51
|
Wu Y, Xv Y, Zhao L, Zhou Z, Wang M, Xi J, Liming Y, Gao J, Deng B, Zheng D. PSTK exerts protective role in cisplatin-tubular cell injury via BAX/BCL2/Caspase3 pathway. Physiol Rep 2025; 13:e70162. [PMID: 39794890 PMCID: PMC11723822 DOI: 10.14814/phy2.70162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 01/13/2025] Open
Abstract
Cisplatin is a widely used anticancer drug, but its accumulation in renal tubular epithelial cells (TECs) can cause acute kidney injury. Phosphoseryl-tRNA kinase (PSTK) is an intermediate product produced under oxidative stress conditions. This study aimed to elucidate whether PSTK could protect TECs and its possible mechanisms. We found that PSTK levels decreased after cisplatin treatment, but PSTK overexpression using lentivirus vectors protected TEC viability. Overexpression of PSTK increased selenoprotein concentrations and reduced intracellular ROS levels. Additionally, PSTK overexpression inhibited the BAX/BCL2/Caspase 3 pathway after cisplatin stimulation, suggesting its potential role in preventing cell apoptosis. Taken together, this study suggests that PSTK could protect TEC viability from cisplatin-induced injury, possibly by inhibiting mitochondrial apoptosis. The study is significant for developing therapeutic strategies that could manipulate PSTK to delay AKI progression.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Pathology and PathophysiologyMedical College of Soochow UniversitySuzhouJiangsuChina
| | - Yuanyuan Xv
- Department of Thoracic Surgery, School of MedicineShanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Limei Zhao
- Department of Pathology and PathophysiologyMedical College of Soochow UniversitySuzhouJiangsuChina
| | - Ziqi Zhou
- School of Biology & Basic Medical Sciences, Medical College of Soochow UniversitySuzhouJiangsuChina
| | - Miaomiao Wang
- Department of OncologyEastern Hepatobiliary Surgery HospitalShanghaiChina
| | - Jima Xi
- Liangxi Taihu Hospital of Traditional Chinese MedicineWuxiJiangsuChina
| | - Ying Liming
- National Heart and Lung Institute Imperial College LondonLondonUK
| | - Jianling Gao
- Department of Intensive CareSuzhou 4th People's Hospital, Soochow UniversitySuzhouChina
| | - Bingqing Deng
- The Department of NephrologySuzhou 4th People's Hospital, Soochow UniversitySuzhouChina
| | - Dong Zheng
- Department of Pathology and PathophysiologyMedical College of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
52
|
Rose O, Croonenberg T, Clemens S, Hinteregger T, Eppacher S, Huber-Cantonati P, Garcia-Miralles M, Liuni R, Dossena S. Cisplatin-Induced Hearing Loss, Oxidative Stress, and Antioxidants as a Therapeutic Strategy-A State-of-the-Art Review. Antioxidants (Basel) 2024; 13:1578. [PMID: 39765905 PMCID: PMC11673797 DOI: 10.3390/antiox13121578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025] Open
Abstract
Cisplatin is an established component of treatment protocols for various solid malignancies but carries a significant potential for serious adverse effects. Ototoxicity from cisplatin treatment is an important dose-limiting toxicity that manifests as bilateral, progressive, irreversible, dose-dependent sensorineural hearing loss, ear pain, tinnitus, and vestibular dysfunction. Despite the recent approval of sodium thiosulphate for the prevention of cisplatin-induced hearing loss (CIHL) in pediatric patients, structured prevention programs are not routinely implemented in most hospitals, and reducing platinum-induced ototoxicity in adults remains an important clinical problem without established treatment options. Cochlear oxidative stress plays a fundamental role in CIHL. Here, we review the molecular mechanisms leading to oxidative stress in CIHL and the clinical and preclinical studies testing antioxidants in CIHL to guide future clinical trials in assessing the efficacy and safety of candidate antioxidant compounds in this clinical setting.
Collapse
Affiliation(s)
- Olaf Rose
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
- Center of Public Health and Health Services Research, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Tim Croonenberg
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
| | - Stephanie Clemens
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
- Center of Public Health and Health Services Research, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Tobias Hinteregger
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
| | - Stefanie Eppacher
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
| | - Petra Huber-Cantonati
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
| | - Marta Garcia-Miralles
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University, 5020 Salzburg, Austria (S.C.)
| | - Raffaella Liuni
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria
- Research and Innovation Center Regenerative Medicine & Novel Therapies (FIZ RM&NT), Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
53
|
Zhi H, Fu H, Zhang Y, Fan N, Zhao C, Li Y, Sun Y, Li Y. Progress of cGAS-STING signaling pathway-based modulation of immune response by traditional Chinese medicine in clinical diseases. Front Immunol 2024; 15:1510628. [PMID: 39737190 PMCID: PMC11683013 DOI: 10.3389/fimmu.2024.1510628] [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: 10/13/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
The cGAS-STING signaling pathway is a critical component of the innate immune response, playing a significant role in various diseases. As a central element of this pathway, STING responds to both endogenous and exogenous DNA stimuli, triggering the production of interferons and pro-inflammatory cytokines to enhance immune defenses against tumors and pathogens. However, dysregulated activation of the STING pathway is implicated in the pathogenesis of multiple diseases, including autoinflammation, viral infections, and cancer. Traditional Chinese Medicines (TCMs), which have a long history of use, have been associated with positive effects in disease prevention and treatment. TCM formulations (e.g., Lingguizhugan Decoction, Yi-Shen-Xie-Zhuo formula) and active compounds (e.g., Glabridin, Ginsenoside Rd) can modulate the cGAS-STING signaling pathway, thereby influencing the progression of inflammatory, infectious, or oncological diseases. This review explores the mechanisms by which TCMs interact with the cGAS-STING pathway to regulate immunity, focusing on their roles in infectious diseases, malignancies, and autoimmune disorders.
Collapse
Affiliation(s)
- Hui Zhi
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Fu
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunxin Zhang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ni Fan
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chengcheng Zhao
- Experimental Teaching and Practical Training Center, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yujiao Sun
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
54
|
Airik M, Clayton K, Wipf P, Airik R. JP4-039 Mitigates Cisplatin-Induced Acute Kidney Injury by Inhibiting Oxidative Stress and Blocking Apoptosis and Ferroptosis in Mice. Antioxidants (Basel) 2024; 13:1534. [PMID: 39765862 PMCID: PMC11727076 DOI: 10.3390/antiox13121534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/15/2025] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent in the treatment of a wide array of cancers. Due to its active transport into the kidney proximal tubule cells, cisplatin treatment can cause a buildup of this nephrotoxic compound in the kidney, resulting in acute kidney injury (AKI). About 30% of patients receiving cisplatin chemotherapy develop cisplatin-induced AKI. JP4-039 is a mitochondria-targeted reactive oxygen species (ROS) and electron scavenger. Recent studies have shown that JP4-039 mitigates a variety of genotoxic insults in preclinical studies in rodents by suppressing oxidative stress-mediated tissue damage and blocking apoptosis and ferroptosis. However, the benefits of JP4-039 treatment have not been tested in the setting of AKI. In this study, we investigated the potential renoprotective effect of JP4-039 on cisplatin-induced AKI. To address this goal, we treated mice with JP4-039 before or after cisplatin administration and analyzed them for functional and molecular changes in the kidney. JP4-039 co-administration attenuated cisplatin-induced renal dysfunction and histopathological changes. Upregulation of tubular injury markers was also suppressed by JP4-039. Mechanistically, JP4-039 suppressed lipid peroxidation, prevented tissue oxidative stress, and preserved the glutathione levels in cisplatin-injected mice. An increase in cisplatin-induced apoptosis and ferroptosis was also alleviated by the compound. Moreover, JP4-039 inhibited cytokine overproduction in cisplatin-injected mice. Together, our findings demonstrate that JP4-039 is a promising therapeutic agent against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Merlin Airik
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kacian Clayton
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Rannar Airik
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
55
|
Oh CJ, Choi W, Lee HY, Lee IK, Kim MJ, Jeon JH. Sodium Phenylbutyrate Attenuates Cisplatin-Induced Acute Kidney Injury Through Inhibition of Pyruvate Dehydrogenase Kinase 4. Biomedicines 2024; 12:2815. [PMID: 39767721 PMCID: PMC11672979 DOI: 10.3390/biomedicines12122815] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Cisplatin nephrotoxicity is a significant clinical issue, and currently, no approved drug exists to prevent cisplatin-induced acute kidney injury (AKI). This study investigated whether sodium phenylbutyrate (4-PBA), a chemical chaperone, can prevent cisplatin-induced AKI. Methods: Six consecutive days of intraperitoneal injections of 4-PBA were administered in a murine model before and after the cisplatin challenge. This study evaluated tubular injury, serum blood urea nitrogen (BUN) and creatinine levels, and inflammatory markers such as tumor necrosis factor-alpha (TNF-α) and intercellular adhesion molecule 1 (ICAM-1). Additionally, apoptosis, mitochondrial membrane potential, oxygen consumption ratio, and reactive oxygen species (ROS) were assessed in renal tubular cells. The expression levels of pyruvate dehydrogenase kinase 4 (Pdk4) were also analyzed. Results: 4-PBA prevented tubular injury and normalized serum BUN and creatinine levels. Inflammatory markers TNF-α and ICAM-1 were suppressed. In renal tubular cells, 4-PBA reduced apoptosis, restored mitochondrial membrane potential and oxygen consumption ratio, and reduced ROS production. Mechanistically, 4-PBA suppressed the expression of Pdk4, which is known to be induced during cisplatin-induced renal injury. The protective effect of 4-PBA was abolished in Pdk4-overexpressing renal tubular cells, indicating that the efficacy of 4-PBA partially depends on the suppression of Pdk4 expression. In cancer cells, 4-PBA did not interfere with the anti-cancer efficacy of cisplatin. Conclusions: These findings suggest that 4-PBA effectively prevents cisplatin-induced acute kidney injury by suppressing Pdk4.
Collapse
Affiliation(s)
- Chang Joo Oh
- Research Institute of Aging and Metabolism, School of Medicine, Kyungpook National University, Daegu 41404, Republic of Korea
| | - Wooyoung Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ha Young Lee
- Research Institute of Aging and Metabolism, School of Medicine, Kyungpook National University, Daegu 41404, Republic of Korea
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, School of Medicine, Kyungpook National University, Daegu 41404, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| | - Jae-Han Jeon
- Research Institute of Aging and Metabolism, School of Medicine, Kyungpook National University, Daegu 41404, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| |
Collapse
|
56
|
Predarska I, Kaluđerović GN, Hey-Hawkins E. Nanostructured mesoporous silica carriers for platinum-based conjugates with anti-inflammatory agents. BIOMATERIALS ADVANCES 2024; 165:213998. [PMID: 39236581 DOI: 10.1016/j.bioadv.2024.213998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 08/09/2024] [Accepted: 08/17/2024] [Indexed: 09/07/2024]
Abstract
This review discusses the relationship between inflammation and cancer initiation and progression, which has prompted research into anti-inflammatory approaches for cancer prevention and treatment. Specifically, it focuses on the use of inflammation-reducing agents to enhance the effectiveness of tumor treatment methods. These agents are combined with platinum(II)-based antitumor drugs to create multifunctional platinum(IV) prodrugs, allowing for simultaneous delivery to tumor cells in a specific ratio. Once inside the cells and subjected to intracellular reduction, both components can act in parallel through distinct pathways. Motivated by the objective of reducing the systemic toxicity associated with contemporary chemotherapy, and with the aim of leveraging the passive enhanced permeability and retention effect exhibited by nanostructured materials to improve their accumulation within tumor tissues, the platinum(IV) complexes have been efficiently loaded into mesoporous silica SBA-15 material. The resulting nanostructured materials are capable of providing controlled release of the conjugates when subjected to simulated plasma conditions. This feature suggests the potential for extended circulation within the body in vivo, with minimal premature release of the drug before reaching the intended target site. The primary emphasis of this review is on research that integrates these two approaches to develop chemotherapeutic treatments that are both more efficient and less harmful.
Collapse
Affiliation(s)
- Ivana Predarska
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany; Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Goran N Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany.
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany.
| |
Collapse
|
57
|
Xu N, Mu R, Deng S, Han Y, Shi Y, Fu X, Li H, Yao Q. Reserpine alleviates cisplatin-induced acute kidney injury via anti-ferroptosis and cGAS/STING pathway. Ren Fail 2024; 46:2406395. [PMID: 39377110 PMCID: PMC11463010 DOI: 10.1080/0886022x.2024.2406395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Cisplatin plays a pivotal role in the chemotherapy treatment of various cancers, but its use is often limited due to its nephrotoxic side effects. Identifying compounds that can mitigate cisplatin-induced nephrotoxicity is therefore of great importance. This study focused on evaluating the protective effects of reserpine against cisplatin-induced acute kidney injury. Reserpine was found to significantly safeguard against kidney damage caused by cisplatin, as indicated by the decreased levels of serum creatinine, blood urea nitrogen, and lactate dehydrogenase induced by cisplatin. Moreover, reserpine improved kidney histology damage caused by cisplatin treatment, with hematoxylin-eosin and periodic acid-Schiff staining revealing notable recovery from renal injury. Mechanistically, reserpine mitigated oxidative stress triggered by cisplatin and exhibits the ability to inhibit ferroptosis both in vivo and in vitro. Additionally, reserpine blocked the activation of the cGAS/STING signaling pathway and the subsequent expression of inflammatory genes, thus reducing inflammation-driven kidney damage. In summary, the findings suggest that reserpine offers a promising new strategy for preventing nephrotoxicity induced by cisplatin.
Collapse
Affiliation(s)
- Nahua Xu
- Zunyi Medical University, Zunyi, China
| | - Rong Mu
- Zunyi Medical University, Zunyi, China
| | - Siyuan Deng
- School of Medicine, Chongqing University, Chongqing, China
| | - Ye Han
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children’s Hospital of Chongqing Medical University), Chongqing, China
| | - Yanyun Shi
- Medical College of Guizhou University, Guiyang, China
| | - Xuemei Fu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children’s Hospital of Chongqing Medical University), Chongqing, China
| | - Hui Li
- Zunyi Medical University, Zunyi, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children’s Hospital of Chongqing Medical University), Chongqing, China
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qi Yao
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
58
|
Jen CW, Chan HC, Chiang CJ, Lee WC, Lu TP, Cheng SHC. Evaluating the Efficacy of Different Treatment Intensities in Nasopharyngeal Carcinoma Patients: A Nationwide Cancer Registry-Based Study. Ann Surg Oncol 2024; 31:9125-9133. [PMID: 39240395 DOI: 10.1245/s10434-024-16145-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE The aim of this study was to evaluate the efficacy of different treatment intensities (TIs) in patients with nasopharyngeal carcinoma (NPC). METHODS The study assessed newly diagnosed, non-metastatic NPC patients from the Taiwan Cancer Registry between 2010 and 2017. TIs were divided into four groups: TI1 [radiotherapy (RT) alone or induction chemotherapy (IC) followed by RT); TI2 (concurrent chemoradiotherapy (CRT) alone); TI3 (IC followed by CRT or CRT followed by adjuvant chemotherapy (AC)]; and TI4 (IC followed by CRT followed by AC). The primary outcome was cancer-specific survival (CSS). RESULTS The study included 9863 patients. For stage I-II NPC patients, there was no significant difference in CSS among the different TI groups. For stage III patients, those receiving TI3 had better CSS (hazard ratio [HR] 0.69) compared with those receiving TI1. No significant differences in CSS were noted among those receiving TI2, TI3, and TI4. For stage IVA-B patients, those receiving TI2 (HR 0.70), TI3 (HR 0.49), and TI4 (HR 0.43) had better CSS compared with those receiving TI1. Compared with stage IVA-B patients receiving TI2, those receiving TI3 (HR 0.70) and TI4 (HR 0.61) had significantly better CSS. No differences in CSS were noted between those receiving TI3 and TI4. CONCLUSIONS For stage I-II NPC patients, RT alone is appropriate. For stage III and IVA-B patients, IC + CRT or CRT + AC may be needed to achieve optimal outcomes. No advantage of IC + CRT + AC over IC + CRT or CRT + AC was observed.
Collapse
Affiliation(s)
- Chung-Wen Jen
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei City, Taiwan
- Department of Radiation Oncology, Taitung Christian Hospital, Taitung City, Taitung County, Taiwan
| | - Han-Ching Chan
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei City, Taiwan
| | - Chun-Ju Chiang
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei City, Taiwan
- Taiwan Cancer Registry, Taipei City, Taiwan
| | - Wen-Chung Lee
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei City, Taiwan
- Taiwan Cancer Registry, Taipei City, Taiwan
- Institute of Health Data Analytics and Statistics, National Taiwan University, Taipei City, Taiwan
| | - Tzu-Pin Lu
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei City, Taiwan.
- Institute of Health Data Analytics and Statistics, National Taiwan University, Taipei City, Taiwan.
| | - Skye Hung-Chun Cheng
- Department of Radiation Oncology, Taitung Christian Hospital, Taitung City, Taitung County, Taiwan.
- Department of Radiation Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei City, Taiwan.
| |
Collapse
|
59
|
Nam D, Park J, Lee J, Son J, Kim JE. mTOR potentiates senescent phenotypes and primary cilia formation after cisplatin-induced G2 arrest in retinal pigment epithelial cells. Cell Signal 2024; 124:111402. [PMID: 39251051 DOI: 10.1016/j.cellsig.2024.111402] [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: 02/27/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cisplatin, a platinum-based anticancer drug, is used to treat several types of cancer. Despite its effectiveness, cisplatin-induced side effects have often been reported. Although cisplatin-induced toxicities, such as apoptosis and/or necrosis, have been well studied, the fate of cells after exposure to sublethal doses of cisplatin needs further elucidation. Treatment with a sublethal dose of cisplatin induced cell cycle arrest at the G2 phase in retinal pigment epithelial cells. Following cisplatin withdrawal, the cells irreversibly exited the cell cycle and became senescent. Notably, the progression from the G2 to the G1 phase occurred without mitotic entry, a phenomenon referred to as mitotic bypass, resulting in the accumulation of cells containing 4N DNA content. Cisplatin-exposed cells exhibited morphological changes associated with senescence, including an enlarged size of cell and nucleus and increased granularity. In addition, the senescent cells possessed primary cilia and persistent DNA lesions. Senescence induced by transient exposure to cisplatin involves mTOR activation. Although transient co-exposure with an mTORC1 inhibitor rapamycin did not prevent mitotic bypass and entry into senescence, it delayed the progression of senescence and attenuated senescent phenotypes, resulting in shorter primary cilia formation. Conclusively, cisplatin induces senescence in retinal pigment epithelial cells by promoting mTOR activation.
Collapse
Affiliation(s)
- Dajeong Nam
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaejung Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaehong Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juyoung Son
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
60
|
Huang Y, Li Y, Xu X, Teng J, Ding X, Xu J. Protective effect of reduced glutathione on acute kidney injury in lung cancer patients treated with cisplatin: a retrospective cohort study. Ren Fail 2024; 46:2411359. [PMID: 39392127 PMCID: PMC11486057 DOI: 10.1080/0886022x.2024.2411359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/04/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Cisplatin is a common cause of acute kidney injury (AKI) during chemotherapy for lung cancer, and the nephrotoxicity limits its clinical use. Reduced glutathione (GSH) is a major component of the cellular antioxidant defense system and performs important physiological functions. The aim of this study was to analyze the protective effect of GSH on AKI in lung cancer patients treated with cisplatin. METHODS The clinical data were retrospectively collected from lung cancer patients treated with cisplatin at our hospital between 1 January and 31 December 2019. The patients were divided into AKI group and non-AKI group based on whether AKI occurred, and into GSH group and non-GSH group based on whether GSH was used. Univariate and multivariate logistic regressions were used to analyze the risk factors for AKI. RESULTS A total of 1372 lung cancer patients treated with cisplatin were enrolled. Of these patients, 231 patients (16.8%) developed AKI. The incidence of AKI was lower in the GSH group compared with the non-GSH group (10.6% vs. 18%, p = 0.009). Multivariate logistic regression analysis indicated that older age (OR = 1.045, 95% CI 1.025-1.065, p < 0.001), anemia (OR = 2.436, 95% CI 1.800-3.298, p < 0.001), higher SUA levels (OR = 1.002, 95% CI 1.000-1.004, p = 0.012), higher total amount of cisplatin per cycle (OR = 1.015, 95% CI 1.004-1.025, p = 0.005), and combination with paclitaxel (OR = 2.099, 95% CI 1.435-3.070, p < 0.001) were independent risk factors for AKI in lung cancer patients treated with cisplatin, whereas GSH (OR = 0.573, 95% CI 0.353-0.931, p = 0.025) and mannitol (OR = 0.229, 95% CI 0.055-0.963, p = 0.044) reduced the risk of AKI. CONCLUSION GSH was an independent protective factor against AKI in lung cancer patients treated with cisplatin and could be considered for clinical use in these patients to better protect renal function.
Collapse
Affiliation(s)
- Ying Huang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Nephrology Clinical Quality Control Center of Xiamen, China
| | - Yang Li
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Nephrology Clinical Quality Control Center of Xiamen, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Nephrology Clinical Quality Control Center of Xiamen, China
| | - Jiarui Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
61
|
Zhu J, Yuan A, Le Y, Chen X, Guo J, Liu J, Chen H, Wang CY, Lu D, Lu K. Yi-Qi-Jian-Pi-Xiao-Yu formula inhibits cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated ferritinophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156189. [PMID: 39515100 DOI: 10.1016/j.phymed.2024.156189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/28/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The kidneys are the primary excretory organs for platinum drugs, making them susceptible to damage from these drugs. Cisplatin-induced acute kidney injury (CIAKI) is the most common side effect observed in patients undergoing clinical cisplatin treatment. A traditional Chinese medicinal preparation, the Yi-Qi-Jian-Pi-Xiao-Yu formula (YQJPXY), which is a modified formulation of the classical Chinese medicine formula Buyang Huanwu Decoction, has long been used in the treatment of clinical kidney diseases. It is expected to be used to ameliorate cisplatin-induced acute kidney injury. However, the mechanism of this YQJPXY for the treatment of cisplatin-induced acute kidney injury remains unclear. PURPOSE The objective of this study is to examine the impact of the YQJPXY on the inhibition of ferroptosis in cisplatin-induced acute kidney injury and to elucidate the underlying mechanisms. METHODS The active components of YQJPXY were analysed using UPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of YQJPXY on CIAKI and ferroptosis in mice subjected to acute cisplatin treatment and in mice receiving cisplatin treatment after STING expression was inhibited using the STING inhibitor C176. The renoprotective effect of YQJPXY on cisplatin-treated mice was evaluated by measuring tissue damage, inflammation and pro-fibrosis. In addition, we employed network pharmacology and molecular docking methodologies to analyse the principal regulatory targets of YQJPXY. Furthermore, the expression of key proteins and markers of ferroptosis and iron metabolism, as well as the levels of key indicators related to STING-associated ferritinophagy, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes. RESULTS The results demonstrated that YQJPXY reduced the levels of indicators of injury, inflammation and pro-fibrosis in CIAKI mice, with renoprotective effects. Network pharmacological analyses revealed that ferroptosis might be the main biological process regulated by YQJPXY. Furthermore, molecular docking results indicated that STING might be a potential regulatory target of YQJPXY. Furthermore, YQJPXY treatment resulted in a significant reduction in MDA and 4-HNE levels, as well as the inhibition of ferroptosis and improvement in iron metabolic processes. Concomitantly, YQJPXY exhibited a robust protective effect on ferroptosis and iron metabolism homeostasis, as evidenced by its inhibitory action on ferritinophagy. Validation experiments utilising the cisplatin inhibitor C176 demonstrated that YQJPXY inhibits cisplatin-induced ferroptosis in kidney via STING-mediated ferritinophagy. CONCLUSION These suggest that YQJPXY alleviates cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated Ferritinophagy.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Aini Yuan
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yifei Le
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaohui Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jianan Guo
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jing Liu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hang Chen
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Cai-Yi Wang
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Dezhao Lu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China.
| |
Collapse
|
62
|
Guo S, Zhao J, Zhang Y, Qin Y, Yuan J, Yu Z, Xing Y, Zhang Y, Hui Y, Wang A, Han M, Zhao Y, Ning X, Sun S. Histone deacetylases: potential therapeutic targets in cisplatin-induced acute kidney injury. Ann Med 2024; 56:2418958. [PMID: 39450927 PMCID: PMC11514411 DOI: 10.1080/07853890.2024.2418958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/11/2024] [Accepted: 07/20/2024] [Indexed: 10/26/2024] Open
Abstract
Aim: Chemotherapy has been well shown to enhance life expectancy in patients with malignancy. However, conventional chemotherapy drugs, particularly cisplatin, are highly associated with nephrotoxicity, which limits therapeutic efficacy and impairs quality of life. Histone deacetylases (HDACs) are proteases that play significant roles in diseases by influencing protein post-translational modification and gene expression. Agents that inhibit HDAC enzymes have been developed and approved by the FDA as anticancer drugs. It is worth noting that in certain preclinical studies with tumour cell lines, the integration of HDAC modulators and cisplatin not only exerts synergistic or additive tumour-killing effects but also alleviates cisplatin nephrotoxicity. The aim of this review is to discuss the role of HDACs in cisplatin nephrotoxicity. Methods: After searching in PubMed and Web of Science databases using 'Histone deacetylase', 'nephrotoxicity', 'cisplatin', and 'onconpehrology' as keywords, studies related was compiled and examined. Results: HDAC inhibitors exert renal protective effects by inhibiting inflammation, apoptosis, oxidative stress, and promoting autophagy; whereas sirtuins play a renal protective role by regulating lipid metabolism, inhibiting inflammation and apoptosis, and protecting mitochondrial biosynthesis and mitochondrial dynamics. These potential interactions provide clues concerning targets for molecular treatment. Conclusion: This review encapsulates the function and molecular mechanisms of HDACs in cisplatin nephrotoxicity, providing the current view by which HDACs induce different biological signaling in the regulation of chemotherapy-associated renal injury. More importantly, this review exhaustively elucidates that HDACs could be targeted to develop a new therapeutic strategy in treating cisplatin nephrotoxicity, which will extend the knowledge of the biological impact and clinical implications of HDACs.
Collapse
Affiliation(s)
- Shuxian Guo
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Jin Zhao
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yuzhan Zhang
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yunlong Qin
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Jinguo Yuan
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Zixian Yu
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yan Xing
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yumeng Zhang
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yueqing Hui
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Anjing Wang
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Mei Han
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yueru Zhao
- School of Clinical Medicine, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Xiaoxuan Ning
- Department of Geriatric, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
63
|
Barakat N, Ismail E, Zahran F. The integrated effect of roflumilast and selenium nanoparticles on nephrotoxicity generated by cisplatin through the regulation of the antioxidant and apoptotic pathways. J Trace Elem Med Biol 2024; 86:127555. [PMID: 39442470 DOI: 10.1016/j.jtemb.2024.127555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
AIM The current investigations aimed to investigate the potential synergistic effect of Roflumilast (ROF) and Selenium nanoparticles (SeNPs) administration on Cisplatin (Cis) -induced nephrotoxicity. MATERIALS AND METHODS Fifty male rats were divided into five groups; Control group: animals were administered 0.9 % saline solution. Cis group: animals were injected with a single dose of 6 mg/kg. ROF group: Rats received a dosage of 1.2 mg/kg orally daily for 11 days. SeNPs group: animals orally received 0.5 mg/kg of ROF daily for 11 days. The ROF + SeNPs group was administered both after receiving a Cis injection for 11 days. Animals were sacrificed at 5 and 11 days, and the urine and blood samples were collected on day 5 and day 11 for chemical analysis, while kidney samples were obtained for molecular, histological, and immunohistochemical studies. RESULTS The levels of serum creatinine, Blood urea nitrogen (BUN), and total protein were elevated in the Cis group compared to the control group (p < 0.05). While the combination of ROF and SeNPs dramatically decreased these values after 5 and 11 days (p < 0.05). In addition, Cis caused renal oxidative stress by elevating MDA levels and suppressing the activities of SOD, GSH, and CAT. Similarly, these effects were modulated by ROF and SeNPs after 11 days (p < 0.05). Furthermore, the concurrent administration of ROF and SeNPs resulted in a significant increase in the expression of HO-1, Nrf2, and Bcl2, while decreasing the expression of BAX and IL-6 compared to the Cis group after 11 days (P < 0.05). CONCLUSION The study showed that both ROF and SeNPs had significant therapeutic potential in reducing the pathological alterations caused by Cis.
Collapse
Affiliation(s)
- Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - Ehab Ismail
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Faten Zahran
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
64
|
Sahara Y, Fukui C, Kuniyoshi Y, Takasato M. Proximal tubule cell maturation rate and function are controlled by PPARα signaling in kidney organoids. Commun Biol 2024; 7:1532. [PMID: 39604738 PMCID: PMC11603349 DOI: 10.1038/s42003-024-07069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/14/2024] [Indexed: 11/29/2024] Open
Abstract
Human pluripotent stem cell-derived kidney organoids are expected to be a useful tool for new drug discoveries, however, the immaturation of kidney organoids causes difficulties in recapitulating renal pharmacokinetics using organoids. Here, we performed time-course single-cell RNA sequencing of kidney organoids and revealed cell heterogeneity in the maturation rate of the proximal tubule. An unbiased analysis to identify upstream targets of genes that are expressed differentially between cells with low and high maturation rates revealed a higher activation of PPARα signaling in rapidly maturing cells. Treatment with a combination of a PPARα agonist and an RXRα agonist induced genes related to proximal tubule maturation and increased the capacity for protein uptake as well as the sensitivity to nephrotoxicity by cisplatin. This method to promote the maturation rate of proximal tubule cells has the potential to be utilized in microphysiological systems to recapitulate proximal tubule functions and to screen nephrotoxic drugs.
Collapse
Affiliation(s)
- Yoshiki Sahara
- RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
- Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Drug Modality Development, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co. Ltd., Minoh, 562-0029, Japan
| | - Chie Fukui
- RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Yuki Kuniyoshi
- Office of Bioinformatics, Department of Drug Discovery Strategy, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co. Ltd., Minoh, 562-0029, Japan
| | - Minoru Takasato
- RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan.
- Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
- Department of Development and Regeneration, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan.
| |
Collapse
|
65
|
Ighofose E, Garrett SH, Al-Marsoummi S, Mehus AA, Sens DA, Singhal SK, Singhal S, Somji S. Effect of Long-Term Cisplatin Exposure on the Proliferative Potential of Immortalized Renal Progenitor Cells. Int J Mol Sci 2024; 25:12553. [PMID: 39684265 DOI: 10.3390/ijms252312553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Cisplatin (CisPt) is a widely used chemotherapeutic agent. However, its nephrotoxic effects pose significant risks, particularly for the development of acute kidney injury (AKI) and potential progression to chronic kidney disease (CKD). The present study investigates the impact of non-lethal exposure of CisPt to immortalized human renal epithelial precursor TERT cells (HRTPT cells) that co-express PROM1 and CD24, markers characteristic of renal progenitor cells. Over eight serial passages, HRTPT cells were exposed to 1.5 µM CisPt, leading to an initial growth arrest, followed by a gradual recovery of proliferative capacity. Despite maintaining intracellular platinum (Pt) levels, the cells exhibited normal morphology by passage eight (P8), with elevated expression of renal stress and damage markers. However, the ability to form domes was not restored. RNA-seq analysis revealed 516 differentially expressed genes between CisPt-exposed and control cells, with significant correlations to cell cycle and adaptive processes, as determined by the Reactome, DAVID, and Panther analysis programs. The progenitor cells treated with CisPt displayed no identity, or close identity, with cells of the normal human nephron. Additionally, several upregulated genes in P8 cells were linked to cancer cell lines, suggesting a complex interaction between CisPt exposure and cellular repair mechanisms. In conclusion, our study demonstrates that renal progenitor cells can recover from CisPt exposure and regain proliferative potential in the continued presence of both extracellular CisPt and intracellular Pt.
Collapse
Affiliation(s)
- Eloho Ighofose
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Scott H Garrett
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Sarmad Al-Marsoummi
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Aaron A Mehus
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Donald A Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Sandeep K Singhal
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Sonalika Singhal
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| | - Seema Somji
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Stop 9037, Grand Forks, ND 58203, USA
| |
Collapse
|
66
|
Song B, Wu X, Zeng Y. Methyltransferase-like 3 represents a prospective target for the diagnosis and treatment of kidney diseases. Hum Genomics 2024; 18:125. [PMID: 39538346 PMCID: PMC11562609 DOI: 10.1186/s40246-024-00692-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Kidney disease is marked by complex pathological mechanisms and significant therapeutic hurdles, resulting in high morbidity and mortality rates globally. A deeper understanding of the fundamental processes involved can aid in identifying novel therapeutic targets and improving treatment efficacy. Current comprehensive data analyses indicate the involvement of methyltransferase-like 3 (METTL3) and its role in RNA N6-methyladenosine methylation in various renal pathologies, including acute kidney injury, renal fibrosis, and chronic kidney disease. However, there is a paucity of thorough reviews that clarify the functional mechanisms of METTL3 and evaluate its importance in enhancing therapeutic outcomes. This review seeks to systematically examine the roles, mechanisms, and potential clinical applications of METTL3 in renal diseases. The findings presented suggest that METTL3 is implicated in the etiology and exacerbation of kidney disorders, affecting their onset, progression, malignancy, and responsiveness to chemotherapeutic agents through the regulation of specific genetic pathways. In conclusion, this review underscores a detrimental correlation between METTL3 and kidney diseases, highlighting the therapeutic promise of targeting METTL3. Additionally, it offers critical insights for researchers concerning the diagnosis, prognosis, and treatment strategies for renal conditions.
Collapse
Affiliation(s)
- Bin Song
- Department of Nephrology, People's Hospital of Deyang City, Deyang, 618000, China
| | - Xiaolong Wu
- Department of Nephrology, People's Hospital of Deyang City, Deyang, 618000, China
| | - Yan Zeng
- Department of Pediatrics, People's Hospital of Deyang City, No. 173, Section 1, Taishan North Road, Deyang, Sichuan Province, 618000, China.
| |
Collapse
|
67
|
Milutinović M, Ćujić Nikolić N, Cvetković T, Šavikin K, Djordjević I, Janković Veličković L, Randjelović M, Miladinović B, Branković S, Kitić D. Chokeberry Products and By-Products as the Potential Pharmaceuticals for Kidney Protection-An Experimental Study in Rats. PLANTS (BASEL, SWITZERLAND) 2024; 13:3136. [PMID: 39599345 PMCID: PMC11597244 DOI: 10.3390/plants13223136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/27/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
The study aimed to investigate the protective effects of chokeberry fruit products and by-products against cisplatin-induced acute nephrotoxicity in rats. Potential mechanisms involving oxidative stress and inflammatory responses were examined through biochemical and histopathological analyses of kidney tissue. Chokeberry waste, along with the whole fruit extract and juice, was evaluated as a potential raw material for pharmaceutical use. The chemical composition of chokeberry juice and extracts was analyzed using spectrophotometry and HPLC. Rats were treated with chokeberry preparations via intragastric tube for ten days, with a single intraperitoneal dose of cisplatin (8 mg/kg BW) administered on the third day. Post-sacrifice, plasma samples were analyzed for biochemical nephrotoxicity markers, oxidative stress, and inflammatory markers. Kidneys were removed for histopathological and biochemical analysis. Cisplatin-induced acute nephrotoxicity was confirmed by elevated plasma creatinine and blood urea nitrogen levels. Additionally, lipid peroxidation was significantly elevated, while reduced glutathione and catalase activity were significantly reduced. Pro-inflammatory mediators IL-1β, TNF-α, and IL-6 levels were significantly increased in the cisplatin group. Treatment with chokeberry extracts and juice significantly mitigated these nephrotoxic effects, as confirmed by histopathological examination and biochemical marker analysis. Notably, the waste extract demonstrated greater efficacy than the whole fruit extract, likely due to its higher concentration of polyphenolic compounds, especially anthocyanins. These results highlight the potential of chokeberry as a therapeutic and preventive agent for kidney protection, emphasizing the value of by-products rich in biologically active compounds.
Collapse
Affiliation(s)
- Milica Milutinović
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| | - Nada Ćujić Nikolić
- Institute of Medicinal Plants Research, Dr Josif Pančić, Tadeuša Koščuška 1, 11000 Beograd, Serbia; (N.Ć.N.); (K.Š.)
| | - Tatjana Cvetković
- Institute of Biochemistry, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia;
- Centre of Medical and Clinical Biochemistry, University Clinical Centre, Bul. Dr Zorana Đinđića 48, 18000 Niš, Serbia
| | - Katarina Šavikin
- Institute of Medicinal Plants Research, Dr Josif Pančić, Tadeuša Koščuška 1, 11000 Beograd, Serbia; (N.Ć.N.); (K.Š.)
| | - Ivana Djordjević
- Pathology and Pathological Anatomy Center, University Clinical Centre, Bul. Dr Zorana Đinđića 48, 18000 Niš, Serbia; (I.D.); (L.J.V.)
| | - Ljubinka Janković Veličković
- Pathology and Pathological Anatomy Center, University Clinical Centre, Bul. Dr Zorana Đinđića 48, 18000 Niš, Serbia; (I.D.); (L.J.V.)
| | - Milica Randjelović
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| | - Bojana Miladinović
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| | - Suzana Branković
- Department of Physiology, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia;
| | - Dušanka Kitić
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| |
Collapse
|
68
|
Hu JW, Xiao JJ, Cai S, Zhong Y, Wang S, Liu S, Wu X, Cai Y, Zhang BF. Inhibition of mitochondrial over-division by (+)-14,15-Dehydrovincamine attenuates cisplatin-induced acute kidney injury via the JNK/Mff pathway. Free Radic Biol Med 2024; 224:190-203. [PMID: 39197599 DOI: 10.1016/j.freeradbiomed.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/01/2024]
Abstract
Cisplatin-induced acute kidney injury (AKI) is characterized by mitochondrial damage and apoptosis, and safe and effective therapeutic agents are urgently needed. Renal tubular epithelial cells, the main site of AKI, are enriched with a large number of mitochondria, which are crucial for the progression of AKI with an impaired energy supply. Vincamine has anti-inflammatory and antioxidant effects in mouse AKI models. As a natural compound derived from Tabernaemontana pandacaqui, (+)-14, 15-Dehydrovincamine and Vincamine differ in structure by only one double bond, and the role and exact mechanism of (+)-14, 15-Dehydrovincamine remains to be elucidated in AKI. The present study demonstrated that (+)-14,15-Dehydrovincamine significantly ameliorated mitochondrial dysfunction and maintained mitochondrial homeostasis in a cisplatin-induced AKI model. Furthermore, (+)-14,15-Dehydrovincamine ameliorates cytochrome C-dependent apoptosis in renal tubular epithelial cells. c-Jun NH2-terminal kinase (JNK) was identified as a potential target protein of (+)-14,15-Dehydrovincamine attenuating AKI by network pharmacological analysis. (+)-14,15-Dehydrovincamine inhibited cisplatin-induced JNK activation, mitochondrial fission factor (Mff) phosphorylation, and dynamin-related protein 1 (Drp1) translocation to the mitochondria in renal tubular epithelial cells. Meanwhile, the JNK activator anisomycin restored Mff phosphorylation and Drp1 translocation, counteracting the protective effect of (+)-14,15-Dehydrovincamine on mitochondrial dysfunction in cisplatin-induced TECs injury. In conclusion, (+)-14,15-Dehydrovincamine reduced mitochondrial fission, maintained mitochondrial homeostasis, and attenuated apoptosis by inhibiting the JNK/Mff/Drp1 pathway, which in turn ameliorated cisplatin-induced AKI.
Collapse
Affiliation(s)
- Jun-Wei Hu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China
| | - Jing-Jie Xiao
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China; Department of Cardiology, Zhongnan Hospital of Wuhan University, Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430071, China
| | - ShiQi Cai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - YuTing Zhong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - ShenTao Wang
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China
| | - ShuYe Liu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China
| | - XiaoYan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - YouSheng Cai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Bai-Fang Zhang
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, 430071, China.
| |
Collapse
|
69
|
Sponfeldner MI, Andrikyan W, Maas R, Fromm MF. Pseudo-Worsening of Kidney Function Due to Inhibition of Renal Creatinine Secretion: Quality of Information Provided in Prescribing Information/SmPC. Clin Pharmacol Ther 2024; 116:1259-1268. [PMID: 38989645 DOI: 10.1002/cpt.3374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
Determination of serum creatinine concentrations and subsequent calculation of estimated glomerular filtration rates (eGFR) is a cornerstone of clinical medicine. Crucial clinical decisions such as drug treatment discontinuations are based on eGFR calculated from serum creatinine measurements. However, creatinine is not only filtered in the kidneys, but also actively secreted into urine. Creatinine transporters such as OCT2, OCT3, MATE1, MATE2-K, and OAT2 expressed in proximal tubular cells are responsible for active renal secretion of creatinine. Multiple drugs (e.g., oral antitumor drugs) inhibit these transporters thereby causing a pseudo-worsening of kidney function with an increase in serum creatinine concentrations and a decrease in eGFR while other methods for eGFR determination (e.g., by cystatin C) reveal normal kidney function. Since US Prescribing Information (PI) and European Summaries of Product Characteristics (SmPCs) are the most relevant source of information for physicians, we investigated the quality of information in US PI/German SmPCs of drugs with clear evidence for pseudo-worsening of kidney function. 514 drugs putatively interacting with creatinine transporters were identified. For 149 of those drugs, an increase in serum creatinine concentrations has been described. Available data confirmed the existence of pseudo-worsening of kidney function for 30 of those drugs, for the remaining 119 drugs existing data are insufficient. Only 23.5% (12/51) of the 30 drugs' PI/SmPCs contained unambiguous statements on this proven pseudo-worsening of kidney function and gave clear recommendations for clinical management. Taken together, inadequate information provided in PI or SmPCs on the pseudo-worsening of kidney function poses patients at unnecessary risks.
Collapse
Affiliation(s)
- Michael I Sponfeldner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Wahram Andrikyan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
70
|
Li Y, Luo C, Cai Y, Wu Y, Shu T, Wei J, Wang H, Niu H. IGF2BP3/NCBP1 complex inhibits renal tubular senescence through regulation of CDK6 mRNA stability. Transl Res 2024; 273:1-15. [PMID: 38945255 DOI: 10.1016/j.trsl.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
Renal aging and the subsequent rise in kidney-related diseases are attributed to senescence in renal tubular epithelial cells (RTECs). Our study revealed that the abnormal expression of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), a reader of RNA N6-methyladenosine, is critically involved in cisplatin-induced renal tubular senescence. In cisplatin-induced senescence of RTECs, the promoter activity and transcription of IGF2BP3 is markedly suppressed. It was due to the down regulation of MYC proto-oncogene (MYC), which regulates IGF2BP3 transcription by binding to the putative site at 1852-1863 of the IGF2BP3 promoter. Overexpression of IGF2BP3 ameliorated cisplatin-induced renal tubular senescence in vitro. Mechanistic studies revealed that IGF2BP3 inhibits cellular senescence in RTECs by enhancing cyclin-dependent kinase 6 (CDK6) mRNA stability and increasing its expression. The inhibition effect of IGF2BP3 on tubular senescence is partially reversed by the knockdown of CDK6. Further, IGF2BP3 recruits nuclear cap binding protein subunit 1 (NCBP1) and inhibits CDK6 mRNA decay, by recognizing m6A modification. Specifically, IGF2BP3 recognizes m6A motif "GGACU" at nucleotides 110-114 in the 5' untranslated region (UTR) field of CDK6 mRNA. The involvement of IGF2BP3/CDK6 in alleviating tubular senescence was confirmed in a cisplatin-induced acute kidney injury (AKI)-to-chronic kidney disease (CKD) model. Clinical data also suggests an age-related decrease in IGF2BP3 and CDK6 levels in renal tissue or serum samples from patients. These findings suggest that IGF2BP3/CDK6 may be a promising target in cisplatin-induced tubular senescence and renal failure.
Collapse
Affiliation(s)
- Yaqin Li
- Department of General Practice, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yating Cai
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Wu
- Department of General Practice, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Shu
- Department of General Practice, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingyan Wei
- Department of General Practice, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongsheng Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Hongxin Niu
- Department of General Practice, Special Medical Service Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
71
|
Zhang H, Deng Z, Wang Y, Zheng X, Zhou L, Yan S, Wang Y, Dai Y, Kanwar YS, Chen F, Deng F. CHIP drives proteasomal degradation of NUR77 to alleviate oxidative stress and intrinsic apoptosis in cisplatin-induced nephropathy. Commun Biol 2024; 7:1403. [PMID: 39462094 PMCID: PMC11513124 DOI: 10.1038/s42003-024-07118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Carboxy-terminus of Hsc70-interacting protein (CHIP), an E3 ligase, modulates the stability of its targeted proteins to alleviate various pathological perturbations in various organ systems. Cisplatin is a widely used chemotherapeutic agent, but it is also known for its alarming renal toxicity. The role of CHIP in the pathogenesis of cisplatin-induced acute kidney injury (AKI) has not been adequately investigated. Herein, we demonstrated that CHIP was abundantly expressed in the renal proximal tubular epithelia, and its expression was downregulated in cisplatin-induced AKI. Further investigation revealed that CHIP overexpression or activation alleviated, while its gene disruption promoted, oxidative stress and apoptosis in renal proximal tubular epithelia induced by cisplatin. In terms of mechanism, CHIP interacted with and ubiquitinated NUR77 to promote its degradation, which consequently shielded BCL2 to maintain mitochondrial permeability of renal proximal tubular cells in the presence of cisplatin. Also, we demonstrated that CHIP interacted with NUR77 via its central coiled-coil (CC) domain, a non-canonical interactive pattern. In conclusion, these findings indicated that CHIP ubiquitinated and degraded its substrate NUR77 to attenuate intrinsic apoptosis in cisplatin-treated renal proximal tubular epithelia, thus providing a novel insight for the pathogenesis of cisplatin-induced AKI.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zebin Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Yilong Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Zheng
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Lizhi Zhou
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Shu Yan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Yashpal S Kanwar
- Departments of Pathology & Medicine, Northwestern University, Chicago, IL, USA
| | - Fangzhi Chen
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
72
|
Jogdeo CM, Panja S, Kumari N, Tang W, Kapoor E, Siddhanta K, Das A, Boesen EI, Foster KW, Oupický D. Inulin-based nanoparticles for targeted siRNA delivery in acute kidney injury. J Control Release 2024; 376:577-592. [PMID: 39419450 DOI: 10.1016/j.jconrel.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
RNA interference has emerged as a promising therapeutic strategy to tackle acute kidney injury (AKI). Development of targeted delivery systems is highly desired for selective renal delivery of RNA and improved therapeutic outcomes in AKI. Inulin is a plant polysaccharide traditionally employed to measure glomerular filtration rate. Here, we describe the synthesis of inulin modified with α-cyclam-p-toluic acid (CPTA) to form a novel renal-targeted polymer, Inulin-CPTA (IC), which is capable of selective siRNA delivery to the injured kidneys. We show that conjugating CPTA to inulin imparts IC with targeting properties for cells that overexpress the C-X-C chemokine receptor 4 (CXCR4). Self-assembled IC/siRNA nanoparticles (polyplexes) demonstrated rapid accumulation in the injured kidneys with selective uptake and prolonged retention in injured renal tubules overexpressing the CXCR4 receptor. Tumor-suppressor protein p53 contributes significantly to the pathogenesis of AKI. siRNA-induced silencing of p53 has shown therapeutic potential in several preclinical studies, making it an important target in the treatment of AKI. Systemically administered nanoparticles formulated using IC and siRNA against p53 selectively accumulated in the injured kidneys and potently silenced p53 expression. Selective p53 knockdown led to positive therapeutic outcomes in mice with cisplatin-induced AKI, as seen by reduced tubular cell death, renal injury, inflammation, and overall improved renal function. These findings indicate that IC is a promising new carrier for renal-targeted delivery of RNA for the treatment of AKI.
Collapse
Affiliation(s)
- Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ashish Das
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Erika I Boesen
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
73
|
Iwhiwhu SA, Kumar R, Khan AH, Afolabi JM, Williams JD, de la Cruz JE, Adebiyi A. A low-dose pemetrexed-cisplatin combination regimen induces significant nephrotoxicity in mice. BMC Nephrol 2024; 25:370. [PMID: 39434019 PMCID: PMC11494951 DOI: 10.1186/s12882-024-03822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Pemetrexed is combined with cisplatin to treat cancer. Whether pemetrexed-cisplatin combination chemotherapy exacerbates cisplatin nephrotoxicity is unclear. Here, we investigated kidney injury in mice administered a non-lethal low-dose regimen of pemetrexed or cisplatin alone and compared it with a pemetrexed-cisplatin combination. METHODS Mice were randomly divided into four groups and administered intraperitoneally the experimental drugs solubilized in captisol (sulfobutylether β-cyclodextrin). Group 1 received captisol, Group 2 pemetrexed (10 mg/kg), Group 3 cisplatin (1 mg/kg), and Group 4 pemetrexed (10 mg/kg) plus cisplatin (1 mg/kg). The mice were treated every other day for two weeks, three times per week. Glomerular filtration rate (GFR) was determined on the third day after the last treatment, followed by a necropsy. RESULTS Whereas the relative kidney weight was comparable in the control vs. pemetrexed or cisplatin alone group, it was significantly increased in the combination group. Mice treated with cisplatin and pemetrexed-cisplatin combination exhibited reduced GFR. The pemetrexed-cisplatin combination caused significant increases in the plasma or urinary levels of kidney injury biomarkers, renal lipid peroxidation, and nitrosative stress compared with pemetrexed or cisplatin alone. Histopathology revealed that pemetrexed or cisplatin alone had minimal effects on the kidneys. By contrast, the pemetrexed-cisplatin combination caused tubular degeneration, dilatation, and granular casts. Live-cell imaging showed that the pemetrexed-cisplatin combination caused more severe apoptosis of primary renal epithelial cells than individual concentrations. CONCLUSIONS These findings suggest that combining pemetrexed and cisplatin causes oxidative kidney damage at individual doses that do not cause significant nephrotoxicity. Hence, the renal function of patients undergoing treatment with the pemetrexed-cisplatin combination needs extensive monitoring.
Collapse
Affiliation(s)
- Samson A Iwhiwhu
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ravi Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri Columbia, MO, 65211, USA
| | - Abdul H Khan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri Columbia, MO, 65211, USA
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, MO, USA
| | - Jeremiah M Afolabi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jada D Williams
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri Columbia, MO, 65211, USA
| | - Julia E de la Cruz
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri Columbia, MO, 65211, USA
| | - Adebowale Adebiyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri Columbia, MO, 65211, USA.
- NextGen Precision Health, University of Missouri, Columbia, MO, USA.
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
74
|
Gueutin V, Cardineau A, Mathian A, Lanot A, Comoz F, Brocheriou I, Izzedine H. Renal involvement in solid cancers: epidemiological, clinical and histological characteristics study of 154 onconephrology patients. BMC Nephrol 2024; 25:367. [PMID: 39427142 PMCID: PMC11490999 DOI: 10.1186/s12882-024-03812-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Onconephrology is a growing discipline that aims to improve the management of patients with cancer and kidney disease. If kidney histology is an essential key, the anatomopathological data remain weak although essential to this complex management. METHODS Patients with active cancer who had a kidney biopsy (KB) between 2014 and 2020 were included, and their clinicobiological and histological data were analyzed retrospectively. RESULTS Our cohort consisted of 154 patients (83 women) with a mean age of 58 years. One hundred twelve patients presented with proteinuria, 95 with acute kidney injury, and 59 with arterial hypertension. Histologically, interstitial fibrosis was found in 74% of KBs, tubular atrophy in 55.1%, arteriolar hyalinosis in 58.4%, and fibrous endarteritis in 54.4%. Regarding the main acute lesions, thrombotic microangiopathy (TMA) was found in 29.9% of biopsies, acute tubular necrosis (ATN) in 51.3%, and acute interstitial nephritis in 24.8%. The etiological diagnosis most often made was the nephrotoxicity of anticancer drugs (87 patients), followed by a pre-renal (15 patients) and kidney disease unrelated to cancer (13 patients). Sixty-seven patients presented with at least 2 associated diagnoses reflecting the complexity of kidney damage in cancer. Different clusters were found, highlighting that immunotherapy and anti-VEGF were the most commonly involved drugs. CONCLUSIONS During onconephrology practice, kidney toxicity of treatments is the most common etiology. Several mechanisms can be involved, underscoring the importance of kidney biopsy and the complexity of its management. Chronic histological lesions were very common.
Collapse
Affiliation(s)
- Victor Gueutin
- Service de néphrologie-dialyse-transplantation CHU de CAEN, Côte de Nacre, Caen, France.
- Service de néphrologie-dialyse, hôpital Jacques-Monod, Rue Eugène-Garnier, Flers, France.
| | - Aurore Cardineau
- Service de néphrologie-dialyse CH Mémorial France Etats-Unis, 715 rue Henri Dunant, Saint Lô, France
| | - Alexis Mathian
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié- Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des anti-phospholipides et autres maladies auto-immunes rares, Service de Médecine Interne 2, Institut E3M, Paris, France
| | - Antoine Lanot
- Service de néphrologie-dialyse-transplantation CHU de CAEN, Côte de Nacre, Caen, France
- Service de néphrologie-dialyse, hôpital Jacques-Monod, Rue Eugène-Garnier, Flers, France
| | - François Comoz
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Caen, France
| | - Isabelle Brocheriou
- Service d'Anatomie et Cytologie Pathologiques, La Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
- UMRS 1155, Institut National de la Santé et de la Recherche Médicale, Hôpital Tenon, Sorbonne Université, Paris, France
| | - Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Paris, France
| |
Collapse
|
75
|
Guo L, Ding Y, Li L, Gao C, Su J, Zhang Z. Molecular Logic Gate for Sensing pH/Peroxynitrite with Potential Applications in Cisplatin Treatment. Anal Chem 2024; 96:15950-15959. [PMID: 39327258 DOI: 10.1021/acs.analchem.4c03020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Cisplatin is a common chemotherapy drug for multiple solid tumors; however, due to the nitrification of peroxynitrite (ONOO-), a series of side effects seriously affect its dose and efficacy. Considering that the reactivity of ONOO- is significantly affected by pH in vitro, revealing their roles in living cells contributes to understanding the side-effect process induced by cisplatin. Herein, we present a near-infrared fluorescent logic gate for sensing pH/ONOO-, which can accurately discriminate four scenarios (no analyte, analyte H+ alone, analyte ONOO- alone, and H+ + ONOO-) and which one comes first. With this probe, the significant roles of pH and ONOO- in cisplatin treatment are disclosed, in which the cell account shows a dramatic reduction accompanied by decreased pH and upregulated ONOO- levels. By artificially recovering the pH, the ONOO- content and cell account can maintain a stable state, possibly due to the protection from acidification and nitration. This work provides an ideal pH/ONOO- logical sensor for revealing their potential roles under cisplatin, which is expected to proffer new insights into more related diseases and drug research.
Collapse
Affiliation(s)
- Lifang Guo
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yuxi Ding
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Lu Li
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Caifang Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Jianhua Su
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zhiyun Zhang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, East China University of Science & Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
76
|
Muramoto K, Urabe F, Koike Y, Yamamoto S, Suzuki H, Miyajima K, Fukuokaya W, Iwatani K, Imai Y, Igarashi T, Mori K, Aikawa K, Kimura S, Tashiro K, Yamada Y, Sasaki T, Sato S, Yuen SKK, Shimomura T, Furuta A, Tsuzuki S, Miki J, Kimura T. Perioperative renal function change and oncological outcomes of radical nephroureterectomy in patients with upper tract urothelial carcinoma: A multicenter retrospective study. Urol Oncol 2024; 42:332.e21-332.e32. [PMID: 38729868 DOI: 10.1016/j.urolonc.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/29/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND The effect of radical nephroureterectomy (RNUx) on postoperative renal function in patients diagnosed with upper tract urothelial carcinoma (UTUC) has not been thoroughly explored. METHODS We conducted a retrospective analysis including 785 patients who underwent RNUx for UTUC. We assessed the preoperative and postoperative estimated glomerular filtration rates (eGFRs) and factors related to the decline in eGFR. Additionally, we examined the effect of comorbidities (diabetes or hypertension) on the postoperative eGFR at 1 year. Cox proportional hazard models were employed to investigate the clinical effect of RNUx on oncological outcomes, including non-urothelial tract recurrence-free survival (NUTRFS), cancer-specific survival (CSS), and overall survival (OS). RESULTS The median preoperative and postoperative eGFR levels were 54.7 and 40.6 ml/min/1.73 m2 respectively. The proportions of patients with preoperative and postoperative eGFR ≥60 mL/min/1.73 m2 were 35.9% and 5.1%, respectively. The median decline in the eGFR after surgery was 26.8%. Patients with preoperative eGFR <60 ml/min/1.73 m2 demonstrated significantly lower odds of a postoperative decline in eGFR of 25% or more. The effect of comorbidities on postoperative eGFR at 1 year was significant (P = 0.048). The 3-year NUTRFS, CSS, and OS rates were 72.9%, 85.2%, and 81.5%, respectively. Preoperative chronic kidney disease was an independent factor associated with inferior NUTRFS, CSS, and OS. CONCLUSION Different degrees of impairment of renal function occur among UTUC patients. Only 5.1% of patients retain a postoperative eGFR ≥60 ml/min/1.73 m2. Preoperative renal impairment was linked to reduced odds of postoperative eGFR decrease and associated with survival. In addition, the presence of comorbidities had a significant effect on the decline in eGFR. These findings emphasize the importance of developing evidence-based perioperative treatment strategies for UTUC patients with impaired renal function.
Collapse
Affiliation(s)
- Katsuki Muramoto
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan.
| | - Yuhei Koike
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shutaro Yamamoto
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirotaka Suzuki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiichiro Miyajima
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Wataru Fukuokaya
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kosuke Iwatani
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan; Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Yu Imai
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Taro Igarashi
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiichiro Mori
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Koichi Aikawa
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shoji Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan; Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Kojiro Tashiro
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan; Department of Urology, Jikei Katsushika Medical Center, Tokyo, Japan
| | - Yuta Yamada
- Department of Urology, University of Tokyo, Tokyo, Japan
| | - Takaya Sasaki
- Department of Nephrology and Hypertension, The Jikei University School of Medicine, Tokyo, Japan
| | - Shun Sato
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Steffi Kar Kei Yuen
- S.H.Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Tatsuya Shimomura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Akira Furuta
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shunsuke Tsuzuki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan; Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
77
|
Li P, Li D, Lu Y, Pan S, Cheng F, Li S, Zhang X, Huo J, Liu D, Liu Z. GSTT1/GSTM1 deficiency aggravated cisplatin-induced acute kidney injury via ROS-triggered ferroptosis. Front Immunol 2024; 15:1457230. [PMID: 39386217 PMCID: PMC11461197 DOI: 10.3389/fimmu.2024.1457230] [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: 06/30/2024] [Accepted: 08/23/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Cisplatin is a widely used chemotherapeutic agent prescribed to treat solid tumors. However, its clinical application is limited because of cisplatin- induced nephrotoxicity. A known complication of cisplatin is acute kidney injury (AKI). Deletion polymorphisms of GSTM1 and GSTT1, members of the glutathione S-transferase family, are common in humans and are presumed to be associated with various kidney diseases. However, the specific roles and mechanisms of GSTM1 and GSTT1 in cisplatin induced AKI remain unclear. Methods To investigate the roles of GSTM1 and GSTT1 in cisplatin-induced AKI, we generated GSTM1 and GSTT1 knockout mice using CRISPR-Cas9 technology and assessed their kidney function under normal physiological conditions and cisplatin treatment. Using ELISA kits, we measured the levels of oxidative DNA and protein damage, along with MDA, SOD, GSH, and the GSH/GSSG ratio in wild-type and GSTM1/GSTT1 knockout mice following cisplatin treatment. Additionally, oxidative stress levels and the expression of ferroptosis-related proteins in kidney tissues were examined through Western blotting, qPCR, immunohistochemistry, and immunofluorescence techniques. Results Here, we found that GSTT1 and GSTM1 were downregulated in the renal tubular cells of AKI patients and cisplatin-treated mice. Compared with WT mice, Gstm1/Gstt1-DKO mice were phenotypically normal but developed more severe kidney dysfunction and exhibited increased ROS levels and severe ferroptosis after injecting cisplatin. Discussion Our study revealed that GSTM1 and GSTT1 can protect renal tubular cells against cisplatin-induced nephrotoxicity and ferroptosis, and genetic screening for GSTM1 and GSTT1 polymorphisms can help determine a standard cisplatin dose for cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Peipei Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Duopin Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yanfang Lu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Fei Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shen Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaonan Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jinling Huo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
78
|
Belletto D, Vigna V, Barretta P, Ponte F, Mazzone G, Scoditti S, Sicilia E. Computational assessment of the use of graphene-based nanosheets as Pt II chemotherapeutics delivery systems. J Comput Chem 2024; 45:2059-2070. [PMID: 38741357 DOI: 10.1002/jcc.27394] [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: 01/31/2024] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Graphene is the newest form of elemental carbon and it is becoming rapidly a potential candidate in the framework of nano-bio research. Many reports confirm the successful use of graphene-based materials as carriers of anticancer drugs having relatively high loading capacities compared with other nanocarriers. Here, the outcomes of a systematic study of the adsorption behavior of FDA approved PtII drugs cisplatin, oxaliplatin, and carboplatin on surface models of pristine, holey, and nitrogen-doped holey graphene are reported. DFT investigations in water solvent have been carried out considering several initial orientations of the drugs with respect to the surfaces. Adsorption free energies, calculated including basis set superposition error (BSSE) corrections, result to be significantly negative for many of the drug@carrier adducts indicating that tested layers could be used as potential carriers for the delivery of anticancer PtII drugs. The reduced density gradient (RDG) analysis allows to show that many kinds of non-covalent interactions, including canonical H-bond, are responsible for the stabilization of the formed adducts.
Collapse
Affiliation(s)
- Daniele Belletto
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| | - Vincenzo Vigna
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| | - Pierraffaele Barretta
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| | - Gloria Mazzone
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| | - Stefano Scoditti
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
79
|
Zhang L, Dong Z, Jiang F, Huang H, Ding H, Liu M. Ferrostatin-1 ameliorates Cis-dichlorodiammineplatinum(II)-induced ovarian toxicity by inhibiting ferroptosis. Mol Med 2024; 30:150. [PMID: 39272008 PMCID: PMC11401273 DOI: 10.1186/s10020-024-00923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Cis-dichlorodiammineplatinum(II) (CDDP), while widely utilized in tumor therapy, results in toxic side effects that patients find intolerable. The specific mechanism by which CDDP inflicts ovarian damage remains unclear. This study aimed to explore the involvement of ferrostatin-1 (FER-1) and ferroptosis in CDDP-induced ovarian toxicity. This study established models of CDDP-induced injury in granulosa cells (GCs) and rat model of premature ovarian failure (POF). CCK-8 assessed the effects of CDDP and FER-1 on GC viability. FerroOrange and Mito-FerroGreen, DCFH-DA and MitoSox-Red, Rhodamine 123 and Transmission electron microscopy (TEM) measured Fe2+, reactive oxygen species (ROS), mitochondrial membrane potential and the mitochondrial morphology in GC cells, respectively. Serum hormone levels; organ indices; malondialdehyde, superoxide dismutase, and glutathione analyses; and western blotting were performed to examine ferroptosis's role in vitro. Molecular docking simulation was evaluated the interaction between FER-1 and GPX4 or FER-1 and NRF2. Molecular docking simulations were conducted to evaluate the interactions between FER-1 and GPX4, as well as FER-1 and NRF2. The findings revealed that CDDP-induced ovarian toxicity involved iron accumulation, increased ROS accumulation, and mitochondrial dysfunction, leading to endocrine disruption and tissue damage in rats. These changes correlated with NRF2, HO-1, and GPX4 levels. However, FER-1 decreased the extent of ferroptosis. Thus, ferroptosis appears to be a crucial mechanism of CDDP-induced ovarian injury, with GPX4 as potential protective targets.
Collapse
Affiliation(s)
- Lu Zhang
- Harbin Medical University, Harbin, 150086, Heilongjiang, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Zhe Dong
- Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Fan Jiang
- Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Huaju Huang
- Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Hui Ding
- Harbin Medical University, Harbin, 150086, Heilongjiang, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Meimei Liu
- Harbin Medical University, Harbin, 150086, Heilongjiang, China.
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.
| |
Collapse
|
80
|
Yang Y, Xiong T, Wang T, Chen X, Ma Z, Zuo B, Ning D, Song R, Liu X, Wang D. Small GTP-binding protein GDP dissociation stimulator influences cisplatin-induced acute kidney injury via PERK-dependent ER stress. Commun Biol 2024; 7:1091. [PMID: 39237614 PMCID: PMC11377573 DOI: 10.1038/s42003-024-06792-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
Cisplatin is a common anticancer drug, but its frequent nephrotoxicity limits its clinical use. Small GTP-binding protein GDP dissociation stimulator (smgGDS), a small GTPase chaperone protein, was considerably downregulated during cisplatin-induced acute kidney injury (CDDP-AKI), especially in renal tubular epithelial cells. SmgGDS-knockdown mice was established and found that smgGDS knockdown promoted CDDP-AKI, as demonstrated by an increase in serum creatine, blood urea nitrogen levels and the appearance of tubular patterns. RNA sequencing suggested that protein kinase RNA-like ER kinase (PERK), which bridges mitochondria-associated ER membranes, was involved in smgGDS knockdown following CDDP-AKI, and then identified that smgGDS knockdown increased phosphorylated-PERK in vivo and in vitro. Furthermore, we confirmed that smgGDS deficiency aggravated apoptosis and ER stress in vivo and in vitro. And the ER stress inhibitor 4-Phenylbutyric acid and the inhibition of PERK phosphorylation mitigated smgGDS deficiency-induced ER stress related apoptosis following cisplatin treatment, while the eIF2α phosphorylation inhibitor could not reverse the smgGDS deficiency accelerated cell death. Furthermore, the over-expression of smgGDS could reverse the ER stress and apoptosis caused by CDDP. Overall, smgGDS regulated PERK-dependent ER stress and apoptosis, thereby influencing renal damage. This study identified a target for diagnosing and treating cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Yuxue Yang
- The Hospital Affiliated to the Medical School of Yangzhou University (Taizhou People's Hospital), No. 366 Taihu Road, Taizhou, Jiangsu, 225300, China
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Ting Xiong
- Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Ti Wang
- The Hospital Affiliated to the Medical School of Yangzhou University (Taizhou People's Hospital), No. 366 Taihu Road, Taizhou, Jiangsu, 225300, China
| | - Xiwei Chen
- The Hospital Affiliated to the Medical School of Yangzhou University (Taizhou People's Hospital), No. 366 Taihu Road, Taizhou, Jiangsu, 225300, China
| | - Ziwei Ma
- Clinical Medical College, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Bangyun Zuo
- The Hospital Affiliated to the Medical School of Yangzhou University (Taizhou People's Hospital), No. 366 Taihu Road, Taizhou, Jiangsu, 225300, China
| | - Dong Ning
- School of Medicine, National University of Ireland Galway, University Road, Galway, 999014, Ireland
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, #88 South University Avenue, Yangzhou, Jiangsu, 225009, China
| | - Xuesong Liu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Daxin Wang
- The Hospital Affiliated to the Medical School of Yangzhou University (Taizhou People's Hospital), No. 366 Taihu Road, Taizhou, Jiangsu, 225300, China.
| |
Collapse
|
81
|
Yang A, Ding Y, Guo C, Liu C, Xiong Z, Quan M, Bai P, Cai R, Li B, Li G, Deng Y, Wu C, Sun Y. PARVB deficiency alleviates cisplatin-induced tubular injury by inhibiting TAK1 signaling. Cell Mol Life Sci 2024; 81:385. [PMID: 39235496 PMCID: PMC11377400 DOI: 10.1007/s00018-024-05422-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024]
Abstract
Cisplatin-induced renal tubular injury largely restricts the wide-spread usage of cisplatin in the treatment of malignancies. Identifying the key signaling pathways that regulate cisplatin-induced renal tubular injury is thus clinically important. PARVB, a focal adhesion protein, plays a crucial role in tumorigenesis. However, the function of PARVB in kidney disease is largely unknown. To investigate whether and how PARVB contributes to cisplatin-induced renal tubular injury, a mouse model (PARVB cKO) was generated in which PARVB gene was specifically deleted from proximal tubular epithelial cells using the Cre-LoxP system. In this study, we found depletion of PARVB in proximal tubular epithelial cells significantly attenuates cisplatin-induced renal tubular injury, including tubular cell death and inflammation. Mechanistically, PARVB associates with transforming growth factor-β-activated kinase 1 (TAK1), a central regulator of cell survival and inflammation that is critically involved in mediating cisplatin-induced renal tubular injury. Depletion of PARVB promotes cisplatin-induced TAK1 degradation, inhibits TAK1 downstream signaling, and ultimately alleviates cisplatin-induced tubular cell damage. Restoration of PARVB or TAK1 in PARVB-deficient cells aggravates cisplatin-induced tubular cell injury. Finally, we demonstrated that PARVB regulates TAK1 protein expression through an E3 ligase ITCH-dependent pathway. PARVB prevents ITCH association with TAK1 to block its ubiquitination. Our study reveals that PARVB deficiency protects against cisplatin-induced tubular injury through regulation of TAK1 signaling and indicates targeting this pathway may provide a novel therapeutic strategy to alleviate cisplatin-induced kidney damage.
Collapse
Affiliation(s)
- Aihua Yang
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yanyan Ding
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chen Guo
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chengmin Liu
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zailin Xiong
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Meiling Quan
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Panzhu Bai
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Renwei Cai
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Binbin Li
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guizhen Li
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yi Deng
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chuanyue Wu
- Department of Pathology, School of Medicine and University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Ying Sun
- Department of Systems Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China.
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
82
|
Yu JB, Padanilam BJ, Kim J. Activation of Yes-Associated Protein Is Indispensable for Transformation of Kidney Fibroblasts into Myofibroblasts during Repeated Administration of Cisplatin. Cells 2024; 13:1475. [PMID: 39273045 PMCID: PMC11393901 DOI: 10.3390/cells13171475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cisplatin is a potent chemotherapy medication that is used to treat various types of cancer. However, it can cause nephrotoxic side effects, which lead to acute kidney injury (AKI) and subsequent chronic kidney disease (CKD). Although a clinically relevant in vitro model of CKD induced by repeated administration of low-dose cisplatin (RAC) has been established, its underlying mechanisms remain poorly understood. Here, we compared single administration of high-dose cisplatin (SAC) to repeated administration of low-dose cisplatin (RAC) in myofibroblast transformation and cellular morphology in a normal rat kidney fibroblast NRK-49F cell line. RAC instead of SAC transformed the fibroblasts into myofibroblasts as determined by α-smooth muscle actin, enlarged cell size as represented by F-actin staining, and increased cell flattening as expressed by the semidiameter ratio of attached cells to floated cells. Those phenomena, as well as cellular senescence, were significantly detected from the time right before the second administration of cisplatin. Interestingly, inhibition of the interaction between Yes-associated protein (YAP) and the transcriptional enhanced associated domain (TEAD) using Verteporfin remarkedly reduced cell size, cellular senescence, and myofibroblast transformation during RAC. These findings collectively suggest that YAP activation is indispensable for cellular hypertrophy, senescence, and myofibroblast transformation during RAC in kidney fibroblasts.
Collapse
Affiliation(s)
- Jia-Bin Yu
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea;
| | - Babu J. Padanilam
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Jinu Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea;
- Department of Anatomy, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
83
|
Ning X, Zhong Y, Cai Q, Wang Y, Jia X, Hsieh JT, Zheng J, Yu M. Gold Nanoparticle Transport in the Injured Kidneys with Elevated Renal Function Biomarkers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402479. [PMID: 39073056 PMCID: PMC11410533 DOI: 10.1002/adma.202402479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/01/2024] [Indexed: 07/30/2024]
Abstract
Renal function biomarkers such as serum blood urea nitrogen (BUN) and creatinine (Cr) serve as key indicators for guiding clinical decisions before administering kidney-excreted small-molecule agents. With engineered nanoparticles increasingly designed to be renally clearable to expedite their clinical translation, understanding the relationship between renal function biomarkers and nanoparticle transport in diseased kidneys becomes crucial to their biosafety in future clinical applications. In this study, renal-clearable gold nanoparticles (AuNPs) are used as X-ray contrast agents to noninvasively track their transport and retention in cisplatin-injured kidneys with varying BUN and Cr levels. The findings reveal that AuNP transport is significantly slowed in the medulla of severely injured kidneys, with BUN and Cr levels elevated to 10 times normal. In mildly injured kidneys, where BUN and Cr levels only four to five times higher than normal, AuNP transport and retention are not predictable by BUN and Cr levels but correlate strongly with the degree of tubular injury due to the formation of gold-protein casts in the Henle's loop of the medulla. These results underscore the need for caution when employing renal-clearable nanomedicines in compromised kidneys and highlight the potential of renal-clearable AuNPs as X-ray probes for assessing kidney injuries noninvasively.
Collapse
Affiliation(s)
- Xuhui Ning
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Yuncheng Zhong
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Qi Cai
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yaohong Wang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Xun Jia
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Jer-Tsong Hsieh
- Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jie Zheng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Mengxiao Yu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, 75080, USA
| |
Collapse
|
84
|
Moreno-Gordaliza E, González-Nicolás MÁ, Lázaro A, Barbas C, Gómez-Gómez MM, López-Gonzálvez Á. Untargeted metabolomics analysis of serum and urine unveils the protective effect of cilastatin on altered metabolic pathways during cisplatin-induced acute kidney injury. Biochem Pharmacol 2024; 227:116435. [PMID: 39025411 DOI: 10.1016/j.bcp.2024.116435] [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: 03/15/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Acute kidney injury (AKI) is one of the most serious complications of cisplatin anticancer therapies. Cilastatin is a highly promising nephroprotective agent to eventually enter clinical use, but its biochemical mechanism is still not fully understood. We have employed an untargeted metabolomics approach based on capillary electrophoresis mass spectrometry (CE-MS) analysis of serum and urine from an in vivo rat model, to explore the metabolic pathways involved in cisplatin-induced AKI and cilastatin nephroprotection. A total of 155 and 76 identified metabolites were found to be significantly altered during cisplatin treatment in urine and serum, respectively. Most of these altered metabolites were either partially or totally recovered by cilastatin and cisplatin co-treatment. The main metabolic pathways disturbed by cisplatin during AKI involved diverse amino acids metabolism and biosynthesis, tricarboxylic acids (TCA) cycle, nicotinate and nicotinamide metabolism, among others. Cilastatin was proved to protect diverse cisplatin-altered pathways involving metabolites related to immunomodulation, inflammation, oxidative stress and amino acid metabolism in proximal tubules. However, cisplatin-altered mitochondrial metabolism (especially, the energy-producing TCA cycle) remained largely unprotected by cilastatin, suggesting an unresolved mitochondrial direct damage. Multivariate analysis allowed effective discrimination of cisplatin-induced AKI and cilastatin renoprotection based on metabolic features. A number of potential serum and urine biomarkers could also be foreseen for cisplatin-induced AKI detection and cilastatin nephroprotection.
Collapse
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Computense s/n, 28040 Madrid, Spain.
| | - M Ángeles González-Nicolás
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Alberto Lázaro
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; Department of Physiology, School of Medicine, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Complutense s/n, 28040 Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Faculty of Pharmacy, Universidad San Pablo-CEU (CEU Universities), Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - M Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Computense s/n, 28040 Madrid, Spain
| | - Ángeles López-Gonzálvez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Faculty of Pharmacy, Universidad San Pablo-CEU (CEU Universities), Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| |
Collapse
|
85
|
Amini N, Nejaddehbashi F, Badavi M, Bayati V, Zahra Basir. Combined effect of naringin and adipose tissue-derived mesenchymal stem cell on cisplatin nephrotoxicity through Sirtuin1/Nrf-2/HO-1 signaling pathway: a promising nephroprotective candidate. Cell Tissue Res 2024; 397:193-204. [PMID: 38953985 DOI: 10.1007/s00441-024-03902-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Cisplatin nephrotoxicity is a well-known emergency clinical condition caused by oxidative stress and inflammation. Naringin (NAR) is considered an antioxidant agent with renoprotective effects capable of removing reactive oxygen species. Adipose tissue-derived mesenchymal stem cells (AD-MSCs) are reported to have anti-inflammatory and antioxidant properties. The present research examined the renoprotective effect of the combination of NAR and AD-MSCs as opposed to each one alone on cisplatin-induced nephrotoxicity through SIRT-1/Nrf-2/HO-1 pathway. This study included five groups (n = 8 each) of male Sprague-Dawley rats (200 - 220 g): sham, cisplatin: rats receiving cisplatin (6.5 mg/kg, i.p.) on the 4th day; NAR+cisplatin: rats pretreated with NAR (1 week, i.p.) + cisplatin on the 4th day; AD-MSCs: rats receiving AD-MSCs (1 × 106) by injection through the tail vein on the 5th day + cisplatin on the 4th day; and NAR+AD-MSCs+cisplatin. On the 8th day, the animals were anesthetized to obtain tissue and blood samples. Biochemical factors, inflammation, oxidative stress, and gene expression were explored. Cisplatin increased blood urea nitrogen, creatinine, inflammation, and oxidative stress. Moreover, mRNA expression of Sirtuin1, nuclear factor erythroid 2-related factor 2 (Nrf-2), and heme oxygenase-1 (HO-1) remarkably reduced. Furthermore, cisplatin led to a disturbance in kidney structure (glomerular atrophy, cell infiltrations, and tubular dysfunction) as confirmed by histology findings. However, NAR pretreatment, AD-MSC administration, or a combination of both significantly reversed these changes. Overall, when used together, NAR and AD-MSCs had stronger cisplatin-induced effects on kidney dysfunction by inhibiting inflammation, reducing oxidative stress, and increasing the Sirtuin1/Nrf-2/HO-1 pathway.
Collapse
Affiliation(s)
- Negin Amini
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Fereshteh Nejaddehbashi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Basir
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
86
|
Zang YD, Wu HJ, Chen XY, Ma ZL, Li CJ, Ma J, Chen XG, Sheng L, Zhang S, Zhang DM. Synthesis and Biological Evaluation of Novel Psidium Meroterpenoid Derivatives against Cisplatin-Induced Acute Kidney Injury. J Med Chem 2024; 67:14234-14255. [PMID: 39137258 DOI: 10.1021/acs.jmedchem.4c01099] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Cisplatin is a widely used drug for the clinical treatment of tumors. However, nephrotoxicity limits its widespread use. A series of compounds including eight analogs (G3-G10) and 40 simplifiers (G11-G50) were synthesized based on the total synthesis of Psiguamer A and B, which were novel meroterpenoids with unusual skeletons from the leaves of Psidium guajava. Among these compounds, (d)-G8 showed the strongest protective effect on cisplatin-induced acute kidney injury (AKI) in vitro and vivo, and slightly enhanced the antitumor efficacy of cisplatin. A mechanistic study showed that (d)-G8 promoted the efflux of cisplatin via upregulating the copper transporting efflux proteins ATP7A and ATP7B. It enhanced autophagy through the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway. (d)-G8 showed no acute toxicity or apparent pathological damage in the healthy mice at a single dose of 1 g/kg. This study provides a promising lead against cisplatin-induced AKI.
Collapse
Affiliation(s)
- Ying-Da Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Hai-Jie Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xin-Yi Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Zhi-Ling Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Chuang-Jun Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Jie Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xiao-Guang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Li Sheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Sen Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Dong-Ming Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
87
|
Bui DT, Nagasaki Y. Developing poly(ethylene glycol)- b-poly(β-hydroxybutyrate)-based self-assembling prodrug for the management of cisplatin-induced acute kidney injury. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2382084. [PMID: 39166178 PMCID: PMC11334744 DOI: 10.1080/14686996.2024.2382084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024]
Abstract
Although β-hydroxybutyrate (BHB), one of the endogenous body ketones, possesses high bioactivities, it is rapidly consumed, metabolized, and eliminated from the body. In this study, we designed new self-assembling nanoparticles that sustainably released BHB to improve bioavailability and evaluated their efficacy in in vivo experiments using rodent animal models. Since poly(β-hydroxybutyrate) [poly(BHB)] is regarded as a polymeric prodrug that is hydrolyzed by endogenous enzymes and releases BHB in a sustained manner, our idea was to engineer hydrophobic poly(BHB) in one of the segments in the amphiphilic block copolymer, of which self-assembles in water to form nanoparticles of tens of nanometers in size (abbreviated as NanoBHB). Here, methoxy-poly(ethylene glycol) was employed as the hydrophilic segment of the block copolymer to stabilize the nanoparticles in aqueous environments, thus enabling NanoBHBs to be administrable both orally and through injection. Experimental results showed that NanoBHB has low toxicity and releases free BHB for an extended period in vitro and in vivo. Moreover, NanoBHB exhibits superior nephroprotective effects in cisplatin-induced acute kidney injury mouse models compared to low-molecular-weight (LMW) sodium BHB, suggesting the potential of NanoBHB as a sustainable release formulation to supply BHB for medicinal applications.
Collapse
Affiliation(s)
- Duc Tri Bui
- Degree Program of Pure and Applied Sciences, Graduate School of Science and Technology, University of Tsukuba, Ibaraki, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Ibaraki, Japan
- Master’s School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
- Center for Research in Radiation and Earth System Science (CRiES), University of Tsukuba, Ibaraki, Japan
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- High-Value Biomaterials Research and Commercialization Center (HBRCC), National Taipei University of Technology, Taipei, Taiwan
| |
Collapse
|
88
|
Luo H, Yi G, Tang H, Chen L, Hu L, Yang D, Chen Z, Li H, Zhan D, Yu Y, Zeng Y, Cai Y, Wu J, Liu H. Proton pump inhibitors may increase the risk of cisplatin-induced acute kidney injury in patients with nasopharyngeal carcinoma: a prospective cohort study. Sci Rep 2024; 14:18839. [PMID: 39138312 PMCID: PMC11322290 DOI: 10.1038/s41598-024-69821-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
Cisplatin is the most commonly used platinum-based treatment for nasopharyngeal carcinoma (NPC). However, its clinical application is limited owing to its nephrotoxicity and gastrointestinal reactions. Proton pump inhibitors (PPIs) have been reported to increase nephrotoxicity risk in previous studies. We aimed to evaluate whether PPIs increase cisplatin-induced nephrotoxicity in patients with NPC. In total, 295 patients were included in this prospective cohort study: 145 in the PPIs group and 150 in the non-PPIs group. All patients underwent cisplatin-based induction chemotherapy, followed by cisplatin-based concurrent chemoradiotherapy. The PPIs group received 40 mg of intravenous esomeprazole sodium for 7 days in each chemotherapy cycle. Chi-squared test and logistic regression analyses with odds ratios and 95% confidence intervals were applied to assess the association between PPIs and the risk of acute kidney injury (AKI). AKI incidence in the PPIs group was significantly higher than that in the non-PPIs group (P = 0.005). After adjusting for various confounders including demographic features, clinical features, and renal function indices, PPIs use was significantly associated with a higher AKI risk (odds ratio: 2.775; 95% confidence interval 1.280-6.020; P = 0.010). The incidences of acute and chronic kidney diseases were similar between both groups (P > 0.05), whereas the incidence of nausea was lower in the PPIs group than in the non-PPIs group (P = 0.029). This study has shown that PPIs use may increase the risk of cisplatin-induced acute nephrotoxicity in patients with NPC.
Collapse
Affiliation(s)
- Haiqing Luo
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Guihua Yi
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Haifeng Tang
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Lingli Chen
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Liren Hu
- School of Public Health of Guangdong Medical University, Zhanjiang, 524023, China
| | - Donghong Yang
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Zihong Chen
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Haiwen Li
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Dechao Zhan
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Ying Yu
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Ying Zeng
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Yilin Cai
- Specialty of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China
| | - Jiayuan Wu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China.
| | - Huafeng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524002, China.
| |
Collapse
|
89
|
Gutgarts V, Gerardine S, Shingarev RA, Knezevic A, Zabor EC, Latcha S, Joy MS, Aleksunes LM, Jaimes EA. Evaluation of Cisplatin-Induced Acute Kidney Injury in Patients Coprescribed Serotonin Receptor Antagonists: A Retrospective Analysis. KIDNEY360 2024; 5:1094-1100. [PMID: 38814726 PMCID: PMC11371355 DOI: 10.34067/kid.0000000000000464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 04/26/2024] [Indexed: 06/01/2024]
Abstract
Key Points Serotonin receptor antagonists reduce the incidence of AKI in patients receiving cisplatin as chemotherapy. New-generation serotonin receptors do not offer any additional advantage in terms of protection from cisplatin induced AKI. Background Cisplatin is an effective first-line therapy for a variety of cancers. Cisplatin is highly emetogenic and resulting volume depletion can contribute to AKI. Antiemetic drugs, such as 5-hydroxytryptamine type 3 receptor antagonists (5-HT3RAs), are commonly prescribed to prevent this complication. Preclinical studies suggest first-generation 5-HT3RAs may alter the renal clearance and increase cisplatin toxicity. This retrospective study evaluated whether different 5-HT3RAs modify the risk of AKI in patients receiving cisplatin. Methods Patients with cancer who received cisplatin between January 1, 2010, and December 31, 2016, were included. Patients older than 18 years with available data for baseline and post-treatment serum creatinine, cisplatin cumulative dose, and administration of 5-HT3RAs, including first-generation (ondansetron, granisetron, and ramosetron) and second-generation (palonosetron), were analyzed. AKI was defined as 1.5× increase in serum creatinine. Fisher exact and Wilcoxon rank-sum tests were used to assess univariable associations between baseline covariates and AKI and logistic regression for multivariable associations with AKI. Results Of 8703 patients identified with cisplatin exposure, 6889 were included. A total of 3881 patients (56.3%) received at least one 5-HT3RA, including palonosetron (3750, 54.4%), ondansetron (1399, 20.3%), and granisetron (11, 0.2%). AKI developed in 1666 patients (24.2%) after cisplatin therapy. Patients who received any 5-HT3RAs were less likely to experience AKI as compared with patients who did not (22.6% versus 26.2%, P = 0.001). Older age, male sex, African ethnicity, and cumulative cisplatin dose were univariably associated with higher risk of AKI (P < 0.001). After adjusting for these variables, use of any of these antiemetic drugs was protective for AKI (odds ratio, 0.84; 95% confidence interval, 0.75 to 0.94; P = 0.003) with no difference detected between type of 5-HT3RA. Conclusions Nephrotoxicity continues to be a concern after cisplatin therapy. Given its emetogenic nature, use of antiemetic drugs, such as 5-HT3RAs, can lessen emesis and lower risk of kidney injury. This retrospective analysis supports use of any 5-HT3RAs to lower risk of AKI.
Collapse
Affiliation(s)
- Victoria Gutgarts
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | | | | | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily C. Zabor
- Department of Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sheron Latcha
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Melanie S. Joy
- Division of Nephrology, Cancer Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, School of Medicine, University of Colorado, Denver, Colorado
| | | | - Edgar A. Jaimes
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| |
Collapse
|
90
|
Zhao X, Meng Y, Dang C, Xue L, Zhang J, Ma S, Li H. VALD-2 mitigates cisplatin-induced acute kidney injury: Mechanistic insights into oxidative stress modulation and inflammation suppression. J Biochem Mol Toxicol 2024; 38:e23786. [PMID: 39072927 DOI: 10.1002/jbt.23786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
This study explores the compelling antitumor properties of VALD-2, a synthetic Schiff base ligand known for its low toxicity. The focus is on investigating VALD-2's protective role against cisplatin-induced acute kidney injury (AKI) in mice, with a specific emphasis on mitigating oxidative stress and inflammation. The study involves daily intraperitoneal injections of amifostine or VALD-2 over 7 days to establish an AKI model. Subsequently, mice were assigned to normal control, cisplatin group, cisplatin + amifostine group, and cisplatin + VALD-2 10 mg/kg group, cisplatin + VALD-2 20 mg/kg, and cisplatin + VALD-2 40 mg/kg. Kidney injury is assessed through serum blood urea nitrogen (BUN) and creatinine (Cr) activity assays. Levels of inflammatory factors, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), in kidney tissue of mice were assessed through enzyme-linked immunosorbent assay (ELISA). The protective effect of VALD-2 is further examined through HE staining to observe pathological changes in kidney injury. The ultrastructural changes of renal cells and tubular epithelial cells were observed by electron microscopy under experimental conditions, indicating the effect of VALD-2 on reversing cisplatin-induced renal injury. The study delves into VALD-2's protective mechanisms against cisplatin-induced kidney injury by using western blot analysis to assess the expression levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA) in kidney tissues. VALD-2 demonstrates significant improvement in cisplatin-induced AKI, as evidenced by increased BUN and Cr levels. It effectively protects kidney tissue from oxidative damage, enhancing SOD and GSH-Px activities while reducing MDA levels. The study also reveals a decrease in TNF-α and IL-6 levels, supported by ELISA results, and histological findings confirm anti-nephrotoxic effects. Western blot analysis shows an upregulation of antioxidant enzymes (SOD, GSH-Px) and a reduction in MDA production. VALD-2 emerges as a promising mitigator of cisplatin-induced AKI, showcasing its ability to enhance oxidative stress-related protein expression. The findings suggest VALD-2 as a potential therapeutic agent for protecting against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Xuhui Zhao
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yuna Meng
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Chunyan Dang
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Li Xue
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jing Zhang
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Shuping Ma
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hongling Li
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
91
|
Wang L, Hao X, Li X, Li Q, Fang X. Effects of ginsenoside Rh2 on cisplatin-induced nephrotoxicity in renal tubular epithelial cells by inhibiting endoplasmic reticulum stress. J Biochem Mol Toxicol 2024; 38:e23768. [PMID: 39015062 DOI: 10.1002/jbt.23768] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 05/23/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Nephrotoxicity remains a major adverse reaction of the anticancer drug cisplatin (CDDP) chemotherapy, which is an important risk factor for chronic renal disease. Ginsenoside Rh2 from Panax ginseng has been shown to protect against CDDP-induced nephrotoxicity in vivo, but its pharmacological effect on renal tubular epithelial cells is not clearly understood. This study examined the molecular mechanisms underlying the nephroprotective effects of Rh2 on CDDP-induced HK-2 cells and acute kidney injury (AKI) mice. As a result of Rh2 treatment, CDDP-induced HK-2 cells showed increased cell viability and reduced lactate dehydrogenase release. Moreover, Rh2 ameliorated CDDP-induced mitochondrial membrane potential, increased antioxidant enzyme activities, and reduced pro-inflammatory cytokine expression to reduce damage. Rh2 inhibited apoptosis and enhanced the antioxidant capacity of HK-2 cells by reducing proteins associated with endoplasmic reticulum (ER) stress, as well as by attenuating tunicamycin-induced ER stress. In addition, treatment of CDDP-induced AKI mice with Rh2 substantially reduced blood urea nitrogen and serum creatinine levels, attenuated histological damage of kidney. Further, Rh2 also improved kidney function by inhibiting ER stress to support in vitro findings. These results consistently demonstrated that Rh2 protects renal tubular epithelial cells from CDDP-induced nephrotoxicity and apoptosis by restoring ER homeostasis, which might suggest a therapeutic potential and providing new insights into AKI alternative therapies.
Collapse
Affiliation(s)
- Lianping Wang
- School of Life Sciences, Jilin University, Changchun, China
- School of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Changchun, China
| | - Xiaogang Hao
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xiangxin Li
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Qingjie Li
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
92
|
Taguchi H, Sumi D, Himeno S, Fujishiro H. Ferroptosis is involved in cisplatin sensitivity of the S3 segment of immortalized proximal tubule cells. Toxicology 2024; 506:153840. [PMID: 38830481 DOI: 10.1016/j.tox.2024.153840] [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: 03/25/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Cisplatin (CDDP) is administered as an anticancer drug across a broad spectrum of cancer treatments, but it causes severe renal damage. Several studies have attempted to elucidate the cause of CDDP-induced renal injury, but the detailed mechanism remains unclear. We previously found that S3 cells are more sensitive to CDDP than S1 and S2 cells by using immortalized cells derived from S1, S2, and S3 segments of proximal tubules. In this study, we investigated the potential contribution of reactive oxygen species (ROS) to the sensitivity of S3 cells to CDDP. The results showed that S3 cells have high sensitivity to CDDP, paraquat (PQ) and three ROS substances. To examine the mechanisms underlying the sensitivity to ROS in S3 cells, we compared the cellular responses of CDDP- and PQ-exposed S3 cells. The results indicated that the levels of intracellular ROS and lipid peroxides were increased in S3 cells after CDDP and PQ exposure. The intracellular levels of antioxidant proteins such as thioredoxin, thioredoxin reductase 1 and glutathione peroxidase 4 were also increased by exposure to PQ, but these proteins were decreased by CDDP exposure in S3 cells. Furthermore, the levels of intracellular free Fe2+ were increased by CDDP exposure only in S3 cells but not S1 or S2 cells, and cytotoxicity by exposure to CDDP in S3 cells was suppressed by ferroptosis inhibitors. These results suggested that the induction of ferroptosis due to the ROS production through attenuation of the antioxidant system and elevated free Fe2+ is partly responsible for the sensitivity of S3 cells to CDDP.
Collapse
Affiliation(s)
- Hiroki Taguchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Daigo Sumi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Seiichiro Himeno
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| |
Collapse
|
93
|
Li J, Yao Y, Lei X, Bao J, An S, Hu H, Sha T, Huang Q, Li T, Zeng Z, Wang X, Cai S. SIRTUIN 5 ALLEVIATES EXCESSIVE MITOCHONDRIAL FISSION VIA DESUCCINYLATION OF ATPASE INHIBITORY FACTOR 1 IN SEPSIS-INDUCED ACUTE KIDNEY INJURY. Shock 2024; 62:235-244. [PMID: 38754030 DOI: 10.1097/shk.0000000000002392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Sepsis-induced acute kidney injury (SAKI) poses a significant clinical challenge with high morbidity and mortality. Excessive mitochondrial fission has been identified as the central pathogenesis of sepsis-associated organ damage, which is also implicated in the early stages of SAKI. Sirtuin 5 (SIRT5) has emerged as a central regulator of cellular mitochondrial function; however, its role in the regulation of sepsis-induced excessive mitochondrial fission in kidney and the underlying mechanism remains unclear. In this study, SAKI was modeled in mice through cecal ligation and puncture, and in human renal tubular epithelial (HK-2) cells stimulated with lipopolysaccharide (LPS), to mimic the cell SAKI model. Our findings revealed that septic mice with a SIRT5 knockout exhibited shortened survival times and elevated levels of renal injury compared to wild-type mice, suggesting the significant involvement of SIRT5 in SAKI pathophysiology. Additionally, we observed that SIRT5 depletion led to increased renal mitochondrial fission, while the use of a mitochondrial fission inhibitor (Mdivi-1) reversed the detrimental effects caused by SIRT5 depletion, emphasizing the pivotal role of SIRT5 in preventing excessive mitochondrial fission. In vitro experiments demonstrated that the overexpression of SIRT5 effectively mitigated the adverse effects of LPS on HK-2 cells viability and mitochondrial fission. Conversely, downregulation of SIRT5 decreased HK-2 cells viability and exacerbated LPS-induced mitochondrial fission. Mechanistically, the protective function of SIRT5 may be in part, ascribed to its desuccinylating action on ATPase inhibitory factor 1. In conclusion, this study provides novel insights into the underlying mechanisms of SAKI, suggesting the possibility of identifying future drug targets in terms of improved mitochondrial dynamics by SIRT5.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tao Li
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiang Wang
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
94
|
Qian Y, Zhao N, Wang M, Zou Z, Xie K. P2X7 receptor deficiency attenuates cisplatin-induced kidney injury via inhibiting NLRP3 inflammasome activation. Biochem Pharmacol 2024; 226:116369. [PMID: 38880358 DOI: 10.1016/j.bcp.2024.116369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Nephrotoxicity is a major constraint of cisplatin application in many solid tumors. Since the lack of preventive strategies, the necessity exists to identify critical molecular targets involved in cisplatin nephrotoxicity. The Purinergic ligand-gcotedion channel 7 receptor (P2X7R) is a ligand-gated ion channel that is predominantly implicated in inflammation and cell death. Our aim is to investigate the role P2X7R in cisplatin-induced acute and chronic kidney injury, as well as the underlying mechanism. In this study, we found that cisplatin can cause an increase in the expression of P2X7R in mouse kidney tissue, and P2X7R knockout can alleviate acute renal function damage caused by cisplatin, as well as the expression of kidney injury molecule 1 (KIM-1) and interleukin-18 (IL-18). Cisplatin can cause an increase in the expression of nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome in mouse kidney tissue. Compared with wild-type mice, P2X7R -/- mice showed decreased expression of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), cleaved Caspase-1, and cleaved IL-1β in kidney tissue after cisplatin administration, and the apoptosis of renal tubular epithelial cells were also decreased. In addition, we also found that NLRP3 knockout can improve cisplatin induced degeneration, detachment, and necrosis of renal tubular epithelial cells. Furthermore, P2X7R -/- mice also showed reduced renal fibrosis and better long-term renal prognosis. In conclusion, our study identified that P2X7R knockout can improve cisplatin induced acute renal injury and chronic renal fibrosis by inhibiting the activation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Yingying Qian
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ning Zhao
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ming Wang
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Kewei Xie
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Uremia Diagnosis and Treatment Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
95
|
Ahmed HA, Shaaban AA, Makled MN, Ibrahim TM. G protein-coupled estrogen receptor selective agonist, G1, improves the molecular and biochemical markers in a cisplatin mouse model of CKD. Chem Biol Interact 2024; 398:111065. [PMID: 38795875 DOI: 10.1016/j.cbi.2024.111065] [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: 02/02/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Multiple cycles of cisplatin result in a permanent loss of kidney function with severe and life-limited chronic kidney disease (CKD) after successful cisplatin therapy. Recently, studies have showed that the activation of G-protein coupled estrogen receptor (GPER) could protect against kidney disease. This study aimed to test the potential of the G1 compound, a GPER selective agonist, to prevent CKD development after cisplatin therapy. Male C57BL/6 mice were exposed to 2 cycles of 2.5 mg/kg cisplatin in a regimen miming clinical exposure (1 injection daily for 5 days, followed by a 16-day recovery period between cycles). G1 (50 or 100 μg/kg) was administered daily for 6 weeks. G1 dose-dependently improved kidney function biomarkers (serum creatinine, creatinine clearance, and protein excretion) and histopathological changes compared to the cisplatin-treated group. Collagen 3 expression was dose-dependently decreased in G1-treated groups that was parallel to the reduction of fibrosis in Masson's trichrome-stained sections. G1 administration also increased total antioxidant capacity (TAC) and nuclear factor erythroid 2-related factor 2 (Nrf2) and reduced the level of malondialdehyde and the proinflammatory cytokine, tumor necrosis factor-α. In addition, G1 downregulated the expression of inflammasome NLRP3 and nuclear factor kappa B p65 (NF-κB p65) in a dose-dependent manner. In conclusion, these data suggest that G1 could be a new therapeutic tool for CKD prevention post cisplatin therapy. These effects might be mediated through the activation of Nrf2 and the inhibition of NF-κB/NLRP3 signaling.
Collapse
MESH Headings
- Animals
- Cisplatin/pharmacology
- Male
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/metabolism
- Mice, Inbred C57BL
- Mice
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/chemically induced
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Disease Models, Animal
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Biomarkers/metabolism
- Receptors, Estrogen/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/agonists
- NF-kappa B/metabolism
- Oxidative Stress/drug effects
Collapse
Affiliation(s)
- Hala A Ahmed
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt
| | - Ahmed A Shaaban
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt; Faculty of Pharmacy, Jerash University, Jerash, 26150, Jordan
| | - Mirhan N Makled
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt.
| | - Tarek M Ibrahim
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt
| |
Collapse
|
96
|
Oliveira CA, Mercês ÉAB, Portela FS, Malheiro LFL, Silva HBL, De Benedictis LM, De Benedictis JM, Silva CCDE, Santos ACL, Rosa DP, Velozo HS, de Jesus Soares T, de Brito Amaral LS. An integrated view of cisplatin-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity: characteristics, common molecular mechanisms, and current clinical management. Clin Exp Nephrol 2024; 28:711-727. [PMID: 38678166 DOI: 10.1007/s10157-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin (CP) is a chemotherapy drug widely prescribed to treat various neoplasms. Although fundamental for the therapeutic action of the drug, its cytotoxic mechanisms trigger adverse effects in several tissues, such as the kidney, liver, and heart, which limit its clinical use. In this sense, studies point to an essential role of damage to nuclear and mitochondrial DNA associated with oxidative stress, inflammation, and apoptosis in the pathophysiology of tissue injuries. Due to the limitation of effective preventive and therapeutic measures against CP-induced toxicity, new strategies with potential cytoprotective effects have been studied. Therefore, this article is timely in reviewing the characteristics and main molecular mechanisms common to renal, hepatic, and cardiac toxicity previously described, in addition to addressing the main validated strategies for the current management of these adverse events in clinical practice. We also handle the main promising antioxidant substances recently presented in the literature to encourage the development of new research that consolidates their potential preventive and therapeutic effects against CP-induced cytotoxicity.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | | | | | | | | | | | | | - Helloisa Souza Velozo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
| |
Collapse
|
97
|
Takemura M, Ikemura K, Okuda M. Effect of Hydration with Bicarbonate Ringer's Solution on Cisplatin-Induced Acute Kidney Injury in Patients with Esophageal Cancer: A Retrospective Cohort Study. Oncology 2024; 103:22-29. [PMID: 39074453 DOI: 10.1159/000540637] [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: 02/05/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Cisplatin (CDDP) often causes acute kidney injury (AKI), and magnesium supplementation has been suggested to be important in preventing CDDP-induced AKI. Sodium bicarbonate Ringer's solution (BRS) is a crystalloid solution composed of various electrolytes, including Mg2+, and can be generally used to supplement missing extracellular fluid and correct metabolic acidosis; however, the clinical outcomes of hydration with BRS for CDDP-induced AKI remain unclear. In this study, we retrospectively compared the effects of BRS and normal saline for hydration in patients undergoing CDDP treatment. METHODS We analyzed the incidence rate of AKI (grade ≥ 1), the severity of AKI, the serum magnesium level, and the incidence rate of grade ≥ 3 hematological toxicities (leukopenia, neutropenia, anemia, or thrombocytopenia) following CDDP and fluorouracil (5-FU) administration in 131 in-patients who received CDDP and 5-FU for the first time to treat esophageal cancer. RESULTS Fifty-six patients (43%) received saline alone, while 75 patients (57%) received BRS for hydration. The incidence rate of AKI (grade ≥ 1) was significantly lower in the BRS group (11%) than that in the saline group (39%, p < 0.001). Moreover, severe AKI (grade ≥ 2) was significantly less common in the BRS group than in the saline group. Although the serum magnesium levels before CDDP administration were not significantly different between the two groups (p = 0.939), the serum magnesium levels on days 2-3 after CDDP administration in the BRS group were significantly higher than those in the saline group (p < 0.001). In contrast, there were no significant differences in the incidence rates of hematological toxicity between the two groups. Multivariate analysis revealed that BRS use was an independent factor that significantly contributed to AKI prevention (odds ratio = 0.061, p < 0.001). CONCLUSION Hydration with BRS could prevent CDDP-induced AKI in patients with esophageal cancer.
Collapse
Affiliation(s)
- Miho Takemura
- Department of Clinical Pharmacy Research and Education, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Kenji Ikemura
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
- Department of Hospital Pharmacy, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masahiro Okuda
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
- Department of Hospital Pharmacy, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
98
|
Pinard L, Adam JP, Chagnon M, Bollée G, Soulières D. Hypokalemia, hypomagnesemia, and hyponatremia are associated with acute kidney injury in patients treated with cisplatin. J Oncol Pharm Pract 2024:10781552241262248. [PMID: 39051634 DOI: 10.1177/10781552241262248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Cisplatin-associated acute kidney injury (C-AKI) is common. Predictive factors include age >60 years, hypertension, cisplatin dose, diabetes, and serum albumin < 3.5 g/L. The association between C-AKI and hypokalemia, hypomagnesemia or hyponatremia has not been well characterized. METHODS Data from a previous retrospective observational study was obtained. Patients were separated into three groups with similar cisplatin doses and schedules. Group A received cisplatin 60-100 mg/m2 every three weeks with laboratory assessments before treatment, group B received cisplatin 60-75 mg/m2 every three weeks with laboratory assessments before days 1 and 8 and group C had weekly cisplatin 40 mg/m2 with weekly laboratories assessments. The association between hypomagnesemia, hypokalemia, hyponatremia, and risk of AKI was determined using a counting process specification of Cox's regression models. RESULTS A total of 1301 patients were separated into groups A (n = 713), B (n = 204), and C (n = 384). The proportion of patients with at least one event of hypokalemia, hypomagnesemia, or hyponatremia was lower in group A (29.2%, 57.6%, 36.2%) compared to groups B (43.6%, 67.2%, 59.8%) and C (49.0%, 78.7%, 51.0%). The incidence of all grade C-AKI was 35.6% (group A), 46.6% (group B), and 18.2% (group C). In group A, the risk of AKI doubled with hyponatremia or hypomagnesemia and tripled with hypokalemia. This association was not seen with other groups. CONCLUSION Among patients with the highest doses of cisplatin, the presence of one electrolyte disorder was associated with an increased risk of C-AKI. Other studies are needed to characterize the presence of an electrolyte disorder as a predictive risk factor of C-AKI in this subpopulation.
Collapse
Affiliation(s)
- Louis Pinard
- Division of Nephrology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Jean-Philippe Adam
- Department of Pharmacy, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Miguel Chagnon
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | - Guillaume Bollée
- Division of Nephrology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Denis Soulières
- Axe cancer, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Division of Medical Oncology/Hematology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| |
Collapse
|
99
|
Yoon SY, Lee S, Lee K, Kim JS, Hwang HS, Kronbichler A, Jacob L, Shin JY, Lee JA, Park J, Lee H, Lee H, Jeong K, Yon DK. Global burden of anticancer drug-induced acute kidney injury and tubulointerstitial nephritis from 1967 to 2023. Sci Rep 2024; 14:16124. [PMID: 38997405 PMCID: PMC11245615 DOI: 10.1038/s41598-024-67020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
This study aims to figure out the worldwide prevalence of anticancer therapy-associated acute kidney injury (AKI) and tubulointerstitial nephritis (TIN) and the relative risk of each cancer drug. We conducted an analysis of VigiBase, the World Health Organization pharmacovigilance database, 1967-2023 via disproportionate Bayesian reporting method. We further categorized the anticancer drugs into four groups: cytotoxic therapy, hormone therapy, immunotherapy, and targeted therapy. Reporting odds ratio (ROR) and information component (IC) compares observed and expected values to investigate the associations of each category of anticancer drugs with AKI and TIN. We identified 32,722 and 2056 reports (male, n = 17,829 and 1,293) of anticancer therapy-associated AKI and TIN, respectively, among 4,592,036 reports of all-drug caused AKI and TIN. There has been a significant increase in reports since 2010, primarily due to increased reports of targeted therapy and immunotherapy. Immunotherapy exhibited a significant association with both AKI (ROR: 8.92; IC0.25: 3.06) and TIN (21.74; 4.24), followed by cytotoxic therapy (7.14; 2.68), targeted therapy (5.83; 2.40), and hormone therapy (2.59; 1.24) for AKI, and by cytotoxic therapy (2.60; 1.21) and targeted therapy (1.54; 0.61) for TIN. AKI and TIN were more prevalent among individuals under 45 years of age, with a female preponderance for AKI and males for TIN. These events were reported in close temporal relationship after initiation of the respective drug (16.53 days for AKI and 27.97 days for TIN), and exhibited a high fatality rate, with 23.6% for AKI and 16.3% for TIN. These findings underscore that kidney-related adverse drug reactions are of prognostic significance and strategies to mitigate such side effects are required to optimize anticancer therapy.
Collapse
Affiliation(s)
- Soo-Young Yoon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Sooji Lee
- Department of Medicine, Kyung Hee University College of Medicine, Seoul, South Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Kyeongmin Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Jin Sug Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hyeon Seok Hwang
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Andreas Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | - Louis Jacob
- Research and Development Unit, Parc Sanitari Sant Joan de Déu, CIBERSAM, ISCIII, Barcelona, Spain
- Department of Physical Medicine and Rehabilitation, Lariboisière-Fernand Widal Hospital, AP-HP, Université Paris Cité, Paris, France
- Epidemiology of Ageing and Neurodegenerative Diseases (EpiAgeing), Inserm U1153, Université Paris Cité, Paris, France
| | - Ju-Young Shin
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jin A Lee
- Department of Biomedical Engineering, Kyung Hee University, Yongin, Korea
| | - Jaeyu Park
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Hyeri Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Hayeon Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea.
- Department of Biomedical Engineering, Kyung Hee University, Yongin, Korea.
| | - Kyunghwan Jeong
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea.
| | - Dong Keon Yon
- Department of Medicine, Kyung Hee University College of Medicine, Seoul, South Korea.
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea.
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea.
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, 23 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
100
|
Jang KW, Kim YS, Kim MJ, Kim SR, Lee DW, Lee SB, Kim IY. Time-restricted feeding protects against cisplatin-induced acute kidney injury in mice. Kidney Res Clin Pract 2024; 43:444-456. [PMID: 38934035 PMCID: PMC11237335 DOI: 10.23876/j.krcp.23.351] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/22/2024] [Accepted: 02/04/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Time-restricted feeding (TRF), devoid of calorie restriction, is acknowledged for promoting metabolic health and mitigating various chronic metabolic diseases. While TRF exhibits widespread benefits across multiple tissues, there is limited exploration into its impact on kidney function. In this study, our aim was to investigate the potential ameliorative effects of TRF on kidney damage in a mouse model of cisplatin-induced acute kidney injury (AKI). METHODS Cisplatin-induced AKI was induced through intraperitoneal injection of cisplatin into C57BL/6 male mice. Mice undergoing TRF were provided unrestricted access to standard chow daily but were confined to an 8-hour feeding window during the dark cycle for 2 weeks before cisplatin injection. The mice were categorized into four groups: control, TRF, cisplatin, and TRF + cisplatin. RESULTS The tubular damage score and serum creatinine levels were significantly lower in the TRF + cisplatin group compared to the cisplatin group. The TRF + cisplatin group exhibited reduced expression of phosphorylated nuclear factor kappa B, inflammatory cytokines, and F4/80-positive macrophages compared to the cisplatin group. Furthermore, oxidative stress markers for DNA, protein, and lipid were markedly decreased in the TRF + cisplatin group compared to the cisplatin group. TUNEL-positive tubular cells, cleaved caspase-3 expression, and the Bax/Bcl-2 ratio in the TRF + cisplatin group were lower than those in the cisplatin group. CONCLUSION TRF, without calorie restriction, effectively mitigated kidney damage by suppressing inflammatory reactions, oxidative stress, and tubular apoptosis in a mouse model of cisplatin-induced AKI. TRF holds promise as a novel dietary intervention for preventing cisplatin-induced AKI.
Collapse
Affiliation(s)
- Kyu Won Jang
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Young Suk Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Min Jeong Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Seo Rin Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Dong Won Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Soo Bong Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Il Young Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|