1
|
Gao X, Gao J. Investigation of the efficacy and pharmacological mechanism of Danhong injections for treating chronic obstructive pulmonary disease: A PRISMA-compliant meta-analysis and network pharmacology analysis. Medicine (Baltimore) 2023; 102:e32846. [PMID: 36749263 PMCID: PMC9901954 DOI: 10.1097/md.0000000000032846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Accumulating evidence supported the clinical efficacy of Danhong injection (DHI) on chronic obstructive pulmonary disease (COPD). It is urgent to summarize the effects of DHI on various outcomes in COPD patients and to elucidate the molecular mechanisms of DHI in treating COPD. METHODS Eligible studies were retrieved from 6 databases including China national knowledge infrastructure, Wangfang, VIP, web of science, PubMed, and Embase. The heterogeneity across studies was tested using the I2 statistic and the quality of studies was assessed. The pooled evaluation of outcomes was calculated using a fix- or random-effect model according to the heterogeneity. The underlying mechanism of DHI in treating COPD was analyzed using network pharmacology. RESULTS A total of 34 eligible studies with a general medium quality were included in the meta-analysis. The pooled data showed that DHI intervention significantly increased clinical efficacy as compared to routine treatment. Meanwhile, our data also revealed that the addition of DHI markedly improved hemorheological indicators, lung function index, arterial blood gas index, and as well as blood coagulation functions. However, the current meta-analysis lacked sufficient data to support the significant effect of DHI on prothrombin time and activated partial thromboplastin time. Network pharmacology found 59 candidate targets of DHI in treating COPD, and enrichment analysis found these targets were associated with lymphocyte proliferation and activation, glucocorticoid receptor signaling, TREM1 signaling, IL-12 signaling and production in macrophages, and aryl hydrocarbon receptor signaling. Multiple core targets including AKT1, TNF, and IL1B, etc. Were identified and might play an important role in the action of DHI against COPD. CONCLUSION Taken together, this study suggested that DHI could ameliorate hemorheological indicators, lung function, arterial blood gas, and as well as coagulation functions of COPD patients and elucidate the underlying mechanism of DHI against COPD.
Collapse
Affiliation(s)
- Xiaoyu Gao
- Department of Pharmacy, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Jinsong Gao
- Intensive Care Unit, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
- * Correspondence: Jinsong Gao, Intensive Care Unit, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine Xiaoshan Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310016, China (e-mail: )
| |
Collapse
|
2
|
Profound Effect of Pulmonary Surfactant on the Treatment of Preterm Infants with Respiratory Distress Syndrome. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:4166994. [PMID: 36262981 PMCID: PMC9550487 DOI: 10.1155/2022/4166994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Accepted: 09/07/2022] [Indexed: 01/26/2023]
Abstract
Inherited diseases caused by dysfunction of pulmonary surfactant metabolism or surfactant dysfunction have recently been considered the underlying causes of neonatal and pediatric respiratory diseases. Respiratory distress syndrome in premature infants is a common respiratory disease in pediatrics. It is caused by underdeveloped lungs in infants and a lack of active substances on the surface of the alveoli, which leads to insufficiency of lung function, which can lead to difficulty breathing, increased heart rate, facial bruising, and more. Neonatal Respiratory Distress Syndrome is a very dangerous disease with a high mortality rate and a great threat to children's lives and health. Therefore, enough attention and treatment should be caused in clinical practice. Natural pulmonary surfactant (PS) has achieved positive effects in the treatment of neonatal respiratory distress syndrome (RDS), reducing neonatal mortality, the application of mechanical ventilation, and the occurrence of late complications. To further explore the role of pulmonary surfactants in the treatment of neonatal respiratory distress syndrome, to analyze the best time to use PS to prevent RDS, this paper has selected premature infants with RDS received by the neonatal department of a hospital in a province from March 2019 to October 2020 to compare the efficacy of pulmonary surfactant (PS) in preterm infants with respiratory distress syndrome (RDS). The experiment has found that the average mechanical ventilation time (5.1 d) and oxygen therapy time (7.3 d) in the early group are shorter than the average mechanical ventilation time (6.4 d) and oxygen therapy time (10.6 d) in the late group. It has been demonstrated that early administration of pulmonary surfactant (PS) therapy is of great help in improving respiratory distress syndrome in premature infants.
Collapse
|
3
|
Abou Baker DH. Can natural products modulate cytokine storm in SARS-CoV2 patients? BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2022; 35:e00749. [PMID: 35702395 PMCID: PMC9181898 DOI: 10.1016/j.btre.2022.e00749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/24/2022] [Accepted: 06/08/2022] [Indexed: 01/08/2023]
Abstract
Currently, the number of cases and deaths of SARS-CoV2, especially among the chronic disease groups, due to aggressive SARS-CoV2 infection is increasing day by day. Various infections, particularly viral ones, cause a cytokine storm resulting in shortness of breath, bleeding, hypotension, and ultimately multi-organ failure due to over-expression of certain cytokines and necrosis factors. The most prominent clinical feature of SARS-CoV2 is the presence of elevated proinflammatory cytokines in the serum of patients with SARS-CoV2. Severe cases exhibit higher levels of cytokines, leading to a "cytokine storm" that further increases disease severity and causes acute respiratory distress syndrome, multiple organ failure, and death. Therefore, targeted cytokine production could be a potential therapeutic option for patients severely infected with SARS-CoV2. Given the current scenario, great scientific progress has been made in understanding the disease and its forms of treatment. Because of natural ingredients properties, they have the potential to be used as potential agents with the ability to modulate immune responses. Moreover, they can be used safely because they have no toxic effects, are biodegradable and biocompatible. However, these natural substances can continue to be used in the development of new therapies and vaccines. Finally, the aim and approach of this review article is to highlight current research on the possible use of natural products with promising potential as immune response activators. Moreover, consider the expected use of natural products when developing potential therapies and vaccines.
Collapse
Affiliation(s)
- Doha H. Abou Baker
- Medicinal and Aromatic Plants Department, National Research Centre, Pharmaceutical and Drug Industries Institute, Dokki, Giza, PO 12622, Egypt
| |
Collapse
|
4
|
Feng Z, Zhou P, Wu X, Zhang J, Zhang M. Hydroxysafflor yellow A protects against ulcerative colitis via suppressing TLR4/NF-κB signaling pathway. Chem Biol Drug Des 2022; 99:897-907. [PMID: 35319164 DOI: 10.1111/cbdd.14045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/24/2022] [Accepted: 03/20/2022] [Indexed: 11/27/2022]
Abstract
Hydroxysafflower yellow A (HSYA) protects against acute kidney injury through TLR4/NF-κB pathway. However, the effect and potential mechanism of HSYA in ulcerative colitis (UC) have been rarely reported, which is thus investigated in this research. An in vivo UC model was established by oral administration of 5% dextran sulfate sodium (DSS) in Sprague-Dawley rats. After HSYA treatment, the daily body weight and colon length of rats were measured. Then rat colon tissues, myeloperoxidase (MPO) activity, and the levels of inflammatory cytokines were examined by histopathological examination (HE) staining, immunohistochemistry, ultraviolet spectrophotometry, and enzyme-linked immune sorbent assay (ELISA) respectively. The activated TLR4/NF-κB pathway was detected by Western blot. RAW 264.7 cell viability was detected by MTT assay after lipopolysaccharide (LPS) treatment, and ELISA and Western blot were performed again to investigate the effects of HSYA on LPS-treated cells. DSS administration increased body weight and colon length of rats and induced colon tissue injury. DSS or LPS treatment up-regulated the levels of TNF-α, IL-1β, and IL-6 and activated TLR4/NF-κB pathway of colon tissues and cells, respectively. HSYA partially reversed the above effect of DSS and LPS treatment, and the effects of the drug were improved with the dosage. Taken together, HSYA alleviates UC by suppressing TLR4/NF-κB signaling pathway, which may provide a new insight for the treatment of UC.
Collapse
Affiliation(s)
- Zhibing Feng
- Department of Anorectal, Jiangxi Province Hospital of Integrated Chinese Western Medicine, Nanchang, China
| | - Ping Zhou
- Department of Anorectal, Jiangxi Province Hospital of Integrated Chinese Western Medicine, Nanchang, China
| | - Xiao Wu
- Department of Anorectal, Jiangxi Province Hospital of Integrated Chinese Western Medicine, Nanchang, China
| | - Junbiao Zhang
- Department of Anorectal, Jiangxi Province Hospital of Integrated Chinese Western Medicine, Nanchang, China
| | - Min Zhang
- Department of Anorectal, Jiangxi Province Hospital of Integrated Chinese Western Medicine, Nanchang, China
| |
Collapse
|
5
|
Hansur L, Louisa M, Wuyung PE, Fadilah F. Daphnoretin from Carthamus tinctorius as a Potential Inflammatory Inhibitor in COVID-19 by Binding to Toll-like Receptor-4: An in silico Molecular Docking Study. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: Cytokine storm in COVID-19 patients has contributed to many morbidities and mortalities in patients. Studies have found that toll-like receptors (TLRs) and some Fc receptors play essential roles in the hyperactivation of the immune system. Up to date, researchers are still in progress to discover effective and safe drugs to alleviate the hyperinflammatory state in COVID-19. The previous studies had shown that Carthamus tinctorius and its bioactive compounds might have anti-inflammatory activities in animal models.
AIM: We aimed to investigate the possible interactions of several flavonoids from C. tinctorius with several immune system components using a biocomputational approach.
METHODS: Molecular docking was done using the AutoDock program based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) COVID-19 pathway. The most suitable receptors found were studied to study the interactions with several flavonoids from C. tinctorius.
RESULTS: TLR4, TLR8, and FcγRIIa were found to bind with SARS CoV2 inflammatory pathway and further selected as macromolecules for potential interactions study with 22 flavonoids from C. tinctorius. Of the 22 flavonoids studied, daphnoretin showed the best binding affinity with TLR4 and Rutin was shown to attach best with FcγRIIa. Unlike its excellent binding to TLR4, daphnoretin showed weak binding to TLR8.
CONCLUSION: Daphnoretin showed an excellent affinity with TLR4 and might be a good candidate as an inhibitor in hyperinflammatory reactions in COVID-19 DTLR8.
Collapse
|
6
|
Pharmacological Actions, Molecular Mechanisms, Pharmacokinetic Progressions, and Clinical Applications of Hydroxysafflor Yellow A in Antidiabetic Research. J Immunol Res 2021; 2021:4560012. [PMID: 34938814 PMCID: PMC8687819 DOI: 10.1155/2021/4560012] [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: 07/29/2021] [Revised: 11/01/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022] Open
Abstract
Hydroxysafflor yellow A (HSYA), a nutraceutical compound derived from safflower (Carthamus tinctorius), has been shown as an effective therapeutic agent in cardiovascular diseases, cancer, and diabetes. Our previous study showed that the effect of HSYA on high-glucose-induced podocyte injury is related to its anti-inflammatory activities via macrophage polarization. Based on the information provided on PubMed, Scopus and Wanfang database, we currently aim to provide an updated overview of the role of HSYA in antidiabetic research from the following points: pharmacological actions, molecular mechanisms, pharmacokinetic progressions, and clinical applications. The pharmacokinetic research of HSYA has laid foundations for the clinical applications of HSYA injection in diabetic nephropathy, diabetic retinopathy, and diabetic neuropathy. The application of HSYA as an antidiabetic oral medicament has been investigated based on its recent oral delivery system research. In vivo and in vitro pharmacological research indicated that the antidiabetic activities of HSYA were based mainly on its antioxidant and anti-inflammatory mechanisms via JNK/c-jun pathway, NOX4 pathway, and macrophage differentiation. Further anti-inflammatory exploration related to NF-κB signaling, MAPK pathway, and PI3K/Akt/mTOR pathway might deserve attention in the future. The anti-inflammatory activities of HSYA related to diabetes and diabetic complications will be a highlight in our following research.
Collapse
|
7
|
Lu X, Ma W, Fan B, Li P, Gao J, Liu Q, Hu C, Li Y, Yao M, Ning H, Xing L. Integrating Network Pharmacology, Transcriptome and Artificial Intelligence for Investigating Into the Effect and Mechanism of Ning Fei Ping Xue Decoction Against the Acute Respiratory Distress Syndrome. Front Pharmacol 2021; 12:731377. [PMID: 34803679 PMCID: PMC8595141 DOI: 10.3389/fphar.2021.731377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/20/2021] [Indexed: 01/19/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a high-mortality disease and lacks effective pharmacotherapy. A traditional Chinese medicine (TCM) formula, Ning Fei Ping Xue (NFPX) decoction, was demonstrated to play a critical role in alleviating inflammatory responses of the lung. However, its therapeutic effectiveness in ARDS and active compounds, targets, and molecular mechanisms remain to be elucidated. The present study investigates the effects of NFPX decoction on ARDS mice induced by lipopolysaccharides (LPS). The results revealed that NFPX alleviated lung edema evaluated by lung ultrasound, decreased lung wet/Dry ratio, the total cell numbers of bronchoalveolar lavage fluid (BALF), and IL-1β, IL-6, and TNF-α levels in BALF and serum, and ameliorated lung pathology in a dose-dependent manner. Subsequently, UPLC-HRMS was performed to establish the compounds of NFPX. A total of 150 compounds in NFPX were characterized. Moreover, integrating network pharmacology approach and transcriptional profiling of lung tissues were performed to predict the underlying mechanism. 37 active components and 77 targets were screened out, and a herbs-compounds-targets network was constructed. Differentially expressed genes (DEGs) were identified from LPS-treated mice compared with LPS combined with NFPX mice. GO, KEGG, and artificial intelligence analysis indicated that NFPX might act on various drug targets. At last, potential targets, HRAS, SMAD4, and AMPK, were validated by qRT-PCR in ARDS murine model. In conclusion, we prove the efficacy of NFPX decoction in the treatment of ARDS. Furthermore, integrating network pharmacology, transcriptome, and artificial intelligence analysis contributes to illustrating the molecular mechanism of NFPX decoction on ARDS.
Collapse
Affiliation(s)
- Xiaoxiao Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wentao Ma
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baofeng Fan
- Air Force General Hospital PLA, Beijing, China
| | - Peng Li
- Department of Basic Sciences, Shanxi Agricultural University, Taigu, China
| | - Jing Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiuhong Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunling Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengying Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hanbing Ning
- Department of Digestive Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lihua Xing
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Amaral-Machado L, Oliveira WN, Rodrigues VM, Albuquerque NA, Alencar ÉN, Egito EST. Could natural products modulate early inflammatory responses, preventing acute respiratory distress syndrome in COVID-19-confirmed patients? Biomed Pharmacother 2021; 134:111143. [PMID: 33360048 PMCID: PMC7832252 DOI: 10.1016/j.biopha.2020.111143] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The ARDS (Acute Respiratory Distress Syndrome) is a severe respiratory syndrome that was recently associated as the main death cause in the COVID-19 pandemic outbreak. Hence, in order to prevent ARDS, the pulmonary function maintenance has been the target of several pharmacological approaches. However, there is a lack of reports regarding the use of effective pharmaceutical active natural products (PANPs) for early treatment and prevention of COVID-19-related ARDS. Therefore, the aim of this work was to conduct a systematic review regarding the PANPs that could be further studied as alternatives to prevent ARDS. Consequently, this work can pave the way to spread the use of PANPs on the prevention of ARDS in COVID-19-confirmed or -suspected patients. METHODS The search strategy included scientific studies published in English from 2015 to 2020 that promoted the elucidation of anti-inflammatory pathways targeting ARDS by in vitro and/or in vivo experiments using PANPs. Then, 74 studies regarding PANPs, able to maintain or improve the pulmonary function, were reported. CONCLUSIONS The PANPs may present different pulmonary anti-inflammatory pathways, wherein (i) reduction/attenuation of pro-inflammatory cytokines, (ii) increase of the anti-inflammatory mediators' levels, (iii) pulmonary edema inhibition and (iv) attenuation of lung injury were the most observed biological effects of such products in in vitro experiments or in clinical studies. Finally, this work highlighted the PANPs with promising potential to be used on respiratory syndromes, allowing their possible use as alternative treatment at the prevention of ARDS in COVID-19-infected or -suspected patients.
Collapse
Affiliation(s)
- Lucas Amaral-Machado
- Department of Pharmacy, Dispersed Systems Laboratory (LaSiD), Federal University of Rio Grande Do Norte (UFRN), 59012-570, Natal, RN, Brazil
| | | | | | | | - Éverton N Alencar
- Department of Pharmacy, Dispersed Systems Laboratory (LaSiD), Federal University of Rio Grande Do Norte (UFRN), 59012-570, Natal, RN, Brazil
| | - Eryvaldo S T Egito
- Department of Pharmacy, Dispersed Systems Laboratory (LaSiD), Federal University of Rio Grande Do Norte (UFRN), 59012-570, Natal, RN, Brazil; Graduate Program in Health Sciences, UFRN, 59012-570, Natal, RN, Brazil.
| |
Collapse
|
9
|
Zhao F, Wang P, Jiao Y, Zhang X, Chen D, Xu H. Hydroxysafflor Yellow A: A Systematical Review on Botanical Resources, Physicochemical Properties, Drug Delivery System, Pharmacokinetics, and Pharmacological Effects. Front Pharmacol 2020; 11:579332. [PMID: 33536906 PMCID: PMC7849182 DOI: 10.3389/fphar.2020.579332] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
Hydroxysafflower yellow A (HSYA), as a principal natural ingredient extracted from safflower (Carthamus tinctorius L.), has significant pharmacological activities, such as antioxidant, anti-inflammatory, anticoagulant, and anticancer effects. However, chemical instability and low bioavailability have been severely hampering the clinical applications of HSYA during the treatment of cardiovascular and cerebrovascular disease. Therefore, this present review systematically summarized the materials about HSYA, including acquisition methods, extraction and detection methods, pharmacokinetics, pharmacological effects and molecular mechanism, especially focus on the possible causes and resolutions about the chemical instability and low bioavailability of HSYA, in order to provide relatively comprehensive basic data for the related research of HSYA.
Collapse
Affiliation(s)
- Feng Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanyuan Jiao
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoxiao Zhang
- Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Postdoctoral Management Office, China Academy of Chinese Medical Sciences, Beijing, China
- China Association of Chinese Medicine, Beijing, China
| | - Daquan Chen
- School of Pharmacy, Yantai University, Yantai, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Shaanxi Institute of International Trade and Commerce, Xianyang, China
| |
Collapse
|
10
|
A Metabolic Perspective and Opportunities in Pharmacologically Important Safflower. Metabolites 2020; 10:metabo10060253. [PMID: 32560514 PMCID: PMC7344433 DOI: 10.3390/metabo10060253] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Safflower (Carthamus tinctorius L.) has long been grown as a crop due to its commercial utility as oil, animal feed, and pharmacologically significant secondary metabolites. The integration of omics approaches, including genomics, transcriptomics, metabolomics, and proteomics datasets, has provided more comprehensive knowledge of the chemical composition of crop plants for multiple applications. Knowledge of a metabolome of plant is crucial to optimize the evolution of crop traits, improve crop yields and quality, and ensure nutritional and health factors that provide the opportunity to produce functional food or feedstuffs. Safflower contains numerous chemical components that possess many pharmacological activities including central nervous, cardiac, vascular, anticoagulant, reproductive, gastrointestinal, antioxidant, hypolipidemic, and metabolic activities, providing many other human health benefits. In addition to classical metabolite studies, this review focuses on several metabolite-based working techniques and updates to provide a summary of the current medical applications of safflower.
Collapse
|
11
|
Three Ingredients of Safflower Alleviate Acute Lung Injury and Inhibit NET Release Induced by Lipopolysaccharide. Mediators Inflamm 2020; 2020:2720369. [PMID: 32189992 PMCID: PMC7066412 DOI: 10.1155/2020/2720369] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/13/2020] [Accepted: 02/05/2020] [Indexed: 01/28/2023] Open
Abstract
Xuebijing injection is a Chinese herb compound to treat sepsis in China, but it contains many different kinds of components, and each component may have different effects in treating sepsis. The present study was performed to investigate the effect of three ingredients of Xuebijing, safflor yellow A (SYA), hydroxysafflor yellow A (HSYA), and anhydrosafflor yellow B (AHSYB), in lipopolysaccharide- (LPS-) induced acute lung injury (ALI). LPS (10 mg/kg) was injected intratracheally to induce acute lung injury in mice, which were then treated with SYA, HSYA, and AHSYB. The blood, bronchoalveolar lavage fluid (BALF), and lung tissues were collected to detect degree of lung injury, level of inflammation, and neutrophil extracellular traps (NETs). In vitro experiments were performed using HL-60 cells stimulated with phorbol myristate acetate (PMA). Lung injury induced by LPS was alleviated by SYA, HSYA, and AHSYB as demonstrated by the histopathologic test. The three components inhibit LPS-induced elevation of the levels of inflammatory factors and wet-to-dry weight ratio as well as the amount of protein and cells in the BALF. They also induced a remarkably less overlay of myeloperoxidase (MPO) and histone in the immunofluorescence assay and reduced level of MPO-DNA complex in plasma. The in vitro assay showed a similar trend that the three components inhibited PMA-induced NET release in neutrophil-like HL-60 cells. Western blot demonstrated that phosphorylation of c-rapidly accelerated fibrosarcoma (c-Raf), mitogen-activated protein kinase ERK kinase (MEK), and extracellular signal-regulated kinase (ERK) in the lungs of LPS-challenged mice, and PMA-treated HL-60 cells were all significantly reduced by SYA, HSYA, and AHSYB. Therefore, our data demonstrated that three components of XBJ, including SYA, HSYA, and AHSYB, showed a protective effect against LPS-induced lung injury and NET release.
Collapse
|
12
|
Wang Y, Wang C. microRNA-211-3p has a Role in the Effects of Lipopolysaccharide on Endoplasmic Reticulum Stress in Cultured Human Skin Fibroblasts. Med Sci Monit Basic Res 2019; 25:164-168. [PMID: 31221950 PMCID: PMC6607940 DOI: 10.12659/msmbr.915379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Lipopolysaccharide (LPS) in bacterial infection of skin wounds delays wound healing. This study aimed to investigate the effects of LPS and endoplasmic reticulum stress in cultured skin fibroblasts and microRNA-211-3p (miR-211-3p) signaling. Material/Methods Human skin fibroblasts were cultured in increasing concentrations of LPS at 0 ng/ml, 5 ng/ml, 10 ng/ml, and 20 ng/ml for 0, 12 h, 24 h, 36 h, and 48 h. Cell proliferation was determined using the MTT assay. Protein expression levels of the transcription factors GRP78, CHOP, p-JNK, and the endoplasmic reticulum stress apoptosis proteins, caspase-12 and Bcl-2, were determined by Western blot. The expression of miR-211-3p in human skin fibroblasts was detected by quantitative polymerase chain reaction (qPCR). Results Cell proliferation of human skin fibroblasts decreased with increasing concentrations of LPS in a dose-dependent and time-dependent way. Protein levels of GRP78, CHOP, p-JNK, caspase-12, and Bcl-2 were increased 8 h and 12 h after LPS treatment compared with 0 h and 4 h after treatment. However, the expression of miR-211-3p was decreased in human skin fibroblasts after treatment with LPS. When miR-211-3p was overexpressed, the endoplasmic reticulum stress/CHOP related proteins, including GRP78, CHOP, p-JNK, caspase-12, and Bcl-2 were unchanged after the addition of LPS. Overexpression of miR-211-3p also reduced inhibitory effects of LPS on the growth of human skin fibroblasts. Conclusions This study showed that microRNA-211-3p had a role in the effects of LPS on endoplasmic reticulum stress and CHOP activation in cultured human skin fibroblasts.
Collapse
Affiliation(s)
- Yongxiang Wang
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Chunyan Wang
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| |
Collapse
|
13
|
Zheng M, Guo X, Pan R, Gao J, Zang B, Jin M. Hydroxysafflor Yellow A Alleviates Ovalbumin-Induced Asthma in a Guinea Pig Model by Attenuateing the Expression of Inflammatory Cytokines and Signal Transduction. Front Pharmacol 2019; 10:328. [PMID: 31024302 PMCID: PMC6459898 DOI: 10.3389/fphar.2019.00328] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 03/19/2019] [Indexed: 12/26/2022] Open
Abstract
Hydroxysafflor yellow A (HSYA) is an effective ingredient of the Chinese herb Carthamus tinctorius L. In this study, we aimed to evaluate the effects of HSYA on ovalbumin (OVA)-induced asthma in guinea pigs, and to elucidate the underlying mechanisms. We established a guinea pig asthma model by intraperitoneal injection and atomized administration OVA. Guinea pigs were injected intraperitoneally with HSYA (50, 75, 112.5 mg/kg) once daily from days 2 to 22 before OVA administration. We examined biomarkers including lung function, pulmonary histopathology, immunoglobulin E (IgE), Th1/Th2 relative inflammatory mediators, and related pathways. Pathological changes in lung tissues were detected by hematoxylin and eosin and periodic acid-Schiff staining. Phosphorylation levels of JNK mitogen-activated protein kinase (MAPK), p38 MAPK, ERK MAPK, and inhibitor of nuclear factor κBα (IκBα) were detected by western blot. plasma levels of total IgE, platelet-activating factor (PAF), and interleukin (IL)-3 were detected by enzyme-linked immunosorbent assay (ELISA). Expression levels of tumor necrosis factor (TNF)-α, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-13, and interferon (IFN)-γ were detected by ELISA and real-time quantitative polymerase chain reaction. HSYA significantly reduced airway resistance, improved dynamic lung compliance, and attenuated the pathologic changes. HSYA also inhibited the phosphorylation of JNK MAPK, p38 MAPK, ERK MAPK, and IκBα, and inhibited the OVA-induced elevations of IgE, PAF, IL-1β, IL-6, IL-4, IL-5, and IL-13 and the decreases in TNF-α, IFN-γ, IL-2, and IL-3. These findings suggest that HSYA has a protective effect on OVA-induced asthma through inhibiting the Th1/Th2 cell imbalance and inhibiting activation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Meng Zheng
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Xinjing Guo
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Ruiyan Pan
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Jianwei Gao
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Baoxia Zang
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Ming Jin
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
14
|
Hydroxysafflor Yellow A: A Promising Therapeutic Agent for a Broad Spectrum of Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8259280. [PMID: 30356354 PMCID: PMC6176289 DOI: 10.1155/2018/8259280] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/12/2018] [Indexed: 01/13/2023]
Abstract
Hydroxysafflor yellow A (HSYA) is one of the major bioactive and water-soluble compounds isolated from Carthami Flos, the flower of safflower (Carthamus tinctorius L.). As a natural pigment with favorable medical use, HSYA has gained extensive attention due to broad and effective pharmacological activities since first isolation in 1993. In clinic, the safflor yellow injection which mainly contains about 80% HSYA was approved by the China State Food and Drug Administration and used to treat cardiac diseases such as angina pectoris. In basic pharmacology, HSYA has been proved to exhibit a broad spectrum of biological effects that include, but not limited to, cardiovascular effect, neuroprotection, liver and lung protection, antitumor activity, metabolism regulation, and endothelium cell protection. Although a great number of studies have been carried out to prove the pharmacological effects and corresponding mechanisms of HYSA, a systemic review of HYSA has not yet been seen. Here, we provide a comprehensive summarization of the pharmacological effects of HYSA. Together with special attention to mechanisms of actions, this review can serve as the basis for further researches and developments of this medicinal compound.
Collapse
|
15
|
Zou J, Wang N, Liu M, Bai Y, Wang H, Liu K, Zhang H, Xiao X, Wang K. Nucleolin mediated pro-angiogenic role of Hydroxysafflor Yellow A in ischaemic cardiac dysfunction: Post-transcriptional regulation of VEGF-A and MMP-9. J Cell Mol Med 2018; 22:2692-2705. [PMID: 29512890 PMCID: PMC5908102 DOI: 10.1111/jcmm.13552] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 01/02/2018] [Indexed: 01/22/2023] Open
Abstract
Hydroxysafflor Yellow A (HSYA), a most representative ingredient of Carthamus tinctorius L., had long been used in treating ischaemic cardiovascular diseases in China and exhibited prominently anticoagulant and pro-angiogenic activities, but the underlying mechanisms remained largely unknown. This study aimed to further elucidate the pro-angiogenic effect and mechanism of HSYA on ischaemic cardiac dysfunction. A C57 mouse model of acute myocardial infarction (AMI) was firstly established, and 25 mg/kg HSYA was intraperitoneally injected immediately after operation and given once, respectively, each morning and evening for 2 weeks. It was found that HSYA significantly improved ischaemia-induced cardiac haemodynamics, enhanced the survival rate, alleviated the myocardial injury and increased the expressions of CD31, vascular endothelial growth factor-A (VEGF-A) and nucleolin in the ischaemic myocardium. In addition, HSYA promoted the migration and tube formation of human umbilical vein endothelial cells (HUVECs), enhanced the expressions of nucleolin, VEGF-A and matrix metalloproteinase-9 (MMP-9) in a dose- and time-dependent manner. However, down-regulation of nucleolin expression sharply abrogated the effect mentioned above of HSYA. Further protein-RNA coimmunoprecipitation and immunoprecipitation-RT-PCR assay showed that nucleolin binded to VEGF-A and MMP-9 mRNA and overexpression of nucleolin up-regulated the mRNA expressions of VEGF-A and MMP-9 in the HUVECs through enhancing the stability of VEGF-A and MMP-9 mRNA. Furthermore, HSYA increased the mRNA expressions of VEGF-A and MMP-9 in the extract of antinucleolin antibody-precipitated protein from the heart of AMI mice. Our data revealed that nucleolin mediated the pro-angiogenic effect of HSYA through post-transcriptional regulation of VEGF-A and MMP-9 expression, which contributed to the protective effect of HSYA on ischaemic cardiac dysfunction.
Collapse
Affiliation(s)
- Jiang Zou
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Nian Wang
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Manting Liu
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Yongping Bai
- Department of Geriatric MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Hao Wang
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Ke Liu
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Huali Zhang
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Xianzhong Xiao
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
| | - Kangkai Wang
- Department of PathophysiologyXiangya School of MedicineCentral South UniversityChangshaChina
- Translational Medicine Center of SepsisKey Lab of Hunan ProvinceCentral South UniversityChangshaChina
- Department of Laboratory AnimalsXiangya School of MedicineCentral South UniversityChangshaChina
| |
Collapse
|