1
|
Yu D, Zhang R, Zhou J, Guo P, Li P, Ye M, Liu Y, Shi S. Pharmacokinetics and safety of pirfenidone in individuals with chronic kidney disease stage G2 and G3a: A single-dose, Phase I, bridging study. J Pharm Sci 2025; 114:1087-1094. [PMID: 39694267 DOI: 10.1016/j.xphs.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND AND OBJECTIVE Pirfenidone is an inhibitor of transforming growth factor-beta 1 (TGF-β1) and is being developed for the treatment of diabetic kidney disease (DKD). We assessed the pharmacokinetics (PK) and safety of a single dose of pirfenidone in individuals with CKD stages G2/G3a. METHODS In this phase I bridging study, patients with CKD stages G2 or G3a, aged 18-70 years, with a body mass index of 18-26 kg/m2, and glomerular filtration rate (eGFR) ranging from 45 to 89 ml/min/1.73 m2, received a single oral dose of 400 mg pirfenidone capsules 30 min after a standard breakfast. The pharmacokinetic parameters of the two groups were measured and compared after blood and urine collection. The co-primary endpoints were the area under the plasma concentration-time curve from time zero to 36 h (AUC0-36) and the maximum observed plasma concentration (Cmax) of pirfenidone. Safety was a secondary endpoint. The trial has been registered on ClinicalTrials.gov (ChiCTR2300077297). RESULTS A total of 20 subjects participated in this study. There were no significant differences between the control group and the patient group (CKD stages G2/G3a) in terms of plasma Cmax, the time to reach the maximum observed concentration (Tmax), and elimination half-life(t1/2). However, the Vz/F of the patient group (CKD G2 stage) was significantly higher than that of the control group. Renal accumulation rate, renal clearance rate (CLr), and urine drug concentration also showed no significant differences. No severe adverse events occurred during the trial. CONCLUSIONS These results indicate that the PK and safety of pirfenidone are not influenced by renal function. Individuals with renal impairment may not require dose adjustments.
Collapse
Affiliation(s)
- Dianwen Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Rui Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Jinping Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Pengpeng Guo
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Peixia Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Menghan Ye
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Yani Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| | - Shaojun Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| |
Collapse
|
2
|
Abdulaal WH, Omar UM, Zeyadi M, El-Agamy DS, Alhakamy NA, Ibrahim SRM, Almalki NAR, Asfour HZ, Al-Rabia MW, Mohamed GA, Elshal M. Pirfenidone ameliorates ANIT-induced cholestatic liver injury via modulation of FXR, NF-кB/TNF-α, and Wnt/GSK-3β/β-catenin signaling pathways. Toxicol Appl Pharmacol 2024; 490:117038. [PMID: 39019095 DOI: 10.1016/j.taap.2024.117038] [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: 04/14/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Cholestasis is a hepatobiliary disorder characterized by the excessive accumulation of toxic bile acids in hepatocytes, leading to cholestatic liver injury (CLI) through multiple pathogenic inflammatory pathways. Currently, there are limited therapeutic options for the management of cholestasis and associated CLI; therefore, new options are urgently needed. Pirfenidone (PF), an oral bioavailable pyridone analog, is used for the treatment of idiopathic pulmonary fibrosis. PF has recently demonstrated diverse potential therapeutic activities against different pathologies. Accordingly, the present study adopted the α-naphthyl isothiocyanate (ANIT)-induced CLI model in mice to explore the potential protective impact of PF and investigate the underlying mechanisms of action. PF intervention markedly reduced the serum levels of ALT, AST, LDH, total bilirubin, and total bile acids, which was accompanied by a remarkable amelioration of histopathological lesions induced by ANIT. PF also protected the mice against ANIT-induced redox imbalance in the liver, represented by reduced MDA levels and elevated GSH and SOD activities. Mechanistically, PF inhibited ANIT-induced downregulated expressions of the farnesoid X receptor (FXR), as well as the bile salt export pump (BSEP) and the multidrug resistance-associated protein 2 (MRP2) bile acid efflux channels. PF further repressed ANIT-induced NF-κB activation and TNF-α and IL-6 production. These beneficial effects were associated with its ability to dose-dependently inhibit Wnt/GSK-3β/β-catenin/cyclin D1 signaling. Collectively, PF protects against ANIT-induced CLI in mice, demonstrating powerful antioxidant and anti-inflammatory activities as well as an ability to oppose BA homeostasis disorder. These protective effects are primarily mediated by modulating the interplay between FXR, NF-κB/TNF-α/IL-6, and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Wesam H Abdulaal
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ulfat M Omar
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Princess Dr. Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mustafa Zeyadi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Dina S El-Agamy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Nabil A Alhakamy
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Sabrin R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah 21442, Saudi Arabia.
| | - Naif A R Almalki
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Experimental Biochemistry Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Hani Z Asfour
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mohammed W Al-Rabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Mahmoud Elshal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
3
|
Torre A, Martínez‐Sánchez FD, Narvaez‐Chávez SM, Herrera‐Islas MA, Aguilar‐Salinas CA, Córdova‐Gallardo J. Pirfenidone use in fibrotic diseases: What do we know so far? Immun Inflamm Dis 2024; 12:e1335. [PMID: 38967367 PMCID: PMC11225083 DOI: 10.1002/iid3.1335] [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: 02/04/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Pirfenidone has demonstrated significant anti-inflammatory and antifibrotic effects in both animal models and some clinical trials. Its potential for antifibrotic activity positions it as a promising candidate for the treatment of various fibrotic diseases. Pirfenidone exerts several pleiotropic and anti-inflammatory effects through different molecular pathways, attenuating multiple inflammatory processes, including the secretion of pro-inflammatory cytokines, apoptosis, and fibroblast activation. OBJECTIVE To present the current evidence of pirfenidone's effects on several fibrotic diseases, with a focus on its potential as a therapeutic option for managing chronic fibrotic conditions. FINDINGS Pirfenidone has been extensively studied for idiopathic pulmonary fibrosis, showing a favorable impact and forming part of the current treatment regimen for this disease. Additionally, pirfenidone appears to have beneficial effects on similar fibrotic diseases such as interstitial lung disease, myocardial fibrosis, glomerulopathies, aberrant skin scarring, chronic liver disease, and other fibrotic disorders. CONCLUSION Given the increasing incidence of chronic fibrotic conditions, pirfenidone emerges as a potential therapeutic option for these patients. However, further clinical trials are necessary to confirm its therapeutic efficacy in various fibrotic diseases. This review aims to highlight the current evidence of pirfenidone's effects in multiple fibrotic conditions.
Collapse
Affiliation(s)
- Aldo Torre
- Metabolic UnitInstituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubiran”Mexico CityMexico
| | - Froylan David Martínez‐Sánchez
- Facultad de MedicinaUniversidad Nacional Autonoma de MexicoMexico CityMexico
- Department of Internal MedicineHospital General “Dr. Manuel Gea González”Mexico CityMexico
| | | | | | | | - Jacqueline Córdova‐Gallardo
- Facultad de MedicinaUniversidad Nacional Autonoma de MexicoMexico CityMexico
- Department of HepatologyHospital General “Dr. Manuel Gea González”Mexico CityMexico
| |
Collapse
|
4
|
Frąk W, Dąbek B, Balcerczyk-Lis M, Motor J, Radzioch E, Młynarska E, Rysz J, Franczyk B. Role of Uremic Toxins, Oxidative Stress, and Renal Fibrosis in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:687. [PMID: 38929126 PMCID: PMC11200916 DOI: 10.3390/antiox13060687] [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: 04/14/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
Affecting millions of people worldwide, chronic kidney disease is a serious medical problem. It results in a decrease in glomerular filtration rate below 60 mL/min/1.73 m, albuminuria, abnormalities in urine sediment and pathologies detected by imaging studies lasting a minimum of 3 months. Patients with CKD develop uremia, and as a result of the accumulation of uremic toxins in the body, patients can be expected to suffer from a number of medical consequences such as progression of CKD with renal fibrosis, development of atherosclerosis or increased incidence of cardiovascular events. Another key element in the pathogenesis of CKD is oxidative stress, resulting from an imbalance between the production of antioxidants and the production of reactive oxygen species. Oxidative stress contributes to damage to cellular proteins, lipids and DNA and increases inflammation, perpetuating kidney dysfunction. Additionally, renal fibrogenesis involving the accumulation of fibrous tissue in the kidneys occurs. In our review, we also included examples of forms of therapy for CKD. To improve the condition of CKD patients, pharmacotherapy can be used, as described in our review. Among the drugs that improve the prognosis of patients with CKD, we can include: GLP-1 analogues, SGLT2 inhibitors, Finerenone monoclonal antibody-Canakinumab and Sacubitril/Valsartan.
Collapse
Affiliation(s)
- Weronika Frąk
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Marta Balcerczyk-Lis
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Motor
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
5
|
Manawy SM, Faruk EM, Hindawy RF, Hassan MM, Farrag DMG, Bashar MAE, Fouad H, Bagabir RA, Hassan DAA, Zaazaa AM, Hablas MGA, Kamal KM. Modulation of the Sirtuin-1 signaling pathway in doxorubicin-induced nephrotoxicity (synergistic amelioration by resveratrol and pirfenidone). Tissue Cell 2024; 87:102330. [PMID: 38412579 DOI: 10.1016/j.tice.2024.102330] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024]
Abstract
The current study was conducted to determine the precise mechanisms of Sirtuin-1 (Sirt-1), TGF- β (Transforming Growth Factor-β), and long non-coding RNA Metastasis Associated Lung Adenocarcinoma Transcript 1 (LncRNA MALAT-1) in signaling pathways in doxorubicin (DOX)-induced nephrotoxicity. The potential therapeutic effect of Resveratrol and Pirfenidone in DOX toxicity was also assessed. Thirty-six male adult rats were evenly distributed into four groups: Group 1: control rats. Group 2: DOX exposed rats' group, each animal received 7.5 mg/kg DOX as a single intravenous dose, Group 3: DOX exposed group subjected to oral resveratrol (20 mg/kg/daily for two weeks), Group 4: DOX exposed group subjected to oral Pirfenidone (200 mg/kg once daily for 10 days). At the planned time, animals were sacrificed. Renal tissue was collected to assess matrix metalloproteinase-9 (MMP9), inflammatory and apoptotic markers: tumor necrosis factor-alpha (TNF- β, caspase-3, cyclo-oxygenase-2 (COX-2), and oxidative stress markers: nitric oxide (NO), Glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD). Sirtuin-1 (Sirt-1), TGF-β, and LncRNA MALAT-1 were quantitatively assessed by real-time RT-PCR in the whole blood. Results showed that the DOX group exhibited a significant increase in oxidative stress markers, and inflammatory, and apoptotic markers in the renal tissue. Histologically, the renal tubule lining cells exhibited vacuolar alterations in the cytoplasm, glomerular atrophy, and vascular congestion. Furthermore, renal degeneration was evident, as confirmed by the heightened immuno-expression of MMP9. Exposure to DOX resulted in a significant decrease in Sirtuin-1 (Sirt-1) with a significant increase in the TGFβ, and LncRNA MALAT-1 gene expression. However, pre-treatment with either resveratrol/or Pirefenidone ameliorated the histological renal alterations, regulated the pathways of Sirt-1, TGFβ, and LncRNA MALAT-1, and decreased all oxidative stress, inflammatory and apoptotic markers. In conclusion, DOX exposure leads to renal toxicity by inducing renal degeneration, oxidative stress, and apoptosis. Administration of either resveratrol or Pirfenidone counteracted these changes and protected the kidney against DOX-induced renal damage.
Collapse
Affiliation(s)
- Samia Mahmoud Manawy
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Eman Mohamed Faruk
- Anatomy Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia; Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Rabab Fawzy Hindawy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Mahmoud M Hassan
- Department of Physiology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Diaa M G Farrag
- Marine Biology Branch, Zoology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt.
| | - Mansour A E Bashar
- Marine Biology Branch, Zoology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt.
| | - Hanan Fouad
- Basic Medical Sciences, Faculty of Medicine, Galala University, Galala City, POB 43711, ATTAKA, Suez Governorate, Egypt; Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo POB 12613, Egypt.
| | - Rania Abubaker Bagabir
- College of Medicine, Hematology and Immunology Department, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Ahmed Mohammed Zaazaa
- Students at Faculty of Medicine, Benha National University, Benha Colleges in Cairo, Egypt
| | | | - K Mostafa Kamal
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
6
|
Asker ME, Abdelmeged AA, Shaheen MA, Eissa RG. Sunitinib displays pulmonary fibrosis in experimental rats: Role of IL-17A dependent pathway. Int Immunopharmacol 2023; 119:110173. [PMID: 37058748 DOI: 10.1016/j.intimp.2023.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/28/2023] [Accepted: 04/07/2023] [Indexed: 04/16/2023]
Abstract
Sunitinib (SUN) is an FDA approved first line drug for management of metastatic renal cancers and advanced cancerous states of gastrointestinal tract, however, side effects including fibrosis has been reported. Secukinumab (Secu) is an immunoglobulin G1 monoclonal antibody that exhibits anti-inflammatory activity by inhibiting several cellular signaling molecules. This study aimed to examine pulmonary protective potential of Secu in SUN-induced pulmonary fibrosis mediated through inhibition of inflammation via targeting IL-17A associated signaling pathway and using pirfenidone (PFD), an antifibrotic drug approved in 2014 for treatment of pulmonary fibrosis with IL-17A as one of its targets, as a reference drug. Wistar rats (160-200 g) were divided randomly into 4 groups (n = 6); Group 1 served as normal control; Group 2 served as disease control where it was exposed to SUN (25 mg/kg; 3 times weekly orally for 28 days); Group 3 was administered SUN and Secu (3 mg/kg subcutaneous at 0,14 and 28 days) and Group 4 was administered SUN and PFD (100 mg/kg/day orally for 28 days). Pro-inflammatory cytokines IL-1β, IL-6 and TNF-α were measured in addition to components of IL-17A signaling pathway (TGF-β, collagen, hydroxyproline). Results revealed that IL-17A-associated signaling pathway was activated in fibrotic lung tissue induced by SUN. Relative to normal control, SUN administration significantly elevated lung organ coefficient, IL-1β, IL-6, TNF-α, IL-17A, TGF-β, hydroxyproline and collagen expression. Secu or PFD treatment restored the altered levels to nearly normal values. Our study indicates that IL-17A participates in the development and progression of pulmonary fibrosis in a TGF-β dependent manner. Hence, components of IL-17A signaling pathway represent potential therapeutic targets for protection and treatment of fibro-proliferative lung disease.
Collapse
Affiliation(s)
- Mervat E Asker
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Asmaa A Abdelmeged
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A Shaheen
- Department of Histology & Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Rana G Eissa
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
7
|
Wang G, Zhou X, Guo Z, Huang N, Li J, Lv Y, Han L, Zheng W, Xu D, Chai D, Li H, Li L, Zheng J. The Anti-fibrosis drug Pirfenidone modifies the immunosuppressive tumor microenvironment and prevents the progression of renal cell carcinoma by inhibiting tumor autocrine TGF-β. Cancer Biol Ther 2022; 23:150-162. [PMID: 35130111 PMCID: PMC8824226 DOI: 10.1080/15384047.2022.2035629] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-β (TGF-β) plays a critical role in regulating cell growth and differentiation. Epithelial to mesenchymal transition (EMT) induced by TGF-β promotes cancer cell migration, invasion, and proliferation. Pirfenidone (5-methyl-1-phenyl-2(1 H)-pyridone, PFD), an approved drug for treating pulmonary and renal fibrosis, is a potent TGF-β inhibitor and found reduced incidence of lung cancer and alleviated renal function decline. However, whether PFD plays a role in controlling renal cancer progression is largely unknown. In the present study, we demonstrated that high TGF-β1 expression was negatively associated with ten-year overall survival of patients with renal cancer. Functionally, blockade of TGF-β signaling with PFD significantly suppressed the progression of renal cancer in a murine model. Mechanistically, we revealed that PFD significantly decreased the expression and secretion of TGF-β both in vitro and in vivo tumor mouse model, which further prevented TGF-β-induced EMT and thus cell proliferation, migration, and invasion. Importantly, the downregulation of TGF-β upon PFD treatment shaped the immunosuppressive tumor microenvironment by limiting the recruitment of tumor-infiltrating MDSCs. Therefore, our study demonstrated that PFD prevents renal cancer progression by inhibiting TGF-β production of cancer cells and downstream signaling pathway, which might be presented as a therapeutic adjuvant for renal cancer.
Collapse
Affiliation(s)
- Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaowan Zhou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zengli Guo
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Nan Huang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Juan Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanfang Lv
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dandan Xu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liantao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
8
|
Antar SA, Saleh MA, Al-Karmalawy AA. Investigating the possible mechanisms of pirfenidone to be targeted as a promising anti-inflammatory, anti-fibrotic, anti-oxidant, anti-apoptotic, anti-tumor, and/or anti-SARS-CoV-2. Life Sci 2022; 309:121048. [PMID: 36209833 PMCID: PMC9536875 DOI: 10.1016/j.lfs.2022.121048] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/23/2022] [Accepted: 10/02/2022] [Indexed: 01/10/2023]
Abstract
Pirfenidone (PFD) is a non-peptide synthetic chemical that inhibits the production of transforming growth factor-beta 1 (TGF-β1), tumor necrosis factor-alpha (TNF-α), platelet-derived growth factor (PDGF), Interleukin 1 beta (IL-1β), and collagen 1 (COL1A1), all of which have been linked to the prevention or removal of excessive scar tissue deposition in many organs. PFD has been demonstrated to decrease apoptosis, downregulate angiotensin-converting enzyme (ACE) receptor expression, reduce inflammation through many routes, and alleviate oxidative stress in pneumocytes and other cells while protecting them from COVID-19 invasion and cytokine storm. Based on the mechanism of action of PFD and the known pathophysiology of COVID-19, it was recommended to treat COVID-19 patients. The use of PFD as a treatment for a range of disorders is currently being studied, with an emphasis on outcomes related to reduced inflammation and fibrogenesis. As a result, rather than exploring the molecule's chemical characteristics, this review focuses on innovative PFD efficacy data. Briefly, herein we tried to investigate, discuss, and illustrate the possible mechanisms of actions for PFD to be targeted as a promising anti-inflammatory, anti-fibrotic, anti-oxidant, anti-apoptotic, anti-tumor, and/or anti-SARS-CoV-2 candidate.
Collapse
Affiliation(s)
- Samar A Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.
| | - Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, the United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| |
Collapse
|
9
|
Zanin-Silva DC, Santana-Gonçalves M, Kawashima-Vasconcelos MY, Oliveira MC. Management of Endothelial Dysfunction in Systemic Sclerosis: Current and Developing Strategies. Front Med (Lausanne) 2021; 8:788250. [PMID: 35004754 PMCID: PMC8727451 DOI: 10.3389/fmed.2021.788250] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic Sclerosis (SSc) is an autoimmune disease marked by dysregulation of the immune system, tissue fibrosis and dysfunction of the vasculature. Vascular damage, remodeling and inadequate endothelial repair are hallmarks of the disease. Since early stages of SSc, damage and apoptosis of endothelial cells (ECs) can lead to perivascular inflammation, oxidative stress and tissue hypoxia, resulting in multiple clinical manifestations. Raynaud's phenomenon, edematous puffy hands, digital ulcers, pulmonary artery hypertension, erectile dysfunction, scleroderma renal crisis and heart involvement severely affect quality of life and survival. Understanding pathogenic aspects and biomarkers that reflect endothelial damage in SSc is essential to guide therapeutic interventions. Treatment approaches described for SSc-associated vasculopathy include pharmacological options to improve blood flow and tissue perfusion and, more recently, cellular therapy to enhance endothelial repair, promote angiogenesis and heal injuries. This mini-review examines the current knowledge on cellular and molecular aspects of SSc vasculopathy, as well as established and developing therapeutic approaches for improving the vascular compartment.
Collapse
Affiliation(s)
- Djúlio César Zanin-Silva
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Basic and Applied Immunology Graduate Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maynara Santana-Gonçalves
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Oncology, Stem Cell and Cell-Therapy Graduate Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marianna Yumi Kawashima-Vasconcelos
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Internal Medicine Graduate Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
10
|
Bai X, Nie P, Lou Y, Zhu Y, Jiang S, Li B, Luo P. Pirfenidone is a renal protective drug: Mechanisms, signalling pathways, and preclinical evidence. Eur J Pharmacol 2021; 911:174503. [PMID: 34547247 DOI: 10.1016/j.ejphar.2021.174503] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/20/2021] [Accepted: 09/10/2021] [Indexed: 11/21/2022]
Abstract
Renal fibrosis, a characteristic of all chronic kidney diseases, lacks effective therapeutic drugs currently. Pirfenidone (PFD), a small molecule drug with good oral bioavailability, is widely used in idiopathic pulmonary fibrosis and exerts anti-fibrotic, anti-inflammatory, antioxidant, and anti-apoptotic effects. These effects have been attributed to the suppression of cell growth factors (in particular, but not exclusively, transforming growth factor-β) and the epithelial-mesenchymal transition, as well as the possible down-regulation of pro-inflammatory mediators (such as tumour necrosis factor-α), the protection of mitochondrial function, and the regulation of inflammatory cells. Considering the activation of similar anti-fibrotic pathways in lung and kidney disease and the broad activity of PFD, this drug has improved the treatment of the renal fibrotic disease. In this review, we briefly summarize the pharmacokinetics and safety of PFD as well as the mechanisms of PFD focusing on kidney disease. We summarize the effects of PFD on renal function and pathological alterations based on animal experiments, as well as changes in growth factors based on both animal and renal cell experiments. Moreover, given the activation of similar profibrotic pathways in pulmonary diseases and other disorders, we reviewed in-depth the possible signalling pathways targeted by PFD to attenuate renal fibrosis and protect renal function. Finally, we provide an overview of the current clinical trials of PFD for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Yan Lou
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Shan Jiang
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China.
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
11
|
Wang J, Xiang H, Lu Y, Wu T, Ji G. New progress in drugs treatment of diabetic kidney disease. Biomed Pharmacother 2021; 141:111918. [PMID: 34328095 DOI: 10.1016/j.biopha.2021.111918] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 02/08/2023] Open
Abstract
Diabetic kidney disease (DKD) is not only one of the main complications of diabetes, but also the leading cause of the end-stage renal disease (ESRD). The occurrence and development of DKD have always been a serious clinical problem that leads to the increase of morbidity and mortality and the severe damage to the quality of life of human beings. Controlling blood glucose, blood pressure, blood lipids, and improving lifestyle can help slow the progress of DKD. In recent years, with the extensive research on the pathological mechanism and molecular mechanism of DKD, there are more and more new drugs based on this, such as new hypoglycemic drugs sodium-glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) inhibitors, and dipeptidyl peptidase-4 (DPP-4) inhibitors with good efficacy in clinical treatment. Besides, there are some newly developed drugs, including protein kinase C (PKC) inhibitors, advanced glycation end product (AGE) inhibitors, aldosterone receptor inhibitors, endothelin receptor (ETR) inhibitors, transforming growth factor-β (TGF-β) inhibitors, Rho kinase (ROCK) inhibitors and so on, which show positive effects in animal or clinical trials and bring hope for the treatment of DKD. In this review, we sort out the progress in the treatment of DKD in recent years, the research status of some emerging drugs, and the potential drugs for the treatment of DKD in the future, hoping to provide some directions for clinical treatment of DKD.
Collapse
Affiliation(s)
- Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
12
|
Inflammation and Oxidative Damage in Ischaemic Renal Disease. Antioxidants (Basel) 2021; 10:antiox10060845. [PMID: 34070611 PMCID: PMC8227971 DOI: 10.3390/antiox10060845] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Ischaemic renal disease as result of atherosclerotic renovascular disease activates a complex biological response that ultimately leads to fibrosis and chronic kidney disease. Large randomised control trials have shown that renal revascularisation in patients with atherosclerotic renal artery disease does not confer any additional benefit to medical therapy alone. This is likely related to the activation of complex pathways of oxidative stress, inflammatory cytokines and fibrosis due to atherosclerotic disease and hypoxic injury due to reduced renal blood flow. New evidence from pre-clinical trials now indicates a role for specific targeted therapeutic interventions to counteract this complex pathogenesis. This evidence now suggests that the focus for those with atherosclerotic renovascular disease should be a combination of revascularisation and renoprotective therapies that target the renal tissue response to ischaemia, reduce the inflammatory infiltrate and prevent or reduce the fibrosis.
Collapse
|
13
|
Beck L, Pinilla E, Arcanjo DDR, Hernanz R, Prat-Duran J, Petersen AG, Köhler R, Sheykhzade M, Comerma-Steffensen S, Simonsen U. Pirfenidone Is a Vasodilator: Involvement of K V7 Channels in the Effect on Endothelium-Dependent Vasodilatation in Type-2 Diabetic Mice. Front Pharmacol 2021; 11:619152. [PMID: 33643042 PMCID: PMC7906977 DOI: 10.3389/fphar.2020.619152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022] Open
Abstract
Endothelial cell dysfunction and fibrosis are associated with worsening of the prognosis in patients with cardiovascular disease. Pirfenidone has a direct antifibrotic effect, but vasodilatation may also contribute to the effects of pirfenidone. Therefore, in a first study we investigated the mechanisms involved in the relaxant effect of pirfenidone in rat intrapulmonary arteries and coronary arteries from normal mice. Then in a second study, we investigated whether pirfenidone restores endothelial function in the aorta and mesenteric arteries from diabetic animals. From 16–18-week old normal male C57BL/6 mice and normoglycemic (db/db+), and type 2 diabetic (db/db) male and female mice, arteries were mounted in microvascular isometric myographs for functional studies, and immunoblotting was performed. In rat pulmonary arteries and mouse coronary arteries, pirfenidone induced relaxations, which were inhibited in preparations without endothelium. In mouse coronary arteries, pirfenidone relaxation was inhibited in the presence of a nitric oxide (NO) synthase inhibitor, NG-nitro-l-arginine (L-NOARG), a blocker of large-conductance calcium-activated potassium channels (BKCa), iberiotoxin, and a blocker of KV7 channels, XE991. Patch clamp studies in vascular smooth muscle revealed pirfenidone increased iberiotoxin-sensitive current. In the aorta and mesenteric small arteries from diabetic db/db mice relaxations induced by the endothelium-dependent vasodilator, acetylcholine, were markedly reduced compared to db/db + mice. Pirfenidone enhanced the relaxations induced by acetylcholine in the aorta from diabetic male and female db/db mice. An opener of KV7 channels, flupirtine, had the same effect as pirfenidone. XE991 reduced the effect of pirfenidone and flupirtine and further reduced acetylcholine relaxations in the aorta. In the presence of iberiotoxin, pirfenidone still increased acetylcholine relaxation in aorta from db/db mice. Immunoblotting for KV7.4, KV7.5, and BKCa channel subunits were unaltered in aorta from db/db mice. Pirfenidone failed to improve acetylcholine relaxation in mesenteric arteries, and neither changed acetylcholine-induced transient decreases in blood pressure in db/db+ and db/db mice. In conclusion, pirfenidone vasodilates pulmonary and coronary arteries. In coronary arteries from normal mice, pirfenidone induces NO-dependent vasodilatation involving BKCa and KV7 channels. Pirfenidone improves endothelium-dependent vasodilatation in aorta from diabetic animals by a mechanism involving voltage-gated KV7 channels, a mechanism that may contribute to the antifibrotic effect of pirfenidone.
Collapse
Affiliation(s)
- Lilliana Beck
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Physiology, Faculty of Pharmacy, Universidad Complutense, Madrid, Spain
| | - Daniel Dias Rufino Arcanjo
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biophysics and Physiology, Laboratory of Functional and Molecular Studies in Physiopharmacology, Federal University of Piauí, Teresina, Brazil
| | - Raquel Hernanz
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Judit Prat-Duran
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Asbjørn Graver Petersen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Ralf Köhler
- Aragón Agency for Research and Development (ARAID), Zaragoza, Spain
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biomedical Sciences/Animal Physiology, Faculty of Veterinary, Central University of Venezuela, Maracay, Venezuela
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Kölükçü E, Firat F, Deresoy FA, Katar M, Atılgan D. The effects of pirfenidone on ischaemia-reperfusion injury in testicular torsion-induced rat model. Andrologia 2020; 53:e13922. [PMID: 33244780 DOI: 10.1111/and.13922] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to analyse the effect of pirfenidone against ischaemia-reperfusion injury occurring after detorsion in rats with induced testicular torsion model. Group 1 was assigned as the control group. Group 2 first had testis torsion performed, and then, testicular detorsion was performed. Group 3 had similar procedures to the rats in Group 2. Rats in Group 3 additionally had 325 mg/kg pirfenidone administered immediately after ischaemia. The blood samples were analysed spectrophotometrically. To determine the intensity of tissue injury, haemorrhage, oedema and congestion levels were evaluated with direct microscopic investigation of testis. Seminiferous tubule architecture, spermatogenesis processes and germ cell maturation were graded by Johnsen and Cosentino scoring systems. In Group 3, superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities increased compared with Group 2 (p:.03 and p:.049 respectively). Additionally, the mean malondialdehyde (MDA) value was higher in Group 2 compared with the other groups (p:.001). Histopathological investigation of rats in Group 3 identified positive changes in haemorrhage, oedema and congestion levels compared with Group 2 (p:.031, p:.048, p:.044 respectively). Similarly, Johnsen and Cosentino scores were positively affected in Group 3 (p:.033, p:.032 respectively). Pirfenidone is protective against testicular oxidative damage.
Collapse
Affiliation(s)
- Engin Kölükçü
- Department of Urology, Gaziosmanpasa University, Tokat, Turkey
| | - Fatih Firat
- Department of Urology, Tokat State Hospital, Tokat, Turkey
| | | | - Muzaffer Katar
- Department of Biochemistry, Gaziosmanpasa University, Tokat, Turkey
| | - Doğan Atılgan
- Department of Urology, Gaziosmanpasa University, Tokat, Turkey
| |
Collapse
|
15
|
Sharawy MH, Serrya MS. Pirfenidone attenuates gentamicin-induced acute kidney injury by inhibiting inflammasome-dependent NLRP3 pathway in rats. Life Sci 2020; 260:118454. [PMID: 32950575 DOI: 10.1016/j.lfs.2020.118454] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 11/19/2022]
Abstract
Acute kidney injury (AKI) is an abrupt and usually reversible decline in renal function. AKI is considered one of the main drawbacks of the use of gentamicin that critically limits its clinical use. In this study, pirfenidone, an oral antifibrotic drug, was given to rats (200 mg/kg, p.o., daily) for seven days alone before the initiation of gentamicin treatment and continued for seven days alongside daily gentamicin injections. In gentamicin group, gentamicin was given to Wistar rats (100 mg/kg, i.p., daily) for seven days to induce AKI. Pirfenidone managed to alleviate gentamicin-induced AKI by improving kidney function parameters including serum creatinine, blood urea nitrogen (BUN), proteinuria, relative kidney-to-body weight ratio and creatinine clearance. Pirfenidone decreased cytotoxicity induced by gentamicin by decreasing lactate dehydrogenase (LDH) activity and improving histologic picture of tubules and glomeruli. Pirfenidone also alleviated oxidative stress induced by gentamicin by reducing malondialdehyde (MDA) and elevating reduced glutathione (GSH). Pirfenidone prevented the upregulated inflammasome pathway markers in the kidney. It succeeded in decreasing toll like recpetor-4 (TLR4), nuclear factor-kappa B (NF-κB), nucleotide-binding oligomerization domain [NOD]-like pyrin domain containing protein 3 (NLRP3), caspase-1, interleukin-1β (IL-1β) and IL-18 levels. Additionally, Pirfenidone caused a decrease in macrophage infiltration displayed by reduction in renal monocyte chemoattractant protein-1 (MCP-1) levels. To sum up, pirfenidone can effectively mitigate gentamicin-induced AKI by inhibiting oxidative stress, macrophage infiltration and inflammasome-dependent NLRP3 pathway-induced inflammation.
Collapse
Affiliation(s)
- Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Marwa S Serrya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
16
|
Sharma N, Gaikwad AB. Effects of renal ischemia injury on brain in diabetic and non-diabetic rats: Role of angiotensin II type 2 receptor and angiotensin-converting enzyme 2. Eur J Pharmacol 2020; 882:173241. [PMID: 32565336 DOI: 10.1016/j.ejphar.2020.173241] [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] [Received: 04/22/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 01/19/2023]
Abstract
Clinically, patients with diabetes mellitus (DM) are more susceptible to ischemic renal injury (IRI) than non-diabetic (ND) patients. Besides, IRI predisposes distant organ dysfunctions including, neurological dysfunction, in which the major contributor remains renin-angiotensin system (RAS). Interestingly, the role of depressor arm of RAS on IRI-associated neurological sequalae remains unclear. Hence, this study aimed to delineate the role of angiotensin II type 2 receptor (AT2R) and angiotensin-converting enzyme 2 (ACE2) under the same. ND and Streptozotocin-induced DM rats with bilateral IRI were treated with AT2R agonist-Compound 21 (C21) (0.3 mg/kg/day, i.p.) or ACE2 activator-Diminazene Aceturate (Dize), (5 mg/kg/day, p.o.) either alone or as combination therapy. Effect of IRI on neurological functions were assessed by behavioural, biochemical, and histopathological analysis. Immunohistochemistry, ELISA and qRT-PCR experiments were conducted for evaluation of the molecular mechanisms. We found that in ND and DM rats, IRI causes increased hippocampal MDA and nitrite levels, augmented inflammatory cytokines (granulocyte-colony stimulating factor, glial fibrillary acidic protein), altered protein levels of Ang II, Ang-(1-7) and mRNA expressions of At1r, At2r and Masr. Treatment with C21 and Dize effectively normalised above-mentioned pathological alterations. Moreover, the protective effect of C21 and Dize combination therapy was better than respective monotherapies, and more likely, exerted via augmentation of protein and mRNA levels of depressor arm components. Thus, AT2R agonist and ACE2 activator therapy prevents the development of IRI-associated neurological dysfunction by attenuating oxidative stress and inflammation, upregulating depressor arm of RAS in brain under ND and DM conditions.
Collapse
Affiliation(s)
- Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, 333031, India.
| |
Collapse
|
17
|
Pirfenidone alleviates lipopolysaccharide-induced lung injury by accentuating BAP31 regulation of ER stress and mitochondrial injury. J Autoimmun 2020; 112:102464. [DOI: 10.1016/j.jaut.2020.102464] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022]
|
18
|
Bennett D, Lanzarone N, Fossi A, Perillo F, De Vita E, Luzzi L, Paladini P, Bargagli E, Sestini P, Rottoli P. Pirfenidone in chronic lung allograft dysfunction: a single cohort study. Panminerva Med 2020; 62. [DOI: 10.23736/s0031-0808.19.03840-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
19
|
Li F, Bahnson EM, Wilder J, Siletzky R, Hagaman J, Nickekeit V, Hiller S, Ayesha A, Feng L, Levine JS, Takahashi N, Maeda-Smithies N. Oral high dose vitamin B12 decreases renal superoxide and post-ischemia/reperfusion injury in mice. Redox Biol 2020; 32:101504. [PMID: 32182573 PMCID: PMC7078436 DOI: 10.1016/j.redox.2020.101504] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/27/2020] [Accepted: 03/09/2020] [Indexed: 01/25/2023] Open
Abstract
Renal ischemia/reperfusion injury (IRI) is a leading cause of acute kidney injury (AKI), a potentially fatal syndrome characterized by a rapid decline in kidney function. Excess production of superoxide contributes to the injury. We hypothesized that oral administration of a high dose of vitamin B12 (B12 - cyanocobalamin), which possesses a superoxide scavenging function, would protect kidneys against IRI and provide a safe means of treatment. Following unilateral renal IR surgery, C57BL/6J wild type (WT) mice were administered B12 via drinking water at a dose of 50 mg/L. After 5 days of the treatment, plasma B12 levels increased by 1.2-1.5x, and kidney B12 levels increased by 7-8x. IRI mice treated with B12 showed near normal renal function and morphology. Further, IRI-induced changes in RNA and protein markers of inflammation, fibrosis, apoptosis, and DNA damage response (DDR) were significantly attenuated by at least 50% compared to those in untreated mice. Moreover, the presence of B12 at 0.3 μM in the culture medium of mouse proximal tubular cells subjected to 3 hr of hypoxia followed by 1 hr of reperfusion in vitro showed similar protective effects, including increased cell viability and decreased reactive oxygen species (ROS) level. We conclude that a high dose of B12 protects against perfusion injury both in vivo and in vitro without observable adverse effects in mice and suggest that B12 merits evaluation as a treatment for I/R-mediated AKI in humans.
Collapse
Affiliation(s)
- Feng Li
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Edward M Bahnson
- Department of Surgery, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jennifer Wilder
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Robin Siletzky
- Department of Surgery, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - John Hagaman
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Volker Nickekeit
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA; Division of Nephropathy, School of Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Sylvia Hiller
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Azraa Ayesha
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lanfei Feng
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago and Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Jerrold S Levine
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago and Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Nobuyuki Takahashi
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School, Sendai, Japan
| | - Nobuyo Maeda-Smithies
- Dept of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
20
|
Zou C, Zhou Z, Tu Y, Wang W, Chen T, Hu H. Pioglitazone Attenuates Reoxygenation Injury in Renal Tubular NRK-52E Cells Exposed to High Glucose via Inhibiting Oxidative Stress and Endoplasmic Reticulum Stress. Front Pharmacol 2020; 10:1607. [PMID: 32038263 PMCID: PMC6989595 DOI: 10.3389/fphar.2019.01607] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/10/2019] [Indexed: 01/15/2023] Open
Abstract
Renal ischemia-reperfusion injury is a major cause of acute kidney injury. In the present study, we investigated the effects of pioglitazone on hypoxia/reoxygenation (H/R) injury in rat renal tubular epithelial cells (RTECs) under normal- (NG) or high-glucose (HG) culture conditions via evaluating oxidative stress and endoplasmic reticulum stress (ERS). The RTECs (NRK-52E cells) were divided into six groups as follows: NG group, HG group, NG + H/R group, HG + H/R group, NG + Pio + H/R group, and HG + Pio + H/R group, among which cells in H/R groups were subjected to 4 h of hypoxia followed by 12 h of reoxygenation. After that, the cells were evaluated using the Cell Counting Kit-8 assay for the determination of their viability and flow cytometry assay for the detection of apoptosis. The levels of superoxide dismutase (SOD), glutathione reductase (GSH), catalase (CAT), and malondialdehyde (MDA) were determined via colorimetric chemical assays. In addition, the expression of ERS-associated proteins, i.e. ATF4, ATF6, GRP78, and CHOP, was determined via western blotting. A HG environment could reduce the viability and increase the apoptotic rate of NRK-52E cells with increased MDA levels and decreased SOD, CAT, and GSH levels, and upregulate the expression of ERS-associated proteins, i.e. ATF4, ATF6, and GRP78. H/R injury could further aggravate changes in the above indicators, but pioglitazone could significantly reverse such changes and alleviate cell injury. Thus, Pioglitazone exhibits a cytoprotective effect on RTECs against H/R injury under NG or HG culture conditions by inhibiting oxidative stress and ERS.
Collapse
Affiliation(s)
- Cong Zou
- Department of Endocrinology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiyu Zhou
- Department of Pathology, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yunming Tu
- Department of Endocrinology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weichao Wang
- Department of Urology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tongchang Chen
- Department of Urology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglin Hu
- Department of Urology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|