1
|
Hesari M, Mohammadi P, Moradi M, Shackebaei D, Yarmohammadi F. Molecular mechanisms involved in therapeutic effects of natural compounds against cisplatin-induced cardiotoxicity: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8367-8381. [PMID: 38850306 DOI: 10.1007/s00210-024-03207-3] [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: 03/15/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Cisplatin is a widely used chemotherapeutic agent for the treatment of various cancers. However, the clinical use of cisplatin is limited by its cardiotoxic side effects. The primary mechanisms implicated in this cardiotoxicity include mitochondrial dysfunction, oxidative stress, inflammation, and apoptotic. Numerous natural compounds (NCs) have been introduced as promising protective factors against cisplatin-mediated cardiac damage. The current review summarized the potential of various NCs as cardioprotective agents at the molecular levels. These compounds exhibited potent antioxidant and anti-inflammatory effects by interaction with the PI3K/AKT, AMPK, Nrf2, NF-κB, and NLRP3/caspase-1/GSDMD pathways. Generally, the modulation of these signaling pathways by NCs represents a promising strategy for improving the therapeutic index of cisplatin by reducing its cardiac side effects.
Collapse
Affiliation(s)
- Mahvash Hesari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Moradi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Ma Y, Bai B, Liu D, Shi R, Zhou Q. Shenqi Fuzheng Injection Reduces Cisplatin-Induced Kidney Injury via cGAS/STING Signaling Pathway in Breast Cancer Mice Model. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:451-469. [PMID: 39165276 PMCID: PMC11335009 DOI: 10.2147/bctt.s475860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
Background Shenqi Fuzheng Injection (SQFZ) is a traditional Chinese medicine injection consists of extracts of Codonopsis pilosula and Astragalus mongholicus. Combining SQFZ with conventional chemotherapy may improve the therapeutic efficacy and reduce side-effects of chemotherapy. However, the mechanisms of SQFZ reducing cisplatin-induced kidney injury are still unclear. Methods The main compounds of SQFZ were identified via UPLC-Q-TOF-MS technique. Using multiple databases to predict potential targets for SQFZ. We established a breast cancer model by injecting 4T1 cells into mice. Tumor growth and body weight were observed. Serum blood urea nitrogen (BUN), creatinine (CRE), and glutathione (GSH) levels were measured. The extent of their kidney injury was measured by hematoxylin-eosin staining (HE). Cell apoptosis was identified using Hoechst33258 staining, flow cytometry and TUNEL. We evaluated H2AX and stimulator of interferon genes (STING) expression by immunohistochemistry (IHC), and assessed apoptosis-associated proteins by Western blotting analysis. We also evaluated mitochondrial function. The secretion of the inflammatory cytokines in serum was observed using ELISA assay. The effect of the STING pathway in HK-2 renal tubular epithelial cells exposed to cisplatin alone or combined with SQFZ. Results The potential targets of SQFZ on kidney injury mainly related to inflammatory responses, oxidation and antioxidant, apoptosis as well as IFN signaling pathway. Cisplatin significantly reduced animal weight, while there were no changes in the combination SQFZ and cisplatin. SQFZ counteracted cisplatin-induced BUN and CRE elevation. SQFZ ameliorated the oxidative stress induced by cisplatin. It diminished cisplatin-induced apoptosis and mitochondrial DNA damage and reversed cisplatin-induced cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/STING signaling pathway activation. It also improved the mitochondrial dysfunction induced by cisplatin. Conclusions The results of the present study suggested that SQFZ effectively reduced cisplatin-induced kidney injury by inhibiting cGAS/STING signaling pathway.
Collapse
Affiliation(s)
- Yingrui Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Bufan Bai
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Deng Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Rong Shi
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Dongfang Hospital Affiliated to Shanghai Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
El-Shoura EAM, Hassanein EHM, Taha HH, Shalkami AGS, Hassanein MMH, Ali FEM, Bakr AG. Edaravone and obeticholic acid protect against cisplatin-induced heart toxicity by suppressing oxidative stress and inflammation and modulating Nrf2, TLR4/p38MAPK, and JAK1/STAT3/NF-κB signals. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5649-5662. [PMID: 38285279 PMCID: PMC11329704 DOI: 10.1007/s00210-024-02956-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024]
Abstract
Cardiotoxicity is a significant adverse effect of cisplatin (CIS) that necessitates extensive medical care. The current study examines the cardioprotective effects of edaravone (EDV), obeticholic acid (OCA), and their combinations on CIS-induced cardiac damage. Rats were allocated into five groups: the normal control group, the remaining four groups received CIS (7.5 mg/kg, i.p.) as a single dose on the fifth day and were assigned to CIS, OCA (10 mg/kg/day) + CIS, EDV (20 mg/kg/day) + CIS, and the (EDV + OCA) + CIS group. Compared to the CIS-treated group, co-treating rats with EDV, OCA, or their combinations significantly decreased ALP, AST, LDH, CK-MB, and troponin-I serum levels and alleviated histopathological heart abnormalities. Biochemically, EDV, OCA, and EDV plus OCA administration mitigated cardiac oxidative stress as indicated by a marked decrease in heart MDA content with a rise in cardiac antioxidants SOD and GSH associated with upregulating Nrf2, PPARγ, and SIRT1 expression. Besides, it dampened inflammation by decreasing cardiac levels of TNF-α, IL-1β, and IL-6, mediated by suppressing NF-κB, JAK1/STAT3, and TLR4/p38MAPK signal activation. Notably, rats co-administered with EDV plus OCA showed noticeable protection that exceeded that of EDV and OCA alone. In conclusion, our study provided that EDV, OCA, and their combinations effectively attenuated CIS-induced cardiac intoxication by activating Nrf2, PPARγ, and SIRT1 signals and downregulating NF-κB, JAK1/STAT3, and TLR4/p38MAPK signals.
Collapse
Affiliation(s)
- Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hesham H Taha
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Abdel-Gawad S Shalkami
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
- Clinical Pharmacy Program, Faculty of Health Science and Nursing, Al-Rayan Colleges, Medina, Kingdom of Saudi Arabia
| | | | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Adel G Bakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| |
Collapse
|
4
|
Oliveira CA, Mercês ÉAB, Portela FS, Malheiro LFL, Silva HBL, De Benedictis LM, De Benedictis JM, Silva CCDE, Santos ACL, Rosa DP, Velozo HS, de Jesus Soares T, de Brito Amaral LS. An integrated view of cisplatin-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity: characteristics, common molecular mechanisms, and current clinical management. Clin Exp Nephrol 2024; 28:711-727. [PMID: 38678166 DOI: 10.1007/s10157-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin (CP) is a chemotherapy drug widely prescribed to treat various neoplasms. Although fundamental for the therapeutic action of the drug, its cytotoxic mechanisms trigger adverse effects in several tissues, such as the kidney, liver, and heart, which limit its clinical use. In this sense, studies point to an essential role of damage to nuclear and mitochondrial DNA associated with oxidative stress, inflammation, and apoptosis in the pathophysiology of tissue injuries. Due to the limitation of effective preventive and therapeutic measures against CP-induced toxicity, new strategies with potential cytoprotective effects have been studied. Therefore, this article is timely in reviewing the characteristics and main molecular mechanisms common to renal, hepatic, and cardiac toxicity previously described, in addition to addressing the main validated strategies for the current management of these adverse events in clinical practice. We also handle the main promising antioxidant substances recently presented in the literature to encourage the development of new research that consolidates their potential preventive and therapeutic effects against CP-induced cytotoxicity.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | | | | | | | | | | | | | - Helloisa Souza Velozo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
| |
Collapse
|
5
|
Rahman AA, Hegazy A, Elabbasy LM, Shoaeir MZ, Abdel-Aziz TM, Abbas AS, Khella HWZ, Eltrawy AH, Alshaman R, Aloyouni SY, Aldahish AA, Zaitone SA. Leflunomide-induced cardiac injury in adult male mice and bioinformatic approach identifying Nrf2/NF-κb signaling interplay. Toxicol Mech Methods 2024; 34:639-653. [PMID: 38389224 DOI: 10.1080/15376516.2024.2322666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Leflunomide (LFND) is an immunosuppressive and immunomodulatory disease-modifying antirheumatic drug (DMARD) that was approved for treating rheumatoid arthritis. LFND-induced cardiotoxicity was not fully investigated since its approval. We investigated the cardiac injury in male mice and identified the role of nuclear factor erythroid 2-related factor 2/nuclear factor-κ B (Nrf2/NF-κB) signaling. Male albino mice were assigned into five groups as control, vehicle, and LFND (2.5, 5, and 10 mg/kg). We investigated cardiac enzymes, histopathology, and the mRNA expression of Nrf2, NF-κB, BAX, and tumor necrosis factor-α (TNF-α). The bioinformatic study identified the interaction between LFND and Nrf2/NF-κB signaling; this was confirmed by amelioration in mRNA expression (0.5- to 0.34-fold decrease in Nrf2 and 2.6- to 4.61-fold increases in NF-κB genes) and increased (1.76- and 2.625-fold) serum creatine kinase (CK) and 1.38- and 2.33-fold increases in creatine kinase-MB (CK-MB). Histopathological results confirmed the dose-dependent effects of LFND on cardiac muscle structure in the form of cytoplasmic, nuclear, and vascular changes in addition to increased collagen deposits and apoptosis which were increased compared to controls especially with LFND 10 mg/kg. The current study elicits the dose-dependent cardiac injury induced by LFND administration and highlights, for the first time, dysregulation in Nrf2/NF-κB signaling.
Collapse
Affiliation(s)
- Abeer A Rahman
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ann Hegazy
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Lamiaa M Elabbasy
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Basic Medical Sciences, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Mohamed Z Shoaeir
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Tarek M Abdel-Aziz
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Awad S Abbas
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Heba W Z Khella
- Department of Clinical Education, Canadian Memorial Chiropractic College, Toronto, Canada
| | - Amira H Eltrawy
- Department of Anatomy and Embryology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Sheka Yagub Aloyouni
- Research Department, Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Afaf A Aldahish
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
6
|
Xia J, Chen C, Sun Y, Li S, Li Y, Cheng BR, Pang Y, Li Y, Li D, Lin Q. Panax quinquefolius saponins and panax notoginseng saponins attenuate myocardial hypoxia-reoxygenation injury by reducing excessive mitophagy. Cell Biochem Biophys 2024; 82:1179-1191. [PMID: 38713401 DOI: 10.1007/s12013-024-01267-z] [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] [Accepted: 03/29/2024] [Indexed: 05/08/2024]
Abstract
OBJECTIVE Panax quinquefolius saponins (PQS) and Panax notoginseng saponins (PNS) are key bioactive compounds in Panax quinquefolius L. and Panax notoginseng, commonly used in the treatment of clinical ischemic heart disease. However, their potential in mitigating myocardial ischemia-reperfusion injury remains uncertain. This study aims to evaluate the protective effects of combined PQS and PNS administration in myocardial hypoxia/reoxygenation (H/R) injury and explore the underlying mechanisms. METHODS To investigate the involvement of HIF-1α/BNIP3 mitophagy pathway in the myocardial protection conferred by PNS and PQS, we employed small interfering BNIP3 (siBNIP3) to silence key proteins of the pathway. H9C2 cells were categorized into four groups: control, H/R, H/R + PQS + PNS, and H/R + PQS + PNS+siBNIP3. Cell viability was assessed by Cell Counting Kit-8, apoptosis rates determined via flow cytometry, mitochondrial membrane potential assessed with the JC-1 fluorescent probes, intracellular reactive oxygen species detected with 2',7'-dichlorodihydrofluorescein diacetate, mitochondrial superoxide production quantified with MitoSOX Red, and autophagic flux monitored with mRFP-GFP-LC3 adenoviral vectors. Autophagosomes and their ultrastructure were visualized through transmission electron microscopy. Moreover, mRNA and protein levels were analyzed via real-time PCR and Western blotting. RESULTS PQS + PNS administration significantly increased cell viability, reduced apoptosis, lowered reactive oxygen species levels and mitochondrial superoxide production, mitigated mitochondrial dysfunction, and induced autophagic flux. Notably, siBNIP3 intervention did not counteract the cardioprotective effect of PQS + PNS. The PQS + PNS group showed downregulated mRNA expression of HIF-1α and BNIP3, along with reduced HIF-1α protein expression compared to the H/R group. CONCLUSIONS PQS + PNS protects against myocardial H/R injury, potentially by downregulating mitophagy through the HIF-1α/BNIP3 pathway.
Collapse
Affiliation(s)
- Junyan Xia
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Cong Chen
- Department of Cardiology, Guanganmen Hospital, China Academy of Chinese Medical Sciences, 100053, Beijing, China
| | - Yanan Sun
- Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Sinai Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, 100010, Beijing, China
| | - Yuxuan Li
- Department of Cardiology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, 100078, Beijing, China
| | - Bai-Ru Cheng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Yanting Pang
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Yan Li
- Department of Cardiology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, 100078, Beijing, China
| | - Dong Li
- Department of Cardiology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, 100078, Beijing, China.
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, 100700, Beijing, China.
| |
Collapse
|
7
|
Vijayakumar A, Kim JH. Ginseng and ginsenosides on cardiovascular and pulmonary diseases; Pharmacological potentials for the coronavirus (COVID-19). J Ginseng Res 2024; 48:113-121. [PMID: 38465214 PMCID: PMC10920003 DOI: 10.1016/j.jgr.2023.10.002] [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: 07/24/2023] [Revised: 09/20/2023] [Accepted: 10/26/2023] [Indexed: 03/12/2024] Open
Abstract
Since its outbreak in late 2019, the Coronavirus disease 2019 (COVID-19) pandemic has profoundly caused global morbidity and deaths. The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has major complications in cardiovascular and pulmonary system. The increased rate of mortality is due to delayed detection of certain biomarkers that are crucial in the development of disease. Furthermore, certain proteins and enzymes in cellular signaling pathways play an important role in replication of SARS-CoV-2. Most cases are mild to moderate symptoms, however severe cases of COVID-19 leads to death. Detecting the level of biomarkers such as C-reactive protein, cardiac troponin, creatine kinase, creatine kinase-MB, procalcitonin and Matrix metalloproteinases helps in early detection of the severity of disease. Similarly, through downregulating Renin-angiotensin system, interleukin, Mitogen-activated protein kinases and Phosphoinositide 3-kinases pathways, COVID-19 can be effectively controlled and mortality could be prevented. Ginseng and ginsenosides possess therapeutic potential in cardiac and pulmonary complications, there are several studies performed in which they have suppressed these biomarkers and downregulated the pathways, thereby inhibiting the further spread of disease. Supplementation with ginseng or ginsenoside could act on multiple pathways to reduce the level of biomarkers significantly and alleviate cardiac and pulmonary damage. Therefore, this review summarizes the potential of ginseng extract and ginsenosides in controlling the cardiovascular and pulmonary diseases by COVID-19.
Collapse
Affiliation(s)
- Ajay Vijayakumar
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan-city, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan-city, Republic of Korea
| |
Collapse
|
8
|
Su Y, Cheng Z, Liu W, Wu T, Wang W, Lin M. Effects of rumen-protective γ-aminobutyric acid additive on lactation performance and serum biochemistry in heat-stressed cows. Front Vet Sci 2023; 10:1228155. [PMID: 37808113 PMCID: PMC10556515 DOI: 10.3389/fvets.2023.1228155] [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: 05/24/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
In the context of global warming, heat stress has become one of the major stress factors limiting dairy cattle production. Although many methods have been explored to help cows mitigate the negative effects of heat stress during the hot summer months, maintaining the performance of high-yielding cows under heat stress is still a great challenge. The aim of this trial was to investigate the effect of RP-GABA in the diet on milk yield, milk composition and serum biochemical parameters in heat-stressed cows. Twenty Chinese Holstein cows in early lactation (51.00 ± 4.92 kg milk/d, 71 ± 10.94 d in milk and 2.68 ± 0.73 parities) were included in this experiment and randomly divided into four groups (n = 5/group). The four experimental groups consisted of one control group (0 g RP-GABA/d) and three treatment groups, given 5, 7.5 and 10 g RP-GABA/d of dry matter (DM) per cow, respectively. The results showed that supplementing high-yielding cows with 10 g/d of RP-GABA improved milk protein production but had no effect on the improvement of other production performance, the alleviation of heat stress in cows, or the improvement of immune function and antioxidant capacity. Ultimately, we conclude that the supplementation of 10 g/d RP-GABA to heat-stressed, high-yielding dairy cows can provide a degree of performance enhancement. Furthermore, our study provides some reference for nutritional improvement measures for summer heat stress in dairy cows, especially high-yielding cows.
Collapse
Affiliation(s)
| | - Zhiqiang Cheng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wengboyang Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | | | | | - Miao Lin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
9
|
Lu X, Liu X, Liang E, Yang R, Liu Y, Liu X, Yan F, Xing Y. Panax quinquefolius saponin inhibits vascular smooth muscle cell calcification via activation of nuclear factor-erythroid 2-related factor 2. BMC Complement Med Ther 2023; 23:129. [PMID: 37085826 PMCID: PMC10120105 DOI: 10.1186/s12906-023-03961-6] [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: 12/26/2022] [Accepted: 04/14/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Panax quinquefolius saponin (PQS) is the main active component of Panax quinquefolius. Emerging evidence suggests that PQS exerts beneficial effects against cardiovascular diseases. However, the role and mechanism of PQS in vascular calcification are not unclear. The present study investigated the effects of PQS on the calcification of vascular smooth muscle cell (VSMCs). METHODS The present study used calcification medium containing 3 mM inorganic phosphate (Pi) to induce rat VSMCs calcification. We investigated the effects of PQS on VSMCs calcification using alizarin red staining and alkaline phosphatase (ALP) activity assays. The intracellular reactive oxygen species (ROS) levels and the transcriptional activity of nuclear factor-erythroid 2-related factor 2 (Nrf2) were determined. The mRNA and protein expression levels of Nrf2, the antioxidant gene heme oxygenase-1 (HO-1), osteogenic markers, including runt-related transcription factor 2 (Runx2) and bone morphogenetic protein 2 (BMP2), and Kelch-like ECH-associated protein 1 (Keap1) were also measured. RESULTS Treatment with Pi significantly increased intracellular calcium deposition and ALP activity, which were suppressed by PQS in a concentration-dependent manner. During VSMCs calcification, PQS inhibited the mRNA and protein expression of Runx2 and BMP2. PQS treatment reduced intracellular ROS production and significantly upregulated Nrf2 transcriptional activity and the expression of Nrf2 and its target antioxidant gene HO-1. PQS suppressed the Pi-induced protein expression of Keap1, which is an endogenous inhibitor of Nrf2. Keap1 siRNA treatment induced Nrf2 expression and downregulated Runx2 expression in the presence of Pi and PQS. CONCLUSION Taken together, these findings suggest that PQS could effectively inhibit VSMCs calcification by ameliorating oxidative stress and regulating osteogenic genes via the promotion of Nrf2 expression.
Collapse
Affiliation(s)
- Xiaoting Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Xue Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Ershun Liang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Ruixue Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Yan Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Xiaoqiong Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Fangfang Yan
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yifan Xing
- Department of Medical Insurance, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
10
|
Moini Jazani A, Arabzadeh A, Haghi-Aminjan H, Nasimi Doost Azgomi R. The role of ginseng derivatives against chemotherapy-induced cardiotoxicity: A systematic review of non-clinical studies. Front Cardiovasc Med 2023; 10:1022360. [PMID: 36844721 PMCID: PMC9946988 DOI: 10.3389/fcvm.2023.1022360] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/12/2023] [Indexed: 02/11/2023] Open
Abstract
Aims Although chemotherapy agents are used to treating cancers, they have serious side effects, like their harmful effects on the cardiovascular system, limiting the clinical use of these chemotherapy agents. This study aimed to systematically investigate the potential role of ginseng derivatives in the prevention of chemotherapy-induced cardiac toxicity. Methods This systematic review was performed according to PRISMA guidelines strategy in databases till August 2022. First, identify studies related to using search terms in titles and abstracts. After studying and screening 209 articles, 16 articles were selected in this study according to our inclusion and exclusion criteria. Results According to the findings of this study, ginseng derivatives showed significant changes in biochemical, histological, and heart weight loss, as well as a reduction in mortality, which occurred in the groups treated with chemotherapy agents compared to the control groups. Co-administration of ginseng derivatives with chemotherapy agents inhibited or reversed these changes to near-moderate levels. The protective effects of ginseng derivatives can be due to their anti-inflammatory, anti-oxidant, and anti-apoptotic action. Conclusion This systematic review shows evidence that concomitant administration of ginseng derivatives improves chemotherapy-induced cardiac toxicity. However, for better conclusions about the practical mechanisms of ginseng derivatives in reducing the cardiac toxic effects of chemotherapy agents and evaluating the efficacy and safety of the compound simultaneously, it is necessary to design comprehensive studies.
Collapse
Affiliation(s)
- Arezoo Moini Jazani
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - AmirAhmad Arabzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran,*Correspondence: Hamed Haghi-Aminjan,✉
| | - Ramin Nasimi Doost Azgomi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran,Ramin Nasimi Doost Azgomi,✉
| |
Collapse
|
11
|
Ali FEM, Hassanein EHM, Abd El-Ghafar OAM, Rashwan EK, Saleh FM, Atwa AM. Exploring the cardioprotective effects of canagliflozin against cisplatin-induced cardiotoxicity: Role of iNOS/NF-κB, Nrf2, and Bax/cytochrome C/Bcl-2 signals. J Biochem Mol Toxicol 2023; 37:e23309. [PMID: 36645100 DOI: 10.1002/jbt.23309] [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: 03/05/2022] [Revised: 11/14/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023]
Abstract
Cardiotoxicity is a severe considerable side effect of cisplatin (CDDP) that requires much medical attention. The current study investigates the cardioprotective effects of canagliflozin (CA) against CDDP-induced heart toxicity. Rats were allocated to the control group; the CA group was administered CA 10 mg/kg/day orally for 10 days; the CDDP group was injected with 7 mg/kg, intraperitoneal as a single dose on the 5th day, and the CDDP + CA group. Compared to the CDDP-treated group, CA effectively attenuated CDDP-induced heart injury as evidenced by a decrease of serum aspartate aminotransferase, alkaline phosphatase, creatine kinase-MB, and lactate dehydrogenase enzymes and supported by the alleviation of histopathological changes in cardiac tissues. Biochemically, CA attenuated cardiac oxidative injury through upregulation of the nuclear factor-erythroid 2 related factor 2 (Nrf2) signal. CA suppressed inflammation by decreasing cardiac NO2 - , MPO, iNOS, nuclear factor kappa B (NF-κB), tumor necrosis factor-alpha, and interleukin 1-beta levels. Besides, CA significantly upregulated cardiac levels of phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and p-AKT proteins. Moreover, CA remarkably mitigated CDDP-induced apoptosis via modulation of Bax, cytochrome C, and Bcl-2 protein levels. Together, the present study revealed that CA could be a good candidate for preventing CDDP-induced cardiac injury by modulating iNOS/NF-κB, Nrf2, PI3K/AKT, and Bax/cytochrome C/Bcl-2 signals.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni Suef, Egypt
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Al-Azhar University, Assuit, Egypt
| | - Fayez M Saleh
- Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
12
|
Yang W, Shao F, Wang J, Shen T, Zhao Y, Fu X, Zhang L, Li H. Ethyl Acetate Extract from Artemisia argyi Prevents Liver Damage in ConA-Induced Immunological Liver Injury Mice via Bax/Bcl-2 and TLR4/MyD88/NF- κB Signaling Pathways. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227883. [PMID: 36431983 PMCID: PMC9693258 DOI: 10.3390/molecules27227883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Immunological liver injury (ILI) is a common liver disease and lacks potent drugs for treatment. Artemisia argyi Lévl. et Vant. (A. argyi), a medicinal and edible homologous plant usually used in diet therapy to cure various liver diseases, provides a great option for the prevention of ILI. PURPOSE To investigate the effect that ethyl acetate extract of A. argyi (AaEA) on Concanavalin A (ConA)-induced ILI and the mechanism of regulating Bax/Bcl-2 and TLR4/MyD88/NF-κB signaling pathways. METHODS The chemical components of AaEA were studied by LC-MS. In animal experiments, the positive control group was administrated diammonium glycyrrhizinate (DIG, 100 mg/kg), while different doses of AaEA groups (AaEA-H, AaEA-M, AaEA-L) were pretreated with AaEA 2.00, 1.00, and 0.50 g/kg, respectively, by intragastric for seven days, once every day. Then, ConA (12.00 mg/kg) was used through tail intravenous injection to establish the ILI model. The blood samples and livers were collected to test the degree of liver dysfunction, inflammation, oxidative stress, histopathological changes, and cell apoptosis. Real-time PCR and Western blotting analysis were used to explain the mechanism of regulating Bax/Bcl-2 and TLR4/MyD88/NF-κB signaling pathways. RESULTS The way in which AaEA prevents liver damage in immunological liver injury (ILI) mice caused by ConA was investigated for the first time. Pretreatment with AaEA reduced the expression of ALT, AST, and inflammatory factors (TNF-α and IFN-γ). Meanwhile, AaEA also reduced MDA levels but upregulated the contents of IL-4, SOD, and GSH-px, alleviating oxidative stress induced by ILI. Western blotting and real-time PCR analysis demonstrated that AaEA could regulate the expression level and relative mRNA expression of key proteins on Bax/Bcl-2 and TLR4/MyD88/NF-κB signaling pathways. Finally, 504 components from AaEA were identified by LC-MS analysis, mainly including flavones, phenolic acids, and terpenoids with anti-inflammatory and liver protective activities, which highlights the potential of AaEA for diet treatment of ILI. CONCLUSION AaEA can work against ConA-induced ILI in mice by regulating Bax/Bcl-2 and TLR4/MyD88/NF-κB signaling pathways, which has the potential to be a great strategy for the prevention of ILI.
Collapse
Affiliation(s)
- Wenqian Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Fei Shao
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jiexin Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Tong Shen
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Yu Zhao
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Xueyan Fu
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Research Center of Modern Hui Medicine Engineering and Technology, Ningxia Medical University, Yinchuan 750000, China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Liming Zhang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Research Center of Modern Hui Medicine Engineering and Technology, Ningxia Medical University, Yinchuan 750000, China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
- Correspondence: (L.Z.); (H.L.)
| | - Hangying Li
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Research Center of Modern Hui Medicine Engineering and Technology, Ningxia Medical University, Yinchuan 750000, China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
- Correspondence: (L.Z.); (H.L.)
| |
Collapse
|
13
|
Lv XF, Wen RQ, Liu K, Zhao XK, Pan CL, Gao X, Wu X, Zhi XD, Ren CZ, Chen QL, Lu WJ, Bai TY, Li YD. Role and molecular mechanism of traditional Chinese medicine in preventing cardiotoxicity associated with chemoradiotherapy. Front Cardiovasc Med 2022; 9:1047700. [PMID: 36419486 PMCID: PMC9678083 DOI: 10.3389/fcvm.2022.1047700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/20/2022] [Indexed: 08/12/2023] Open
Abstract
Cardiotoxicity is a serious complication of cancer therapy. It is the second leading cause of morbidity and mortality in cancer survivors and is associated with a variety of factors, including oxidative stress, inflammation, apoptosis, autophagy, endoplasmic reticulum stress, and abnormal myocardial energy metabolism. A number of studies have shown that traditional Chinese medicine (TCM) can mitigate chemoradiotherapy-associated cardiotoxicity via these pathways. Therefore, this study reviews the effects and molecular mechanisms of TCM on chemoradiotherapy-related cardiotoxicity. In this study, we searched PubMed for basic studies on the anti-cardiotoxicity of TCM in the past 5 years and summarized their results. Angelica Sinensis, Astragalus membranaceus Bunge, Danshinone IIA sulfonate sodium (STS), Astragaloside (AS), Resveratrol, Ginsenoside, Quercetin, Danggui Buxue Decoction (DBD), Shengxian decoction (SXT), Compound Danshen Dripping Pill (CDDP), Qishen Huanwu Capsule (QSHWC), Angelica Sinensis and Astragalus membranaceus Bunge Ultrafiltration Extract (AS-AM),Shenmai injection (SMI), Xinmailong (XML), and nearly 60 other herbs, herbal monomers, herbal soups and herbal compound preparations were found to be effective as complementary or alternative treatments. These preparations reduced chemoradiotherapy-induced cardiotoxicity through various pathways such as anti-oxidative stress, anti-inflammation, alleviating endoplasmic reticulum stress, regulation of apoptosis and autophagy, and improvement of myocardial energy metabolism. However, few clinical trials have been conducted on these therapies, and these trials can provide stronger evidence-based support for TCM.
Collapse
Affiliation(s)
- Xin-Fang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Ruo-Qing Wen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin-Ke Zhao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Chen-Liang Pan
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Lanzhou University Second Hospital, Lanzhou, China
| | - Xiao-Dong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Chun-Zhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Qi-Lin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Wei-Jie Lu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Ting-Yan Bai
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Ying-Dong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
14
|
Hassanein EHM, Ali FEM, Mohammedsaleh ZM, Atwa AM, Elfiky M. The involvement of Nrf2/HO-1/cytoglobin and Ang-II/NF-κB signals in the cardioprotective mechanism of lansoprazole against cisplatin-induced heart injury. Toxicol Mech Methods 2022; 33:316-326. [PMID: 36258671 DOI: 10.1080/15376516.2022.2137870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Cardiac toxicity is a serious adverse effect of cisplatin (CIS). Lansoprazole (LPZ) is a proton pump inhibitor with promising cardioprotective effects. Our study planned to examine the cardioprotective effect of LPZ against CIS-induced cardiac injury. To achieve this goal, thirty-two male rats were randomly allocated into 4 groups. CIS, 7 mg/kg, was injected i.p. on the 5th day of the experiment. LPZ was administered via oral gavage at a dose of 50 mg/kg. The present study revealed that CIS injection induced a remarkable cardiac injury evidenced by an increase in serum ALP, AST, CK-MB, LDH, and troponin-I levels. The cardiac oxidative damage was also observed after CIS injection and mediated by down-regulation of GSH, SOD, GST, Nrf2, HO-1, PPAR-γ, and cytoglobin levels associated with the up-regulation of MDA content. Besides, CIS injection caused a significant inflammatory reaction mediated by alteration of cardiac NF-κB, STAT-3, p-STAT-3, and IκB expressions. Additionally, cardiac Ang-II expression was significantly increased in CIS control rats, while Ang 1-7 expression was significantly reduced relative to normal rats. In contrast, LPZ administration remarkably ameliorated these changes in the heart of CIS-intoxicated rats. Collectively, LPZ potently attenuated cardiac toxicity induced by CIS via regulation of Nrf2/HO-1, PPAR-γ, cytoglobin, IκB/NF-κB/STAT-3, and Ang-II/Ang 1-7 signals.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Mohamed Elfiky
- Human Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Shebin ElKoum-Menoufia, Egypt.,Anatomy Department, Faculty of Medicine, Batterjee Medical College, Jedda, Saudi Arabia
| |
Collapse
|
15
|
Xie D, Hu G, Chen C, Ahmadinejad F, Wang W, Li PL, Gewirtz DA, Li N. Loss of sphingosine kinase 2 protects against cisplatin-induced kidney injury. Am J Physiol Renal Physiol 2022; 323:F322-F334. [PMID: 35834271 PMCID: PMC9394771 DOI: 10.1152/ajprenal.00229.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 06/23/2022] [Accepted: 07/07/2022] [Indexed: 01/01/2023] Open
Abstract
Cisplatin is an established chemotherapeutic drug for treatment of solid-organ cancers and is the primary drug used in the treatment of head and neck cancer; however, cisplatin-induced nephrotoxicity largely limits its clinical use. Inhibition of sphingosine kinase 2 (SphK2) has been demonstrated to alleviate various kidney diseases. Therefore, we hypothesized that inhibition of SphK2 could also protect against cisplatin-induced nephrotoxicity. Results from the present study showed that the SphK2 inhibitor ABC294640 or knockdown of SphK2 by siRNA blocked the cisplatin-induced increase of cellular injury markers (neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, and cleaved caspase-3) by Western blot analysis in HK-2 cells, a human renal tubular cell line. In addition, SphK2 inhibition blocked cisplatin-induced activation of NF-κB by Western blot analysis and immunostaining analysis. Furthermore, SphK2 inhibition suppressed cisplatin-induced increases of proinflammatory markers (NLR family pyrin domain containing 3, interleukin-1β, and interleukin-6). Genetic deletion of the SphK2 gene in mice further confirmed that inhibition of SphK2 protected against cisplatin-induced kidney damage in vivo. Compared with wild-type mice, SphK2 knockout mice exhibited less renal dysfunction and reduced promotion of kidney injury markers, inflammatory factors, tubular morphology damage, and fibrotic staining. At the same time, the SphK2 inhibitor ABC294640 failed to interfere with the activity of cisplatin or radiation in two cell culture models of head and neck cancer. It is concluded that inhibition of Sphk2 protects against cisplatin-induced kidney injury. SphK2 may be used as a potential therapeutic target for the prevention or treatment of cisplatin-induced kidney injury.NEW & NOTEWORTHY The present study provides new findings that sphingosine kinase 2 (SphK2) is highly expressed in renal tubules, cisplatin treatment increases the expression of SphK2 in proximal tubular cells and kidneys, and inhibition of SphK2 alleviates cisplatin-induced kidney injury by suppressing the activation of NF-κB, production of inflammatory factors, and apoptosis. SphK2 may serve as a potential therapeutic target for the prevention or treatment of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Dengpiao Xie
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gaizun Hu
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chaoling Chen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Fereshteh Ahmadinejad
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Weili Wang
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Ningjun Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
16
|
Meng C, Fan L, Wang X, Wang Y, Li Y, Pang S, Lv S, Zhang J. Preparation and Evaluation of Animal Models of Cardiotoxicity in Antineoplastic Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3820591. [PMID: 35847594 PMCID: PMC9277159 DOI: 10.1155/2022/3820591] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
The continuous development of antineoplastic therapy has significantly reduced the mortality of patients with malignant tumors, but its induced cardiotoxicity has become the primary cause of long-term death in patients with malignant tumors. However, the pathogenesis of cardiotoxicity of antineoplastic therapy is currently unknown, and practical means of prevention and treatment are lacking in clinical practice. Therefore, how to effectively prevent and treat cardiotoxicity while treating tumors is a major challenge. Animal models are important tools for studying cardiotoxicity in antitumor therapy and are of great importance in elucidating pathophysiological mechanisms and developing and evaluating modality drugs. In this paper, we summarize the existing animal models in antitumor therapeutic cardiotoxicity studies and evaluate the models by observing the macroscopic signs, echocardiography, and pathological morphology of the animals, aiming to provide a reference for subsequent experimental development and clinical application.
Collapse
Affiliation(s)
- Chenchen Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Lu Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Xiaoming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| |
Collapse
|
17
|
Xie LY, Yang Z, Wang Y, Hu JN, Lu YW, Zhang H, Jiang S, Li W. 1- O-Actylbritannilactone Ameliorates Alcohol-Induced Hepatotoxicity through Regulation of ROS/Akt/NF-κB-Mediated Apoptosis and Inflammation. ACS OMEGA 2022; 7:18122-18130. [PMID: 35664604 PMCID: PMC9161245 DOI: 10.1021/acsomega.2c01681] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/04/2022] [Indexed: 05/20/2023]
Abstract
1-O-Acetylbritannilactone (ABL) is a marker component of Inula britannica L. and is reported to exhibit multiple pharmacological activities, including antiaging, anti-inflammatory, and antidiabetic properties. Although the protective effect of Inula britannica L. on animal models of liver injury has been widely reported, the effect of ABL on alcohol-induced liver damage has not been confirmed. The present study was designed to investigate the protective effect of ABL against alcohol-induced LO2 human normal liver cell injury and to further clarify the underlying mechanism. Our results revealed that ABL at concentrations of 0.5, 1, and 2 μM could remarkably suppress the decreased viability of LO2 cells stimulated by alcohol. In addition, ABL pretreatment improved alcohol-induced oxidative damage by decreasing the level of reactive oxygen species (ROS) and the excessive consumption of glutathione peroxidase (GSH-Px), while increasing the level of catalase (CAT) in LO2 cells. Moreover, Western blotting analysis showed that ABL pretreatment activated protein kinase B (Akt) phosphorylation, increased downstream antiapoptotic protein Bcl-2 expression, and decreased the phosphorylation level of the caspase family including caspase 9 and caspase 3 proteins, thereby attenuating LO2 cell apoptosis. Importantly, we also found that ABL significantly inhibits the activation of the nuclear factor-kappa B (NF-κB) signaling pathway by reducing the secretion of proinflammatory factors including tumor necrosis factor-α (TNF-α) and interleukin (IL-1β). In conclusion, the current research clearly suggests that the protective effect of ABL on alcohol-induced hepatotoxicity may be achieved in part through regulation of the ROS/Akt/NF-κB signaling pathway to inhibit inflammation and apoptosis in LO2 cells. (The article path map has not been seen.).
Collapse
Affiliation(s)
- Li-ya Xie
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Zhen Yang
- Jilin
Academy of Chinese Medicine Sciences, Changchun 130012, China
| | - Ying Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Jun-nan Hu
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Ya-wei Lu
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Hao Zhang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Shuang Jiang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- E-mail: . Phone/Fax: +86-431-84533304
| | - Wei Li
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- E-mail: . Phone/Fax: +86-431-84533304
| |
Collapse
|
18
|
Wang Y, Zhang JJ, Hou JG, Li X, Liu W, Zhang JT, Zheng SW, Su FY, Li W. Protective Effect of Ginsenosides from Stems and Leaves of Panax ginseng against Scopolamine-Induced Memory Damage via Multiple Molecular Mechanisms. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1113-1131. [PMID: 35475974 DOI: 10.1142/s0192415x22500458] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although growing evidence has shown that ginsenosides from stems and leaves of Panax ginseng (GSLS) exercise a protective impact on the central nervous system, in the model of memory damage induced by scopolamine, it is still rarely reported. Thus, the mechanism of action needs to be further explored. This study was to investigate the effect of GSLS on scopolamine (SCOP)-induced memory damage and the underlying mechanism. Male ICR mice were treated with SCOP (3 mg/kg) for 7 days, with or without GSLS (75 and 150 mg/kg) treatment for 14 days. After GSLS treatment, the memory damage induced by SCOP was significantly ameliorated as shown by the improvement of cholinergic function (AChE and ChAT), brain tissue hippocampus morphology (H&E staining), and oxidative stress (MDA, GSH, and NO). Meanwhile, immunohistochemical assay suggested that GSLS increased the expression of brain-derived neurotrophic factor (BDNF) and Tyrosine Kinase receptor B (TrkB). Further mechanism research indicated that GSLS inhibited the Tau hyperphosphorylation and cell apoptosis by regulating the PI3K/AKT pathway and inhibited neuroinflammation by regulating the NF-κB pathway, thereby exerting a cognitive impairment improvement effect. This work suggested that GSLS could protect against SCOP-induced memory defects possibly through inhibiting oxidative stress, inhibiting neuroinflammation and cell apoptosis.
Collapse
Affiliation(s)
- Ying Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jin-Gang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- Intelligent Synthetic Biology Center, Daejeon 34141, Republic of Korea
| | - Xin Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Wei Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Si-Wen Zheng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Feng-Yan Su
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| |
Collapse
|
19
|
Jia Y, Guo H, Cheng X, Zhang Y, Si M, Shi J, Ma D. Hesperidin protects against cisplatin-induced cardiotoxicity in mice by regulating the p62-Keap1-Nrf2 pathway. Food Funct 2022; 13:4205-4215. [PMID: 35332348 DOI: 10.1039/d2fo00298a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hesperidin (HES) is an abundant and economical dietary bioflavonoid, and it has several pharmacological properties such as antioxidant activity and powerful cardiac protection. However, HES protection against cisplatin (CP)-induced cardiotoxicity and its mechanism have not been fully clarified. The current study was performed to further elucidate the mechanism of HES against CP-induced cardiotoxicity. Mice were orally administered HES (100 or 300 mg kg-1 day-1) for 7 consecutive days and then injected intraperitoneally (i.p.) with CP (5 mg kg-1) on days 3 and 6. On day 8, mice were anaesthetised with sodium pentobarbital (50 mg kg-1, i.p.), and blood and heart samples were collected for analysis. HES treatment reduced CP-induced cardiac pathologic damage and leakage of the myocardial markers cardiac troponin I (cTnI), creatine kinase (CK), and lactate dehydrogenase (LDH). HES treatment reduced levels of reactive oxygen species (ROS) and malondialdehyde (MDA), which is an oxidative product, and increased antioxidant marker levels including superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH). HES also reduced the CP-induced release of the inflammatory factors tumour necrosis factor (TNF)-α and interleukin (IL)-6. Additionally, HES treatment up-regulated the expression of anti-apoptotic protein Bcl-2 and down-regulated the expression of pro-apoptotic proteins Bax and Caspase-3. HES treatment also improved the expression of pathway proteins p62 and Nrf2 and inhibited the increase in CP-induced Keap1 expression. Thus, HES may provide protection against CP cardiotoxicity through inhibiting oxidative stress, inflammation, and apoptosis, which may contribute to activation of the p62-Keap1-Nrf2 signalling pathway. These findings suggest that HES may be a promising protective agent against CP cardiotoxicity in future anticancer clinical practice.
Collapse
Affiliation(s)
- Yuxin Jia
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China.
| | - Hui Guo
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China.
| | - Xizhen Cheng
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China.
| | - Yuling Zhang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China.
| | - Mingdong Si
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China.
| | - Jing Shi
- Department of Scientific Research Management, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
| | - Donglai Ma
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China. .,Hebei Technology Innovation Center of TCM Formula Preparations, Shijiazhuang, 050200, Hebei, China
| |
Collapse
|
20
|
Xing JJ, Mi XJ, Hou JG, Cai EB, Zheng SW, Wang SH, Wang Z, Chen C, Li W. Maltol mitigates cisplatin-evoked cardiotoxicity via inhibiting the PI3K/Akt signaling pathway in rodents in vivo and in vitro. Phytother Res 2022; 36:1724-1735. [PMID: 35174550 DOI: 10.1002/ptr.7405] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 01/12/2023]
Abstract
Our current research aims to evaluate the efficiency of a flavor enhancer, maltol (produced by heating ginseng) against cisplatin-evoked cardiotoxicity by establishing cisplatin-induced heart injury in vivo and H9C2 rat cardiomyocyte model. The cisplatin-treated mice at 3 mg/kg for four times on the 7th, 9th, 11th and 13th day, and in them appeared a serious cardiac damage accompanied with the increase in indicators of heart damage. Multiple exposure of 3 mg/kg for four times of cisplatin increased cardiac cells apoptosis with increased expression of Bax and cleaved-caspase 3, and decreased expression of Bcl-2. Interestingly, supplement of maltol at doses of 50 and 100 mg/kg for 15 days significantly suppressed the cardiac disturbance. In cultured H9C2 cells, maltol enhanced PI3K/Akt expression level during cisplatin treatment, and reduced cisplatin-induced apoptosis. Notably, inhibition of PI3K/Akt by LY294002 and HY-10249A lessened the efficacy of maltol. In mice, maltol apparently induced PI3K/Akt in heart tissues and protected against cisplatin-induced cardiotoxicity. In conclusion, maltol exerted the protective effects against cisplatin-induced cardiotoxicity, at least partially by inhibiting the activation of PI3K/Akt signaling pathways in cardiomyocytes, to ease oxidative stress, and alleviate reactive oxygen species-mediated apoptosis.
Collapse
Affiliation(s)
- Jing-Jing Xing
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Xiao-Jie Mi
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jin-Gang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - En-Bo Cai
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Si-Wen Zheng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Shi-Han Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| |
Collapse
|
21
|
Lin M, Xiong W, Wang S, Li Y, Hou C, Li C, Li G. The Research Progress of Trastuzumab-Induced Cardiotoxicity in HER-2-Positive Breast Cancer Treatment. Front Cardiovasc Med 2022; 8:821663. [PMID: 35097033 PMCID: PMC8789882 DOI: 10.3389/fcvm.2021.821663] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
In recent years, the incidence of breast cancer has been increasing on an annual basis. Human epidermal growth factor receptor-2 (HER-2) is overexpressed in 15-20% human breast cancers, which is associated with poor prognosis and a high recurrence rate. Trastuzumab is the first humanized monoclonal antibody against HER-2. The most significant adverse effect of trastuzumab is cardiotoxicity, which has become an important factor in limiting the safe use of the drug. Unfortunately, the mechanism causing this cardiotoxicity is still not completely understood, and the use of preventive interventions remains controversial. This article focuses on trastuzumab-induced cardiotoxicity, reviewing the clinical application, potential cardiotoxicity, mechanism and discussing the potential interventions through summarizing related researches over the past tens of years.
Collapse
Affiliation(s)
- Mengmeng Lin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiping Xiong
- Department of Cardiology, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Shiyuan Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingying Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunying Hou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyu Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guohui Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Exploring the potential mechanism of Rhodomyrtus tomentosa (Ait.) Hassk fruit phenolic rich extract on ameliorating nonalcoholic fatty liver disease by integration of transcriptomics and metabolomics profiling. Food Res Int 2022; 151:110824. [PMID: 34980375 DOI: 10.1016/j.foodres.2021.110824] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), as the commonest form of chronic liver disease, is accompanied by liver oxidative stress and inflammatory responses. Rhodomyrtus tomentosa (Ait.) Hassk fruit phenolic rich extract (RTE) possesses multiple pharmacological effects in management of chronic diseases. In this study, the liver-protective effect of RTE on mice with high-fat-diet (HFD)-induced NAFLD was investigated for the first time, and the underlying molecular mechanism was explored via integration of transcriptomics and metabolomics. The results showed that RTE mitigated liver damage, which was evidenced by declined inflammatory cell infiltration in liver, decreased liver function markers, oxidative stress indexes, lipid profile levels and inflammatory cytokines levels. The differential metabolites by metabonomics illustrated supplementation of RTE affected metabolomics pathways including tryptophan metabolism, alanine, aspartate and glutamate metabolism, D-glutamine and D-glutamate metabolism, cysteine and methionine metabolism, arginine and proline metabolism, which are all involved in oxidative stress and inflammation. Furthermore, the five differential expression genes (DEGs) through liver transcriptomics were screened and recognized, namely Tnfrsf21, Ifit1, Inhbb, Mapk15 and Gadd45g, which revealed that HFD induced Cytokine-cytokine receptor interaction pathway, NF-κB signaling pathway NOD-like receptor pathway, TNF signaling pathway. Integrated analysis of transcriptomics and metabolomics confirmed the supplementation of RTE had significantly regulatory effects on the metabolic pathways involved in inflammatory responses. Additionally, RT-PCR and western blot authenticated RTE intervention regulated the mRNA levels of liver genes involved in inflammation response and inhibited the liver endotoxin-TLR4-NF-κB pathway triggered by HFD, thus alleviating NAFLD. Our findings strongly support the possibility that RTE can be regarded as a potential therapeutic method for obesity-associated NAFLD.
Collapse
|
23
|
Zhang X, Han L, Li P, Zhang S, Zhang M, Li X, Chu J, Wang L, Tu P, Zhang Y, Liu K. Region-Specific Biomarkers and Their Mechanisms in the Treatment of Lung Adenocarcinoma: A Study of Panax quinquefolius from Wendeng, China. Molecules 2021; 26:molecules26226829. [PMID: 34833921 PMCID: PMC8623508 DOI: 10.3390/molecules26226829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/16/2022] Open
Abstract
Panax quinquefolius, a popular medicinal herb, has been cultivated in China for many years. In this work, the region-specific profiles of metabolites in P. quinquefolius from Wendeng was investigated using liquid-chromatography-quadrupole-time-of-flight-(LC-Q-TOF)-based metabolomics analysis. The three most abundant biomarkers, identified as ginsenoside Rb3, notoginsenoside R1, and ginsenoside Rc, were the representative chemical components employed in the network pharmacology analysis. In addition, molecular docking and western blotting analyses revealed that the three compounds were effective binding ligands with Hsp90α, resulting in the inactivation of SRC and PI3K kinase, which eventually led to the inactivation of the Akt and ERK pathways and lung cancer suppression. The outcomes obtained herein demonstrated the intriguing chemical characteristics and potential functional activities of P. quinquefolius from Wendeng.
Collapse
Affiliation(s)
- Xuanming Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Liwen Han
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 256200, China;
| | - Peihai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Shanshan Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Mengqi Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Xiaobin Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Jie Chu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Lizhen Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China;
| | - Yun Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
- Correspondence: (Y.Z.); (K.L.)
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (X.Z.); (P.L.); (S.Z.); (M.Z.); (X.L.); (J.C.); (L.W.)
- Correspondence: (Y.Z.); (K.L.)
| |
Collapse
|
24
|
Xu J, Zhang B, Chu Z, Jiang F, Han J. Wogonin Alleviates Cisplatin-induced Cardiotoxicity in Mice Via Inhibiting Gasdermin D-mediated Pyroptosis. J Cardiovasc Pharmacol 2021; 78:597-603. [PMID: 34651602 PMCID: PMC8492184 DOI: 10.1097/fjc.0000000000001085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/29/2021] [Indexed: 01/15/2023]
Abstract
ABSTRACT Cardiotoxicity has been well documented as a side effect of cisplatin (CDDP) treatment. The inflammatory response plays a crucial role in the pathological process of CDDP-induced cardiotoxicity. Wogonin is a natural flavonoid compound that possesses cardioprotective and anti-inflammatory qualities. Knowledge of the pharmacological effect and mechanism of wogonin could reveal an efficient way to identify therapeutic strategies. In this study, the potential of wogonin to antagonize CDDP-induced cardiotoxicity was evaluated in C57BL/6 mice in vivo and in H9c2 cells in vitro. The results showed that wogonin protected against CDDP-induced cardiac dysfunction, myocardial injury, and pyroptosis in vivo. Using a Gasdermin D expression plasmid, we revealed that wogonin dramatically reduced CDDP-induced pyroptosis by modulating the Gasdermin D protein in H9c2 cells. In conclusion, wogonin has great potential in attenuating CDDP-induced cardiotoxicity. In addition, greater emphasis should be placed on the antipyroptotic effects of wogonin for the treatment of other diseases.
Collapse
Affiliation(s)
- Jiajun Xu
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bin Zhang
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhenliang Chu
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fenfen Jiang
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
25
|
Bayrak S, Aktaş S, Altun Z, Çakir Y, Tütüncü M, Kum Özşengezer S, Yilmaz O, Olgun N. Antioxidant effect of acetyl-l-carnitine against cisplatin-induced cardiotoxicity. J Int Med Res 2021; 48:300060520951393. [PMID: 32865065 PMCID: PMC7469739 DOI: 10.1177/0300060520951393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective Cisplatin (CDDP) toxicity is a dose-limiting clinical problem in clinical practice, mainly because of nephrotoxicity or ototoxicity. However, the mechanism of CDDP-induced cardiotoxicity is poorly understood. Acetyl-l-carnitine (ALCAR) is an antioxidant agent with protective effects against the side effects of various chemotherapeutics. CDDP-induced cardiotoxicity and the protective role of ALCAR were evaluated in this study. Methods Morphological changes were evaluated in hematoxylin and eosin-stained sections, and immunohistochemistry for caspase-3, superoxide dismutase-2 (SOD-2), inducible nitrite oxide synthase (iNOS), cyclooxygenase-2, and Bcl-2 was performed using the hearts of athymic nude mice carrying xenograft neuroblastoma tumors. Mice were randomized (six/group) to the control, CDDP (16 mg/kg), and ALCAR (200 mg/kg)+CDDP (16 mg/kg) groups. Results were analyzed using nonparametric tests. Results No difference was observed in the rates of cardiac necrosis, dilated/congested blood vessels, hemorrhage, polymorphonuclear leukocyte infiltration, edema, and pyknotic nuclei among the groups. SOD-2 expression was increased in the CDDP group but not in the ALCAR+CDDP group. iNOS, Bcl-2, and caspase-3 levels were not significantly different among the groups. Conclusions ALCAR might be a candidate protective agent for CDDP-induced cardiotoxicity. SOD-2, as a member of the oxidant system, should be evaluated in further studies as a biomarker of cardiotoxicity.
Collapse
Affiliation(s)
- Serdar Bayrak
- Dokuz Eylul University Institute of Oncology, Department of Clinical Oncology, İzmir, Turkey
| | - Safiye Aktaş
- Dokuz Eylul University Institute of Oncology, Department of Basic Oncology, Izmir, Turkey
| | - Zekiye Altun
- Dokuz Eylul University Institute of Oncology, Department of Basic Oncology, Izmir, Turkey
| | - Yasemin Çakir
- Dokuz Eylul University Institute of Oncology, Department of Basic Oncology, Izmir, Turkey
| | - Merve Tütüncü
- Dokuz Eylul University Institute of Oncology, Department of Basic Oncology, Izmir, Turkey
| | - Selen Kum Özşengezer
- Dokuz Eylul University Institute of Oncology, Department of Basic Oncology, Izmir, Turkey
| | - Osman Yilmaz
- Dokuz Eylul University, Department of Laboratory Animal Science, Izmir, Turkey
| | - Nur Olgun
- Dokuz Eylul University Institute of Oncology, Department of Pediatric Oncology, Izmir, Turkey
| |
Collapse
|
26
|
Multidirectional effects of saponin fraction isolated from the leaves of sea buckthorn Elaeagnus rhamnoides (L.) A. Nelson. Biomed Pharmacother 2021; 137:111395. [PMID: 33761611 DOI: 10.1016/j.biopha.2021.111395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 01/11/2023] Open
Abstract
Many studies show that saponins isolated from various plants have a cytotoxic effect on cancer cells inducing apoptosis and autophagy. On the other hand, saponins also exhibit a number of beneficial properties, such as antioxidant properties. Thus, saponins can be considered both in terms of their therapeutic and protective effects during anticancer treatment. In this study, we investigated the effect of the saponin fraction isolated from sea buckthorn (Elaeagnus rhamnoides (L.) A. Nelson) leaves on the viability of HL-60 cancer cells using resazurin assay and its ability to induction of apoptosis with Annexin V-FITC and propidium iodide (PI) double staining. Moreover, we studied its effect on the oxidative stress induced by H2O2, and anti-platelet and anticoagulant potential in whole blood using T-TAS, a microchip-based flow chamber system. We observed that the saponin fraction significantly decreased the viability of HL-60 cells at the concentration above 50 µg/mL and induced apoptosis at the concentration of 100 µg/mL. Moreover, we observed that saponin fraction used at lower concentrations, such as 0.5 and 1 µg/mL, stimulated HL-60 cells and increased their viability. The saponin fraction also decreased the level of free radicals and reduced oxidative DNA damage measured by the comet assay. However, at high concentration of oxidant H2O2 equal 5 mM, we noticed that the saponin fraction at 50 µg/mL increased the level of free radicals in HL-60 cells. We also demonstrated anticoagulant potential of the saponin fraction at the concentration of 50 µg/mL. Our results indicate that the saponin fraction obtained from sea buckthorn leaves can show both chemotherapeutic and chemoprotective potential.
Collapse
|
27
|
Hu JN, Yang JY, Jiang S, Zhang J, Liu Z, Hou JG, Gong XJ, Wang YP, Wang Z, Li W. Panax quinquefolium saponins protect against cisplatin evoked intestinal injury via ROS-mediated multiple mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153446. [PMID: 33387967 DOI: 10.1016/j.phymed.2020.153446] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Cisplatin is one of the most common chemotherapeutic drugs. Cisplatin-induced toxicity gives rise to gastrointestinal cell damage, subsequent diarrhea and vomiting, leading to the discontinuation of its clinical application in long-term cancer chemotherapy. Panax quinquefolium L., also known as American ginseng, has many pharmacological activities such as improving immunity, anti-tumor, anti-radiation and blood sugar lowering. PURPOSE Previously, our laboratory reported that American ginseng berry extract could alleviate chemotherapeutic agents-induced renal damage caused by cisplatin. Hence, this study further explored the protective effect of P. quinquefolium saponins (PQS) on cisplatin-induced intestinal injury in mice and the possible molecular mechanisms. METHODS Biochemical markers, levels of inflammatory factors, histopathological staining and western blotting were used to analyze intestinal injury based on various molecular mechanisms. RESULTS We demonstrated the destruction of the intestinal barrier caused by cisplatin exposure by detecting the activity of diamine oxidase (DAO) and the expression of tight junction proteins zonula occludens-1 (ZO-1) and occludin. Meanwhile, cisplatin exposure changed SOD and MDA levels in the small intestine, causing oxidative damage to the intestinal mucosa. The inflammation associated-intestinal damage was further explored by the measurement of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and analysis of nuclear factor-kappa B (NF-κB) inflammatory pathway protein expression. Moreover, apoptotic cells labeled with TUNEL staining-positive cells and activated caspase family proteins suggest that cisplatin induces intestinal apoptosis. Interestingly, PQS pretreatment significantly reversed these situations. CONCLUSION These evidences clearly suggest that PQS can alleviate cisplatin-induced intestinal damage by inhibiting oxidative stress, reducing the occurrence of inflammation and apoptosis, and improving intestinal barrier function.
Collapse
Affiliation(s)
- Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Jia-Yu Yang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Jing Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Jin-Gang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China
| | - Xiao-Jie Gong
- College of Life Science, Dalian Minzu University, Dalian 116600 China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118 China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China.
| |
Collapse
|
28
|
Ginsenoside Rb3 Alleviates the Toxic Effect of Cisplatin on the Kidney during Its Treatment to Oral Cancer via TGF- β-Mediated Mitochondrial Apoptosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6640714. [PMID: 33510805 PMCID: PMC7826210 DOI: 10.1155/2021/6640714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/03/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022]
Abstract
Objective The research aimed to confirm the role of the transforming growth factor-β (TGF-β) in cisplatin- (CPT-) evoked kidney toxicity and elucidate the mechanism that ginsenoside Rb3 (Rb3) could alleviate the kidney toxicity by CPT during its treatment to oral cancer via TGF-β-mediated mitochondrial apoptosis. Methods The model of xenograft nude mice bearing oral carcinoma cells ACC83 was established and treated with CPT and/or Rb3, respectively. Bodyweights of the treated mice were weighed, and the kidney tissues were collected; following, the histopathology and the expression of TGF-β were examined using H&E staining and immunohistochemistry. Afterward, the renal cells GP-293 were treated with CPT and/or Rb3. The expression and phosphoration of TGF-β, Smad2, Smad3, Bcl-2, and Bax in GP-293 cells were detected by Western blotting. The cellular apoptosis and mitochondrial membrane potential were analyzed using flow cytometry. Results The xenograft nude mice exposure to CPT presented the bodyweight loss, necrotic areas, and the increased expression of TGF in kidney tissue, and Rb3 pretreatment relieved these changes evoked by CPT. In GP-293 cells, CPT administration induced the phosphorylation of Smad2 and Smad3, and Rb3 pretreatment suppressed the induced phosphorylation by CPT. Besides, flow cytometry analysis showed that Rb3 inhibited the CPT-evoked cellular apoptosis ratio and mitochondrial membrane depolarization. The Western blotting test indicated that Rb3 alleviated the cleavage of PARP, caspase 3, caspase 8, and caspase 9, the induction of Bax expression, and inhibition of Bcl-2 expression. Additionally, after treating with the TGF inhibitor of disitertide, Rb3 exhibited no alleviation effects on CPT-evoked cellular apoptosis ratio, inhibition of Bax expression, and induction of Bcl-2 expression in GP-293 cells. Conclusion Rb3 could alleviate CPT-evoked toxic effects on kidney cells during its treatment to oral cancer via TGF-β-mediated mitochondrial apoptosis.
Collapse
|
29
|
Yu P, Li Y, Fu W, Li X, Liu Y, Wang Y, Yu X, Xu H, Sui D. Panax quinquefolius L. Saponins Protect Myocardial Ischemia Reperfusion No-Reflow Through Inhibiting the Activation of NLRP3 Inflammasome via TLR4/MyD88/NF-κB Signaling Pathway. Front Pharmacol 2021; 11:607813. [PMID: 33628178 PMCID: PMC7898550 DOI: 10.3389/fphar.2020.607813] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022] Open
Abstract
At present, many patients who undergo reperfusion immediately after percutaneous coronary intervention will undergo microvascular obstruction and reduction in myocardial blood flow. This phenomenon is called "no-reflow (NR)," and there is still no effective therapy for NR. Studies showed Panax quinquefolius L. saponins (PQS) have effect on MI/R injury, while the effect and mechanism of PQS on MI/R induced NR are not clear. In this study, we established a MI/R model to investigate whether PQS decrease NR phenomenon via suppression of inflammation. We found that PQS significantly alleviated the symptoms of NR by reducing ischemia, infarction, and NR area; improving cardiac function; preventing pathological morphology changes of myocardium; depressing leukocytes' aggregation and adhesion; and suppressing the excessive inflammation. Further study demonstrated that PQS remarkably inhibited TLR4, MyD88, p-NF-κB, and NLRP3 inflammasome-associated protein, and these effects could be reversed by LPS. These results indicated that PQS may protect NR by inhibiting the activation of NLRP3 inflammasome via TLR4/MyD88/NF-κB signaling pathway in part, suggesting that PQS exist potential in preventing NR induced by MI/R.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
30
|
Lin Z, Bao Y, Hong B, Wang Y, Zhang X, Wu Y. Salvianolic acid B attenuated cisplatin-induced cardiac injury and oxidative stress via modulating Nrf2 signal pathway. J Toxicol Sci 2021; 46:199-207. [PMID: 33952797 DOI: 10.2131/jts.46.199] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cardiovascular complications have been well documented as the downside to conventional cancer chemotherapy. As a notable side effect of cisplatin (CDDP), cardiotoxicity represents a major obstacle to the successful treatment of cancer. It has been reported that Salvianolic acid B (SalB) possesses cardioprotective quality. However, the effect of SalB on cardiac damage caused by conventional cancer chemotherapy remains unclear. In this study, we clarified the protective effect of SalB on cisplatin-induced heart injury. Furthermore, in H9c2 cells, SalB dramatically reduced cisplatin-induced apoptosis and oxidative stress by modulating the nuclear factor erythroid-2-related factor 2 (Nrf2) signaling pathway. In conclusion, SalB had great potential in mitigating cisplatin-induced cardiac injury. Furthermore, more attention should be placed on natural active compounds containing SalB with antioxidant effects for the treatment of cardiomyopathy.
Collapse
Affiliation(s)
- Zhenjian Lin
- Department of Pharmacy, Sanmen People's Hospital of Zhejiang, China
| | - Yuyan Bao
- Department of Pharmacy, Sanmen People's Hospital of Zhejiang, China
| | - Bing Hong
- Department of Pharmacy, Sanmen People's Hospital of Zhejiang, China
| | - Yangyang Wang
- Department of Pharmacy, Sanmen People's Hospital of Zhejiang, China
| | - Xiaomin Zhang
- Department of Pharmacy, Sanmen People's Hospital of Zhejiang, China
| | - Yaping Wu
- Department of Pharmacy, Sanmen People's Hospital of Zhejiang, China
| |
Collapse
|
31
|
Yarmohammadi F, Hayes AW, Karimi G. Natural compounds against cytotoxic drug-induced cardiotoxicity: A review on the involvement of PI3K/Akt signaling pathway. J Biochem Mol Toxicol 2020; 35:e22683. [PMID: 33325091 DOI: 10.1002/jbt.22683] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/09/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022]
Abstract
Cardiotoxicity is a critical concern in the use of several cytotoxic drugs. Induction of apoptosis, inflammation, and autophagy following dysregulation of the PI3K/Akt signaling pathway contributes to the cardiac damage induced by these drugs. Several natural compounds (NCs), including ferulic acid, gingerol, salvianolic acid B, paeonol, apigenin, calycosin, rutin, neferine, higenamine, vincristine, micheliolide, astragaloside IV, and astragalus polysaccharide, have been reported to suppress cytotoxic drug-induced cardiac injury. This article reviews these NCs that have been reported to have a protective effect against cytotoxic drug-induced cardiotoxicity through regulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- College of Public Health, University of South Florida, Tampa, Florida, USA.,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Qi Y, Ying Y, Zou J, Fang Q, Yuan X, Cao Y, Cai Y, Fu S. Kaempferol attenuated cisplatin-induced cardiac injury via inhibiting STING/NF-κB-mediated inflammation. Am J Transl Res 2020; 12:8007-8018. [PMID: 33437376 PMCID: PMC7791507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/26/2020] [Indexed: 06/12/2023]
Abstract
Cardiovascular complications have been well documented as the downside to conventional cancer chemotherapy. As a notable side effect of cisplatin, cardiotoxicity represents a major obstacle to the successful treatment of cancer. It has been reported that kaempferol (KPF) possesses cardioprotective and anti-inflammatory qualities. However, the effect of KPF on cardiac damage caused by conventional cancer chemotherapy remains unclear. In this study, we clarified the protective effect of KPF on cisplatin-induced heart injury, and conducted in-depth research on the molecular mechanism underlying this effect. The results showed that KPF protected against cardiac dysfunction and injury induced by cisplatin in vivo. In H9c2 cells, KPF dramatically reduced cispaltin-induced apoptosis and inflammatory response by modulating STING/NF-κB pathway. In conclusion, these results showed that KPF had great potential in attenuating cisplatin-induced cardiac injury. Besides, greater emphasis should be placed in the future on natural active compounds containing KPF with anti-inflammatory effects for the treatment of these diseases.
Collapse
Affiliation(s)
- Yajun Qi
- Department of Pharmacy, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Jie Zou
- Department of Pharmacy, The 903rd Hospital of PLAHangzhou 310000, Zhejiang, China
| | - Qilu Fang
- Department of Pharmacy, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| | - Xiaohong Yuan
- Department of Anesthesiology, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310012, Zhejiang, China
| | - Yingying Cao
- Department of Pharmacy, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| | - Yunfang Cai
- Department of Anesthesiology, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310012, Zhejiang, China
| | - Shuang Fu
- Department of Anesthesiology, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310012, Zhejiang, China
| |
Collapse
|
33
|
Inhibition of p300 by Garcinol Protects against Cisplatin-Induced Acute Kidney Injury through Suppression of Oxidative Stress, Inflammation, and Tubular Cell Death in Mice. Antioxidants (Basel) 2020; 9:antiox9121271. [PMID: 33327548 PMCID: PMC7765028 DOI: 10.3390/antiox9121271] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/03/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence suggests that epigenetic mechanisms such as histone modification are crucially involved in the pathophysiology of acute kidney injury (AKI). The histone acetyltransferase p300 regulates several biological processes through the acetylation of histones or transcription factors. However, the role of p300 in cisplatin-induced AKI remains poorly understood. Therefore, we investigated the effects of garcinol, a potent p300 inhibitor, on cisplatin-induced AKI and explored the mechanisms. Administration of garcinol significantly reversed the upregulation of p300 and increased acetylation of histone H3, along with amelioration of renal dysfunction and histopathological injury in the kidneys of cisplatin-injected mice. Garcinol also attenuated oxidative stress and reduced expression of pro-oxidant enzymes. In addition, garcinol reduced the elevated production of cytokines and chemokines and suppressed immune cell accumulation together with downregulation of vascular adhesion molecules. These beneficial effects of garcinol were associated with a reduction in acetylation of the p65 subunit of nuclear factor kappa-B. Further, garcinol significantly inhibited apoptosis and caspase-3 activation, with a decrease in p53 acetylation in cisplatin-injected mice. Taken together, we demonstrated that the inhibition of p300 by garcinol ameliorated cisplatin-induced renal injury, presumably through epigenetic mechanisms. These results suggest that garcinol might be a potential preventive agent for cisplatin-induced AKI.
Collapse
|
34
|
Saponins Extracted from Tea ( Camellia Sinensis) Flowers Induces Autophagy in Ovarian Cancer Cells. Molecules 2020; 25:molecules25225254. [PMID: 33187244 PMCID: PMC7696525 DOI: 10.3390/molecules25225254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 12/25/2022] Open
Abstract
Tea flower saponins (TFS) possess effective anticancer properties. The diversity and complexity of TFS increases the difficulty of their extraction and purification from tea flowers. Here, multiple methods including solvent extraction, microporous resin separation and preparative HPLC separation were used to obtain TFS with a yield of 0.34%. Furthermore, we revealed that TFS induced autophagy—as evidenced by an increase in MDC-positive cell populations and mCherry-LC3B-labeled autolysosomes and an upregulation of LC3II protein levels. 3-MA reversed the decrease in cell viability induced by TFS, showing that TFS induced autophagic cell death. TFS-induced autophagy was not dependent on the Akt/mTOR/p70S6K signaling pathway. TFS-induced autophagy in OVCAR-3 cells was accompanied by ERK pathway activation and reactive oxygen species (ROS) generation. This paper is the first report of TFS-mediated autophagy of ovarian cancer cells. These results provide new insights for future studies of the anti-cancer effects of TFS.
Collapse
|
35
|
Żuchowski J, Skalski B, Juszczak M, Woźniak K, Stochmal A, Olas B. LC/MS Analysis of Saponin Fraction from the Leaves of Elaeagnus rhamnoides (L.) A. Nelson and Its Biological Properties in Different In Vitro Models. Molecules 2020; 25:molecules25133004. [PMID: 32630067 PMCID: PMC7411717 DOI: 10.3390/molecules25133004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
This study focuses on saponin fraction from sea buckthorn (Elaeagnus rhamnoides (L.) A. Nelson) leaves. It has known that for example teas from sea buckthorn leaves have anti-obesity properties. The objective of our present experiments was to investigate both the chemical composition of saponin fraction, as well as their biological properties in different in vitro models (using human plasma, blood platelets, and peripheral blood mononuclear cells (PBMCs)). We observed that saponin fraction reduces plasma lipid peroxidation and protein carbonylation induced by H2O2/Fe. This fraction also decreased DNA oxidative damage induced by H2O2 in PBMCs. Regarding the cytotoxicity of saponin fraction (0.5–50 µg/mL) none was found to cause lysis of blood platelets, and PBMCs. Our results, for the first time indicate that saponin fraction from sea buckthorn leaves may be a new promising source of compounds for prophylaxis and treatment of diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Jerzy Żuchowski
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation, State Research Institute, 24-100 Puławy, Poland; (J.Ż); (A.S.)
| | - Bartosz Skalski
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Łódź, Poland;
| | - Michał Juszczak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Łódź, Poland; (M.J.); (K.W.)
| | - Katarzyna Woźniak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Łódź, Poland; (M.J.); (K.W.)
| | - Anna Stochmal
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation, State Research Institute, 24-100 Puławy, Poland; (J.Ż); (A.S.)
| | - Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Łódź, Poland;
- Correspondence:
| |
Collapse
|
36
|
Wang J, Zhang W, Wu L, Mei Y, Cui S, Feng Z, Chen X. New insights into the pathophysiological mechanisms underlying cardiorenal syndrome. Aging (Albany NY) 2020; 12:12422-12431. [PMID: 32561688 PMCID: PMC7343447 DOI: 10.18632/aging.103354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
Communication between the heart and kidney occurs through various bidirectional pathways. The heart maintains continuous blood flow through the kidney while the kidney regulates blood volume thereby allowing the heart to pump effectively. Cardiorenal syndrome (CRS) is a pathologic condition in which acute or chronic dysfunction of the heart or kidney induces acute or chronic dysfunction of the other organ. CRS type 3 (CRS-3) is defined as acute kidney injury (AKI)-mediated cardiac dysfunction. AKI is common among critically ill patients and correlates with increased mortality and morbidity. Acute cardiac dysfunction has been observed in over 50% of patients with severe AKI and results in poorer clinical outcomes than heart or renal dysfunction alone. In this review, we describe the pathophysiological mechanisms responsible for AKI-induced cardiac dysfunction. Additionally, we discuss current approaches in the management of patients with CRS-3 and the development of targeted therapeutics. Finally, we summarize current challenges in diagnosing mild cardiac dysfunction following AKI and in understanding CRS-3 etiology.
Collapse
Affiliation(s)
- Jin Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Weiguang Zhang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Lingling Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Yan Mei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Zhe Feng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| |
Collapse
|
37
|
Yin M, Li N, Makinde EA, Olatunji OJ, Ni Z. N6-2-hydroxyethyl-adenosine ameliorate cisplatin induced acute kidney injury in mice. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1760149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Min Yin
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Na Li
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | | | | | - Ziyuan Ni
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|