1
|
Sun XH, Chai YH, Bai XT, Li HX, Yang PP, Xi YM. Saikosaponin A Mediates the Anti-Acute Myeloid Leukemia Effect via the P-JNK Signaling Pathway Induced by Endoplasmic Reticulum Stress. Drug Des Devel Ther 2025; 19:1983-2001. [PMID: 40124558 PMCID: PMC11928444 DOI: 10.2147/dddt.s498458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Objective This study aims to investigate the antitumor effects of saikosaponin A (SSA) on acute myeloid leukemia (AML) and elucidate its underlying mechanisms, particularly focusing on the endoplasmic reticulum stress (ERS)-mediated MAPK-p-JNK signaling pathway. Methods The inhibitory effects of SSA on the proliferation of AML cell lines K562 and HL60 were evaluated using CCK8 and EdU assays. Apoptotic effects induced by SSA were analyzed via flow cytometry. RNA sequencing was performed to identify differentially expressed genes and enriched signaling pathways. Western blot analysis was utilized to confirm the involvement of ERS and activation of the MAPK-p-JNK signaling pathway. Further validation of the potential mechanism of SSA-induced apoptosis was conducted using SP600125 and 4PBA. The in vivo anti-AML efficacy of SSA was assessed using a xenograft model. Results SSA exhibited significant inhibitory effects on the proliferation of AML cell lines K562 and HL60, with IC50 values at 12, 24, and 48 hours demonstrating time- and dose-dependency (19.84 μM, 17.86 μM, and 15.38 μM for K562; 22.73 μM, 17.02 μM, and 15.25 μM for HL60, respectively). Western blot analysis demonstrated that SSA induces apoptosis in AML cells through the mitochondrial apoptotic pathway. Transcriptomic profiling and Western blot analyses confirmed that SSA activates the ERS-mediated p-JNK signaling pathway to induce apoptosis in AML, a process that can be reversed by the addition of 4PBA or SP600125. Furthermore, SSA significantly reduced tumor volume and weight in a NOD-SCID mouse xenograft model without causing notable toxicity to the liver, kidneys, lungs, or heart, while also activating the ERS and p-JNK signaling pathways in vivo. Conclusion SSA induces apoptosis in AML cells by activating the ERS-mediated p-JNK signaling pathway, exhibiting significant anti-AML effects both in vitro and in vivo, accompanied by a favorable safety profile.
Collapse
MESH Headings
- Saponins/pharmacology
- Humans
- Oleanolic Acid/pharmacology
- Oleanolic Acid/analogs & derivatives
- Endoplasmic Reticulum Stress/drug effects
- Cell Proliferation/drug effects
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Animals
- Mice
- Apoptosis/drug effects
- Dose-Response Relationship, Drug
- MAP Kinase Signaling System/drug effects
- Drug Screening Assays, Antitumor
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Structure-Activity Relationship
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Molecular Structure
- HL-60 Cells
- K562 Cells
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Xiao-Hong Sun
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Yi-Hong Chai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Xiao-Teng Bai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Hong-Xing Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Pan-Pan Yang
- Department of Gynecology and Obstetrics, The First Hospital of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Ya-Ming Xi
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, People’s Republic of China
- Division of Hematology, The First Hospital of Lanzhou University, Lanzhou, 730000, People’s Republic of China
| |
Collapse
|
2
|
Sun XH, Chai YH, Bai XT, Li HX, Xi YM. Pharmacology, medical uses, and clinical translational challenges of Saikosaponin A: A review. Heliyon 2024; 10:e40427. [PMID: 39641011 PMCID: PMC11617869 DOI: 10.1016/j.heliyon.2024.e40427] [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: 04/25/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
Saikosaponin A (SSA), the primary active monomer derived from the Radix bupleuri, demonstrates a diverse array of pharmacological activities, including anti-inflammatory, antitumor, analgesic, anti-fibrotic, antidepressant, and immune-modulating properties. Despite its potential therapeutic impact on various human diseases, comprehensive studies exploring SSA's efficacy in these contexts remain limited. This review synthesizes the current research landscape regarding SSA's therapeutic applications across different diseases, highlighting critical insights to overcome existing limitations and clinical challenges. The findings underscore the importance of further investigations into SSA's mechanisms of action, facilitating the development of targeted therapeutic strategies and their translation into clinical practice.
Collapse
Affiliation(s)
- Xiao-Hong Sun
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Yi-Hong Chai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Xiao-Teng Bai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Hong-Xing Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Ya-Ming Xi
- Division of Hematology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
3
|
Chen Q, Wang J, Sun L, Ba B, Shen D. Mechanism of Astragalus membranaceus (Huangqi, HQ) for treatment of heart failure based on network pharmacology and molecular docking. J Cell Mol Med 2024; 28:e18331. [PMID: 38780500 PMCID: PMC11114218 DOI: 10.1111/jcmm.18331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/23/2024] [Accepted: 03/25/2024] [Indexed: 05/25/2024] Open
Abstract
Heart failure is a leading cause of death in the elderly. Traditional Chinese medicine, a verified alternative therapeutic regimen, has been used to treat heart failure, which is less expensive and has fewer adverse effects. In this study, a total of 15 active ingredients of Astragalus membranaceus (Huangqi, HQ) were obtained; among them, Isorhamnetin, Quercetin, Calycosin, Formononetin, and Kaempferol were found to be linked to heart failure. Ang II significantly enlarged the cell size of cardiomyocytes, which could be partially reduced by Quercetin, Isorhamnetin, Calycosin, Kaempferol, or Formononetin. Ang II significantly up-regulated ANP, BNP, β-MHC, and CTGF expressions, whereas Quercetin, Isorhamnetin, Calycosin, Kaempferol or Formononetin treatment partially downregulated ANP, BNP, β-MHC and CTGF expressions. Five active ingredients of HQ attenuated inflammation in Ang II-induced cardiomyocytes by inhibiting the levels of TNF-α, IL-1β, IL-18 and IL-6. Molecular docking shows Isorhamnetin, Quercetin, Calycosin, Formononetin and Kaempferol can bind with its target protein ESR1 in a good bond by intermolecular force. Quercetin, Calycosin, Kaempferol or Formononetin treatment promoted the expression levels of ESR1 and phosphorylated ESR1 in Ang II-stimulated cardiomyocytes; however, Isorhamnetin treatment had no effect on ESR1 and phosphorylated ESR1 expression levels. In conclusion, our results comprehensively illustrated the bioactives, potential targets, and molecular mechanism of HQ against heart failure. Isorhamnetin, Quercetin, Calycosin, Formononetin and Kaempferol might be the primary active ingredients of HQ, dominating its cardioprotective effects against heart failure through regulating ESR1 expression, which provided a basis for the clinical application of HQ to regulate cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Qiuxiang Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan UniversityHubei Key Laboratory of CardiologyWuhanChina
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Juan Wang
- Department of CardiologyThe Fifth Affiliated Hospital of Xinjiang medical UniversityUrumchiChina
| | - Lihua Sun
- Department of CardiologyThe Fifth Affiliated Hospital of Xinjiang medical UniversityUrumchiChina
| | - Bayinsilema Ba
- Department of CardiologyThe Fifth Affiliated Hospital of Xinjiang medical UniversityUrumchiChina
| | - Difei Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan UniversityHubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
4
|
Reis-Mendes A, Ferreira M, Padrão AI, Duarte JA, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. The Role of Nrf2 and Inflammation on the Dissimilar Cardiotoxicity of Doxorubicin in Two-Time Points: a Cardio-Oncology In Vivo Study Through Time. Inflammation 2024; 47:264-284. [PMID: 37833616 PMCID: PMC10799157 DOI: 10.1007/s10753-023-01908-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023]
Abstract
Doxorubicin (DOX) is a topoisomerase II inhibitor used in cancer therapy. Despite its efficacy, DOX causes serious adverse effects, such as short- and long-term cardiotoxicity. This work aimed to assess the short- and long-term cardiotoxicity of DOX and the role of inflammation and antioxidant defenses on that cardiotoxicity in a mice model. Adult CD-1 male mice received a cumulative dose of 9.0 mg/kg of DOX (2 biweekly intraperitoneal injections (ip), for 3 weeks). One week (1W) or 5 months (5M) after the last DOX administration, the heart was collected. One week after DOX, a significant increase in p62, tumor necrosis factor receptor (TNFR) 2, glutathione peroxidase 1, catalase, inducible nitric oxide synthase (iNOS) cardiac expression, and a trend towards an increase in interleukin (IL)-6, TNFR1, and B-cell lymphoma 2 associated X (Bax) expression was observed. Moreover, DOX induced a decrease on nuclear factor erythroid-2 related factor 2 (Nrf2) cardiac expression. In both 1W and 5M, DOX led to a high density of infiltrating M1 macrophages, but only the 1W-DOX group had a significantly higher number of nuclear factor κB (NF-κB) p65 immunopositive cells. As late effects (5M), an increase in Nrf2, myeloperoxidase, IL-33, tumor necrosis factor-α (TNF-α), superoxide dismutase 2 (SOD2) expression, and a trend towards increased catalase expression were observed. Moreover, B-cell lymphoma 2 (Bcl-2), cyclooxygenase-2 (COX-2), and carbonylated proteins expression decreased, and a trend towards decreased p38 mitogen-activated protein kinase (MAPK) expression were seen. Our study demonstrated that DOX induces adverse outcome pathways related to inflammation and oxidative stress, although activating different time-dependent response mechanisms.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Isabel Padrão
- Research Center in Physical Activity, Faculty of Sport, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - José Alberto Duarte
- Research Center in Physical Activity, Faculty of Sport, University of Porto, Porto, Portugal
- 1H-TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
- Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
5
|
Reis-Mendes A, Ferreira M, Duarte JA, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. The role of inflammation and antioxidant defenses in the cardiotoxicity of doxorubicin in elderly CD-1 male mice. Arch Toxicol 2023; 97:3163-3177. [PMID: 37676301 PMCID: PMC10567829 DOI: 10.1007/s00204-023-03586-1] [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: 06/26/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023]
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent used against several cancer types. However, due to its cardiotoxic adverse effects, the use of this drug may be also life-threatening. Although most cancer patients are elderly, they are poorly represented and evaluated in pre-clinical and clinical studies. Considering this, the present work aims to evaluate inflammation and oxidative stress as the main mechanisms of DOX-induced cardiotoxicity, in an innovative approach using an experimental model constituted of elderly animals treated with a clinically relevant human cumulative dose of DOX. Elderly (18-20 months) CD-1 male mice received biweekly DOX administrations, for 3 weeks, to reach a cumulative dose of 9.0 mg/kg. One week (1W) or two months (2 M) after the last DOX administration, the heart was collected to determine both drug's short and longer cardiac adverse effects. The obtained results showed that DOX causes cardiac histological damage and fibrosis at both time points. In the 1W-DOX group, the number of nuclear factor kappa B (NF-κB) p65 immunopositive cells increased and a trend toward increased NF-κB p65 expression was seen. An increase of inducible nitric oxide synthase (iNOS) and interleukin (IL)-33 and a trend toward increased IL-6 and B-cell lymphoma-2-associated X (Bax) expression were seen after DOX. In the same group, a decrease in IL-1β, p62, and microtubule-associated protein 1A/1B-light chain 3 (LC3)-I, p38 mitogen-activated protein kinase (MAPK) expression was observed. Contrariwise, the animals sacrificed 2 M after DOX showed a significant increase in glutathione peroxidase 1 and Bax expression with persistent cardiac damage and fibrosis, while carbonylated proteins, erythroid-2-related factor 2 (Nrf2), NF-κB p65, myeloperoxidase, LC3-I, and LC3-II expression decreased. In conclusion, our study demonstrated that in an elderly mouse population, DOX induces cardiac inflammation, autophagy, and apoptosis in the heart in the short term. When kept for a longer period, oxidative-stress-linked pathways remained altered, as well as autophagy markers and tissue damage after DOX treatment, emphasizing the need for continuous post-treatment cardiac monitoring.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - José Alberto Duarte
- Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, Research Center in Physical Activity, Health and Leisure (CIAFEL), University of Porto, 4200-450, Porto, Portugal
- 1H-TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116, Gandra, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, 4050-313, Porto, Portugal
- Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, 4450-208, Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
6
|
Xu Z, Zhou H, Zhang Y, Cheng Z, Wan M, Qin W, Li P, Feng J, Shao S, Xue W, Guo H, Liu B. Recent pharmacological advances in the treatment of cardiovascular events with Astragaloside IV. Biomed Pharmacother 2023; 168:115752. [PMID: 37875045 DOI: 10.1016/j.biopha.2023.115752] [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: 08/03/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death and disability globally. A wide range of CVDs have been reported, each of which diverges significantly, exhibiting sophisticated types of pathogenesis (e.g., inflammatory, oxidative stress, and disorders in cardiomyocyte metabolism). Compared with conventional treatments in modern medicine, traditional Chinese medicine (TCM) can exhibit comparative advantages in the treatment of CVDs. TCM can be utilized to develop effective strategies for addressing the challenges of CVD, with fewer side effects and higher therapeutic efficiency. Astragaloside IV (AS-IV) has been confirmed as one of the major active ingredients found in Astragalus membranaceus (a Chinese herbal medicine that has been extensively employed clinically for the treatments of CVDs). Since recent studies have shown that AS-IV in CVD treatments has achieved promising results, the substance has aroused great attention and further discussions in the field. The present review aims to summarize the recent pharmacological advances in employing AS-IV in the treatment of CVDs.
Collapse
Affiliation(s)
- Zehui Xu
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Houle Zhou
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yihan Zhang
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ziji Cheng
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Melisandre Wan
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanting Qin
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peiyu Li
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaming Feng
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuijin Shao
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenlong Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Haidong Guo
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Baonian Liu
- Department of Anatomy, College of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
7
|
Wang Q, Jiang Y, Wei N, Li J, Zhang M, Chen L. Comparative pharmacokinetics of four bioactive components in normal and chronic heart failure rats after oral administration of Qiangxin Lishui Prescription by microdialysis combined with ultra-high-performance liquid chromatography. J Sep Sci 2023; 46:e2300518. [PMID: 37853838 DOI: 10.1002/jssc.202300518] [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: 07/21/2023] [Revised: 09/07/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023]
Abstract
Qiangxin Lishui Prescription (QLP) has been clinically applied for treating heart failure with remarkable curative effects. A multi-component pharmacokinetic research is very necessary for determining active substances in it. This study aims to profile the traits and differences in the pharmacokinetics of salvianolic acid B, astragaloside IV, calycosin-7-O-β-D-glucoside and kaempferol in QLP between normal and chronic heart failure (CHF) rats by microdialysis combined with ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Sensitive, selective, and online microdialysis combined with the UHPLC-MS/MS method was successfully established and applied to study the pharmacokinetics of QLP. The pathological condition of CHF could lead to the enhancement of systematic exposure and reduction of the metabolic rate of four bioactive components for better bioavailability and therapeutic efficacy. The pharmacokinetic results will provide data support for the clinical application of QLP.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Yong Jiang
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Nina Wei
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Jindong Li
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Mei Zhang
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Linwei Chen
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
8
|
Zhang L, Yang C, Zhao Y, Yang Z, Meng X, Yan D. Comparative pharmacokinetic analysis of six major bioactive constituents using UPLC-MS/MS in samples isolated from normal and diabetic nephropathy rats after oral administration of Gushen Jiedu capsule. J Pharm Biomed Anal 2023; 235:115638. [PMID: 37633162 DOI: 10.1016/j.jpba.2023.115638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/07/2023] [Accepted: 08/07/2023] [Indexed: 08/28/2023]
Abstract
Berberine, palmatine, physcion, rhein, calycosin-7-O-glucoside, and ferulic acid are six major active consituents that are present in Gushen Jiedu capsule (GSJD) extracts. The aim of this study was to determine the pharmacokinetics of the six active consituents in vivo by a rapid, sensitive, and precise UPLC-MS/MS method, which were compared between normal and diabetic nephropathy (DN) rats. Good separation of the target analytes and internal standards (ketoprofen and puerarin) was obtained on a Waters BEH C18 UPLC column with a mobile phase of 0.1 % formic acid acetonitrile-0.1 % formic acid water. All the calibration curves showed good linearity with a regression coefficient (r2) of ≥ 0.9908. The lower limits of quantification (LLOQ) for berberine, palmatine, physcion, rhein, calycosin-7-O-glucoside, and ferulic acid were 20, 2.5, 20, 20, 2.5, and 2.5 ng/mL, respectively. The relative standard deviations (RSDs) of intra-day and inter-day precision were all within 12.66 %, and the relative errors of intra-day and inter-day accuracy ranged from - 15.00 to 14.93 %. Good extraction recovery and matrix effects were obtained. The stability study confirmed the stability of the six analytes (RSD < 15 %). Finally, the data showed that the pharmacokinetic parameters (especially CLz/F, AUC and Tmax) of the six target analytes in DN rats were significantly different from those in normal rats. PK studies under pathological conditions could provide new thoughts to elucidate the underlying mechanism of GSJD and promote the clinical development of GSJD to treat DN.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| | - Chunjing Yang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| | - Yidan Zhao
- Department of Pharmacy, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Zhirui Yang
- Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China; Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xintong Meng
- Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Wang X, Liu H, Shu L, Yao Y, Xu Y, Wei J, Li Y. Rapid identification of chemical constituents in Hugan tablets by ultra-performance liquid chromatography-quadrupole-exactive orbitrap mass spectrometry. J Sep Sci 2023; 46:e2300302. [PMID: 37568249 DOI: 10.1002/jssc.202300302] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023]
Abstract
Hugan tablet is a Chinese medicine preparation. It is composed of Bupleuri Radix, Artemisiae Scopariae Herba, Isatidis Radix, Schisandrae Chinensis Fructus, Suis Fellis Pulvis, and Vigna radiata L. It has the effects of dispersing stagnated liver qi, strengthening the spleen and eliminating food to be used for the treatment of chronic hepatitis and early cirrhosis. However, the chemical composition of Hugan tablet is complex and not fully understood, which hampers the research in pharmacology. In this study, a reliable method for the rapid analysis and identification of the chemical components in Hugan tablet by their characteristic fragments and neutral losses using ultra-performance liquid chromatography-quadrupole-exactive orbitrap mass spectrometry was developed. A total of 144 chemical components were tentatively identified, including 57 organic acids, 19 flavonoids, 23 alkaloids, 18 lignans, 7 saponins, and 20 others. These components may be the active ingredients of Hugan tablet. The established method can systematically and rapidly analyze the chemical components in Hugan tablet, which provides a basis for the pharmacodynamic substance study and is meaningful for the quality control of Hugan tablet.
Collapse
Affiliation(s)
- Xiaowen Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Huiru Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Lexin Shu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yaqi Yao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yanyan Xu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Jinxia Wei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| |
Collapse
|
10
|
Chen Y, Wang Y, Li Z, Jing J, Jiang D, Yuan X, Li F. Exploration of the Mechanism of Shengxian Decoction Against Chronic Obstructive Pulmonary Disease Based on Network Pharmacology and Experimental Verification. Assay Drug Dev Technol 2023; 21:258-272. [PMID: 37682969 DOI: 10.1089/adt.2023.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023] Open
Abstract
Shengxian decoction (SXT) is clinically used in chronic obstructive pulmonary disease (COPD) treatment. This study aimed to explore the mechanism and target genes of SXT acting on COPD. Differentially expressed genes (DEGs) between COPD and controls were identified and then performed enrichment analysis. The effective active compounds and corresponding target genes were obtained from the traditional Chinese medicine systems pharmacology database. We also compiled COPD related genes from the GeneCards database. Through the protein-protein interaction (PPI) network and least absolute shrinkage and selection operator (LASSO) regression was performed to identify key genes. Molecular docking was used for docking of key genes and compounds. The expression of key genes was detected by quantitative real-time PCR in COPD patients and bronchial epithelial cells stimulated with cigarette stroke extract (CSE). We identified 1,458 intersected DEGs from GSE47460 and GSE57148 datasets. Compared with intersected DEGs, we obtained 33 SXT target COPD-related genes. PI3K-Akt signaling pathway, MAPK signaling pathway, and focal adhesion were enriched by these 33 genes, as well as intersected DEGs. According to LASSO regression, there were 12 genes considered as signature genes. Then we constructed active compounds and corresponding six target genes. Finally, HIF1A and IL1B were selected as key genes by combining PPI network. HIF1A and IL1B were all upregulated expression in COPD and CSE stimulated cells and recovered in SXT treated CSE stimulated cells. This study provides a scientific basis for the identification of active compounds and target genes of SXT in the treatment of COPD.
Collapse
Affiliation(s)
- Yifei Chen
- Basic Teaching and Research Office of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Shuimogou, Urumqi, China
| | - Yiming Wang
- Department of Acupuncture, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
| | - Zheng Li
- Department of Respiration, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
- Department of Respiration, National Clinical Research Base of Traditional Chinese Medicine in Xinjiang, Shaybagh, Urumqi, China
| | - Jing Jing
- Department of Respiration, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
- Department of Respiration, National Clinical Research Base of Traditional Chinese Medicine in Xinjiang, Shaybagh, Urumqi, China
| | - De Jiang
- Basic Teaching and Research Office of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Shuimogou, Urumqi, China
| | - Xiaoxia Yuan
- Basic Teaching and Research Office of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Shuimogou, Urumqi, China
| | - Fengsen Li
- Department of Respiration, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
| |
Collapse
|
11
|
Yang ZQ, Han YY, Gao F, Tian JY, Bai R, Guo QH, Liu XC. Shengxian decoction protects against chronic heart failure in a rat model via energy regulation mechanisms. BMC Complement Med Ther 2023; 23:200. [PMID: 37330478 PMCID: PMC10276516 DOI: 10.1186/s12906-023-04035-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/10/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Chronic heart failure (CHF) is actually a disease caused by an imbalanced energy metabolism between myocardial energy demand and supply, ultimately resulting in abnormal myocardial cell structure and function. Energy metabolism imbalance plays an important role in the pathological process of chronic heart failure (CHF). Improving myocardial energy metabolism is a new strategy for the treatment of CHF. Shengxian decoction (SXT), a well-known traditional Chinese medicine (TCM) formula, has good therapeutic effects on the cardiovascular system. However, the effects of SXT on the energy metabolism of CHF is unclear. In this study, we probed the regulating effects of SXT on energy metabolism in CHF rats using various research methods. METHODS High-performance liquid chromatography (HPLC) analysis was used to perform quality control of SXT preparations. Then, SD rats were randomly assigned into 6 groups: sham, model, positive control (trimetazidine) and high-, middle-, and low-dose SXT groups. Specific reagent kits were used to detect the expression levels of ALT and AST in rats' serum. Echocardiography was used to evaluate cardiac function. H&E, Masson and TUNEL staining were performed to examine myocardial structure and myocardial apoptosis. Colorimetry was used to determine myocardial ATP levels in experimental rats. Transmission electron microscopy was used to observe the ultrastructure of myocardial mitochondria. ELISA was used to estimate CK, cTnI, and NT-proBNP levels, and LA、FFA、MDA、SOD levels. Finally, Western blotting was used to examine the protein expression of CPT-1, GLUT4, AMPK, p-AMPK, PGC-1α, NRF1, mtTFA and ATP5D in the myocardium. RESULTS HPLC showed that our SXT preparation method was feasible. The results of ALT and AST tests indicate that SXT has no side effect on the liver function of rats. Treatment with SXT improved cardiac function and ventricular remodelling and inhibited cardiomyocyte apoptosis and oxidative stress levels induced by CHF. Moreover, CHF caused decrease ATP synthesis, which was accompanied by a reduction in ATP 5D protein levels, damage to mitochondrial structure, abnormal glucose and lipid metabolism, and changes in the expression of PGC-1α related signal pathway proteins, all of which were significantly alleviated by treatment with SXT. CONCLUSION SXT reverses CHF-induced cardiac dysfunction and maintains the integrity of myocardial structure by regulating energy metabolism. The beneficial effect of SXT on energy metabolism may be related to regulating the expression of the PGC-1α signalling pathway.
Collapse
Affiliation(s)
- Ze-Qi Yang
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| | - Yang-Yang Han
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| | - Fan Gao
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| | - Jia-Ye Tian
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| | - Ran Bai
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| | - Qiu-Hong Guo
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| | - Xing-Chao Liu
- Hebei University of Chinese Medicine, Xinshi South Road No 326, Qiaoxi District, Shijiazhuang, 050091 Hebei China
| |
Collapse
|
12
|
He X, Zhong Z, Wang Q, Jia Z, Lu J, Chen J, Liu P. Pharmacokinetics and tissue distribution of bleomycin-induced idiopathic pulmonary fibrosis rats treated with cryptotanshinone. Front Pharmacol 2023; 14:1127219. [PMID: 36969870 PMCID: PMC10034131 DOI: 10.3389/fphar.2023.1127219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Introduction: Cryptotanshinone(CTS), a compound derived from the root of Salvia miltiorrhiza, has been linked to various of diseases, particularly pulmonary fibrosis. In the current study, we investigated the benefit of CTS on Sprague-Dawley (SD) rats induced by bleomycin (BLM) and established high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) methods to compare pharmacokinetics and tissue distribution in subsequent normal and modulated SD rats.Methods: The therapeutic effect of CTS on BLM-induced SD rats was evaluated using histopathology, lung function and hydroxyproline content measurement, revealing that CTS significantly improved SD rats induced by BLM. Additionally, a simple, rapid, sensitive and specific HPLC-MS/MS method was developed to determine the pharmacokinetics of various components in rat plasma.Results: Pharmacokinetic studies indicated that CTS was slowly absorbed by oral administration and had low bioavailability and a slow clearance rate. The elimination of pulmonary fibrosis in 28-day rats was slowed down, and the area under the curve was increased compared to the control group. Long-term oral administration of CTS did not accumulate in vivo, but the clearance was slowed down, and the steady-state blood concentration was increased. The tissue distribution study revealed that CTS exposure in the lungs and liver.Discussion: The lung CTS exposure was significantly higher in the model group than in the control group, suggesting that the pathological changes of pulmonary fibrosis were conducive to the lung exposure of CTS and served as the target organ of CTS.
Collapse
Affiliation(s)
- Xiangjun He
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zhi Zhong
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Quan Wang
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zhenmao Jia
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Jing Lu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jing Lu, ; Jianwen Chen, ; Peiqing Liu,
| | - Jianwen Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jing Lu, ; Jianwen Chen, ; Peiqing Liu,
| | - Peiqing Liu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jing Lu, ; Jianwen Chen, ; Peiqing Liu,
| |
Collapse
|
13
|
Chen L, Wei N, Jiang Y, Yuan C, Xu L, Li J, Kong M, Chen Y, Wang Q. Comparative pharmacokinetics of seven bioactive components after oral administration of crude and processed Qixue Shuangbu Prescription in chronic heart failure rats by microdialysis combined with UPLC-MS/MS. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:116035. [PMID: 36513265 DOI: 10.1016/j.jep.2022.116035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qixue Shuangbu Prescription (QSP) is a classical traditional Chinese medicine prescription, which has widely used for the treatment of chronic heart failure (CHF). Preliminary clinical studies have shown that the efficacy of processed QSP (P-QSP) in treating CHF is greater than crude QSP (C-QSP). However, the pharmacokinetic characteristics of its major bioactive components under pathological conditions are unclear. AIM OF STUDY This study aims to compare pharmacokinetics of seven bioactive components after oral administration of C-QSP and P-QSP in CHF model rats. MATERIALS AND METHODS Ginsenoside Rb1, ginsenoside Re, ginsenoside Rg1, ferulic acid, astragaloside IV, calycosin-7-O-β-D-glucoside, and paeoniflorin in QSP were used as the target components. CHF model in rats was induced by the intraperitoneal injection of doxorubicin. A microdialysis combined with UPLC-MS/MS method was first established to compare the pharmacokinetics of seven major bioactive components in CHF model rats after oral administration of C-QSP and P-QSP. RESULTS This method was successfully applied to the pharmacokinetic investigation of seven major components of C-QSP and P-QSP following oral administration in CHF model rats. Compared with the C-QSP group, the Cmax, AUC0-t and AUC0-∞ of ginsenoside Rb1, ginsenoside Re, ginsenoside Rg1, ferulic acid, astragaloside IV and paeoniflorin significantly increased (P < 0.05) in the P-QSP group, which suggested that the absorptivity and bioavailability were increased. Lower T1/2, MRT0-t of ginsenoside Rb1, gerulic acid and higher T1/2, MRT0-t of ginsenoside Rb1, astragaloside IV, paeoniflorin in the P-QSP group, which indicated that eliminated more quickly or slowly, respectively. CONCLUSIONS The pharmacokinetic parameters of bioactive components were significantly changed for better bioavailability and absorption, longer lasting time elimination, which were beneficial for enhancing therapeutic efficacy in the P-QSP group. This study will provide a new perspective to explain the pharmacokinetic-pharmacodynamic correlation of P-QSP on the treatment of CHF.
Collapse
Affiliation(s)
- Linwei Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Cardiology, Taizhou Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Nina Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yong Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Chengye Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Cardiology, Taizhou Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Luwei Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Cardiology, Taizhou Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Jindong Li
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China.
| | - Min Kong
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China.
| | - Yan Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Cardiology, Taizhou Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Qin Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Cardiology, Taizhou Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| |
Collapse
|
14
|
Li J, Wang M, Yao L, Lu B, Gui M, Zhou X, Fu D. Yixin Granules Reduce Myocardial Inflammation and Fibrosis in Rats with Heart Failure by Inhibiting the Expression of ADAMTS8. Int Heart J 2023; 64:741-749. [PMID: 37518355 DOI: 10.1536/ihj.22-715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Yixin granules are medications modified from a Chinese prescription (Sheng Xian Tang) that has been used to alleviate shortness of breath. ADAM metallopeptidase with thrombospondin type 1 motif 8 (ADAMTS8) is upregulated in the myocardium of patients with dilated cardiomyopathy. Its high expression is associated with tumor necrosis factor (TNF) -α and myocardial fibrosis. This study aimed to explore the effect of Yixin granules on heart failure (HF) in rats and whether this effect is correlated with ADAMTS8 to provide new ideas for the treatment of HF.HF rat models were established by ligation of the left anterior descending coronary artery. Model rats were injected with adeno-associated virus vectors for the overexpression of ADAMTS8 and/or treated with Yixin granules for 4 weeks. Hematoxylin-eosin and Masson staining were used to detect myocardial injury and fibrosis, respectively. Reverse transcription polymerase chain reaction, western blotting, and/or enzyme-linked immunosorbent assay were used to detect the expression of ADAMTS8, TNF-α, interleukin (IL) -1β, IL-6, collagen I, collagen III, and α-smooth muscle actin in myocardium. The myocardial infarction area of rats was measured using 2,3,5-triphenyltetrazolium chloride staining.ADAMTS8 was upregulated in the myocardium of HF rats. Yixin granule treatment improved left ventricular contractility and reduced ADAMTS8 expression, myocardial injury, inflammation, and fibrosis in HF rats. ADAMTS8 overexpression aggravated myocardial injury, inflammation, and fibrosis. Moreover, ADAMTS8 overexpression counteracted the cardioprotective effects of Yixin granules.Yixin granules may reduce myocardial inflammation and fibrosis in HF rats by inhibiting the expression of ADAMTS8.
Collapse
Affiliation(s)
- Jianhua Li
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| | - Mingzhu Wang
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| | - Lei Yao
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| | - Bo Lu
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| | - Mingtai Gui
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| | - Xunjie Zhou
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| | - Deyu Fu
- Department of Heart Disease, Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai University of Traditional Chinese Medicine
| |
Collapse
|
15
|
Advances in the Bioactivities of Phytochemical Saponins in the Prevention and Treatment of Atherosclerosis. Nutrients 2022; 14:nu14234998. [PMID: 36501028 PMCID: PMC9735883 DOI: 10.3390/nu14234998] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by hardening and narrowing of arteries. AS leads to a number of arteriosclerotic vascular diseases including cardiovascular diseases, cerebrovascular disease and peripheral artery disease, which pose a big threat to human health. Phytochemicals are a variety of intermediate or terminal low molecular weight secondary metabolites produced during plant energy metabolism. Phytochemicals from plant foods (vegetables, fruits, whole grains) and traditional herb plants have been shown to exhibit multiple bioactivities which are beneficial for prevention and treatment against AS. Many types of phytochemicals including polyphenols, saponins, carotenoids, terpenoids, organic sulfur compounds, phytoestrogens, phytic acids and plant sterols have already been identified, among which saponins are a family of glycosidic compounds consisting of a hydrophobic aglycone (sapogenin) linked to hydrophilic sugar moieties. In recent years, studies have shown that saponins exhibit a number of biological activities such as anti-inflammation, anti-oxidation, cholesterol-lowering, immunomodulation, anti-platelet aggregation, etc., which are helpful in the prevention and treatment of AS. This review aims to summarize the recent advances in the anti-atherosclerotic bioactivities of saponins such as ginsenoside, soyasaponin, astra-galoside, glycyrrhizin, gypenoside, dioscin, saikosaponin, etc.
Collapse
|
16
|
Lin P, Wang Q, Chen J, Zhao H, Huang H, Xiao Q, Qin Z, Chen J, Yao X, Yao Z. Kinetic features of Gualou-Xiebai-Banxia decoction, a classical traditional Chinese medicine formula, in rat plasma and intestine content based on its metabolic profile. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
17
|
Ivanova GT. Effect of Doxorubicin on the Reactivity of Rat Mesenteric Arteries. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Wang X, Zhang J, Luo L, Song X, Wang P, Liu D. Comparative pharmacokinetics of 24 major bioactive components in normal and ARDS rats after oral administration of Xuanfei Baidu granules. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115472. [PMID: 35718055 DOI: 10.1016/j.jep.2022.115472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xuanfei Baidu prescription, consisting of 13 Chinese medicines, was formulated by academicians Boli Zhang and Professor Qingquan Liu based on their experience in first-line clinical treatment of COVID-19. Xuanfei Baidu granules (XFBD granules) are a proprietary Chinese medicine preparation developed based on Xuanfei Baidu prescription. It is recommended for the treatment of patients with the common wet toxin and lung stagnation syndrome of COVID-19. However, the pharmacokinetic characteristics of its major bioactive components in rats under different physiological and pathological conditions are unclear. MATERIALS AND METHODS A rapid and sensitive analytical method, ultra-performance liquid chromatography coupled with mass spectrometry (UPLC-MS/MS), was developed and applied to 24 major bioactive components in normal and ARDS rats after oral administration of XFBD granules. We studied the metabolic process of XFBD granules in vivo to compare the differences in pharmacokinetic parameters between normal and model metabolic processes. RESULTS This method was successfully applied to the pharmacokinetic investigation of 24 major components of XFBD granules following oral administration in normal and ARDS rats. Eight components, including ephedrine and amygdalin, were more highly absorbed and had shorter Tmax values than the model group; the absorption of six components, such as rhein, decreased in ARDS rats, and there was no significant difference in the absorption of ten components, such as verbenalin and naringin, between the normal and ARDS rats. The results showed that the peak times of other analytes were very short, and 80% of these target constituents were eliminated in both normal and ARDS rats within 6 h except for liquiritigenin and 18β-glycyrrhetinic acid. CONCLUSIONS In this study, a rapid and sensitive UPLC-MS/MS analytical method was developed and applied to 24 major bioactive components in normal and ARDS rats after the oral administration of XFBD granules. This will serve to form the basis for further studies on the pharmacokinetic-pharmacodynamic correlation of XFBD granules.
Collapse
Affiliation(s)
- Xinrui Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin, China
| | - Jingze Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin, China
| | - Lifei Luo
- Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin, China
| | - Xinbo Song
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ping Wang
- Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin, China
| | - Dailin Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin, China.
| |
Collapse
|
19
|
Plasma Pharmacokinetics and Tissue Distribution of Doxorubicin in Rats following Treatment with Astragali Radix. Pharmaceuticals (Basel) 2022; 15:ph15091104. [PMID: 36145325 PMCID: PMC9505068 DOI: 10.3390/ph15091104] [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: 08/16/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Doxorubicin (DOX) is an essential component in chemotherapy, and Astragali Radix (AR) is a widely used tonic herbal medicine. The combination of DOX and AR offers widespread, well-documented advantages in treating cancer, e.g., reducing the risk of adverse effects. This study mainly aims to uncover the impact of AR on DOX disposition in vivo. Rats received a single intravenous dose of 5 mg/kg DOX following a single-dose co-treatment or multiple-dose pre-treatment of AR (10 g/kg × 1 or × 10). The concentrations of DOX in rat plasma and six tissues, including heart, liver, lung, kidney, spleen, and skeletal muscle, were determined by a fully validated LC-MS/MS method. A network-based approach was further employed to quantify the relationships between enzymes that metabolize and transport DOX and the targets of nine representative AR components in the human protein−protein interactome. We found that short-term (≤10 d) AR administration was ineffective in changing the plasma pharmacokinetics of DOX in terms of the area under the concentration−time curve (AUC, 1303.35 ± 271.74 μg/L*h versus 1208.74 ± 145.35 μg/L*h, p > 0.46), peak concentrations (Cmax, 1351.21 ± 364.86 μg/L versus 1411.01 ± 368.38 μg/L, p > 0.78), and half-life (t1/2, 31.79 ± 5.12 h versus 32.05 ± 6.95 h, p > 0.94), etc. Compared to the isotype control group, DOX concentrations in six tissues slightly decreased under AR pre-administration but only showed statistical significance (p < 0.05) in the liver. Using network analysis, we showed that five of the nine representative AR components were not localized to the vicinity of the DOX disposition-associated module. These findings suggest that AR may mitigate DOX-induced toxicity by affecting drug targets rather than drug disposition.
Collapse
|
20
|
Liu X, Chen Q, Ji X, Yu W, Wang T, Han J, Li S, Liu J, Zeng F, Zhao Y, Zhang Y, Luo Q, Wang S, Wang F. Astragaloside IV promotes pharmacological effect of Descurainia sophia seeds on isoproterenol-induced cardiomyopathy in rats by synergistically modulating the myosin motor. Front Pharmacol 2022; 13:939483. [PMID: 36034815 PMCID: PMC9403516 DOI: 10.3389/fphar.2022.939483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022] Open
Abstract
Descurainia sophia seeds (DS), Astragalus mongholicus (AM), and their formulas are widely used to treat heart failure caused by various cardiac diseases in traditional Chinese medicine practice. However, the molecular mechanism of action of DS and AM has not been completely understood. Herein, we first used mass spectrometry coupled to UPLC to characterize the chemical components of DS and AM decoctions, then applied MS-based quantitative proteomic analysis to profile protein expression in the heart of rats with isoproterenol-induced cardiomyopathy (ISO-iCM) before and after treated with DS alone or combined with AM, astragaloside IV (AS4), calycosin-7-glucoside (C7G), and Astragalus polysaccharides (APS) from AM. We demonstrated for the first time that DS decoction alone could reverse the most of differentially expressed proteins in the heart of the rats with ISO-iCM, including the commonly recognized biomarkers natriuretic peptides (NPPA) of cardiomyopathy and sarcomeric myosin light chain 4 (MYL4), relieving ISO-iCM in rats, but AM did not pronouncedly improve the pharmacological efficiency of DS. Significantly, we revealed that AS4 remarkably promoted the pharmacological potency of DS by complementarily reversing myosin motor MYH6/7, and further downregulating NPPA and MYL4. In contrast, APS reduced the efficiency of DS due to upregulating NPPA and MYL4. These findings not only provide novel insights to better understanding in the combination principle of traditional Chinese medicine but also highlight the power of mass spectrometric proteomics strategy combined with conventional pathological approaches for the traditional medicine research.
Collapse
|
21
|
Fu C, Zhang K, Wang M, Qiu F. Casticin and chrysosplenol D from Artemisia annua L. induce apoptosis by inhibiting topoisomerase IIα in human non-small-cell lung cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154095. [PMID: 35398735 DOI: 10.1016/j.phymed.2022.154095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 03/12/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Artemisia annua L. (A. annua) and its active components exhibit antitumour effects in many cancer cells. However, the biological processes and mechanisms involved are not well understood, especially for the treatment of non-small-cell lung cancer (NSCLC). PURPOSE This study aimed to comprehensively explore the biological processes of A. annua and its active components in NSCLC cells and to identify the mechanism by which these compounds induce apoptosis. STUDY DESIGNS/METHODS Cell viability and flow cytometry assays were used to evaluate the cytotoxicity of A. annua active components casticin (CAS) and chrysosplenol D (CHD) in A. annua in NSCLC cells. After treatment with CAS and CHD, A549 cells were subjected to RNA sequencing (RNA-seq) analysis, differentially expressed genes (DEGs) were screened and subjected to functional enrichment analysis (KEGG and GO analysis) as well as protein interaction network analysis. The key targets associated with apoptosis induction in A549 cells were screened by Cytoscape, and the screened DEGs were validated by qRT-PCR. Immunoblotting, immunofluorescence, and molecular docking assays were used to determine whether CAS and/or CHD could induce apoptosis in NSCLC cells by inducing DNA damage through down-regulation of topoisomerase IIα (topo IIα) expression. The same experiments were verified again in the H1299 lung cancer cell line. RESULTS CAS and CHD inhibited NSCLC cells proliferation in a time- and dose-dependent manner, and significantly induced apoptosis. A total of 115 co-upregulated DEGs and 277 co-downregulated DEGs were identified in A549 cells following treatment with CAS and CHD. Comprehensive and systematic data about biological processes and mechanisms were obtained. DNA damage pathways and topo IIα targets were screened to study the apoptosis effects of CAS and CHD on NSCLC cells. CAS and CHD may be able to induce DNA damage by binding to topo IIα-DNA and reducing topo IIα activity. CONCLUSION This study suggested that CAS and CHD may reduce topo IIα activity by binding to topo IIα-DNA, affecting the replication of DNA, triggering DNA damage, and inducing apoptosis. It described a novel mechanism associated with topo IIα inhibition to reveal a novel role for CAS and CHD in A. annua as potential anticancer agents and/or adjuvants in NSCLC cells.
Collapse
Affiliation(s)
- Chunqing Fu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Keyu Zhang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Manyuan Wang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Feng Qiu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
22
|
Li K, You F, Zhang Q, Yuan R, Yuan Q, Fu X, Ren Y, Wang Q, Li X, Zhang Z, Shichiri M, Yu Y. Chemical and Biological Evidence of the Efficacy of Shengxian Decoction for Treating Human Lung Adenocarcinoma. Front Oncol 2022; 12:849579. [PMID: 35372052 PMCID: PMC8975620 DOI: 10.3389/fonc.2022.849579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/24/2022] [Indexed: 11/23/2022] Open
Abstract
Shengxian Decoction (SXT) is a traditional Chinese medicine prescription comprising several anti-cancer medicinal herbs. However, the anti-cancer effect of SXT has rarely been reported. Herein, we explored the therapeutic potential of SXT for the treatment of lung adenocarcinoma (LUAD). High-performance liquid chromatography analysis of crude SXT extract revealed the abundance of mangiferin, an established anti-cancer compound. The serum pharmacological evaluation revealed that serum SXT suppressed A549 lung cancer cell proliferation in vitro. The tumor-inhibitory activity of SXT was confirmed in vivo via tumor formation assays in nude mice. We applied biochemical, histopathological and imaging approaches to investigate the cellular targets of SXT. The results indicated that the treatment with SXT induced tumor necrosis, and downregulated hypoxia-inducible factor 1 alpha in the serum. In vivo biosafety assessment of SXT revealed low levels of toxicity in mouse models. Our study provides the first scientific evidence that SXT effectively represses cancer cell growth and, thus, may serve as a safe anti-cancer agent for LUAD treatment.
Collapse
Affiliation(s)
- Kejuan Li
- College of Life Science, Sichuan Normal University, Chengdu, China
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Fengming You
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin Zhang
- College of Life Science, Sichuan Normal University, Chengdu, China
| | - Ruijiao Yuan
- College of Life Science, Sichuan Normal University, Chengdu, China
| | - Qianghua Yuan
- Oncology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Fu
- Oncology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yifeng Ren
- Oncology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Wang
- Oncology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Li
- Oncology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenya Zhang
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Mototada Shichiri
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Japan
- DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), AIST, Tsukuba, Japan
| | - Yue Yu
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Japan
- DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), AIST, Tsukuba, Japan
- *Correspondence: Yue Yu,
| |
Collapse
|