1
|
Hu R, Wu F, Zheng YQ. Ivacaftor attenuates gentamicin-induced ototoxicity through the CFTR-Nrf2-HO1/NQO1 pathway. Redox Rep 2024; 29:2332038. [PMID: 38563333 PMCID: PMC10993751 DOI: 10.1080/13510002.2024.2332038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVES Gentamicin is one of the most common ototoxic drugs that can lower patients' quality of life. Oxidative stress is a key factors inducing sensory hair cell death during gentamicin administration. So far, there are no effective drugs to prevent or treat gentamicin- induced hearing loss. A recent study found cystic fibrosis transmembrane conductance regulator (CFTR) as a new target to modulate cellular oxidative balance. The objective of this study was to estimate the effect of the CFTR activator ivacaftor on gentamicin-induced ototoxicity and determine its mechanism. METHODS The hair cell count was analyzed by Myosin 7a staining. Apoptosis was analyzed by TUNEL Apoptosis Kit. Cellular reactive oxygen species (ROS) level was detected by DCFH-DA probes. The Nrf2 related proteins expression levels were analyzed by western blot. RESULTS An in vitro cochlear explant model showed that gentamicin caused ROS accumulation in sensory hair cells and induced apoptosis, and this effect was alleviated by pretreatment with ivacaftor. Western blotting showed that ivacaftor administration markedly increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO1), and NAD(P)H:quinone oxidoreductase 1 (NQO1). The protective effect of ivacaftor was abolished by the Nrf2 inhibitor ML385. DISCUSSION Our results indicate the protective role of the CFTR-Nrf2-HO1/NQO1 pathway in gentamicin-induced ototoxicity. Ivacaftor may be repositioned or repurposed towards aminoglycosides-induced hearing loss.
Collapse
Affiliation(s)
- Rui Hu
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, People’s Republic of China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Fan Wu
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Pathology and Laboratory Medicine, The Medical University of South Carolina, Charleston, SC, USA
| | - Yi-Qing Zheng
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, People’s Republic of China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
2
|
Zheng ZL, Ma JW, Luo Y, Liang GJ, Lei SJ, Yan KJ, Meng HB, Liu XJ. Mechanism of dexmedetomidine protection against cisplatin induced acute kidney injury in rats. Ren Fail 2024; 46:2337287. [PMID: 38627212 PMCID: PMC11022910 DOI: 10.1080/0886022x.2024.2337287] [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: 09/26/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE This study explored the molecular mechanisms by which dexmedetomidine (Dex) alleviates cisplatin (CP)-induced acute kidney injury (AKI) in rats. METHODS CP-induced AKI models were established, and Dex was intraperitoneally injected at different concentrations into rats in the model groups. Subsequently, rats were assigned to the control, CP, CP + Dex 10 μg/kg, and CP + Dex 25 μg/kg groups. After weighing the kidneys of the rats, the kidney arterial resistive index was calculated, and CP-induced AKI was evaluated. In addition, four serum biochemical indices were measured: histopathological damage in rat kidneys was detected; levels of inflammatory factors, interleukin (IL)-1β, IL-18, IL-6, and tumor necrosis factor alpha, in kidney tissue homogenate of rats were assessed through enzyme-linked immunosorbent assay (ELISA); and levels of NLRP-3, caspase-1, cleaved caspase-1, gasdermin D (GSDMD), and GSDMD-N in kidney tissues of rats were determined via western blotting. RESULTS Dex treatment reduced nephromegaly and serum clinical marker upregulation caused by CP-induced AKI. In addition, hematoxylin and eosin staining revealed that Dex treatment relieved CP-induced kidney tissue injury in AKI rats. ELISA analyses demonstrated that Dex treatment reduced the upregulated levels of proinflammatory cytokines in the kidney tissue of AKI rats induced by CP, thereby alleviating kidney tissue injury. Western blotting indicated that Dex alleviated CP-induced AKI by inhibiting pyroptosis mediated by NLRP-3 and caspase-1. CONCLUSION Dex protected rats from CP-induced AKI, and the mechanism may be related to NLRP-3/Caspase-1-mediated pyroptosis.
Collapse
Affiliation(s)
- Zeng-lu Zheng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Jun-wei Ma
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Yi Luo
- Department of Respiratory, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Gui-jin Liang
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Shi-jie Lei
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Ke-jin Yan
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Hai-bing Meng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Xiu-juan Liu
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| |
Collapse
|
3
|
Li X, Xu F, Zhang P, Mao L, Guo Y, Li H, Xie Y, Li Y, Liao Y, Chen J, Wu D, Zhang D. Overexpression of PRDM16 attenuates acute kidney injury progression: genetic and pharmacological approaches. MedComm (Beijing) 2024; 5:e737. [PMID: 39309696 PMCID: PMC11416085 DOI: 10.1002/mco2.737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/25/2024] Open
Abstract
Acute kidney injury (AKI) presents as a condition marked by a sudden and rapid decrease in kidney function over a short timeframe, resulting from diverse causes. As a transcription factor, PR domain-containing 16 (PRDM16), has recently been implicated in brown fat biogenesis and heart diseases. Our recent works indicated that PRDM16 could suppress the occurrence of renal interstitial fibrosis in diabetic kidney disorder. Nonetheless, the effect and regulatory mechanism of PRDM16 in AKI remain elusive. Our study demonstrated that PRDM16 inhibited apoptosis induced by ischemic/reperfusion (I/R) in BUMPT (Boston University mouse kidney proximal tubular) cells and HK-2(Human Kidney-2) cells. Mechanistically, PRDM16 not only bound to the promoter region of S100 Calcium Binding Protein A6 (S100A6)and upregulated its expression but also interacted with its amino acids 945-949, 957-960, and 981-984 to suppress the p38MAPK and JNK axes via inhibition of PKC-η activity and mitochondrial reactive oxygen species (ROS) production. Furthermore, cisplatin- and I/R-stimulated AKI progression were ameliorated in PRDM16 proximal-tubule-specific knockin mice, whereas exacerbated in PRDM16 knockout proximal-tubule-specific mice). Moreover, we observed that formononetin ameliorated I/R- and cisplatin-triggered AKI progression in mice. Taken together, these findings reveal a novel self-protective mechanism in AKI, whereby PRDM16 regulates the S100A6/PKC-η/ROS/p38MAPK and JNK pathways to inhibit AKI progression.
Collapse
Affiliation(s)
- Xiaozhou Li
- Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Emergency Medicine and Difficult Diseases Institute,Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Fang Xu
- Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Emergency Medicine and Difficult Diseases Institute,Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Pan Zhang
- Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Emergency Medicine and Difficult Diseases Institute,Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of Epidemiology and Health StatisticsXiangya School of Public HealthCentral South UniversityChangshaHunanChina
| | - Liufeng Mao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Yong Guo
- Department of Organ Procurement OrganizationThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Huiling Li
- Department of OphthalmologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yuxing Xie
- Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Emergency Medicine and Difficult Diseases Institute,Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yijian Li
- Department of UrologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yingjun Liao
- Department of AnesthesiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Junxiang Chen
- Department of NephrologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Donghai Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences,Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
| | - Dongshan Zhang
- Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Emergency Medicine and Difficult Diseases Institute,Department of Emergency MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of NephrologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
4
|
Huenchuguala S, Segura-Aguilar J. Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson's Disease. Antioxidants (Basel) 2024; 13:1125. [PMID: 39334784 PMCID: PMC11428591 DOI: 10.3390/antiox13091125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recently, a single-neuron degeneration model has been proposed to understand the development of idiopathic Parkinson's disease based on (i) the extremely slow development of the degenerative process before the onset of motor symptoms and during the progression of the disease and (ii) the fact that it is triggered by an endogenous neurotoxin that does not have an expansive character, limiting its neurotoxic effect to single neuromelanin-containing dopaminergic neurons. It has been proposed that aminochrome is the endogenous neurotoxin that triggers the neurodegenerative process in idiopathic Parkinson's disease by triggering mitochondrial dysfunction, oxidative stress, neuroinflammation, dysfunction of both lysosomal and proteasomal protein degradation, endoplasmic reticulum stress and formation of neurotoxic alpha-synuclein oligomers. Aminochrome is an endogenous neurotoxin that is rapidly reduced by flavoenzymes and/or forms adducts with proteins, which implies that it is impossible for it to have a propagative neurotoxic effect on neighboring neurons. Interestingly, the enzymes DT-diaphorase and glutathione transferase M2-2 prevent the neurotoxic effects of aminochrome. Natural compounds present in fruits, vegetables and other plant products have been shown to activate the KEAP1/Nrf2 signaling pathway by increasing the expression of antioxidant enzymes including DT-diaphorase and glutathione transferase. This review analyzes the possibility of searching for natural compounds that increase the expression of DT-diaphorase and glutathione transferase through activation of the KEAP1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Sandro Huenchuguala
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, Instituto de Ciencias Biomédicas (ICBM), Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
5
|
Almohawes ZN, El-Kott A, Morsy K, Shati AA, El-Kenawy AE, Khalifa HS, Elsaid FG, Abd-Lateif AEKM, Abu-Zaiton A, Ebealy ER, Abdel-Daim MM, Ghanem RA, Abd-Ella EM. Salidroside inhibits insulin resistance and hepatic steatosis by downregulating miR-21 and subsequent activation of AMPK and upregulation of PPARα in the liver and muscles of high fat diet-fed rats. Arch Physiol Biochem 2024; 130:257-274. [PMID: 35061559 DOI: 10.1080/13813455.2021.2024578] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023]
Abstract
This study evaluated if salidroside (SAL) alleviates high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) by downregulating miR-21. Rats (n = 8/group) were treated for 12 weeks as normal diet (control/ND), ND + agmoir negative control (NC) (150 µg/kg), ND + SAL (300 mg/kg), HFD, HFD + SAL, HFD + compound C (an AMPK inhibitor) (200 ng/kg), HFD + SAL + NXT629 (a PPAR-α antagonist) (30 mg/kg), and HFD + SAL + miR-21 agomir (150 µg/kg). SAL improved glucose and insulin tolerance and preserved livers in HFD-fed rats. In ND and HFD-fed rats, SAL reduced levels of serum and hepatic lipids and the hepatic expression of SREBP1, SREBP2, fatty acid (FA) synthase, and HMGCOAR. It also activated hepatic Nrf2 and increased hepatic/muscular activity of AMPK and levels of PPARα. All effects afforded by SAL were prevented by CC, NXT629, and miR-21 agmoir. In conclusion, activation of AMPK and upregulation of PPARα mediate the anti-steatotic effect of SAL.
Collapse
Affiliation(s)
- Zakiah N Almohawes
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Attalla El-Kott
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, College of Science, Damanhour University, Damanhour, Egypt
| | - Kareem Morsy
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, College of Science, Cairo University, Cairo, Egypt
| | - Ali A Shati
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ayman E El-Kenawy
- Pathology Department, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Heba S Khalifa
- Zoology Department, College of Science, Damanhour University, Damanhour, Egypt
| | - Fahmy G Elsaid
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | | | | | - Eman R Ebealy
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Pharmaceutical Sciences Department, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Reham A Ghanem
- Oral Biology Department, Faculty of Oral and Dental Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Eman M Abd-Ella
- Zoology Department, College of Science, Fayoum University, Fayoum, Egypt
- Biology Department, College of Science and Art, Al-Baha University, Al-Mandaq, Saudi Arabia
| |
Collapse
|
6
|
Ding M, Bao Y, Liang H, Zhang X, Li B, Yang R, Zeng N. Potential mechanisms of formononetin against inflammation and oxidative stress: a review. Front Pharmacol 2024; 15:1368765. [PMID: 38799172 PMCID: PMC11116718 DOI: 10.3389/fphar.2024.1368765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Formononetin (FMNT) is a secondary metabolite of flavonoids abundant in legumes and graminaceous plants such as Astragalus mongholicus Bunge [Fabaceae; Astragali radix] and Avena sativa L. [Poaceae]. Astragalus is traditionally used in Asia countries such as China, Korea and Mongolia to treat inflammatory diseases, immune disorders and cancers. In recent years, inflammation and oxidative stress have been found to be associated with many diseases. A large number of pharmacological studies have shown that FMNT, an important bioactive metabolite of Astragalus, has a profoundly anti-inflammatory and antioxidant potential. This review focuses on providing comprehensive and up-to-date findings on the efficacy of the molecular targets and mechanisms involve of FMNT and its derivatives against inflammation and oxidative stress in both in vitro and in vivo. Relevant literature on FMNT against inflammation and oxidative stress between 2013 and 2023 were analyzed. FMNT has antioxidant and anti-inflammatory potential and shows mild or no toxicity in various diseases. Moreover, in the medical field, FMNT has shown potential in the prevention and treatment of cancers, neurological diseases, fibrotic diseases, allergic diseases, metabolic diseases, cardiovascular diseases, gastrointestinal diseases and autoimmune diseases. Thus, it is expected to be utilized in more products in the medical, food and cosmetic industries in the future.
Collapse
Affiliation(s)
- Meiling Ding
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiwen Bao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiongwei Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruocong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Yao J, Peng T, Shao C, Liu Y, Lin H, Liu Y. The Antioxidant Action of Astragali radix: Its Active Components and Molecular Basis. Molecules 2024; 29:1691. [PMID: 38675511 PMCID: PMC11052376 DOI: 10.3390/molecules29081691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Astragali radix is a traditional medicinal herb with a long history and wide application. It is frequently used in prescriptions with other medicinal materials to replenish Qi. According to the classics of traditional Chinese medicine, Astragali radix is attributed with properties such as Qi replenishing and surface solidifying, sore healing and muscle generating, and inducing diuresis to reduce edema. Modern pharmacological studies have demonstrated that some extracts and active ingredients in Astragali radix function as antioxidants. The polysaccharides, saponins, and flavonoids in Astragali radix offer beneficial effects in preventing and controlling diseases caused by oxidative stress. However, there is still a lack of comprehensive research on the effective components and molecular mechanisms through which Astragali radix exerts antioxidant activity. In this paper, we review the active components with antioxidant effects in Astragali radix; summarize the content, bioavailability, and antioxidant mechanisms; and offer a reference for the clinical application of Astragalus and the future development of novel antioxidants.
Collapse
Affiliation(s)
- Juan Yao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730101, China; (T.P.); (C.S.); (H.L.)
| | - Ting Peng
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730101, China; (T.P.); (C.S.); (H.L.)
| | - Changxin Shao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730101, China; (T.P.); (C.S.); (H.L.)
| | - Yuanyuan Liu
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730013, China;
| | - Huanhuan Lin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730101, China; (T.P.); (C.S.); (H.L.)
| | - Yongqi Liu
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730013, China;
| |
Collapse
|
8
|
Wang Q, Zhen W, Lippi G, Liu Q. The effect of Astragali Radix-Radix Angelica Sinensis on acute kidney injury: a network pharmacology and molecular docking study. Transl Androl Urol 2024; 13:91-103. [PMID: 38404557 PMCID: PMC10891378 DOI: 10.21037/tau-23-562] [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: 11/04/2023] [Accepted: 01/10/2024] [Indexed: 02/27/2024] Open
Abstract
Background Acute kidney injury (AKI) is a devastating clinical syndrome with high mortality rate attributed to lack of effective treatment. The herbal pair of Astragali Radix (AR) and Radix Angelica Sinensis (RAS) is a commonly prescribed herbal formula or is added to other traditional Chinese medicine (TCM) prescriptions for the treatment of kidney diseases. AR-RAS has certain protective effects on AKI in experiments, but the relevant mechanisms have yet to be clear. So this study aims to explore the mechanism of action of AR-RAS in AKI by combining network pharmacology and molecular docking methods. Methods In Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), the major AR-RAS chemical components and associated action targets were found and screened. The DrugBank and GeneCards databases were used to find AKI-related targets. The targets that are in close relationship with AKI were obtained from Therapeutic Target database (TTD), Online Mendelian Inheritance in Man (OMIM), and PharmGKB databases. The "herb-active ingredient-target" network was drawn by Cytoscape 3.8.0 software. The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database was used to build the protein-protein interaction network. Bioconductor/R was used to examine Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. AR-RAS components and critical targets were docked using the AutoDock Vina program. Results A compound-target network, built by screening and analyzing the results, allowed to identify 19 active components and 101 possible therapeutic targets for AKI. The main ingredients were quercetin, kaempferol, 7-o-methylisocronulatol, formononetin and isorhamnetin. The key targets included AKT serine/threonine kinase 1 (AKT1), nuclear receptor coactivator 1 (NCOA1), JUN, estrogen receptor alpha (ESR1) and mitogen-activated protein kinase 8 (MAPK8). These molecules are targeted by pathways such as the calcium signaling route, the tumor necrosis factor (TNF) signaling pathway and the interleukin-17 (IL-17) signaling pathway, as well as the development of T helper 17 cells. Molecular docking demonstrated that AR-active RAS components exhibited strong binding activities to probable targets of AKI. Conclusions We described here the potential active ingredients, possible targets responsible for the efficacy of AR-RAS in AKI treatment, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Qin Wang
- Suzhou Medical College of Soochow University, Suzhou, China
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wenrui Zhen
- Department of Intervention Therapy, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Qi Liu
- Suzhou Medical College of Soochow University, Suzhou, China
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
9
|
Li Y, Wu J, Yu H, Lu X, Ni Y. Formononetin ameliorates cisplatin-induced hair cell death via activation of the PI3K/AKT-Nrf2 signaling pathway. Heliyon 2024; 10:e23750. [PMID: 38192850 PMCID: PMC10772176 DOI: 10.1016/j.heliyon.2023.e23750] [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: 05/02/2023] [Revised: 10/23/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
Cisplatin (CDDP) stands as a highly effective chemotherapeutic agent; however, its ototoxicity remains a perplexing challenge in the field. Formononetin (FMNT), a potent flavonoid isolated from Astragalus membranaceus, displays a diverse range of promising pharmacological activities, encompassing antioxidant, anti-apoptotic, and anti-inflammatory effects. Nonetheless, the advantageous effects of FMNT on cisplatin-induced cochlear hair cell injury demand further investigation. This study aimed to assess the protective properties of FMNT against cisplatin-induced hair cell damage by conducting in vitro assays on explant-cultured cochlear hair cells. The findings revealed that FMNT exhibited a notable reduction in cisplatin-induced hair cell apoptosis. Also, FMNT effectively mitigated the accumulation of reactive oxygen species and mitochondrial damage in cochlear explants exposed to cisplatin, while also restoring the turnover of the reduced glutathione (GSH)/glutathione disulfide (GSSG) ratio. Furthermore, our study demonstrated that FMNT protects hair cells against CDDP injury through the activation of the PI3K/AKT-Nrf2 signaling pathway. Consequently, formononetin emerges as a potential therapeutic agent for the treatment of cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Yimeng Li
- Otorhinolaryngology Department and ENT Institute of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200031, People's Republic of China
| | - Jingfang Wu
- Otorhinolaryngology Department and ENT Institute of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200031, People's Republic of China
| | - Huiqian Yu
- Otorhinolaryngology Department and ENT Institute of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200031, People's Republic of China
| | - Xiaoling Lu
- Otorhinolaryngology Department and ENT Institute of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200031, People's Republic of China
| | - Yusu Ni
- Otorhinolaryngology Department and ENT Institute of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200031, People's Republic of China
| |
Collapse
|
10
|
Dong K, Sun Y, Gao X, Wang J, Wu X, Guo C. Mixed micelles loaded with hesperidin protect against acetaminophen induced acute liver injury by inhibiting the mtDNA-cGAS-STING pathway. Colloids Surf B Biointerfaces 2024; 233:113656. [PMID: 37984191 DOI: 10.1016/j.colsurfb.2023.113656] [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: 06/28/2023] [Revised: 10/28/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Excessive acetaminophen (APAP) is the main cause of drug-induced acute liver failure, and the pathogenesis has not been elucidated and there is a lack of effective drugs. Hesperidin (Hes), a rich flavanone in citrus peel with excellent biological activities, is a potential agent for treatment liver injury. Due to poor water solubility of Hes, this study prepared mixed micelles using polyvinyl pyrrolidone (PVP K17) and poloxamer 188, and encapsulated Hes (Hes-MMs). The results showed that Hes-MMs exhibited a uniform spherical shape with a particle size of 66.80 ± 0.83 nm, and Hes-MMs significantly improved the dispersibility, antioxidant activity, and cellular uptake of Hes. In vitro results showed that Hes-MMs protected the proliferation inhibition of HepG2 cells induced by APAP, inhibited the production of reactive oxygen species (ROS) and the damage of mitochondrial membrane potential (MMP) induced by APAP. Furthermore, Hes-MMs exerted liver protective effects by inhibiting APAP induced mtDNA release and activating the cGAS-STING pathway. In vivo results demonstrated that Hes-MMs showed protective and therapeutic effects on APAP induced liver injury, and their mechanisms were related to the mtDNA-cGAS-STING signaling pathway. In summary, our study demonstrated that the mtDNA-cGAS-STING pathway was involved in APAP induced acute liver injury, and Hes-MMs might be a potential therapeutic agent for treating APAP induced acute liver injury.
Collapse
Affiliation(s)
- Kehong Dong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yuxuan Sun
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xintao Gao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jing Wang
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou 014030, China
| | - Xiaochen Wu
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chuanlong Guo
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
11
|
Zhang L, Wu Q, Huang Y, Zheng J, Guo S, He L. Formononetin ameliorates airway inflammation by suppressing ESR1/NLRP3/Caspase-1 signaling in asthma. Biomed Pharmacother 2023; 168:115799. [PMID: 37922653 DOI: 10.1016/j.biopha.2023.115799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Since inhaled glucocorticoids are the first-line treatment for asthma, asthma management becomes extremely difficult when asthma does not react well to glucocorticoids. Formononetin, a bioactive isoflavone and typical phytoestrogen, has been shown to have an anti-inflammatory impact while alleviating epithelial barrier dysfunction, which plays a role in the pathogenesis of allergic illnesses like asthma. However, the biological mechanisms behind this impact are unknown. As a result, we set out to investigate the effects of formononetin on airway inflammation and epithelial barrier repair in house dust mite (HDM)-induced asthmatic mice. We further expanded on formononetin's putative mode of action in reducing airway inflammation by modifying epithelial barrier dysfunction. In the current study, researchers discovered that formononetin significantly lowered total IgE levels in serum and interleukin (IL)-4, IL-6, and IL-17A levels in bronchoalveolar lavage fluid (BALF) in HDM-challenged asthmatic mice. Experiments on cell proliferation, migration, and apoptosis were performed in vitro to determine the effect of formononetin on bronchial epithelial barrier repair. Furthermore, in lipopolysaccharide (LPS)-stimulated 16HBE cells, formononetin increased cell proliferation and migration while preventing apoptosis and lowering the Bax/Bcl-2 ratio. In vitro and in vivo, formononetin significantly inhibited toll-like receptor 4 (TLR4) and estrogen receptor (ESR1)/Nod-like receptor family pyrin domain-containing protein 3 (NLRP3)/Caspase-1 signaling. These findings show that formononetin can reduce airway inflammation in HDM-challenged asthmatic mice by promoting epithelial barrier repair and possibly by inhibiting ESR1/NLRP3/Caspase-1 signaling as the underlying mechanism; formononetin could be a promising alternative treatment for asthma.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Wu
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuying Huang
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Zheng
- Department of Respiratory Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Guo
- Department of Endocrine, Genetics and Metabolism, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Li He
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
12
|
Mentese A, Demir S, Mungan SA, Alemdar NT, Demir EA, Aliyazicioglu Y. Gentisic acid ameliorates cisplatin-induced reprotoxicity through suppressing endoplasmic reticulum stress and upregulating Nrf2 pathway. Tissue Cell 2023; 85:102256. [PMID: 37918215 DOI: 10.1016/j.tice.2023.102256] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Reproductive toxicity is a serious side effect of cisplatin (CP) chemotherapy. Gentisic acid (GTA) is a phenolic acid with strong antioxidant properties. Here, we aimed to determine therapeutic effect of GTA against CP-induced testicular toxicity in rats for the first time. Male Sprague-Dawley rats received a single dose of CP (5 mg/kg; intraperitoneal) and treated with GTA (1.5 and 3 mg/kg; intraperitoneal; 3 consecutive days). The levels of oxidative stress (OS), inflammation, endoplasmic reticulum stress (ERS) and apoptosis biomarkers were assessed in the testicular tissue of rats. In addition, how CP affects the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and the effect of GTA on this situation were also addressed in the testicular tissue. CP administration induced histopathological changes in testicular tissue of rats with a significant increase in OS, inflammation, ERS and apoptosis biomarkers and a decrease in antioxidant capacity and Nrf2 expression levels. Administrations of GTA resulted in an amelioration of these altered parameters. These data suggest that GTA may be a potential therapeutic agent against CP-induced testicular toxicity. Activation of the Nrf2 pathway plays a key role of this therapeutic effect of GTA.
Collapse
Affiliation(s)
- Ahmet Mentese
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| | - Selim Demir
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Karadeniz Technical University, 61080 Trabzon, Turkey.
| | - Sevdegul Aydin Mungan
- Department of Medical Pathology, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| | - Nihal Turkmen Alemdar
- Department of Medical Biochemistry, Graduate School of Health Sciences, Karadeniz Technical University, 61080 Trabzon, Turkey; Department of Medical Services and Techniques, Vocational School of Health Services, Recep Tayyip Erdogan University, 53100 Rize, Turkey
| | - Elif Ayazoglu Demir
- Department of Chemistry and Chemical Processing Technologies, Macka Vocational School, Karadeniz Technical University, 61750 Trabzon, Turkey
| | - Yuksel Aliyazicioglu
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| |
Collapse
|
13
|
Li J, Zheng S, Fan Y, Tan K. Emerging significance and therapeutic targets of ferroptosis: a potential avenue for human kidney diseases. Cell Death Dis 2023; 14:628. [PMID: 37739961 PMCID: PMC10516929 DOI: 10.1038/s41419-023-06144-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/24/2023]
Abstract
Kidney diseases remain one of the leading causes of human death and have placed a heavy burden on the medical system. Regulated cell death contributes to the pathology of a plethora of renal diseases. Recently, with in-depth studies into kidney diseases and cell death, a new iron-dependent cell death modality, known as ferroptosis, has been identified and has attracted considerable attention among researchers in the pathogenesis of kidney diseases and therapeutics to treat them. The majority of studies suggest that ferroptosis plays an important role in the pathologies of multiple kidney diseases, such as acute kidney injury (AKI), chronic kidney disease, and renal cell carcinoma. In this review, we summarize recently identified regulatory molecular mechanisms of ferroptosis, discuss ferroptosis pathways and mechanisms of action in various kidney diseases, and describe the protective effect of ferroptosis inhibitors against kidney diseases, especially AKI. By summarizing the prominent roles of ferroptosis in different kidney diseases and the progress made in studying ferroptosis, we provide new directions and strategies for future research on kidney diseases. In summary, ferroptotic factors are potential targets for therapeutic intervention to alleviate different kidney diseases, and targeting them may lead to new treatments for patients with kidney diseases.
Collapse
Affiliation(s)
- Jinghan Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Sujuan Zheng
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yumei Fan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China.
| | - Ke Tan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China.
| |
Collapse
|
14
|
Kim K, Hong HL, Kim GM, Leem J, Kwon HH. Eupatilin Ameliorates Lipopolysaccharide-Induced Acute Kidney Injury by Inhibiting Inflammation, Oxidative Stress, and Apoptosis in Mice. Curr Issues Mol Biol 2023; 45:7027-7042. [PMID: 37754228 PMCID: PMC10530142 DOI: 10.3390/cimb45090444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Acute kidney injury (AKI) is a common complication of sepsis. Eupatilin (EUP) is a natural flavone with multiple biological activities and has beneficial effects against various inflammatory disorders. However, whether EUP has a favorable effect on septic AKI remains unknown. Here, we examined the effect of EUP on lipopolysaccharide (LPS)-evoked AKI in mice. LPS-evoked renal dysfunction was attenuated by EUP, as reflected by reductions in serum creatinine and blood urea nitrogen levels. LPS injection also induced structural damage such as tubular cell detachment, tubular dilatation, brush border loss of proximal tubules, and upregulation of tubular injury markers. However, EUP significantly ameliorated this structural damage. EUP decreased serum and renal cytokine levels, prevented macrophage infiltration, and inhibited mitogen-activated protein kinase and NF-κB signaling cascades. Lipid peroxidation and DNA oxidation were increased after LPS treatment. However, EUP mitigated LPS-evoked oxidative stress through downregulation of NPDPH oxidase 4 and upregulation of antioxidant enzymes. EUP also inhibited p53-mediated apoptosis in LPS-treated mice. Therefore, these results suggest that EUP ameliorates LPS-evoked AKI through inhibiting inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Kiryeong Kim
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea; (K.K.); (H.-L.H.)
| | - Hyo-Lim Hong
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea; (K.K.); (H.-L.H.)
| | - Gyun Moo Kim
- Department of Emergency Medicine, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea;
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Hyun Hee Kwon
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea; (K.K.); (H.-L.H.)
| |
Collapse
|
15
|
Chen H, Ma Y, Qi X, Tian J, Ma Y, Niu T. α-Lactalbumin Peptide Asp-Gln-Trp Ameliorates Hepatic Steatosis and Oxidative Stress in Free Fatty Acids-Treated HepG2 Cells and High-Fat Diet-Induced NAFLD Mice by Activating the PPARα Pathway. Mol Nutr Food Res 2023; 67:e2200499. [PMID: 37354055 DOI: 10.1002/mnfr.202200499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/26/2023] [Indexed: 06/26/2023]
Abstract
SCOPE Dietary intervention has emerged as a promising strategy for the management of nonalcoholic fatty liver disease (NAFLD). The aim of this study is to investigate the ameliorative effects of the α-lactalbumin peptide Asp-Gln-Trp (DQW) against NAFLD and the underlying mechanism. METHODS AND RESULTS The models of lipid metabolism disorders are established both in HepG2 cells and in C57BL/6J mice. The results demonstrate that DQW activates peroxisome proliferator-activated receptor α (PPARα) and subsequently ameliorates lipid deposition and oxidative stress in vitro. Interestingly, GW6471 markedly attenuates the modulatory effects of DQW on the PPARα pathway in HepG2 cells. Moreover, results of in vivo experiments indicate that DQW alleviates body weight gain, dyslipidemia, hepatic steatosis, and oxidative stress in high-fat-diet (HFD)-induced NAFLD mice. At the molecular level, DQW activates PPARα, subsequently enhances fatty acid β-oxidation, and reduces lipogenesis, thereby ameliorating hepatic steatosis. Meanwhile, DQW may ameliorate liver injury and oxidative stress via activating the PPARα/nuclear-factor erythroid 2 (Nrf2)/heme-oxygenase 1 (HO-1) pathway. CONCLUSION Those results indicate that α-lactalbumin peptide DQW may be an effective dietary supplement for alleviating NAFLD by alleviating lipid deposition and oxidative stress.
Collapse
Affiliation(s)
- Haoran Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Yanfeng Ma
- Hainan Mengniu Technology Development Co., Ltd., Haikou, Hainan, 571900, China
- School of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Xiaofen Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Jianjun Tian
- School of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Tianjiao Niu
- Hainan Mengniu Technology Development Co., Ltd., Haikou, Hainan, 571900, China
| |
Collapse
|
16
|
Miao M, Wang X, Liu T, Li YJ, Yu WQ, Yang TM, Guo SD. Targeting PPARs for therapy of atherosclerosis: A review. Int J Biol Macromol 2023:125008. [PMID: 37217063 DOI: 10.1016/j.ijbiomac.2023.125008] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
Atherosclerosis, a chief pathogenic factor of cardiovascular disease, is associated with many factors including inflammation, dyslipidemia, and oxidative stress. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors and are widely expressed with tissue- and cell-specificity. They control multiple genes that are involved in lipid metabolism, inflammatory response, and redox homeostasis. Given the diverse biological functions of PPARs, they have been extensively studied since their discovery in 1990s. Although controversies exist, accumulating evidence have demonstrated that PPAR activation attenuates atherosclerosis. Recent advances are valuable for understanding the mechanisms of action of PPAR activation. This article reviews the recent findings, mainly from the year of 2018 to present, including endogenous molecules in regulation of PPARs, roles of PPARs in atherosclerosis by focusing on lipid metabolism, inflammation, and oxidative stress, and synthesized PPAR modulators. This article provides information valuable for researchers in the field of basic cardiovascular research, for pharmacologists that are interested in developing novel PPAR agonists and antagonists with lower side effects as well as for clinicians.
Collapse
Affiliation(s)
- Miao Miao
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Tian Liu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Yan-Jie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Wen-Qian Yu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Tong-Mei Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
17
|
Jin X, He R, Liu J, Wang Y, Li Z, Jiang B, Lu J, Yang S. An herbal formulation "Shenshuaifu Granule" alleviates cisplatin-induced nephrotoxicity by suppressing inflammation and apoptosis through inhibition of the TLR4/MyD88/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 306:116168. [PMID: 36646160 DOI: 10.1016/j.jep.2023.116168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenshuaifu Granule (SSF) is an in-hospital preparation approved by the Guangdong Food and Drug Administration of China. It has been clinically used against kidney diseases for more than 20 years with a definite curative effect. AIM OF THE STUDY Cisplatin (CDDP) is a first-line chemotherapeutic drug in clinical practice, primarily excreted by the kidney with nephrotoxicity as a common side effect. Approximately 5-20% of cancer patients develop acute kidney injury (AKI) after chemotherapy; however, prevention and control strategies are currently unavailable. Therefore, it is important to identify safe and effective drugs that can prevent the nephrotoxicity of CDDP. SSF is an herbal formulation with 8 herbs, and has been used to protect the kidney in China. Nonetheless, its mechanism in relieving CDDP nephrotoxicity remains unclear. Therefore, this work attempt to prove that SSF can alleviate CDDP nephrotoxicity. We also explore its mechanism. MATERIALS AND METHODS First, Thin Layer Chromatography (TLC) of a few herbs in SSF were performed for quality control. Several open-access databases were used to identify the active ingredients of SSF, their corresponding targets, and CDDP-induced nephrotoxicity targets. We performed Protein-Protein Interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Next, the results of network pharmacology were validated using CDDP-induced nephrotoxicity mouse models. Renal function in the mice was assessed by analyzing the levels of serum creatinine (Scr) and blood urea nitrogen (BUN). On the other hand, renal damage was assessed by determining the level of tubular injury and apoptotic cells using Periodic acid-Schiff (PAS) staining and Terminal Dutp Nick End-Labeling (TUNEL) staining, respectively. The expression of inflammatory and apoptotic-related targets including IL-1β, IL-6, TNF-α, Cox-2, Bax, Bcl-2, Cleaved-caspase 3, and Cleaved-caspase 9 was determined using Western Blot (WB) and Immunohistochemistry (IHC). Furthermore, WB was used to analyze the expression of proteins associated with the TLR4/MyD88/NF-κB pathway in the kidneys of mice with CDDP-induced nephrotoxicity. Finally, molecular docking simulations were performed to evaluate the binding abilities between major active ingredients of SSF and core targets. RESULT Through network pharmacology, we identified 127 active ingredients of SSF and their corresponding 134 targets. Additional screening identified 14 active ingredients and 17 targets for further analysis. In biological process (BP), the targets were enriched in inflammation and apoptosis, among others. In KEGG terms, they were enriched in apoptosis and NF-κB pathways. Animal experiments revealed that SSF significantly reduced the levels of Scr and BUN and prevented renal tubular damage in mice treated with CDDP. In addition, SSF inhibited inflammation and apoptosis by targeting the TLR4/MyD88/NF-κB pathway. Molecular docking revealed good binding capacities of active ingredients and core targets. CONCLUSION In summary, the experimental findings were consistent with the network pharmacological predictions. SSF can inhibit inflammation and apoptosis by targeting the TLR4/MyD88/NF-κB pathway. Taken together, our data suggest that SSF is an alternative agent for the treatment of CDDP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Xiaoming Jin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Riming He
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Jiahui Liu
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Yuzhi Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Zhongtang Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Beibei Jiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Shenzhen, 518033, China.
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| |
Collapse
|
18
|
Wang R, Huang Y, Yu L, Li S, Li J, Han B, Zheng X, Zhang Z. The role of mitochondrial dynamics imbalance in hexavalent chromium-induced apoptosis and autophagy in rat testis. Chem Biol Interact 2023; 374:110424. [PMID: 36849043 DOI: 10.1016/j.cbi.2023.110424] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023]
Abstract
Hexavalent chromium (Cr(VI)) is a ubiquitous environmental pollutant that can cause reproductive toxicity. However, the exact mechanism of Cr(VI)-induced testis toxicity remains largely elusive. This study aims to explore the possible molecular mechanism of Cr(VI)-provoked testicular toxicity. Male Wistar rats were intraperitoneally injected with 0, 2, 4, or 6 mg/kg body weight/day of potassium dichromate (K2Cr2O7), respectively, for 5 weeks. The results revealed that Cr(VI)-treated rat testis presented varying degrees of damage in a dose-dependent manner. Concretely, Cr(VI) administration suppressed Sirtuin 1/Peroxisome proliferator-activated receptor-γ coactivator-1α pathway and led to mitochondrial dynamics disorder, along with the elevation of mitochondrial division and the repression of mitochondrial fusion. Meanwhile, the downstream effector of Sirt1, nuclear factor-erythroid-2-related factor 2 (Nrf2), was downregulated, and correspondingly exacerbated oxidative stress. Mitochondrial dynamics disorder and Nrf2 inhibition collectively contribute to abnormal mitochondrial dynamics in testis, which further promotes apoptosis and autophagy, evidenced by dose-dependently increasing the protein levels and gene expressions of apoptosis-related (including Bcl-2-associated X protein, cytochrome c, and cleaved-caspase 3) and autophagy-related (Beclin-1, ATG4B, and ATG5). Collectively, our results demonstrate that Cr(VI) exposure induced testis apoptosis and autophagy by disrupting the balance of mitochondrial dynamics and the oxidation-reduction process in rats.
Collapse
Affiliation(s)
- Ruonan Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yuxiang Huang
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar, 161000, China
| | - Lu Yu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xiaoyan Zheng
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.
| |
Collapse
|
19
|
Huang J, Liang Y, Zhou L. Natural products for kidney disease treatment: Focus on targeting mitochondrial dysfunction. Front Pharmacol 2023; 14:1142001. [PMID: 37007023 PMCID: PMC10050361 DOI: 10.3389/fphar.2023.1142001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
The patients with kidney diseases are increasing rapidly all over the world. With the rich abundance of mitochondria, kidney is an organ with a high consumption of energy. Hence, renal failure is highly correlated with the breakup of mitochondrial homeostasis. However, the potential drugs targeting mitochondrial dysfunction are still in mystery. The natural products have the superiorities to explore the potential drugs regulating energy metabolism. However, their roles in targeting mitochondrial dysfunction in kidney diseases have not been extensively reviewed. Herein, we reviewed a series of natural products targeting mitochondrial oxidative stress, mitochondrial biogenesis, mitophagy, and mitochondrial dynamics. We found lots of them with great medicinal values in kidney disease. Our review provides a wide prospect for seeking the effective drugs targeting kidney diseases.
Collapse
|
20
|
Chen H, Qi X, Guan K, Wang R, Li Q, Ma Y. Tandem mass tag-based quantitative proteomics analysis reveals the effects of the α-lactalbumin peptides GINY and DQW on lipid deposition and oxidative stress in HepG2 cells. J Dairy Sci 2023; 106:2271-2288. [PMID: 36797178 DOI: 10.3168/jds.2022-22511] [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: 07/12/2022] [Accepted: 09/28/2022] [Indexed: 02/16/2023]
Abstract
The objective of this study was to investigate the mechanism by which the α-lactalbumin peptides Gly-Ile-Asn-Tyr (GINY) and Asp-Gln-Trp (DQW) ameliorate free fatty acid-induced lipid deposition in HepG2 cells. The results show that GINY and DQW reduced triglyceride, total cholesterol, and free fatty acid levels significantly in free fatty acid-treated HepG2 cells. Based on proteomic analysis, GINY and DQW alleviated lipid deposition and oxidative stress mainly through the peroxisome proliferator-activated receptor (PPAR) pathway, fatty acid metabolism, oxidative phosphorylation, and response to oxidative stress. In vitro experiments confirmed that GINY and DQW upregulated the mRNA and protein expression of fatty acid β-oxidation-related and oxidative stress-related genes, and downregulated the mRNA and protein expression of lipogenesis-related genes by activating peroxisome proliferator-activated receptor α (PPARα). Meanwhile, GINY and DQW reduced free fatty acid-induced lipid droplet accumulation and reactive oxygen species generation, and enhanced the mitochondrial membrane potential and ATP levels. Furthermore, GINY and DQW enhanced carnitine palmitoyl-transferase 1a (CPT-1a) and superoxide dismutase activities, and diminished acetyl-coenzyme A carboxylase 1 (ACC1) and fatty acid synthase (FASN) activities in a PPARα-dependent manner. Interestingly, GW6471 (a PPARα inhibitor) weakened the effects of GINY and DQW on the PPARα pathway. Hence, our findings suggest that GINY and DQW have the potential to alleviate nonalcoholic fatty liver disease by activating the PPARα pathway.
Collapse
Affiliation(s)
- Haoran Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Xiaofen Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Qiming Li
- New Hope Dairy Co. Ltd., Chengdu, 610063, Sichuan, China; Dairy Nutrition and Function, Key Laboratory of Sichuan Province, Chengdu, 610000, Sichuan, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
21
|
Demirtas L, Gürbüzel M, Akbas EM, Tahirler H, Karatas O, Kemal Arslan Y. The Effects of Sunitinib in Healthy and Cisplatin-Induced Rats. Chem Biodivers 2023; 20:e202200704. [PMID: 36703598 DOI: 10.1002/cbdv.202200704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/04/2023] [Indexed: 01/28/2023]
Abstract
Sunitinib is a multitargeted kinase inhibitor that inhibits many receptor tyrosine kinases and has been used in the treatment of gastrointestinal stromal tumors, metastatic renal cell carcinoma, and pancreatic neuroendocrine tumors. In this study, the effects of sunitinib given to rats, both alone and after stress with cisplatin, were investigated. The animals were divided into four groups - (1) control group (C) administered interperitoneally with a single dose 0.9 % saline, (2) Cis group administered a single dose (7 mg/kg) of cisplatin, (3) Sun group administered 10 mg/kg sunitinib for seven days, and (4) Cis+Sun group administered 10 mg/kg sunitinib for seven days after a single dose (7 mg/kg) of cisplatin. After these applications, the rats were sacrificed, and blood and tissue samples were taken for biochemical and histopathological evaluations. Sunitinib did not show any effect on urea, creatine, and kidney IL1β and TGF-β3 expression levels when administered alone; it increased ALT, AST, and IL-38 levels. When sunitinib was given to the cisplatin-induced rats, it was observed that the increase in ALT, AST, and IL-38 levels increased more than the rats that was given only sunitinib. According to the data obtained, sunitinib does not cause a significant change in kidney tissue under both normal and stress conditions, while it creates stress in liver tissue. In addition, its toxicity in the liver becomes more certain as a result of its combination with cisplatin.
Collapse
Affiliation(s)
- Levent Demirtas
- Department of Internal Medicine, Faculty of Medicine, Erzincan Binali Yıldırım University, 24100, Erzincan, Türkiye
| | - Mehmet Gürbüzel
- Department of Medical Biology, Faculty of Medicine, Erzincan Binali Yıldırım University, 24100, Erzincan, Türkiye
| | - Emin Murat Akbas
- Department of Endocrinology, Faculty of Medicine, Erzincan Binali Yıldırım University, 24100, Erzincan, Türkiye
| | - Hilal Tahirler
- Department of Internal Medicine, Gülhane Training and Research Hospital, Health Sciences University, 06010, Ankara, Türkiye
| | - Ozhan Karatas
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, 58140, Sivas, Türkiye
| | - Yusuf Kemal Arslan
- Department of Biostatistics, Medical Faculty, Çukurova University, 01330, Adana, Türkiye
| |
Collapse
|
22
|
Qi X, Wang J, Fei F, Gao X, Wu X, Shi D, Guo C. Myricetin-Loaded Nanomicelles Protect against Cisplatin-Induced Acute Kidney Injury by Inhibiting the DNA Damage-cGAS-STING Signaling Pathway. Mol Pharm 2023; 20:136-146. [PMID: 36326450 DOI: 10.1021/acs.molpharmaceut.2c00520] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acute kidney injury (AKI) is the most common side effect of the anti-cancer drug cisplatin, and currently, no effective preventive measures are available in clinical practice. Oxidative stress and DNA damage mechanisms may be involved in cisplatin-induced AKI. In this study, we prepared Kolliphor HS15-based myricetin-loaded (HS15-Myr) nanomicelles and explored the mechanism of protection against cisplatin-induced AKI. In vitro results showed that the HS15-Myr nanomicelles enhanced the antioxidant activity of myricetin (Myr) and inhibited cisplatin-induced proliferation inhibition of HK-2 cells. Moreover, the HS15-Myr nanomicelles inhibited cisplatin-induced reactive oxygen species accumulation, mitochondrial membrane potential reduction, and DNA damage, which might be related to the inhibition of the cyclic GMP-AMP synthase (cGAS)─stimulating interferon gene (STING) signaling pathway. In vivo results in mice showed that the significant reductions in body weight and renal indices and the increased blood urea nitrogen and serum creatinine levels induced by cisplatin could be significantly reversed by pretreating with the HS15-Myr nanomicelles. Furthermore, nanomicelle pretreatment significantly altered the activities of antioxidant enzymes (e.g., GSH, MDA, and SOD) induced by cisplatin. In addition, cisplatin-induced inflammatory responses in mouse kidney tissue were found to be inhibited by pretreatment with HS15-Myr nanomicelles, such as IL-1β and TNF-α expression. The nanomicelles also significantly inhibited cisplatin-induced activation of the DNA damage-cGAS-STING pathway in kidney tissues. Together, our findings suggest that Myr-loaded nanomicelles are potential nephroprotective drugs.
Collapse
Affiliation(s)
- Xueju Qi
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jing Wang
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou 014030, China
| | - Fengshu Fei
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xintao Gao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiaochen Wu
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266273 Shandong, China
| | - Chuanlong Guo
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.,State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266273 Shandong, China
| |
Collapse
|
23
|
Sha Q, Huang Y, Zhou F, Wang Y, Fang W, Li J. Effects of polydatin from Huhuang Shaoshang Liniment on oxidative damage and inflammatory response in rats with cerebral ischemia-reperfusion. Am J Transl Res 2023; 15:995-1006. [PMID: 36915795 PMCID: PMC10006806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 01/02/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVE To analyze the effect of polydatin (PD) from Huhuang Shaoshang Liniment on oxidative damage and inflammatory response in rats with cerebral ischemia-reperfusion (CI/R), and the effect of PD on NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1)/NAD(P)H quinone oxidoreductase 1 (NQO1) pathway. METHODS The rat model of focal CI/R was established using suture-occlusion method and treated with intraperitoneal injection of PD (25 mg/kg and 50 mg/kg). High performance liquid chromatography (HPLC) was used to determine the PD content in three batches of Huhuang Shaoshang Liniment. The neurological function scores of each group were observed at 24 h, 48 h, and 72 h postoperatively. The hematoxylin and eosin (HE) staining was conducted to observe the morphological structure of brain tissue, the triphenyltetrazolium chloride (TTC) staining was used to determine the size of cerebral infarction, and the neuronal apoptotic index was calculated using image analysis system under the optical microscope. The expressions of the Nrf2/HO-1/NQO1 pathway and neuronal apoptosis-related proteins in brain tissue were measured using Western blot. RESULTS The PD content in three batches of Huhuang Shaoshang Liniment was detected by HPLC, and the average result showed that the product contained 0.73 mg PD per 1 mL. The PD 50 mg/kg group and 25 mg/kg group showed lower neurological function scores at 24 h, 48 h, and 72 h postoperatively, lower percentage of cerebral infarction area on the ischemic side and apoptotic index, lower interleukin (IL)-1β, IL-6, malondialdehyde (MDA), and lactate dehydrogenase (LDH) levels, and lower Bax protein expression (P < 0.05), and showed higher IL-4, IL-10, superoxide dismutase (SOD), glutathione peroxidase (GPx), Nrf2, HO-1, NQO1, and c-Myc protein expression than the CI/R group (P < 0.05). CONCLUSION PD from Huhuang Shaoshang Liniment can alleviate neurological damage, improve neuronal morphology and structure, reduce cerebral infarction area, alleviate oxidative damage and inflammatory response, and inhibit neuronal cell apoptosis in CI/R model rats.
Collapse
Affiliation(s)
- Qiankun Sha
- Department of Pharmacy, Chongqing Yangdu Biology Institute Chongqing 408000, China
| | - Yan Huang
- Department of Pharmacy, The People's Hospital of Nanchuan Chongqing 408499, China
| | - Faying Zhou
- Department of General Surgery, Qianjiang Central Hospital of Chongqing Chongqing 409000, China
| | - Yong Wang
- Department of Dermatology, Qijiang District Traditional Chinese Medicine Hospital of Chongqing Chongqing 401420, China
| | - Wei Fang
- Department of Pharmacy, Chongqing Sanxia Central Hospital Chongqing 404100, China
| | - Jing Li
- Department of Pharmacy, People's Hospital of Chongqing Banan District, Chongqing 401320, China
| |
Collapse
|
24
|
Formononetin Inhibits Hepatic I/R-Induced Injury through Regulating PHB2/PINK1/Parkin Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6481192. [PMID: 36506934 PMCID: PMC9734001 DOI: 10.1155/2022/6481192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/03/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022]
Abstract
Formononetin (FN), an isoflavone compound mainly isolated from soy and red clover, had showed its anti-inflammation, antioxidative effects in some degenerative diseases and cholestasis. However, the role of FN in protecting ischemia/reperfusion- (I/R-) induced liver injury and the possible mechanism were unclear. In this study, effects of FN on liver injury were investigated in a rat hepatic I/R model; further, mitophagy-related proteins were measured by immunoblotting or immunofluorescence. The possible roles of PHB2 and PINK1 in regulating mitophagy by FN were verified using adeno-associated virus knockdown. The results showed that FN had protective effects against hepatic I/R injury through regulating PINK1/Parkin-regulated mitophagy. Further, we found that FN inhibited PARL expression and prevented PGAM5 cropped by increasing the expression of PHB2. The knockdown of PINK1 or PHB2 both abolished the protective effects of FN. Taken together, our findings indicated that the isoflavone compound FN promoted PHB2/PINK1/Parkin-mediated mitophagy pathway to protect liver from I/R-induced injury. These results provided novel insights into the potential prevention strategies of FN and its underlying mechanisms.
Collapse
|
25
|
Theaflavin-3,3'-Digallate Inhibits Erastin-Induced Chondrocytes Ferroptosis via the Nrf2/GPX4 Signaling Pathway in Osteoarthritis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3531995. [PMID: 36439689 PMCID: PMC9691334 DOI: 10.1155/2022/3531995] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022]
Abstract
There is evidence that osteoarthritis (OA) is associated with ferroptosis which is a kind of lipid peroxidation-related cell death. Theaflavin-3,3'-digallate(TF3), a polyphenol compound extracted from black tea, possesses antioxidative and anti-inflammatory properties, but its effects on chondrocyte ferroptosis in osteoarthritis (OA) remain unclear. Our present study aims at exploring the protective role and underlying mechanisms of TF3 against erastin-induced chondrocyte ferroptosis in OA. In human primary chondrocytes treated with erastin alone or combined with different doses of TF3, cell viability was assessed by MTS. Ferroptosis-related proteins, including Gpx4, HO-1, and FTH1, were detected by western blot. The levels of lipid peroxidation and Fe2+ were determined by fluorescence staining. Meanwhile, the change of related proteins in the Nrf2/Gpx4 signaling pathway was determined by western blot. siRNA-mediated Nrf2 knockdown and the Gpx4 inhibitor RSL3 were used to explore molecular mechanisms for TF3-induced ferroptosis in OA chondrocyte. The magnetic resonance imaging (MRI), HE staining, Masson's staining, and immunohistochemistry were used to evaluate articular cartilage damages in the rat OA model. The results showed that Gpx4 expression was markedly downregulated in the chondrocytes of OA patients. TF3 reversed erastin-induced ferroptosis of human cultured chondrocytes, lipid ROS, and Fe2+ production in mitochondria. Moreover, the expression of Gpx4, HO-1, FTH1, and Nrf2 was markedly induced by TF3 in the erastin-treated chondrocytes. The antiferroptotic effect of TF3 was related to enhance Nrf2/Gpx4 signaling pathway. Finally, TF3 inhibited OA progression by alleviating in vivo cartilage damage related to chondrocyte ferroptosis. Thus, TF3 significantly inhibits chondrocyte ferroptosis by activating the Nrf2/Gpx4 signaling pathway, suggesting that TF3 serves as a potential therapeutic supplement for OA treatment.
Collapse
|
26
|
Kim DU, Kweon B, Oh JY, Seo CS, Kim DG, Kim HY, Lee HS, Park SJ, Bae GS. Ojeoksan Ameliorates Cisplatin-Induced Acute Kidney Injury in Mice by Downregulating MAPK and NF-κB Pathways. Int J Mol Sci 2022; 23:ijms232012254. [PMID: 36293111 PMCID: PMC9603434 DOI: 10.3390/ijms232012254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022] Open
Abstract
Acute kidney injury (AKI) is a major side effect of cisplatin, a crucial anticancer agent. Therefore, it is necessary to develop drugs to protect against cisplatin-induced nephrotoxicity. Ojeoksan (OJS), a traditional blended herbal prescription, is mostly used in Korea; however, there are no reports on the efficacy of OJS against cisplatin-induced AKI. To investigate the reno-protective effect of OJS on AKI, we orally administered 50, 100, and 200 mg/kg of OJS to mice 1 h before intraperitoneal injection with 20 mg/kg of cisplatin. OJS inhibited the increase of blood urea nitrogen (BUN) and serum creatinine (SCr) levels and reduced histological changes in the kidney, like loss of brush borders, renal tubular necrosis, and cast formation. Administration of OSJ reduced the levels of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. In addition, OJS inhibited the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways in cisplatin-induced AKI. These results suggest that OJS attenuates cisplatin-induced AKI by downregulating the MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Dong-Uk Kim
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
| | - Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
| | - Jin-Young Oh
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
| | - Chang-Seob Seo
- KM Science Research Division, Korea Institute of Oriental Medicine, Yuseong-gu, Daejeon 34054, Korea
| | - Dong-Gu Kim
- Department of Oriental Medicine Resources, Jeonbuk National University, 79 Gobong-ro, Iksan 54596, Korea
| | - Hye-Yoom Kim
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
| | - Ho-Sub Lee
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
| | - Sung-Joo Park
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
| | - Gi-Sang Bae
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan 54538, Korea
- Correspondence: ; Tel.: +82-63-850-6842
| |
Collapse
|
27
|
Zhang J, Zhao Y, Wang S, Li G, Xu K. CREBH alleviates mitochondrial oxidative stress through SIRT3 mediating deacetylation of MnSOD and suppression of Nlrp3 inflammasome in NASH. Free Radic Biol Med 2022; 190:28-41. [PMID: 35926687 DOI: 10.1016/j.freeradbiomed.2022.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/23/2022] [Accepted: 07/24/2022] [Indexed: 02/07/2023]
Abstract
Lipotoxicity and unresolved oxidative stress are key drivers of metabolic inflammation in nonalcoholic steatohepatitis (NASH). cAMP-response element binding protein H(CREBH) is a liver-specific transcription factor and regulates the glucose and lipid metabolism of NASH. However, its role in mitochondrial oxidative stress and its association with sirtuin 3 (SIRT3), a master regulator of deacetylation for mitochondrial proteins, remains elusive. In this study, AML-12 cells were treated with palmitic acid to imitate the pathological changes of NASH in vitro and 8-week-old male C57BL/6J mice were fed with a high-fat (HF) diet or a methionine-choline-deficient (MCD) diet to build the widely accepted in vivo model of NASH. We found that lipid overload induced mitochondrial oxidative stress and stimulated the expression of CREBH and SIRT3. CREBH overexpression alleviated the mitochondrial oxidative stress. Moreover, CREBH promoted SIRT3 expression, which regulated the deacetylation of manganese superoxide dismutase (MnSOD) and inhibited NOD-Like Receptor Pyrin Domain Containing 3 (Nlrp3) inflammasome activation whereas suppression of SIRT3 damaged the protecting ability of CREBH in mitochondrial oxidative stress. CREBH knockout mice were highly susceptible to HF and MCD diet-induced NASH with more severe oxidative stress. Collectively, our results firstly provided the support that CREBH could serve as a protective factor in the progression of NASH by regulating the acetylation of MnSOD and the activation of Nlrp3 inflammasome through SIRT3. These results suggest that CREBH might be a valuable therapeutic candidate for NASH.
Collapse
Affiliation(s)
- Junli Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yajuan Zhao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuhan Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guixin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
28
|
Cisplatin-Induced Kidney Toxicity: Potential Roles of Major NAD +-Dependent Enzymes and Plant-Derived Natural Products. Biomolecules 2022; 12:biom12081078. [PMID: 36008971 PMCID: PMC9405866 DOI: 10.3390/biom12081078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is an FDA approved anti-cancer drug that is widely used for the treatment of a variety of solid tumors. However, the severe adverse effects of cisplatin, particularly kidney toxicity, restrict its clinical and medication applications. The major mechanisms of cisplatin-induced renal toxicity involve oxidative stress, inflammation, and renal fibrosis, which are covered in this short review. In particular, we review the underlying mechanisms of cisplatin kidney injury in the context of NAD+-dependent redox enzymes including mitochondrial complex I, NAD kinase, CD38, sirtuins, poly-ADP ribosylase polymerase, and nicotinamide nucleotide transhydrogenase (NNT) and their potential contributing roles in the amelioration of cisplatin-induced kidney injury conferred by natural products derived from plants. We also cover general procedures used to create animal models of cisplatin-induced kidney injury involving mice and rats. We highlight the fact that more studies will be needed to dissect the role of each NAD+-dependent redox enzyme and its involvement in modulating cisplatin-induced kidney injury, in conjunction with intensive research in NAD+ redox biology and the protective effects of natural products against cisplatin-induced kidney injury.
Collapse
|
29
|
Wang Q, Ma X. Gut microbial sodium butyrate alleviates renal ischemia-reperfusion injury by regulating HES1/PPARα. Mol Immunol 2022; 150:20-28. [PMID: 35930845 DOI: 10.1016/j.molimm.2022.07.009] [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: 01/19/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022]
Abstract
This study investigated the effect of gut microbial sodium butyrate (NaB) on renal ischemia-reperfusion injury (IRI) and its mechanism using a rat model of renal IRI and a HK-2 cell model of hypoxia-reoxygenation (HR) injury. The activity of malondialdehyde, superoxide dismutase, glutathione peroxidase, and catalase in kidney tissues and HK-2 cells was detected. ELISA was performed to measure the concentrations of TNF-α, IL-1β, and IL-6 in serum and cell culture supernatant. TUNEL staining and flow cytometry were used to assess apoptosis in kidney tissues and HK-2 cells, respectively. UCSC and JASPAR predicted the binding sites between HES1 and PPARα promoter, followed by experimental verification of the binding. NaB pretreatment inhibited oxidative stress, inflammation, and apoptosis following renal IRI in vivo and in vitro. NaB suppressed the expression of HES1 and promoted that of PPARα. Overexpression of HES1 or knockdown of PPARα in HR-treated HK-2 cells inhibited the protective effects of NaB. HES1 repressed the expression of PPARα by binding PPARα promoter. In conclusion, NaB may alleviate renal IRI by promoting the transcription of PPARα via downregulation of HES1.
Collapse
Affiliation(s)
- Qiong Wang
- Kidney Disease and Dialysis Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China
| | - Xiaoying Ma
- Kidney Disease and Dialysis Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China; Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China.
| |
Collapse
|
30
|
Tang JL, Xin M, Zhang LC. Protective effect of Astragalus membranaceus and Astragaloside IV in sepsis-induced acute kidney injury. Aging (Albany NY) 2022; 14:5855-5877. [PMID: 35859295 PMCID: PMC9365550 DOI: 10.18632/aging.204189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 07/11/2022] [Indexed: 12/29/2022]
Abstract
Background: Acute kidney injury (AKI) is the most common target organ damage in sepsis. Sepsis-associated AKI (SA-AKI) may be characterized by damage to the renal tubular epithelium. In this study, the pharmacological mechanisms of Astragalus membranaceus and its active monomer Astragaloside IV (AS-IV) were predicted based on a network pharmacology approach and validated both in vitro and in vivo using the SA-AKI model. Method: We constructed an in vivo sepsis model using a mouse cecum ligation puncture (CLP) and HK-2 cells were treated with lipopolysaccharide (LPS) to mimic Gram (–) induced sepsis to assess the renal-protective efficacy of Astragalus membranaceus and AS-IV. Results: The findings demonstrated that Astragalus membranaceus and AS-IV attenuate renal tubular injury in mice with polymicrobial sepsis, including vacuolization, loss of brush border, mitochondrial ultrastructural changes, and increased staining of kidney injury molecule-1 (KIM-1). AS-IV protected human proximal tubular epithelial (HK-2) cells against LPS induced cell viability loss. Both Astragalus membranaceus and AS-IV activated the PI3K/AKT pathway both in vitro and in vivo, as shown by Western blot and immunohistochemistry analysis. Conclusion: The findings demonstrate that Astragalus membranaceus and AS-IV protect against sepsis-induced kidney tubular injury by activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Jia-Long Tang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Xin
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
31
|
Althunibat OY, Abukhalil MH, Aladaileh SH, Qaralleh H, Al-Amarat W, Alfwuaires MA, Algefare AI, Namazi NI, Melebary SJ, Babalghith AO, Conte-Junior CA. Formononetin Ameliorates Renal Dysfunction, Oxidative Stress, Inflammation, and Apoptosis and Upregulates Nrf2/HO-1 Signaling in a Rat Model of Gentamicin-Induced Nephrotoxicity. Front Pharmacol 2022; 13:916732. [PMID: 35712704 PMCID: PMC9197219 DOI: 10.3389/fphar.2022.916732] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022] Open
Abstract
Gentamicin (GEN) is a bactericidal aminoglycoside known to cause nephrotoxicity. Formononetin (FN) is a potent flavonoid that exhibits numerous promising pharmacological activities. In this study, we have assessed the nephroprotective efficacy of FN against GEN-induced renal injury in rats. Rats were orally administered with FN (60 mg/kg/day, for 2 weeks) and were co-treated with intraperitoneal (i.p.) injection of GEN (100 mg/kg/day) during the days 8–14. GEN-treated rats demonstrated increased urea and creatinine levels in serum associated with marked histopathological changes in the kidney. Malondialdehyde (MDA) and protein carbonyl contents were elevated, whereas glutathione concentration and catalase and superoxide dismutase activities were lowered in GEN-administered rats. The FN largely prevented tissue damage, attenuated renal function, reduced MDA and protein carbonyl, and enhanced antioxidant capacity in the kidney of GEN-administrated animals. The kidney of GEN-treated rats demonstrated elevated Bax and caspase-3 protein expression, accompanied by lowered Bcl-2 protein expression, an effect that FN attenuated. Moreover, FN treatment caused upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) expression in renal tissue of GEN-intoxicated animals. Collectively, FN protects against GEN-caused renal damage via exhibiting antioxidant, anti-inflammatory, and antiapoptotic activities and augmenting Nrf2 signaling, suggesting FN as a promising agent for preventing drug-induced organ damage.
Collapse
Affiliation(s)
- Osama Y. Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an, Jordan
- *Correspondence: Osama Y. Althunibat, ; Mohammad H. Abukhalil,
| | - Mohammad H. Abukhalil
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an, Jordan
- Department of Biology, College of Science, Al-Hussein Bin Talal University, Ma’an, Jordan
- *Correspondence: Osama Y. Althunibat, ; Mohammad H. Abukhalil,
| | - Saleem H. Aladaileh
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an, Jordan
| | - Haitham Qaralleh
- Department of Medical Laboratory Sciences, Mutah University, Karak, Jordan
| | - Wesam Al-Amarat
- Department of Medical Support, Al-karak University College, Al-Balqa’ Applied University, As-Salt, Jordan
| | - Manal A. Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abdulmohsen I. Algefare
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Nader Ibrahim Namazi
- Pharmaceutics and Pharmaceutical Technology Department, College of Pharmacy, Taibah University, Al Madinah Al Munawarah, Saudi Arabia
| | - Sahar J. Melebary
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahmad O. Babalghith
- Medical Genetics Department, College of Medicine, Umm al-qura University, Makkah, Saudi Arabia
| | - Carlos Adam Conte-Junior
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Elhady SS, Abdelhameed RFA, Mehanna ET, Wahba AS, Elfaky MA, Koshak AE, Noor AO, Bogari HA, Malatani RT, Goda MS. Metabolic Profiling, Chemical Composition, Antioxidant Capacity, and In Vivo Hepato- and Nephroprotective Effects of Sonchus cornutus in Mice Exposed to Cisplatin. Antioxidants (Basel) 2022; 11:819. [PMID: 35624682 PMCID: PMC9137627 DOI: 10.3390/antiox11050819] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Sonchus cornutus (Asteraceae) is a wild. edible plant that represents a plentiful source of polyphenolic compounds. For the first time, the metabolic analysis profiling demonstrated the presence of anthocyanidin glycosides, coumarins, flavonoids and their corresponding glycosides, and phenolic acids. The total phenolic compounds were determined to be 206.28 ± 14.64 mg gallic acid equivalent/gm, while flavonoids were determined to be 45.56 ± 1.78 mg quercetin equivalent/gm. The crude extract of S. cornutus exhibited a significant 1,1-diphenyl-2-picrylhydrazyl free radical scavenging effect with half-maximal inhibitory concentration (IC50) of 16.10 ± 2.14 µg/mL compared to ascorbic acid as a standard (10.64 ± 0.82 µg/mL). In vitro total antioxidant capacity and ferric reducing power capacity assays revealed a promising reducing potential of S. cornutus extract. Therefore, the possible protective effects of S. cornutus against hepatic and renal toxicity induced by cisplatin in experimental mice were investigated. S. cornutus significantly ameliorated the cisplatin-induced disturbances in liver and kidney functions and oxidative stress, decreased MDA, ROS, and NO levels, and restored CAT and SOD activities. Besides, it reversed cisplatin-driven upregulation in inflammatory markers, including iNOS, IL-6, and IL-1β levels and NF-κB and TNF-α expression, and elevated anti-inflammatory IL-10 levels and Nrf2 expression. Additionally, the extract mitigated cisplatin alteration in apoptotic (Bax and caspase-3) and anti-apoptotic (Bcl-2) proteins. Interestingly, hepatic, and renal histopathology revealed the protective impacts of S. cornutus against cisplatin-induced pathological changes. Our findings guarantee a protective effect of S. cornutus against cisplatin-induced hepatic and renal damage via modulating oxidative stress, inflammation, and apoptotic pathways.
Collapse
Affiliation(s)
- Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.A.E.); (A.E.K.)
| | - Reda F. A. Abdelhameed
- Department of Pharmacognosy, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Eman T. Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.T.M.); (A.S.W.)
| | - Alaa Samir Wahba
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.T.M.); (A.S.W.)
| | - Mahmoud A. Elfaky
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.A.E.); (A.E.K.)
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulrahman E. Koshak
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.A.E.); (A.E.K.)
| | - Ahmad O. Noor
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.O.N.); (H.A.B.); (R.T.M.)
| | - Hanin A. Bogari
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.O.N.); (H.A.B.); (R.T.M.)
| | - Rania T. Malatani
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.O.N.); (H.A.B.); (R.T.M.)
| | - Marwa S. Goda
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| |
Collapse
|
33
|
Ma T, Ma Y, Yu Y, Jia L, Lv L, Song X, Tang J, Xu X, Sheng X, Li T, Gao L. Emodin Attenuates the ECM Degradation and Oxidative Stress of Chondrocytes through the Nrf2/NQO1/HO-1 Pathway to Ameliorate Rat Osteoarthritis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5581346. [PMID: 39346968 PMCID: PMC11427723 DOI: 10.1155/2022/5581346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/06/2021] [Accepted: 12/24/2021] [Indexed: 10/01/2024]
Abstract
Osteoarthritis (OA) substantially reduces the quality of life of the elderly. OA therapy remains a challenge since no treatment options for its causes are so far available. Over recent years, researchers have speculated that emodin may represent a potential treatment strategy for OA. However, it remains unclear whether the mechanism of action of emodin is associated with the inhibition of OA-induced oxidative stress. In the present study, the potential antioxidant mechanism of action of emodin and its protective properties against the development of OA were investigated both in vitro and in vivo. In vitro, emodin inhibited the production of reactive oxygen species (ROS) in chondrocytes induced by hydrogen peroxide (H2O2) and reduced the expression of matrix metalloproteinase (MMP)3 and MMP13 in a concentration-dependent manner. It was found that emodin upregulated the Nrf2/NQO1/HO-1 pathway, thereby attenuating the effects of oxidative stress caused by OA. In a rat model of posttraumatic OA induced by anterior cruciate ligament transection (ACLT), emodin reduced the extent of joint swelling. Emodin attenuated oxidative damage in the cartilage by upregulating superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) activity, reducing malondialdehyde (MDA) concentration and inhibiting the expression of the extracellular matrix (ECM) degradation biomarkers cartilage oligomeric matrix protein (COMP), and C-terminal telopeptide of type I collagen (CTX-I) and type II collagen (CTX-II), thereby reducing cartilage damage. In summary, the present study indicates that emodin reduces ECM degradation and oxidative stress in chondrocytes via the Nrf2/NQO1/HO-1 pathway, thereby ameliorating OA in rats.
Collapse
Affiliation(s)
- Tianwen Ma
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yuanqiang Ma
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yue Yu
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Lina Jia
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Liangyu Lv
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaopeng Song
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jilang Tang
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xinyu Xu
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xuanbo Sheng
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ting Li
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Li Gao
- Heilongjiang Key Laboratory for Animal Disease Pathogenesis and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
34
|
Templeton EM, Lassé M, Kleffmann T, Ellmers LJ, Palmer SC, Davidson T, Scott NJA, Pickering JW, Charles CJ, Endre ZH, Cameron VA, Richards AM, Rademaker MT, Pilbrow AP. Identifying Candidate Protein Markers of Acute Kidney Injury in Acute Decompensated Heart Failure. Int J Mol Sci 2022; 23:ijms23021009. [PMID: 35055195 PMCID: PMC8778509 DOI: 10.3390/ijms23021009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022] Open
Abstract
One-quarter of patients with acute decompensated heart failure (ADHF) experience acute kidney injury (AKI)—an abrupt reduction or loss of kidney function associated with increased long-term mortality. There is a critical need to identify early and real-time markers of AKI in ADHF; however, to date, no protein biomarkers have exhibited sufficient diagnostic or prognostic performance for widespread clinical uptake. We aimed to identify novel protein biomarkers of AKI associated with ADHF by quantifying changes in protein abundance in the kidneys that occur during ADHF development and recovery in an ovine model. Relative quantitative protein profiling was performed using sequential window acquisition of all theoretical fragment ion spectra–mass spectrometry (SWATH–MS) in kidney cortices from control sheep (n = 5), sheep with established rapid-pacing-induced ADHF (n = 8), and sheep after ~4 weeks recovery from ADHF (n = 7). Of the 790 proteins quantified, we identified 17 candidate kidney injury markers in ADHF, 1 potential kidney marker of ADHF recovery, and 2 potential markers of long-term renal impairment (differential abundance between groups of 1.2–2.6-fold, adjusted p < 0.05). Among these 20 candidate protein markers of kidney injury were 6 candidates supported by existing evidence and 14 novel candidates not previously implicated in AKI. Proteins of differential abundance were enriched in pro-inflammatory signalling pathways: glycoprotein VI (activated during ADHF development; adjusted p < 0.01) and acute phase response (repressed during recovery from ADHF; adjusted p < 0.01). New biomarkers for the early detection of AKI in ADHF may help us to evaluate effective treatment strategies to prevent mortality and improve outcomes for patients.
Collapse
Affiliation(s)
- Evelyn M. Templeton
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
- Correspondence: ; Tel.: +64-03-364-12-53
| | - Moritz Lassé
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Torsten Kleffmann
- Research Infrastructure Centre, Division of Health Sciences, University of Otago, Dunedin 9016, New Zealand;
| | - Leigh J. Ellmers
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Suetonia C. Palmer
- Department of Medicine, University of Otago, Christchurch 8014, New Zealand;
| | - Trent Davidson
- Department of Anatomical Pathology, Prince of Wales Hospital, Sydney, NSW 2031, Australia;
| | - Nicola J. A. Scott
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - John W. Pickering
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Christopher J. Charles
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Zoltan H. Endre
- Department of Nephrology, Prince of Wales Hospital, Sydney, NSW 2031, Australia;
| | - Vicky A. Cameron
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - A. Mark Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
- Cardiovascular Research Institute, Department of Cardiology, National University of Singapore, Singapore 119077, Singapore
| | - Miriam T. Rademaker
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Anna P. Pilbrow
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| |
Collapse
|
35
|
Hei B, Yue C, Sun Y. Long Noncoding RNA ZFAS1 Protects HK-2 Cells against Sepsis-Induced Injury through Targeting the miR3723p/PPAR α Axis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7768963. [PMID: 35035856 PMCID: PMC8759900 DOI: 10.1155/2022/7768963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 12/25/2022]
Abstract
In septic acute kidney injury, one of the main purposes of long noncoding RNA (lncRNA) ZFAS1 is still unclear. This study is intended to analyze the effects of lncRNA ZFAS1 on the septic AKI in the HK-2 cell line. Materials and Methods. In order to construct an in vitro model of septic AKI, HK-2 cells have been treated with lipopolysaccharides. CCK-8 assay has been utilized to check the viability of HK-2 cells. The contents of inflammatory cytokines (that includes IL-1β, TNF-α, and IL-6) have been marked with enzyme-linked immune sorbent assay (ELISA). Cell apoptosis was assessed by TUNEL staining. To detect the expression of lncRNA ZFAS1 and microRNA-372-3p, quantitative reverse-transcription PCR has been used. And to confirm the connection among genes, luciferase reporter assay has been applied. Results. Overexpression of ZFAS1 alleviated LPS-induced HK-2 cell injury. ZFAS1 positively regulated expression of α receptor activated by peroxisome proliferation (PPARα) through competitive linkage with miR-372-3p. In addition, over expression of miR-372-3p counteracted the protective effect of upward regulation of ZFAS1 on LPS-induced HK-2 cell damage, which could be reversed by over expression of PPARα. Conclusion. It is concluded that, in LPS-induced HK-2 cell injury, ZFAS1 has a protective role via modulating the miR-372-3p/PPARα axis, suggesting the potential of ZFAS1 as a protective target for septic AKI.
Collapse
Affiliation(s)
- Bingchang Hei
- Intensive Care Unit, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, Heilongjiang, China
| | - Caifang Yue
- Department of Critical Care Medicine, No. 1 Hospital Attached to Jiamusi University in Heilongjiang Province, ICU, Jiamusi 154002, Heilongjiang, China
| | - Yao Sun
- Department of Neurology, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin 150088, Heilongjiang, China
| |
Collapse
|
36
|
Meng X, Huang W, Mo W, Shu T, Yang H, Ning H. ADAMTS-13-regulated nuclear factor E2-related factor 2 signaling inhibits ferroptosis to ameliorate cisplatin-induced acute kidney injuy. Bioengineered 2021; 12:11610-11621. [PMID: 34666603 PMCID: PMC8810018 DOI: 10.1080/21655979.2021.1994707] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 01/20/2023] Open
Abstract
ADAMTS-13 plays an important role in acute kidney injury (AKI), but the mechanism of cisplatin (CP) induced AKI remains unclear. Ferroptosis is increased in CP-induced AKI, and ADAMTS13 levels are associated with ferritin expression. In this article, we will explore the relationship between the three. After CP induction, mice were given 0.1 and 0.3 nmol/kg ADAMTS-13, and then serum creatinine (Scr) and blood urea nitrogen (BUN) were detected by the kits. The pathological changes of renal tissue were observed by staining with HE and PAS staining, and Western blot detected the expressions of KIM1 and NGAL in renal tissu. Perl's staining detected iron deposition in renal tissues, the kits detected iron levels, and western blot detected the expression of ferroptosis related proteins. Then the mechanism was further explored by adding ferroptosis inhibitors Ferrostatin 1 (Fer-1) and iron supplements Fe. The expression of Nrf2 pathway related proteins were detected by Western blot. We found that ADAMTS13 alleviated CP-induced ferroptosis in AKI mice with renal function impairment and tubular damage. Fer-1partially reversed CP-induced AKI, and Fe exacerbated this effect. ADAMTS13 alleviated CP-induced inflammatory response and oxidative stress in AKI mice, during which the Nrf2 signaling pathway was abnormal. Overall, ADAMTS-13-regulated Nrf2 signaling inhibits ferroptosis to ameliorate CP-induced AKI.
Collapse
Affiliation(s)
- Xiaoyan Meng
- Department of Nephrology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Wenjing Huang
- Department of Nephrology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Weiwei Mo
- Department of Nephrology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Tingting Shu
- Department of Nephrology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Haoqiang Yang
- Department of Nephrology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Haibo Ning
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
37
|
Mechanism Prediction of Astragalus membranaceus against Cisplatin-Induced Kidney Damage by Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9516726. [PMID: 34457031 PMCID: PMC8390139 DOI: 10.1155/2021/9516726] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023]
Abstract
Background Cisplatin is a frequently used and effective chemotherapy drug in clinical practice, but severe side effects limit its use, among which nephrotoxicity is considered the most serious and prolonged damage to the body. Astragalus membranaceus (AM) is a well-known herbal medicine, and modern pharmacological studies have confirmed its antioxidant, immunomodulatory, and antiapoptotic effects. Clinical studies have shown that AM and its active components can attenuate cisplatin-induced kidney damage, but the molecular mechanism has not been fully expounded. Materials and Methods First, the components and targets information of AM were collected from the TCMSP, and the relevant targets of cisplatin-induced kidney damage were accessed from the GeneCards and OMIM databases. Then, the core targets were selected by the Venn diagram and network topology analysis, which was followed by GO and KEGG pathway enrichment analysis. Finally, we construct a component-target-pathway network. Furthermore, molecular docking was carried out to identify the binding activity between active components and key targets. Results A total of 20 active components and 200 targets of AM and 646 targets related to cisplatin-induced kidney damage were obtained. 91 intersection targets were found between AM and cisplatin-induced kidney damage. Then, 16 core targets were identified, such as MAPK1, TNF-α, and p53. Furthermore, GO and KEGG pathway enrichment analysis suggested that MAPK, Toll-like receptor, and PI3K-Akt signaling pathways may be of significance in the treatment of cisplatin-induced kidney damage by AM. Molecular docking indicated that quercetin and kaempferol had high binding affinities with many core targets. Conclusion In summary, the active components, key targets, and signaling pathways of AM in the treatment of cisplatin-induced kidney damage were predicted in this study, which contributed to the development and application of AM.
Collapse
|
38
|
Li Z, Lu S, Li X. The role of metabolic reprogramming in tubular epithelial cells during the progression of acute kidney injury. Cell Mol Life Sci 2021; 78:5731-5741. [PMID: 34185125 PMCID: PMC11073237 DOI: 10.1007/s00018-021-03892-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/01/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is one of the most common clinical syndromes. AKI is associated with significant morbidity and subsequent chronic kidney disease (CKD) development. Thus, it is urgent to develop a strategy to hinder AKI progression. Renal tubules are responsible for the reabsorption and secretion of various solutes and the damage to this part of the nephron is a key mediator of AKI. As we know, many common renal insults primarily target the highly metabolically active proximal tubular cells (PTCs). PTCs are the most energy-demanding cells in the kidney. The ATP that they use is mostly produced in their mitochondria by fatty acid β-oxidation (FAO). But, when PTCs face various biological stresses, FAO will shut down for a time that outlives injury. Recent studies have suggested that surviving PTCs can adapt to FAO disruption by increasing glycolysis when facing metabolic constraints, although PTCs do not perform glycolysis in a normal physiological state. Enhanced glycolysis in a short period compensates for impaired energy production and exerts partial renal-protective effects, but its long-term effect on renal function and AKI progression is not promising. Deranged FAO and enhanced glycolysis may contribute to the AKI to CKD transition through different molecular biological mechanisms. In this review, we concentrate on the recent pathological findings of AKI with regards to the metabolic reprogramming in PTCs, confirming that targeting metabolic reprogramming represents a potentially effective therapeutic strategy for the progression of AKI.
Collapse
Affiliation(s)
- Zhenzhen Li
- Medicial Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Shan Lu
- Emergency Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaobing Li
- College of Basic Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
| |
Collapse
|
39
|
PPP1CA/YAP/GS/Gln/mTORC1 pathway activates retinal Müller cells during diabetic retinopathy. Exp Eye Res 2021; 210:108703. [PMID: 34280391 DOI: 10.1016/j.exer.2021.108703] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Accepted: 07/15/2021] [Indexed: 01/04/2023]
Abstract
Diabetic retinopathy (DR) is a vision-loss complication caused by diabetes with high prevalence. During DR, the retinal microvascular injury and neurodegeneration derived from chronic hyperglycemia have attracted global attention to retinal Müller cells (RMCs), the major macroglia in the retina contributes to neuroprotection. Protein Phosphatase 1 Catalytic Subunit Alpha (PPP1CA) dephosphorylates the transcriptional coactivator Yes-associated protein (YAP) to promote the transcription of glutamine synthetase (GS). GS catalyzes the transformation of neurotoxic glutamate (Glu) into nontoxic glutamine (Gln) to activate the mammalian target of rapamycin complex 1 (mTORC1), which promotes the activation of RMCs. In this study, in vitro MIO-M1 cell and in vivo mouse high-fat diet and streptozotocin (STZ)-induced diabetic model to explore the role of the PPP1CA/YAP/GS/Gln/mTORC1 pathway on the activation of MRCs during DR. Results showed that PPP1CA promoted the dephosphorylation and nuclear translocation of YAP in high glucose (HG)-exposed MIO-M1 cells. YAP transcribed GS in HG-exposed MIO-M1 cells in a TEAD1-dependent and PPP1CA-dependent way. GS promoted the biosynthesis of Gln in HG-exposed MIO-M1 cells. Gln activated mTORC1 instead of mTORC2 in HG-exposed MIO-M1 cells. The proliferation and activation of HG-exposed MIO-M1 cells were PPP1CA/YAP/GS/Gln/mTORC1-dependent. Finally, RMC proliferation and activation during DR were inhibited by the PPP1CA/YAP/GS/Gln/mTORC1 blockade. The findings supplied a potential idea to protect RMCs and alleviate the development of DR.
Collapse
|
40
|
Byun EB, Song HY, Kim WS, Han JM, Seo HS, Park SH, Kim K, Byun EH. Protective Effect of Polysaccharides Extracted from Cudrania tricuspidata Fruit against Cisplatin-Induced Cytotoxicity in Macrophages and a Mouse Model. Int J Mol Sci 2021; 22:ijms22147512. [PMID: 34299130 PMCID: PMC8304288 DOI: 10.3390/ijms22147512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022] Open
Abstract
Although cisplatin is one of most effective chemotherapeutic drugs that is widely used to treat various types of cancer, it can cause undesirable damage in immune cells and normal tissue because of its strong cytotoxicity and non-selectivity. This study was conducted to investigate the cytoprotective effects of Cudrania tricuspidata fruit-derived polysaccharides (CTPS) against cisplatin-induced cytotoxicity in macrophages, lung cancer cell lines, and a mouse model, and to explore the possibility of application of CTPS as a supplement for anticancer therapy. Both cisplatin alone and cisplatin with CTPS induced a significant cytotoxicity in A549 and H460 lung cancer cells, whereas cytotoxicity was suppressed by CTPS in cisplatin-treated RAW264.7 cells. CTPS significantly attenuated the apoptotic and necrotic population, as well as cell penetration in cisplatin-treated RAW264.7 cells, which ultimately inhibited the upregulation of Bcl-2-associated X protein (Bax), cytosolic cytochrome c, poly (adenosine diphosphateribose) polymerase (PARP) cleavage, and caspases-3, -8, and -9, and the downregulation of B cell lymphoma-2 (Bcl-2). The CTPS-induced cytoprotective action was mediated with a reduction in reactive oxygen species production and mitochondrial transmembrane potential loss in cisplatin-treated RAW264.7 cells. In agreement with the results obtained above, CTPS induced the attenuation of cell damage in cisplatin-treated bone marrow-derived macrophages (primary cells). In in vivo studies, CTPS significantly inhibited metastatic colonies and bodyweight loss as well as immunotoxicity in splenic T cells compared to the cisplatin-treated group in lung metastasis-induced mice. Furthermore, CTPS decreased the level of CRE and BUN in serum. In summation, these results suggest that CTPS-induced cytoprotective action may play a role in alleviating the side effects induced by chemotherapeutic drugs.
Collapse
Affiliation(s)
- Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Ha-Yeon Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Korea;
| | - Jeong Moo Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Ho Seong Seo
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea; (E.-B.B.); (H.-Y.S.); (J.M.H.); (H.S.S.)
| | - Sang-Hyun Park
- Department of Food Science and Technology, Kongju National University, Yesan 32439, Korea; (S.-H.P.); (K.K.)
| | - Kwangwook Kim
- Department of Food Science and Technology, Kongju National University, Yesan 32439, Korea; (S.-H.P.); (K.K.)
| | - Eui-Hong Byun
- Department of Food Science and Technology, Kongju National University, Yesan 32439, Korea; (S.-H.P.); (K.K.)
- Correspondence: ; Tel.: +82-413-301-481; Fax: +82-413-301-489
| |
Collapse
|
41
|
Son SH, Lee SM, Lee MH, Son YK, Kim SE, An WS. Omega-3 Fatty Acids Upregulate SIRT1/3, Activate PGC-1α via Deacetylation, and Induce Nrf1 Production in 5/6 Nephrectomy Rat Model. Mar Drugs 2021; 19:182. [PMID: 33810216 PMCID: PMC8066610 DOI: 10.3390/md19040182] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial dysfunction contributes to the pathogenesis of kidney injury related with cardiovascular disease. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) protects renal tubular cells by upregulating nuclear factor erythroid 2-related factor 2 (Nrf2). AMP-activated protein kinase (pAMPK)-mediated phosphorylation and sirtuin 1/3 (SIRT1/3)-mediated deacetylation are required for PGC-1α activation. In the present study, we aimed to investigate whether omega-3 fatty acids (FAs) regulate the expression of mediators of mitochondrial biogenesis in 5/6 nephrectomy (Nx) rats. Male Sprague-Dawley rats were assigned to the following groups: sham control, Nx, and Nx treated with omega-3 FA. The expression of PGC-1α, phosphorylated PGC-1α (pPGC-1α), acetylated PGC-1α, and factors related to mitochondrial biogenesis was examined through Western blot analysis. Compared to the control group, the expression of PGC-1α, pAMPK, SIRT1/3, Nrf1, mTOR, and Nrf2 was significantly downregulated, and that of Keap 1, acetylated PGC-1α, and FoxO1/3, was significantly upregulated in the Nx group. These changes in protein expression were rescued in the omega-3 FA group. However, the expression of pPGC-1α was similar among the three groups. Omega-3 FAs may involve mitochondrial biogenesis by upregulating Nrf1 and Nrf2. This protective mechanism might be attributed to the increased expression and deacetylation of PGC-1α, which was triggered by SIRT1/3.
Collapse
Affiliation(s)
- Sung Hyun Son
- Department of Internal Medicine, BHS Han Seo Hospital, Busan 48253, Korea;
| | - Su Mi Lee
- Department of Internal Medicine, Dong-A University, Busan 49201, Korea; (S.M.L.); (Y.K.S.); (S.E.K.)
| | - Mi Hwa Lee
- Department of Anatomy and Cell Biology, Dong-A University, Busan 49201, Korea;
| | - Young Ki Son
- Department of Internal Medicine, Dong-A University, Busan 49201, Korea; (S.M.L.); (Y.K.S.); (S.E.K.)
| | - Seong Eun Kim
- Department of Internal Medicine, Dong-A University, Busan 49201, Korea; (S.M.L.); (Y.K.S.); (S.E.K.)
| | - Won Suk An
- Department of Internal Medicine, Dong-A University, Busan 49201, Korea; (S.M.L.); (Y.K.S.); (S.E.K.)
| |
Collapse
|