1
|
Ujah GA, Ofutet EO, Ukam CIO, Omiunu PE, Ackley EU, Japhet IG, Ntauko JC, Clement QC, Atu R, Nna VU. Protective effect of tert-butylhydroquinone against cisplatin-induced hepatorenal injury via modulating oxidative stress, inflammation, and apoptosis. Arch Physiol Biochem 2024; 130:951-961. [PMID: 38993034 DOI: 10.1080/13813455.2024.2376812] [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: 03/27/2024] [Revised: 06/03/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
CONTEXT Cisplastin (CDDP) is a chemotherapeutic drug frequently used to manage a variety of cancers. However, its use is associated with hepatorenal toxicity resulting from elevated reactive oxygen species production. OBJECTIVE Herein, the hepatorenal protective effect of tert-butylhydroquinone (tBHQ) in cisplatin (CDDP)-treated rats was examined. METHODS Wistar male rats randomly divided into four groups: normal control, tBHQ, CDDP and tBHQ + CDDP received 50 mg/kg b.w./day of tBHQ orally for 14 days while 7 mg/kg b.w of CDDP was administered intraperitoneally on Day 8. RESULTS CDDP increased serum biomarkers of hepatic (AST, ALP, ALT, GGT) and renal (creatinine, urea, uric acid, kidney injury molecule 1) function. The levels of nuclear factor erythroid-2-related factor 2 protein and the activities of superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase activities were decreased in liver and kidney. Also, CDDP increased hepatic and renal levels of NF-κB, TNFα, Bax and caspase-3 proteins and decreased hepatorenal levels of Bcl-2 protein in the liver and kidney. Pre-treatment with tBHQ prevented these negative effects. SIGNIFICANCE Pre-intervention with tBHQ attenuates hepatorenal toxicity of CDDP by dampening oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Godwin Adakole Ujah
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Emmanuel Oleba Ofutet
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
- Department of Physiology, Faculty of Medicine and Pharmaceutical Science, Kampala International University, Tanzania
| | - Catherine Ironya-Ogar Ukam
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, ICGEB, Trieste, Italy
| | - Precious Evangeline Omiunu
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Emaediong Ufot Ackley
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Iboro Godwin Japhet
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Jane Charles Ntauko
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Queen Comfort Clement
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Racheal Atu
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Victor Udo Nna
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| |
Collapse
|
2
|
Kara O. Protective effect of coenzyme Q10 in cyclophosphamide-induced kidney damage in rats. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20230990. [PMID: 38716935 PMCID: PMC11068380 DOI: 10.1590/1806-9282.20230990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/22/2023] [Indexed: 05/12/2024]
Abstract
OBJECTIVE We aimed to investigate the effect of coenzyme q10 on cyclophosphamide-induced kidney damage in rats. METHODS A total of 30 female Wistar-Albino rats were utilized to form three groups. In group 1 (control group) (n=10), no drugs were given. In group 2 (cyclophosphamide group) (n=10), 30 mg/kg intraperitoneal cyclophosphamide was administered for 7 days. In group 3 (cyclophosphamide+coenzyme q10 group) (n=10), 30 mg/kg cyclophosphamide and 10 mg/kg coenzyme q10 were given for 7 days via intraperitoneal route. Right kidneys were removed in all groups. Blood malondialdehyde levels and activities of catalase and superoxide dismutase were measured. Histopathological damage was evaluated by examining the slides prepared from kidney tissue using a light microscope. RESULTS Tissue damage was significantly higher in the cyclophosphamide group than in the cyclophosphamide+coenzyme q10 group (p<0.05). The malondialdehyde levels were significantly higher and the activities of superoxide dismutase and catalase were lower in the cyclophosphamide group than in the cyclophosphamide+coenzyme q10 group (p<0.05). CONCLUSION Coenzyme q10 may be a good option to prevent cyclophosphamide-induced kidney damage.
Collapse
Affiliation(s)
- Ozlem Kara
- Kirsehir Ahi Evran University, School of Medicine, Department of Histology and Embryology – Kırşehir, Turkey
| |
Collapse
|
3
|
Yao J, Chen Y, Zhang L, Cheng Y, Chen Z, Zhang Y, Zheng X, Lv Y, Wang S, Li Z, Zhao J. pH-responsive CuS/DSF/EL/PVP nanoplatform alleviates inflammatory bowel disease in mice via regulating gut immunity and microbiota. Acta Biomater 2024; 178:265-286. [PMID: 38417643 DOI: 10.1016/j.actbio.2024.02.034] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
The clinical treatment of inflammatory bowel disease (IBD) is challenging. We developed copper sulfate (CuS)/disulfiram (DSF)/methacrylic acid-ethyl acrylate copolymer (EL)/polyvinylpyrrolidone (PVP) nanoplatform (CuS/DSF/EL/PVP) and evaluated its efficiency for treating IBD. After oral administration, the pH-sensitive EL protected the CuS/DSF/EL/PVP against degradation by acidic gastric juices. Once the colon was reached, EL was dissolved, releasing DSF and Cu2+. Further, the main in vivo metabolite of DSF can bind to Cu2+ and form copper (II) N, N-diethyldithiocarbamate (CuET), which significantly alleviated acute colitis in mice. Notably, CuS/DSF/EL/PVP outperformed CuS/EL/PVP and DSF/EL/PVP nanoplatforms in reducing colonic pathology and improving the secretion of inflammation-related cytokines (such as IL-4 and IL-10) in the colonic mucosa. RNA-seq analysis revealed that the nanoplatform reduced colonic inflammation and promoted intestinal mucosal repair by upregulating C-type lectin receptor (CLR)-related genes and signaling pathways. Furthermore, CuS/DSF/EL/PVP showed potential for improving colitis Th1/Th17 cells through innate immunity stimulation, down-regulation of inflammatory cytokines, and upregulation of anti-inflammatory cytokines. Additionally, the intervention with CuS/DSF/EL/PVP led to increased intestinal flora diversity, decreased Escherichia-Shigella abundance, and elevated levels of short-chain fatty acid (SCFA)-producing bacteria Prevotella, Lactobacillus, and Bifidobacterium, indicating their potential to modulate the dysregulated intestinal flora and suppress inflammation. STATEMENT OF SIGNIFICANCE: Our study introduces the CuS/DSF/EL/PVP nanoplatform as a therapeutic strategy for treating inflammatory bowel disease (IBD). This approach demonstrates significant efficacy in targeting the colon and alleviating acute colitis in mice. It uniquely modulates gut immunity and microbiota, exhibiting a notable impact on inflammation-related cytokines and promoting intestinal mucosal repair. The nanoplatform's ability to regulate gut flora diversity, combined with its cost-effective and scalable production, positions it as a potentially transformative treatment for IBD, offering new avenues for personalized medical interventions.
Collapse
Affiliation(s)
- Jinpeng Yao
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yu Chen
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China
| | - Liang Zhang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yuancun Cheng
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China
| | - Zheng Chen
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China
| | - Yanhui Zhang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Xiaoyi Zheng
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yanwei Lv
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Shige Wang
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China.
| | - Zhaoshen Li
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China.
| | - Jiulong Zhao
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China.
| |
Collapse
|
4
|
Jiayi H, Ziyuan T, Tianhua X, Mingyu Z, Yutong M, Jingyu W, Hongli Z, Li S. Copper homeostasis in chronic kidney disease and its crosstalk with ferroptosis. Pharmacol Res 2024; 202:107139. [PMID: 38484857 DOI: 10.1016/j.phrs.2024.107139] [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: 01/15/2024] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024]
Abstract
Chronic kidney disease (CKD) has become a global public health problem with high morbidity and mortality. Renal fibrosis can lead to end-stage renal disease (ESRD). However, there is still no effective treatment to prevent or delay the progression of CKD into ESRD. Therefore, exploring the pathogenesis of CKD is essential for preventing and treating CKD. There are a variety of trace elements in the human body that interact with each other within a complex regulatory network. Iron and copper are both vital trace elements in the body. They are critical for maintaining bodily functions, and the dysregulation of their metabolism can cause many diseases, including kidney disease. Ferroptosis is a new form of cell death characterized by iron accumulation and lipid peroxidation. Studies have shown that ferroptosis is closely related to kidney disease. However, the role of abnormal copper metabolism in kidney disease and its relationship with ferroptosis remains unclear. Here, our current knowledge regarding copper metabolism, its regulatory mechanism, and the role of abnormal copper metabolism in kidney diseases is summarized. In addition, we discuss the relationship between abnormal copper metabolism and ferroptosis to explore the possible pathogenesis and provide a potential therapeutic target for CKD.
Collapse
Affiliation(s)
- Huang Jiayi
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Tong Ziyuan
- China Medical University, Shenyang 110122, People's Republic of China
| | - Xu Tianhua
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Zhang Mingyu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Ma Yutong
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Wang Jingyu
- Renal Division, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Zhou Hongli
- Department of Nephrology, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 110004, People's Republic of China
| | - Sun Li
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
5
|
Ewees MGED, Orfali R, Rateb EE, Hassan HM, Hozzein WN, Alkhalfah DHM, Sree HTA, Abdel Rahman FEZS, Rateb ME, Mahmoud NI. Modulation of mi-RNA25/Ox-LDL/NOX4 signaling pathway by polyphenolic compound Hydroxytyrosol as a new avenue to alleviate cisplatin-induced acute kidney injury, a mechanistic study in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 103:104262. [PMID: 37699441 DOI: 10.1016/j.etap.2023.104262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/16/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023]
Abstract
Acute kidney injury (AKI) caused by Cis is considered one of the most severe adverse effects, which restricts its use and efficacy. This study seeks to examine the potential reno-protective impact of phenolic compound Hydroxytyrosol (HT) against Cis-induced AKI and the possible involvement of the mi-RNA25/Ox-LDL/NOX4 pathway elucidating the probable implicated molecular mechanisms. Forty rats were placed into 5 groups. Group I received saline only. Group II received Cis only. Group III, IV, and V received 20, 50, and 100 mg/kg b.w, of HT, respectively, with Cis delivery. NOX4, Ox-LDL, and gene expression of mi-RNA 25, TNF-α, and HO-1 in renal tissue were detected. HT showed reno-protective effect and significantly upregulated mi-RNA 25 and HO-1 as well as decreased the expression of NOX4, Ox-LDL, and TNF-α. In conclusion, HT may be promising in the fight against Cis-induced AKI through modulation of mi-RNA25/Ox-LDL/NOX4 pathway.
Collapse
Affiliation(s)
- Mohamed Gamal El-Din Ewees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt.
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia.
| | - Enas Ezzat Rateb
- Department of Physiology, Faculty of Medicine, Beni-Suef University, Beni-Suef 62521, Egypt.
| | - Hossam M Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt.
| | - Wael N Hozzein
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| | - Dalal Hussien M Alkhalfah
- Department of Biology. College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia.
| | - Haidy Tamer Abo Sree
- Department of Basic Sciences Department, Biochemistry, Faculty of Oral and Dental Medicine, Nahda University, Beni-Suef 11787, Egypt.
| | - Fatema El-Zahraa S Abdel Rahman
- Department of Basic Sciences Department, Physiology, Faculty of Oral and Dental Medicine, Nahda University, Beni-Suef 11787, Egypt.
| | - Mostafa E Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley PA1 2BE, UK.
| | - Nesreen Ishak Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt.
| |
Collapse
|
6
|
Su S, Wei Z, Huang H, Yoshizawa T, Inui T, Funahashi M. Conditioned nausea induced by cisplatin and emetine identified by a taste reactivity test in rats. Physiol Behav 2023:114278. [PMID: 37352906 DOI: 10.1016/j.physbeh.2023.114278] [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: 04/05/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
No prior studies have shown that gaping reactions are produced with the avoidance of conditioned taste caused by cisplatin and emetine. Therefore, we tried to demonstrate it using a taste reactivity test in rats and found the gaping reactions induced when saccharin is readministered after gustatory conditioning that paired saccharin with cisplatin or emetine. Since conditioned gaping reactions indicate the aversion to saccharin taste and conditioned nausea, the present study suggest that the taste aversion is induced by cisplatin and emetine. It was also found that with intraperitoneal injections of emetine alone, gaping almost never occurs.
Collapse
Affiliation(s)
- Shaoyi Su
- Oral Physiology, Department of Oral Functional Science, Division of Oral Medical Science, Faculty of Dental Medicine and Graduate school of Dental Medicine, Hokkaido University
| | - Zimo Wei
- Oral Physiology, Department of Oral Functional Science, Division of Oral Medical Science, Faculty of Dental Medicine and Graduate school of Dental Medicine, Hokkaido University
| | - Helai Huang
- Oral Physiology, Department of Oral Functional Science, Division of Oral Medical Science, Faculty of Dental Medicine and Graduate school of Dental Medicine, Hokkaido University
| | - Tomohiko Yoshizawa
- Oral Physiology, Department of Oral Functional Science, Division of Oral Medical Science, Faculty of Dental Medicine and Graduate school of Dental Medicine, Hokkaido University
| | - Tadashi Inui
- Oral Physiology, Department of Oral Functional Science, Division of Oral Medical Science, Faculty of Dental Medicine and Graduate school of Dental Medicine, Hokkaido University
| | - Makoto Funahashi
- Oral Physiology, Department of Oral Functional Science, Division of Oral Medical Science, Faculty of Dental Medicine and Graduate school of Dental Medicine, Hokkaido University.
| |
Collapse
|
7
|
Ahmad S, Pandey AR, Rai AK, Singh SP, Kumar P, Singh S, Gulzar F, Ahmad I, Sashidhara KV, Tamrakar AK. Moringa oleifera impedes protein glycation and exerts reno-protective effects in streptozotocin-induced diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116117. [PMID: 36584917 DOI: 10.1016/j.jep.2022.116117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/26/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Moringa oleifera is a valued plant with wide distribution in tropical and subtropical regions of the world. It is traditionally used for the treatment of fever, infections, rheumatism, cancer, improving cardiac, renal and hepatic functions, and regulating blood glucose level. The plant has been scientifically reported for the anti-inflammatory, antioxidant, renoprotective, and anti-diabetic properties. Diabetic patients are prone to develop end-stage renal diseases due to incidence of diabetes-induced renal dysfunctions. Given that, increased production and accumulation of advanced glycation end-products (AGEs) play a conspicuous role in the development of diabetes-linked renal dysfunctions, nature-based interventions with AGEs inhibitory activity can prevent renal dysfunctions leading to renoprotection. AIM OF THE STUDY The study aimed to demonstrate the preventive effects of the ethanolic extract of the leaves of Moringa oleifera (EEMO) on protein glycation and its further assessment for the renoprotective effect in diabetic rats. MATERIALS AND METHODS Antiglycation activity of EEMO was assessed in vitro using bovine serum albumin. For reno-protective activity assessment, streptozotocin (STZ)-induced diabetic rats were orally treated with EEMO (100 mg/kg) or standard antiglycation agent aminoguanidine (100 mg/kg) for consecutive 8 weeks. The effects on glucose homeostasis, renal functions, and renal morphology were assessed by clinical biochemistry, molecular and histological examination. RESULTS Presence of EEMO efficiently prevented glucose-, fructose- or methylglyoxal-mediated glycation of protein. Under in vivo set-up, compared to diabetic control rats, EEMO treatment effectively improved the glucose tolerance and body weight, and reduced the serum levels of triglycerides and total cholesterol. Additionally, EEMO administration significantly ameliorated renal dysfunctions in diabetic rats characterized by improved levels of creatinine, urea nitrogen, and uric acid in serum, and total protein level in urine, accompanied by improved kidney morphology. The diabetes-associated pro-inflammatory response characterized by upregulated expression of the inducible nitric oxide synthase (iNos), activation of nuclear factor kappa B (NF-κB) and the raised levels of inflammatory factors, interleukin-1 beta (IL-1β) and interleukin-6 (IL-6) in renal tissue was significantly attenuated in EEMO-treated rats. Moreover, EEMO treatment diminished renal reactive oxygen species (ROS) levels in diabetic animals. CONCLUSIONS Our study demonstrated that EEMO prevented AGEs formation and ameliorated renal dysfunctions in diabetic rats by blocking inflammatory/oxidative pathways. Our observations justify M. oleifera as a potential source of therapeutic interventions for diabetic nephropathy management.
Collapse
Affiliation(s)
- Shadab Ahmad
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Alka Raj Pandey
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Amit K Rai
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Suriya P Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Pawan Kumar
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Sushmita Singh
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Farah Gulzar
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Ishbal Ahmad
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India
| | - Akhilesh K Tamrakar
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, UP, India.
| |
Collapse
|
8
|
Memudu AE, Dongo GA. A study to demonstrate the potential of Anabolic Androgen Steroid to activate oxidative tissue damage, nephrotoxicity and decline endogenous antioxidant system in renal tissue of Adult Wistar Rats. Toxicol Rep 2023; 10:320-326. [PMID: 36911166 PMCID: PMC9996376 DOI: 10.1016/j.toxrep.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/30/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Anabolic Androgenic steroids (AAS) are abused and reports have been made on their deleterious effects on various organs. It is imperative to report the mechanism of inducing oxidative tissue damage even in the presence of an intracellular antioxidant system by the interaction between lipid peroxidation and the antioxidant system in the kidney. Twenty (20) adult male Wistar rats used were grouped into: A- Control, BOlive oil vehicle, C- 120 mg/kg of AAS orally for three weeks, and D- 7 days withdrawal group following 120 mg/kg/ 21days of AAS intake. Serum was assayed for lipid peroxidation marker Malondialdehyde (MDA) and antioxidant enzyme -superoxide Dismutase (SOD). Sectioned of kidneys were stained to see the renal tissue, mucin granules, and basement membrane. AAS-induced oxidative tissue damage, in the presence of an endogenous antioxidant, is characterized by increased lipid peroxidation and decreased SOD level which resulted in the loss of renal tissue cells membrane integrity which is a characteristic of the pathophysiology of nephron toxicity induced by a toxic compound. However, this was progressively reversed by a period of discontinuation of AAS drug exposure.
Collapse
Affiliation(s)
- Adejoke Elizabeth Memudu
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Medical Sciences, Edo State University Uzairue, Edo State Nigeria
- Corresponding author.
| | - Gambo A. Dongo
- Department of Anatomy, Faculty of Basic Medical Sciences, Bingham University Karu Nasarawa State Nigeria
| |
Collapse
|
9
|
Lin H, Wu D, Xiao J. Identification of key cuproptosis-related genes and their targets in patients with IgAN. BMC Nephrol 2022; 23:354. [PMID: 36329405 PMCID: PMC9635123 DOI: 10.1186/s12882-022-02991-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND IgA nephropathy (IgAN) is one of the most common forms of chronic glomerulonephritis, but the aetiology and pathogenesis remain unclear. Cuproptosis is a newly identified form of cell death that plays an important role in many diseases. Researchers have not clearly determined whether the expression of cuproptosis-related genes (CRGs) is involved in the pathogenesis of IgAN. METHODS The GSE93798, GSE50469 and GSE37460 datasets containing microarray data from patients with IgAN (63) and healthy controls (31) were downloaded from the GEO database. Immune cells and immune-related functions were analysed in patients with IgAN and controls, and genes were identified that may be related to cuproptosis. A logistic regression model was established according to the results, and then GO and KEGG enrichment analyses were performed. Finally, possible drugs were selected using the DSigDB database. RESULTS The subjects in the different groups showed significantly different fractions of immune cells and immune-related functions, and 11 genes related to cuproptosis may be involved in these processes. Based on these 11 genes, the ROC curve was plotted, and the AUC value was calculated (0.898, 95% CI: 0.839-0.958). The result revealed good predictability. Then, genes with P < 0.05 (lipoyltransferase 1, LIPT1) were selected to plot an ROC curve, and the AUC value was calculated (0.729, 95% CI: 0.636-0.821). Enrichment analyses showed that the TCA cycle and multiple metabolic pathways may also be involved in the occurrence of IgAN. Finally, 293 potential drugs that may be used to treat IgAN were identified based on these genes. CONCLUSION In this study, we identified some novel CRGs that may be involved in IgAN, among which LIPT1 was significantly differentially expressed. It may predict the risk of IgAN and provides a possible target for the treatment of IgAN. Further experimental studies are needed to explore how these CRGs mediate the occurrence and development of IgAN.
Collapse
Affiliation(s)
- Huagang Lin
- grid.413597.d0000 0004 1757 8802Department of Nephrology, Huadong Hospital Affiliated to Fudan University, 200040 Shanghai, P.R. China ,grid.8547.e0000 0001 0125 2443Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, P.R. China
| | - Deping Wu
- grid.413597.d0000 0004 1757 8802Department of Nephrology, Huadong Hospital Affiliated to Fudan University, 200040 Shanghai, P.R. China ,grid.8547.e0000 0001 0125 2443Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, P.R. China
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, 200040, Shanghai, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, P.R. China.
| |
Collapse
|
10
|
Chi F, Zhang G, Ren N, Zhang J, Du F, Zheng X, Zhang C, Lin Z, Li R, Shi X, Zhu Y. The anti-alcoholism drug disulfiram effectively ameliorates ulcerative colitis through suppressing oxidative stresses-associated pyroptotic cell death and cellular inflammation in colonic cells. Int Immunopharmacol 2022; 111:109117. [PMID: 35969897 DOI: 10.1016/j.intimp.2022.109117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/06/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Oxidative stress, cell pyroptosis and inflammation are considered as important pathogenic factors for ulcerative colitis (UC) development, and the traditional anti-alcoholism drug disulfiram (DSF) has recently been reported to exert its regulating effects on all the above cellular functions, which makes DSF as ideal therapeutic agent for UC treatment, but this issue has not been fully studied. METHODS Dextran sulfate sodium (DSS)-induced animal models in C57BL/6J mice and lipopolysaccharide (LPS)-induced cellular models in colonic cell lines (HT-29 and Caco-2) for UC were respectively established. Cytokine secretion was determined by ELISA. Cell viability and proliferation were evaluated by MTT assay and EdU assay. Real-Time qPCR, Western Blot, immunofluorescent staining assay and immunohistochemistry (IHC) were employed to evaluate gene expressions. The correlations of the genes in the clinical tissues were analyzed by using the Pearson Correlation analysis. RESULTS DSF restrained oxidative stress, pyroptotic cell death and cellular inflammation in UC models in vitro and in vivo, and elimination of Reactive Oxygen Species (ROS) by N-acetyl-l-cysteine (NAC) rescued cell viability in LPS-treated colonic cells (HT-29 and Caco-2). Further experiments suggested that a glycogen synthase kinase-3β (GSK-3β)/Nrf2/NLRP3 signaling cascade played critical role in this process. Mechanistically, DSF downregulated GSK-3β and NLRP3, whereas upregulated Nrf2 in LPS-treated colonic cells. Also, the regulating effects of DSF on Nrf2 and NLRP3 were abrogated by upregulating GSK-3β. Moreover, upregulation of GSK-3β abolished the protective effects of DSF on LPS-treated colonic cells. CONCLUSIONS Taken together, data of this study indicated that DSF restrained oxidative damages-related pyroptotic cell death and inflammation via regulating the GSK-3β/Nrf2/NLRP3 pathway, leading to the suppression of LPS-induced UC development.
Collapse
Affiliation(s)
- Fengxu Chi
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Qunli Seventh Street No. 2075, Daoli District, Harbin 150001, Heilongjiang, China
| | - Guangquan Zhang
- Department of Hepatobiliary Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shennan Middle Road No. 3025, Shenzhen 518033, China.
| | - Niansheng Ren
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Qunli Seventh Street No. 2075, Daoli District, Harbin 150001, Heilongjiang, China.
| | - Jian Zhang
- Department of Tumor Laparoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Road No. 23, Nangang District, Harbin 150001, Heilongjiang, China.
| | - Fei Du
- Department of Hepatobiliary Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shennan Middle Road No. 3025, Shenzhen 518033, China
| | - Xiyan Zheng
- Department of Hepatobiliary Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shennan Middle Road No. 3025, Shenzhen 518033, China
| | - Cong Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Qunli Seventh Street No. 2075, Daoli District, Harbin 150001, Heilongjiang, China
| | - Zhiqun Lin
- Department of Hepatobiliary Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shennan Middle Road No. 3025, Shenzhen 518033, China
| | - Ruixi Li
- Department of Hepatobiliary Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shennan Middle Road No. 3025, Shenzhen 518033, China.
| | - Xianjie Shi
- Department of Hepatobiliary Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shennan Middle Road No. 3025, Shenzhen 518033, China.
| | - Yuekun Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Qunli Seventh Street No. 2075, Daoli District, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
11
|
Balzer MS, Doke T, Yang YW, Aldridge DL, Hu H, Mai H, Mukhi D, Ma Z, Shrestha R, Palmer MB, Hunter CA, Susztak K. Single-cell analysis highlights differences in druggable pathways underlying adaptive or fibrotic kidney regeneration. Nat Commun 2022; 13:4018. [PMID: 35821371 PMCID: PMC9276703 DOI: 10.1038/s41467-022-31772-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/01/2022] [Indexed: 01/14/2023] Open
Abstract
The kidney has tremendous capacity to repair after acute injury, however, pathways guiding adaptive and fibrotic repair are poorly understood. We developed a model of adaptive and fibrotic kidney regeneration by titrating ischemic injury dose. We performed detailed biochemical and histological analysis and profiled transcriptomic changes at bulk and single-cell level (> 110,000 cells) over time. Our analysis highlights kidney proximal tubule cells as key susceptible cells to injury. Adaptive proximal tubule repair correlated with fatty acid oxidation and oxidative phosphorylation. We identify a specific maladaptive/profibrotic proximal tubule cluster after long ischemia, which expresses proinflammatory and profibrotic cytokines and myeloid cell chemotactic factors. Druggability analysis highlights pyroptosis/ferroptosis as vulnerable pathways in these profibrotic cells. Pharmacological targeting of pyroptosis/ferroptosis in vivo pushed cells towards adaptive repair and ameliorates fibrosis. In summary, our single-cell analysis defines key differences in adaptive and fibrotic repair and identifies druggable pathways for pharmacological intervention to prevent kidney fibrosis.
Collapse
Affiliation(s)
- Michael S Balzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ya-Wen Yang
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel L Aldridge
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hailong Hu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hung Mai
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ziyuan Ma
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rojesh Shrestha
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew B Palmer
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
12
|
Potential Protective Effects of Antioxidants against Cyclophosphamide-Induced Nephrotoxicity. Int J Nephrol 2022; 2022:5096825. [PMID: 35469319 PMCID: PMC9034963 DOI: 10.1155/2022/5096825] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/11/2022] [Accepted: 03/24/2022] [Indexed: 11/18/2022] Open
Abstract
Cyclophosphamide is an alkylating antineoplastic agent, and it is one of the most successful drugs with wide arrays of clinical activity. It has been in use for several types of cancer treatments and as an immunosuppressive agent for the management of autoimmune and immune-mediated diseases. Nowadays, its clinical use is limited due to various toxicities, including nephrotoxicity. Even though the mechanisms are not well understood, cyclophosphamide-induced nephrotoxicity is reported to be mediated through oxidative stress. This review focuses on the potential role of natural and plant-derived antioxidants in preventing cyclophosphamide-induced nephrotoxicity.
Collapse
|
13
|
Sun F, Wang H, Nie J, Hong B. Repurposing disulfiram as a chemo-therapeutic sensitizer: molecular targets and mechanisms. Anticancer Agents Med Chem 2022; 22:2920-2926. [PMID: 35430981 DOI: 10.2174/1871520621666220415102553] [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: 10/14/2021] [Revised: 12/20/2021] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Currently, chemo-therapy is still the main strategy for cancer treatment. However, chemo-therapy resistance remains its main challenge. Disulfiram [DSF] is a drug approved by FDA for the treatment of alcohol addiction, but it is later discovered that it has the anticancer activity. Importantly, there have been many literatures reporting that DSF can be used as a chemo-therapeutic sensitizer to enhance the anticancer activity of chemo-drugs in a variety of cancers. Furthermore, the combinations of DSF and chemo-drugs have been tested in clinic trials. In the review, we summarized the possible molecular targets and mechanisms of DSF to reverse chemo-resistance. We also further discussed the opportunities and challenges of DSF as a chemo-therapeutic sensitizer. In conclusion, DSF could be a potential repurposed drug to sensitize cancer cells to chemo-therapy in clinic.
Collapse
Affiliation(s)
- Feilong Sun
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Hefei, Anhui, China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jinfu Nie
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Bo Hong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| |
Collapse
|
14
|
Guo W, Chen S, Li C, Xu J, Wang L. Application of Disulfiram and its Metabolites in Treatment of Inflammatory Disorders. Front Pharmacol 2022; 12:795078. [PMID: 35185542 PMCID: PMC8848744 DOI: 10.3389/fphar.2021.795078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 12/27/2022] Open
Abstract
Disulfiram has been used clinically for decades as an anti-alcoholic drug. Recently, several studies have demonstrated the anti-inflammatory effects of disulfiram and its metabolism, which can alleviate the progression of inflammation in vivo and in vitro. In the current study, we summarize the anti-inflammatory mechanisms of disulfiram and its metabolism, including inhibition of pyroptosis by either covalently modifying gasdermin D or inactivating nod-like receptor protein 3 inflammasome, dual effects of intracellular reactive oxygen species production, and inhibition of angiogenesis. Furthermore, we review the potential application of disulfiram and its metabolism in treatment of inflammatory disorders, such as inflammatory bowel disease, inflammatory injury of kidney and liver, type 2 diabetes mellitus, sepsis, uveitis, and osteoarthritis.
Collapse
Affiliation(s)
- Wenyi Guo
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shihong Chen
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chengqing Li
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianwei Xu
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Wang
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Shandong University, China
| |
Collapse
|
15
|
Liu Y, Zhang W, Nan JB, Lei KX, Zou HL. Two Cu(II)-Based Coordination Polymers: Magnetic Properties and Treatment Activity on the Restless Leg Syndrome. J Inorg Organomet Polym Mater 2022. [DOI: 10.1007/s10904-021-02092-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
16
|
L-carnitine ameliorates bile duct ligation induced liver fibrosis via reducing the nitrosative stress in experimental animals: preclinical evidences. Heliyon 2021; 7:e08488. [PMID: 34901512 PMCID: PMC8642613 DOI: 10.1016/j.heliyon.2021.e08488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/23/2021] [Accepted: 11/24/2021] [Indexed: 11/22/2022] Open
Abstract
Bile duct ligation (BDL) has been extensively used in studying the mechanisms of fibrogenesis and anti-fibrotic drugs. Considering the liver regenerative capacity and the diverse results from BDL, the present study aimed to evaluate the protective effect of L-carnitine on bile duct ligation-induced liver fibrosis in experimental rats. Rats were randomly divided into seven groups (n = 6). The bile duct was ligated and serum aspartate transaminase (AST), alanine transaminase (ALT), total bilirubin and albumin, hepatic hydroxyproline (HP), reduced glutathione (GSH), and malondialdehyde (MDA) and cytokines were measured. iNOS expression was measured by using Western blot and finally, liver tissue was processed for histopathological analysis (H&E staining)". The level of iNOS was increased in the control group, whereas a decrease in the level of iNOS was found in the L-carnitine treated group. In the present study, we found that bile duct ligation in rats showed an increase in body and liver weight, while treatment with carnitine showed normal body and liver weight. Serum AST, ALT, total bilirubin, HP, GSH, MDA, and cytokines were increased in bile duct ligated rats. In addition, L-carnitine treated rats showed a reduction in oxidative stress as well as inhibiting the release of cytokines in a dose-dependent manner and showed protection against bile duct ligation. The study concludes that L-carnitine has a protective effect against the liver fibrosis induced by bile duct ligation.
Collapse
|
17
|
Regression Modeling of the Antioxidant-to-Nephroprotective Relation Shows the Pivotal Role of Oxidative Stress in Cisplatin Nephrotoxicity. Antioxidants (Basel) 2021; 10:antiox10091355. [PMID: 34572987 PMCID: PMC8464812 DOI: 10.3390/antiox10091355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
The clinical utility of the chemotherapeutic drug cisplatin is significantly limited by its nephrotoxicity, which is characterized by electrolytic disorders, glomerular filtration rate decline, and azotemia. These alterations are consequences of a primary tubulopathy causing injury to proximal and distal epithelial cells, and thus tubular dysfunction. Oxidative stress plays a role in cisplatin nephrotoxicity and cytotoxicity, but its relative contribution to overall toxicity remains unknown. We studied the relation between the degree of oxidative reduction (provided by antioxidant treatment) and the extent of nephrotoxicity amelioration (i.e., nephroprotection) by means of a regression analysis of studies in animal models. Our results indicate that a linear relation exists between these two parameters, and that this relation very nearly crosses the value of maximal nephroprotection at maximal antioxidant effect, suggesting that oxidative stress seems to be a pivotal and mandatory mechanism of cisplatin nephrotoxicity, and, hence, an interesting, rationale-based target for clinical use. Our model also serves to identify antioxidants with enhanced effectiveness by comparing their actual nephroprotective power with that predicted by their antioxidant effect. Among those, this study identified nanoceria, erythropoietin, and maltol as highly effective candidates affording more nephroprotection than expected from their antioxidant effect for prospective clinical development.
Collapse
|
18
|
Zhou S, Zhou Y, Yu J, Jiang L, Xiang Y, Wang J, Du Y, Cui X, Ge F. A neutral polysaccharide from Ophiocordyceps lanpingensis restrains cisplatin-induced nephrotoxicity. Food Sci Nutr 2021; 9:3602-3616. [PMID: 34262721 PMCID: PMC8269674 DOI: 10.1002/fsn3.2317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 04/18/2021] [Indexed: 12/11/2022] Open
Abstract
Ophiocordyceps lanpingensis is an edible mushroom distributed over the south-eastern part of the Tibet Plateau, which is also recognized as an effective ethnomedicine to alleviate diseases. This study explored the effects of a kind of Ophiocordyceps lanpingensis neutral polysaccharide (ONP) on RAW264.7 macrophages and cisplatin-induced nephrotoxicity. The results showed that ONP relieved the inflammatory response of RAW264.7 macrophages by increasing the expression level of anti-inflammatory factor IL-10. Furthermore, ONP treatment significantly prolonged the survival of the mice treated by cisplatin through decelerating pathological progress and alleviating damaged functions of the kidneys. Compared with the cisplatin group, ONP reduced the oxidative stress of the renal cells and the expression levels of pro-inflammatory factors. Apoptosis of renal cells was also weakened in the ONP treatment group. These findings indicated that ONP alleviated cisplatin nephrotoxicity mainly by inhibiting oxidative stress, inflammation, and apoptosis in the kidneys, underscoring the potential of ONP supplementation to alleviate the side effects of cisplatin chemotherapy.
Collapse
Affiliation(s)
- Shubo Zhou
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Yongchun Zhou
- Yunnan Cancer Center Molecular Diagnostics CenterYunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Jiaji Yu
- Department of Microbiology, Immunology & Molecular GeneticsUniversity of CaliforniaLos AngelesCAUSA
| | - Li Jiang
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Yingying Xiang
- Department of StomatologyYan’an Hospital Affiliated to Kunming Medical UniversityKunmingChina
| | - Juan Wang
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Yaxi Du
- Yunnan Cancer Center Molecular Diagnostics CenterYunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Xiuming Cui
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| | - Feng Ge
- Yunnan Provincial Key Laboratory of Panax notoginsengFaculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingChina
| |
Collapse
|
19
|
Xie HQ, Li YB, Tian QC, Fan QL. Two Cu(II)-bearing coordination polymers: magnetic properties and application values on vaginitis by balancing the microbial ecology in the vagina. JOURNAL OF POLYMER RESEARCH 2021. [DOI: 10.1007/s10965-021-02556-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
Sarkar AA, Allyn DM, Delay RJ, Delay ER. Cyclophosphamide-Induced Inflammation of Taste Buds and Cytoprotection by Amifostine. Chem Senses 2021; 46:bjab031. [PMID: 34161570 PMCID: PMC8345827 DOI: 10.1093/chemse/bjab031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Taste buds in the oral cavity have a complex immune system regulating normal functions and inflammatory reactions. Cyclophosphamide (CYP), a chemotherapy drug, has wide-ranging disruptive effects on the taste system including loss of taste function, taste sensory cells, and capacity for taste cell renewal. In bladder epithelium, CYP also induces inflammation. To determine if CYP induces inflammation in taste buds, we used immunohistochemistry to examine tumor necrosis factor alpha (TNF-α) (a proinflammatory cytokine) expression over a 72-hour period. Expression of TNF-α increased in a subset of PLCβ2 labeled (Type II) cells, but not SNAP-25 labeled (Type III) cells, between 8 and 24 h postinjection and declined slowly thereafter. This inflammatory response may play an important role in the disruptive effects of CYP on the taste system. Further, pretreatment with amifostine, a sulfhydryl drug known to protect normal tissues during chemo- or radiation therapy, reduced the amount of CYP-induced TNF-α expression in taste buds, suggesting this drug is capable of protecting normal cells of the taste system from adverse effects of CYP. Amifostine, used as a pretreatment to CYP and possibly other chemotherapy drugs, may offer clinical support for preventing negative side effects of chemotherapy on the taste system.
Collapse
Affiliation(s)
- Anish A Sarkar
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| | - David M Allyn
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
- Program of Biotechnology, School of Engineering and Applied Sciences, University of Pennsylvania, 107 Towne Building, 220 South 33rd Street, Philadelphia, PA 19104-6391, USA
| | - Rona J Delay
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| | - Eugene R Delay
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| |
Collapse
|
21
|
Frączek K, Kowalczyk A, Pekala M, Kasarello K, Sygitowicz G, Sulejczak D, Zaremba M, Konop M, Frankowska M, Filip M, Bujalska-Zadrozny M, Kleczkowska P. The Positive and Negative Outcome of Morphine and Disulfiram Subacute Co-Administration in Rats in the Absence of Ethanol Challenge. Pharmaceutics 2020; 13:pharmaceutics13010029. [PMID: 33375279 PMCID: PMC7823937 DOI: 10.3390/pharmaceutics13010029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/13/2020] [Accepted: 12/22/2020] [Indexed: 01/21/2023] Open
Abstract
Recently, a well-known anti-alcohol agent, disulfiram (DSF), has gain much interest, as it was found to be effective in the treatment of cocaine abusers, thus also giving hope for patients addicted to opioids and other illicit drugs. Therefore, this study was aimed to investigate the possible outcome that might occur within the subacute co-administration of both morphine (MRF) and DSF in rats, but in the absence of ethanol challenge. As observed, intraperitoneal DSF dose-dependently enhanced MRF-mediated analgesia with the maximal efficacy at a dose of 100 mg/kg. Furthermore, MRF-induced tolerance and aggressive behavior were significantly reduced by DSF (100 mg/kg, i.p.) in comparison to MRF solely. Nonetheless, significant blood biochemical markers of hepatotoxicity were found (i.e., alteration in the levels of glutathione, blood urea nitrogen, etc.), following a combination of both drugs. Likewise, histological analysis of liver tissue revealed severe changes in the group of DSF + MRF, which includes swelling, cell death, damage to certain vessels, and hemorrhages into the liver parenchyma. Our findings indicate that DSF should be used with extreme caution, especially within the course of subacute concomitant use with MRF, as several possible side effects may take place.
Collapse
Affiliation(s)
- Karolina Frączek
- Centre for Preclinical Research (CBP), Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (K.F.); (A.K.); (M.P.); (M.B.-Z.)
| | - Agnieszka Kowalczyk
- Centre for Preclinical Research (CBP), Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (K.F.); (A.K.); (M.P.); (M.B.-Z.)
| | - Martyna Pekala
- Centre for Preclinical Research (CBP), Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (K.F.); (A.K.); (M.P.); (M.B.-Z.)
| | - Kaja Kasarello
- Centre for Preclinical Research (CBP), Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
| | - Grażyna Sygitowicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland;
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland;
| | - Malgorzata Zaremba
- Centre for Preclinical Research (CBP), Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
| | - Marek Konop
- Centre for Preclinical Research (CBP), Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
| | - Malgorzata Frankowska
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (M.F.); (M.F.)
| | - Malgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (M.F.); (M.F.)
| | - Magdalena Bujalska-Zadrozny
- Centre for Preclinical Research (CBP), Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (K.F.); (A.K.); (M.P.); (M.B.-Z.)
| | - Patrycja Kleczkowska
- Centre for Preclinical Research (CBP), Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; (K.F.); (A.K.); (M.P.); (M.B.-Z.)
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-1166127
| |
Collapse
|
22
|
Casanova AG, Hernández-Sánchez MT, Martínez-Salgado C, Morales AI, Vicente-Vicente L, López-Hernández FJ. A meta-analysis of preclinical studies using antioxidants for the prevention of cisplatin nephrotoxicity: implications for clinical application. Crit Rev Toxicol 2020; 50:780-800. [PMID: 33170047 DOI: 10.1080/10408444.2020.1837070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cisplatin is an effective chemotherapeutic drug whose clinical use and efficacy are limited by its nephrotoxicity, which affects mainly the renal tubules and vasculature. It accumulates in proximal and distal epithelial tubule cells and causes oxidative stress-mediated cell death and malfunction. Consequently, many antioxidants have been tested for their capacity to prevent cisplatin nephrotoxicity. In this study, we made a systematic review of the literature and meta-analyzed 152 articles, which tested the nephroprotective effect of isolated compounds or mixtures of natural origin on cisplatin nephrotoxicity in preclinical models. This meta-analysis identified the most effective candidates and examined the efficacy obtained by antioxidants administered by the oral and intraperitoneal routes. By comparing with a recent, similar meta-analysis performed on clinical studies, this article identifies a disconnection between preclinical and clinical research, and contextualizes, discusses, and integrates the existing preclinical information toward the optimized selection of candidates to be further explored (clinical level). Despite proved efficacy, this article discusses the barriers limiting the clinical development of natural mixtures, such as those in extracts from Calendula officinalis flowers and Heliotropium eichwaldii roots. On the contrary, isolated compounds are more straightforward candidates, among which arjunolic acid and quercetin stand out in this meta-analysis.
Collapse
Affiliation(s)
- Alfredo G Casanova
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - M Teresa Hernández-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martínez-Salgado
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I Morales
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| | - Laura Vicente-Vicente
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J López-Hernández
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| |
Collapse
|
23
|
Jain PG, Nayse PG, Patil DJ, Shinde SD, Surana SJ. The possible antioxidant capabilities of formononetin in guarding against streptozotocin-induced diabetic nephropathy in rats. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00071-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Abstract
Background
Oxidative stress has been considered as a contributory aspect for major complications of diabetes mellitus consisting of diabetic nephropathy. This study aimed to examine the therapeutic effect of formononetin in streptozotocin (STZ)-induced diabetic nephropathy through measuring biochemical parameters, oxidative indicators, and histopathological examination of renal tissues.
Results
Administration of a dose of STZ (55 mg/kg of body weight) intraperitoneal induced diabetic nephropathy in rats as indicated by an increase in serum glucose, creatinine, triglyceride, cholesterol, and BUN levels related to the depletion of serum albumin level. Besides, STZ treatment led to the depletion of antioxidant enzymes together with superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT). Administration of formononetin at the dose of 10, 20, and 40 mg/kg extensively decreased biochemical parameters with a rise in serum albumin level. Formononetin was observed to improved antioxidant enzyme ranges and offered protection against lipid peroxidation (LPO). STZ administered rats show an elevated level of TNF-α and IL-6. Meanwhile, formononetin-treated rats inhibited the elevated level of cytokine.
Conclusion
This study concluded that formononetin may additionally modulate oxidative stress and protected renal tissues from STZ injury. It also showed improvement in renal histopathological architecture in STZ-induced diabetic nephropathy.
Collapse
|
24
|
Taghizadeh F, Hosseinimehr SJ, Zargari M, Karimpour Malekshah A, Talebpour Amiri FB. Gliclazide attenuates cisplatin-induced nephrotoxicity through inhibiting NF-κB and caspase-3 activity. IUBMB Life 2020; 72:2024-2033. [PMID: 32687680 DOI: 10.1002/iub.2342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022]
Abstract
Cisplatin (CP), as a chemotherapeutic drug, causes nephrotoxicity that has limited the clinical utility of CP. Gliclazide (GLZ), as an antihyperglycemic drug, at low dose has antioxidant property. In this study, we aimed to investigate the protective effect of GLZ against CP-induced acute renal injury. Sixty-four BALB/c mice were randomly divided into eight groups. The groups were included as control, GLZ (5, 10, and 25 mg/kg), CP, and GLZ (5, 10, and 25 mg/kg) + CP. Renal function markers (serum creatinine and blood urea nitrogen), oxidative stress markers (malondialdehyde and glutathione), apoptotic marker (caspase-3), and NF-κB were histopathologically evaluated. The results of our study showed that increased urea and creatinine were evidence of CP-induced nephrotoxicity. Histopathological examination revealed tubular epithelial and Bowman degeneration, edema, and cytoplasmic vacuolation in renal tissue structure. Administration of GLZ reduced oxidative stress, caspase-3, and NF-κB activity, and improved kidney function markers in CP-treated mice compared with CP alone group. Also, we observed that the histological tissue structure of the kidney was maintained. GLZ at dose of 25 mg/kg had higher protective effect as compared with other doses. Overall, our study suggests that GLZ with antioxidant, antiapoptotic, and anti-inflammatory properties may be a promising new therapeutic agent to prevent CP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Fatemeh Taghizadeh
- Department of Anatomy, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Department of Biochemistry, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | | |
Collapse
|
25
|
Lu W, Shi J, Nie Y, Yang L, Chen J, Zhao F, Yang S, Xu L, Chi X. Synthesis, crystal structure, antiproliferative activity, DNA binding and density functional theory calculations of 3‐(pyridin‐2‐yl)‐8‐
tert
‐butylcoumarin and its copper(II) complex. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Wen Lu
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Jiuzhou Shi
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - YingFang Nie
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Lu Yang
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Jichao Chen
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Fengyi Zhao
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Shilong Yang
- Advanced Analysis and Testing Center Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Li Xu
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
- Co‐Innovation Center for Sustainable Forestry in Southern China Nanjing Forestry University Nanjing Jiangsu 210037 China
| | - Xingwei Chi
- College of Science Nanjing Forestry University Nanjing Jiangsu 210037 China
| |
Collapse
|
26
|
Jangra A, Choi SA, Yang J, Koh EJ, Phi JH, Lee JY, Wang KC, Kim SK. Disulfiram potentiates the anticancer effect of cisplatin in atypical teratoid/rhabdoid tumors (AT/RT). Cancer Lett 2020; 486:38-45. [PMID: 32428661 DOI: 10.1016/j.canlet.2020.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/16/2020] [Accepted: 05/05/2020] [Indexed: 01/04/2023]
Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is the most malignant tumor of the central nervous system that generally occurs in young children. Despite the use of intensive multimodal therapy for AT/RT, the prognosis is still poor. The brain tumor initiating cells in AT/RT cells has been suggested as one of the challenges in AT/RT treatment. These cells have high expression of aldehyde dehydrogenase (ALDH). We investigated the combination effect of the ALDH inhibitor, disulfiram and cisplatin in the treatment of AT/RT cells. Isobologram analysis revealed that the combination therapy synergistically increases AT/RT cell death. The enzyme activity of ALDH AT/RT cells was effectively reduced by the combination therapy. We proposed that the synergistic augmentation occurs, at least partially through an increase in cleaved Poly (ADP-ribose) polymerase (PARP)-dependent apoptosis mediated by activating transcription factor 3 (ATF3). In the AT/RT mouse model, the combination therapy decreased tumor volume and prolonged survival. Immunofluorescence assay in mouse brain tissues were consistent with the expression of ATF3 and cleaved PARP. Our study demonstrates enhanced anti-cancer effect of combination therapy of disulfiram and cisplatin. This combination might provide a viable therapeutic strategy for AT/RT patients.
Collapse
Affiliation(s)
- Anshika Jangra
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeyul Yang
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Eun Jung Koh
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Regional Emergency Medical Center, Seoul National University Hospital, Seoul, South Korea
| | - Ji Hoon Phi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Department of Anatomy, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyu-Chang Wang
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, South Korea; Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
27
|
Li H, Wang J, Wu C, Wang L, Chen ZS, Cui W. The combination of disulfiram and copper for cancer treatment. Drug Discov Today 2020; 25:1099-1108. [PMID: 32320854 DOI: 10.1016/j.drudis.2020.04.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022]
Abstract
Disulfiram (DSF) is a thiuram derivative that was developed to treat alcoholism but was also found to have antitumor activity. Copper (Cu), as a trace metal, has important roles in the body. Numerous studies have shown that the combination of DSF and copper (DSF/Cu) greatly enhances its antitumor efficacy. Given that the efficacy of DSF is well established and its safety profile is understood, repurposing DSF as a new anticancer drug is a promising strategy. Here, we summarize the pharmacological effects of DSF and the role of Cu in cancer, and focus on the antitumor effect of DSF/Cu, especially the mechanisms involved in enhancing drug sensibility by targeting specific molecules. We also provide rational strategies for using DSF as a cancer therapy.
Collapse
Affiliation(s)
- Hong Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Jingyu Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, NY 11439, USA.
| | - Wei Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China.
| |
Collapse
|
28
|
Rocha-Pereira C, Silva V, Costa VM, Silva R, Garcia J, Gonçalves-Monteiro S, Duarte-Araújo M, Santos-Silva A, Coimbra S, Dinis-Oliveira RJ, Lopes C, Silva P, Long S, Sousa E, de Lourdes Bastos M, Remião F. Histological and toxicological evaluation, in rat, of a P-glycoprotein inducer and activator: 1-(propan-2-ylamino)-4-propoxy-9 H-thioxanthen-9-one (TX5). EXCLI JOURNAL 2019; 18:697-722. [PMID: 31611753 PMCID: PMC6785774 DOI: 10.17179/excli2019-1675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022]
Abstract
P-glycoprotein (P-gp) is an ATP-binding cassette transporter involved in the efflux of numerous compounds that influences the pharmacokinetics of xenobiotics. It reduces intestinal absorption and exposure of target cells to toxicity. Thioxanthones are compounds able to induce and/or activate P-gp in vitro. Particularly, 1-(propan-2-ylamino)-4-propoxy-9H-thioxanthen-9-one (TX5) behaves as a P-gp inducer and activator in vitro. The aims of this study were: i) to perform a histological characterization, by testing a single high dose of TX5 [30 mg/kg, body weight (b.w.), gavage], administered to Wistar Han rats, 24 hours after administration; and ii) to perform both a complete histological characterization and a preliminary safety evaluation, in distinct target organs, 24 hours after administration of a single lower dose of TX5 (10 mg/kg, b.w., gavage) to Wistar Han rats. The results showed a relevant histological toxicity for the higher dose of TX5 administered (30 mg/kg, b.w.), manifested by extensive hepatic necrosis and splenic toxicity (parenchyma with hyperemia, increased volume of both white and red pulp, increased follicles marginal zone). Moreover, in the kidneys, a slight hyperemia and tubular edema were observed in TX5-treated animals, as well as an inflammation of the small intestine. On the contrary, for the lower tested dose (10 mg/kg, b.w.), we did not observe any relevant histological toxicity in the evaluated organs. Additionally, no significant differences were found in the ATP levels between TX5-exposed and control animals in any of the evaluated organs, with the exception of the intestine, where ATP levels were significantly higher in TX5-treated rats. Similarly, TX5 caused a significant increase in the ratio GSH/GSSG only in the lungs. TX5 (10 mg/kg, b.w.) did not induce any change in any of the hematological and biochemical circulating evaluated parameters. However, TX5 was able to significantly reduce the activated partial thromboplastin time, without affecting the prothrombin time. The urine biochemical analysis revealed a TX5-mediated increase in both creatinine and sodium. Taken together, our results show that TX5, at a dose of 10 mg/kg, does not induce considerable toxicity in the biological matrices studied. Given this adequate safety profile, TX5 becomes a particularly interesting compound for ex vivo and in vivo studies, regarding the potential for induction and activation of P-gp at the intestinal barrier.
Collapse
Affiliation(s)
- Carolina Rocha-Pereira
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Vera Silva
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Vera Marisa Costa
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Juliana Garcia
- CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Department of Agronomy, University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal
| | - Salomé Gonçalves-Monteiro
- LAQV/REQUIMTE, Laboratório de Farmacologia, Departamento de Ciências do Medicamento, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Alice Santos-Silva
- UCIBIO/REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Susana Coimbra
- UCIBIO/REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.,Instituto de Investigação e Formação Avançada em Ciências e Tecnologias Saúde (IINFACTS), Departamento de Ciências, Instituto Universitário de Ciências da Saúde (IUCS-CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.,Instituto de Investigação e Formação Avançada em Ciências e Tecnologias Saúde (IINFACTS), Departamento de Ciências, Instituto Universitário de Ciências da Saúde (IUCS-CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal.,Departamento de Saúde Pública e Ciências Forenses e Educação Médica, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Catarina Lopes
- Molecular Oncology and Viral Pathology Group, Centro de Investigação do IPO-Porto
| | - Paula Silva
- Departamento de Microscopia, Laboratório de Histologia e Embriologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Solida Long
- CIIMAR, Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR, Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fernando Remião
- UCIBIO/REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
29
|
Abidi A, Kourda N, Feki M, Ben Khamsa S. Protective Effect of Tunisian Flaxseed Oil against Bleomycin-Induced Pulmonary Fibrosis in Rats. Nutr Cancer 2019; 72:226-238. [PMID: 31155950 DOI: 10.1080/01635581.2019.1622741] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/17/2019] [Accepted: 05/19/2019] [Indexed: 12/17/2022]
Abstract
The aims of this study are to investigate the preventive effect of flaxseed oil (FO) on bleomycin (BLM)-induced pulmonary fibrosis (PF). Thirty adult male Wistar rats (180-220 g) were randomly divided into three groups. The control group (G1) received no treatment, the group (G2) received only intratracheally BLM, and the group (G3) received FO (2 mL/kg body weight) once a day for 60 days + BLM (4 mg/kg body weight "bw"). Our results demonstrated that FO protected against BLM-induced PF, by increasing proline, fructose, glucose, glyceride, choline, lactate, and malate metabolites in bronchoalveolar lavage fluid (Balf) which are involved in anti-inflammatory reactions. Also, FO-treatment reduced the score of fibrosis and the inflammatory index and revealed a decrease in tumor growth factor beta (TGFβ) density in alveoli, inflammatory infiltrate and fibrocytes, comparatively to the BLM group. As well, our data demonstrated that acute BLM-induced fibrosis was accompanied by an oxidative stress in lung tissue as assessed by an increase of lipid peroxidation as well as antioxidant enzyme activities depletion such as superoxide dismutase (SOD) and catalase (CAT). The FO treatment reversed all disturbances of BLM-induced oxidative stress parameters, and increased fatty acids levels promoting anti-inflammatory reactions especially in erythrocytes (linoleic, α-linolenic, docosapentaenoic acids).
Collapse
Affiliation(s)
- Anouar Abidi
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Nadia Kourda
- Department of Anatomy and Pathology, Charles Nicole Hospital, Tunis, Tunisia
| | - Moncef Feki
- Departement of Biochemistry, Hospital La Rabta Tunis, Tunisia
| | - Saloua Ben Khamsa
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|