyakawa J, Akiyama Y, Yamada Y, Sato Y, Yamada D, Nakagawa T, Fukuhara H, Kume H. Comparison of full-dose gemcitabine/cisplatin, dose-reduced gemcitabine/cisplatin, and gemcitabine/carboplatin in real-world patients with advanced urothelial carcinoma. BMC Urol 2022; 22:177. [DOI: 10.1186/s12894-022-01139-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
While gemcitabine/cisplatin (GC) is the gold standard regimen for patients with advanced urothelial carcinoma (aUC), either dose-reduced GC or gemcitabine/carboplatin (GCa) is an alternative option for “cisplatin-unfit” patients. However, few studies have compared outcomes with these commonly used regimens in the real-world setting.
Methods
We retrospectively reviewed patients with aUC who received full-dose GC, dose-reduced GC, or GCa as first-line salvage chemotherapy at two university hospitals between 2016 and 2020. Progression-free survival, cancer-specific survival, and overall survival, as well as best overall response and adverse event profiles, were compared among these three regimens.
Results
Of 105 patients, 41, 27, and 37 patients received full-dose GC, dose-reduced GC, and GCa, respectively. Significant differences were noted in the patients’ baseline age, primary site, and renal function among the three regimens. Sixty-nine (65.7%) patients died during a median follow-up period of 14 months. There was no significant difference among the three regimens for all survival outcomes and best overall response. However, the complete response rate of dose-reduced GC (2/27, 7.4%) appeared inferior to that of full-dose GC (9/41, 22.0%) or GCa (6/37, 16.2%). Regarding adverse event profiles, no significant difference was observed among the three regimens, except for significantly fewer cases with elevated alanine aminotransferase in the GCa group compared with the other groups.
Conclusions
This study compared the oncological and toxicological outcomes of full-dose GC, dose-reduced GC, and GCa in real-world patients with aUC. Unlike in the clinical trial setting, there were almost no significant differences among the three regimens.
Collapse
|
249
|
Tae JH, Ha MS, Chi BH, Chang IH, Kim TH, Myung SC, Nguyen TT, Kim M, Lee KE, Kim Y, Woo HK, Kyoung DS, Kim H, Choi SY. Neoadjuvant versus adjuvant chemotherapy in upper tract urothelial carcinoma: A nationwide cohort study. BMC Urol 2022; 22:175. [PMID: 36352437 PMCID: PMC9644507 DOI: 10.1186/s12894-022-01112-6] [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: 04/21/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
Purpose This study aimed to evaluate the trend of adjuvant chemotherapy (AC) and neoadjuvant chemotherapy (NAC) in patients who underwent radical nephroureterectomy with bladder cuff excision (NUx) for upper tract urothelial carcinoma (UTUC) to compare the perioperative outcomes and overall survival (OS) between AC and NAC using nationwide population-based data. Materials and methods We collected data on patients diagnosed with UTUC and treated with NUx between 2004 and 2016 using the National Health Insurance Service database, and evaluated the overall treatment trends. The AC and NAC groups were propensity score-matched. Cox proportional hazard and Kaplan-Meier analyses were used to assess survival. Results Of the 8,705 enrolled patients, 6,627 underwent NUx only, 94 underwent NAC, and 1,984 underwent AC. The rate of NUx without perioperative chemotherapy increased from 70.8 to 78.2% (R2 = 0.632; p < 0.001). The rates of dialysis (p = 0.398), TUR-BT (p = 1.000), and radiotherapy (p = 0.497) after NUx were similar. In the Kaplan-Meier curve, the NAC and AC groups showed no significant difference (p = 0.480). In multivariate analysis, treatment with AC or NAC was not associated with OS (hazard ratio 0.83, 95% confidence interval 0.49–1.40, p = 0.477). Conclusion The use of NUx without perioperative chemotherapy has tended to increase in South Korea. Dialysis, TUR-BT, and radiotherapy rates after NUx were similar between the NAC and AC groups. There was no significant difference in OS between the NAC and AC groups. Proper perioperative chemotherapy according to patient and tumor conditions should be determined by obtaining more evidence of UTUC.
Collapse
|
250
|
Yu JB, Lee DS, Padanilam BJ, Kim J. Repeated Administration of Cisplatin Transforms Kidney Fibroblasts through G2/M Arrest and Cellular Senescence. Cells 2022; 11:cells11213472. [PMID: 36359868 PMCID: PMC9655665 DOI: 10.3390/cells11213472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Cisplatin is a potent chemotherapeutic used for the treatment of many types of cancer, but it has nephrotoxic side effects leading to acute kidney injury and subsequently chronic kidney disease (CKD). Previous work has focused on acute kidney tubular injury induced by cisplatin, whereas the chronic sequelae post-injury has not been well-explored. In the present study, we established a kidney fibroblast model of CKD induced by repeated administration of cisplatin (RAC) as a clinically relevant model. In NRK-49F rat kidney fibroblasts, RAC upregulated α-smooth muscle actin (α-SMA) and fibronectin proteins, suggesting that RAC induces kidney fibroblast-to-myofibroblast transformation. RAC also enhanced cell size, including the cell attachment surface area, nuclear area, and cell volume. Furthermore, RAC induced p21 expression and senescence-associated β-galactosidase activity, suggesting that kidney fibroblasts exposed to RAC develop a senescent phenotype. Inhibition of p21 reduced cellular senescence, hypertrophy, and myofibroblast transformation induced by RAC. Intriguingly, after RAC, kidney fibroblasts were arrested at the G2/M phase. Repeated treatment with paclitaxel as an inducer of G2/M arrest upregulated p21, α-SMA, and fibronectin in the kidney fibroblasts. Taken together, these data suggest that RAC transforms kidney fibroblasts into myofibroblasts through G2/M arrest and cellular senescence.
Collapse
Affiliation(s)
- Jia-Bin Yu
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea
| | - Dong-Sun Lee
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea
- Jeju Microbiome Research Center, Jeju National University, Jeju 63243, Korea
- Faculty of Biotechnology, College of Applied Life Sciences, SARI, Jeju National University, Jeju 63243, Korea
| | - Babu J. Padanilam
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence: (B.J.P.); (J.K.)
| | - Jinu Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea
- Department of Anatomy, Jeju National University College of Medicine, Jeju 63243, Korea
- Correspondence: (B.J.P.); (J.K.)
| |
Collapse
|