1
|
Wu R, Guo J, Liu Y, Huang S, Wu P, Liu W. Angiogenesis promotion of the transplantation of human amniotic mesenchymal stem cells via the Ang-1/Tie-2 signaling pathways in Alzheimer's disease model. J Alzheimers Dis 2025:13872877251338687. [PMID: 40326004 DOI: 10.1177/13872877251338687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
BackgroundAlzheimer's disease (AD) is a progressive degenerative disease of the central nervous system, leading to cognitive decline, mental symptoms, and behavioral disorders. The comorbidity of cerebrovascular disease in AD patients will accelerate the development of cognitive impairment and dementia. Since the dysfunction of the cerebral vasculature is closely related to neuropathology in AD patients, the protection of cerebral microvascular function and the improvement of cerebral microcirculation may bring a potential path for AD treatment. Human amniotic mesenchymal cells (hAMSCs) as a more advantageous cellular therapy for AD are proven to improve AD model mice's learning and memory abilities significantly, but fewer studies on angiogenesis and blood-brain barrier recovery have been found.ObjectiveThe study aimed to analyze the changes in angiogenesis-related factors of hAMSCs transplantation in the AD model and explore the underlying molecular mechanism.MethodshAMSCs were injected into APP/PS1 and wild type (WT) mice via tail vein, and the hAMSCs distribution in the cerebral tissue and angiogenesis in the hippocampal tissues were observed.ResultshAMSCs were found in the cortex and hippocampal areas of APP/PS1 and WT mice. hAMSCs transplantation significantly increased CD31 and Tie-2 expression in AD mice compared with the control group.ConclusionsThe study indicates that hAMSCs can cross the blood-brain barrier and enter the cerebral tissue of the mouse, transplantation of hAMSCs may promote angiogenesis in the AD model. The Ang-1/Tie-2 signaling pathway may be a therapeutically attractive target for the hAMSCs treatment of AD.
Collapse
Affiliation(s)
- Rui Wu
- Department of Brain Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi Province, China
| | - Jing Guo
- Department of Brain Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi Province, China
| | - Yang Liu
- Department of Brain Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi Province, China
| | - Sirou Huang
- The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shanxi Province, China
| | - Pingping Wu
- The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shanxi Province, China
| | - Weiping Liu
- Department of Brain Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi Province, China
| |
Collapse
|
2
|
Huo X, Peng Y, Li H, Li C, Liao F, Miao C, Huang Y. The emerging role of vascular endothelial cell-mediated angiogenesis in the imbalance of RA synovial microenvironment and its clinical relevance. Front Pharmacol 2025; 16:1481089. [PMID: 40255565 PMCID: PMC12006175 DOI: 10.3389/fphar.2025.1481089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 03/24/2025] [Indexed: 04/22/2025] Open
Abstract
Vascular endothelial cells (VEC) play a key role in the occurrence and progression of vascular inflammation. VEC activation secretes powerful inflammatory mediators and aggravates the development of rheumatoid arthritis (RA). Angiogenesis plays a key role in the pathological processes of inflammation and synovial infiltration, driving RA progression. A substantial amount of evidence suggests that the VEC at the inflammatory site of RA is both an active participant and a regulator of the inflammatory process. At present, the research progress of VEC and inflammation in RA is still incomplete. In this review, we summarize the role of VEC and angiogenesis in the development of RA, describe the relevant cells, cytokines and signaling pathways involved in regulation, and provide research clues on the role of post-translational modification (PTMs) in VEC function and angiogenesis in RA, and classify and integrate these mechanisms and therapeutic strategies. This review aims to synthesize current evidence to support the established link between VEC and RA-related pathology, provide a theoretical basis for clinical studies, and provide valuable insights into the development of therapeutic drugs against RA.
Collapse
Affiliation(s)
- Xingxing Huo
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Yanhui Peng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chen Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Faxue Liao
- Department of Orthopaedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, China
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, China
| | - Yurong Huang
- Department of Respiratory Medicine and Center of Infection and Immunity, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Yuan M, Wu Y, Zhou X, Cai Y, Li H, Xia A, Wang X, Wen J, Duan Q, Xu C, Cao H, Miao C. Clematichinenoside AR alleviates rheumatoid arthritis by inhibiting synovial angiogenesis through the HIF-1α/VEGFA/ANG2 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156552. [PMID: 40020629 DOI: 10.1016/j.phymed.2025.156552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Clematichinenoside AR (CAR) is an effective monomer component of Clematis chinensis Osbeck, which has therapeutic effects on rheumatoid arthritis (RA), but its specific mechanism is still not fully elucidated. PURPOSE This study elucidated whether CAR alleviated RA by inhibiting synovial angiogenesis and revealed its molecular mechanism. METHODS Arthritis indicators and H&E staining were used to evaluate the therapeutic effects of CAR on collagen-induced arthritis (CIA) rats, and the IHC, IF, EdU-Hoechst, tunel, flow cytometry, wound healing and transwell assay were used to investigate the effects of CAR on synovial angiogenesis. The co-culture model of RA fibroblast-like synoviocytes (FLSs) and human umbilical vein endothelial cells (HUVECs) was established. Tube formation, western blot, RT-qPCR and other related methods were used to evaluate the specific mechanism of CAR. RESULTS CAR alleviated arthritis pathology and inhibited angiogenesis in CIA rats. CAR inhibited the proliferation, migration and invasion of RA FLSs, and promoted their apoptosis. Importantly, overexpression of HIF-1α inversed the inhibitory impact of CAR on the expression of HIF-1α, VEGFA, VEGFR2, and ANG2, as well as the inhibitory effects of CAR on the expression of CD31/34 and the HUVEC tube formation. Molecular docking, molecular dynamics, and experimental verification confirmed that CAR has a strong binding affinity with HIF-1α, further indicating that HIF-1α was a target of CAR for anti-angiogenesis. CONCLUSION CAR had a good inhibitory effect on RA, and its mechanism was inhibition of synovial angiogenesis through the HIF-1α/VEGF/ANG2 axis.
Collapse
Affiliation(s)
- Meiling Yuan
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Xinyue Zhou
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Yikang Cai
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Aixin Xia
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Jianting Wen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, 117 Meishan Road, Hefei 230031, Anhui Province, China
| | - Qiangjun Duan
- Department of Experimental Teaching Center, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Chenhao Xu
- Department of Experimental Teaching Center, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Huibo Cao
- Department of Endocrinology, Chuzhou Integrated Traditional Chinese and Western Medicine Hospital, Anhui University of Chinese Medicine, 788 Huifeng East Road, Chuzhou 239000, Anhui Province, China
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; Institute of Prevention and Treatment of Rheumatoid Arthritis, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China; School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China.
| |
Collapse
|
4
|
Guan Y, Zhao X, Lu Y, Zhang Y, Lu Y, Wang Y. New bitongling regulates gut microbiota to predict angiogenesis in rheumatoid arthritis via the gut-joint axis: a deep neural network approach. Front Microbiol 2025; 16:1528865. [PMID: 39963498 PMCID: PMC11830818 DOI: 10.3389/fmicb.2025.1528865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is a persistent autoimmune disorder marked by inflammation and joint damage. Although current treatments, such as disease-modifying antirheumatic drugs (DMARDs), help control symptoms, they frequently cause substantial side effects, highlighting the urgent need for safer and more effective alternatives. Recent research indicates that gut microbiota might be pivotal in RA development through the "gut-joint axis," presenting novel therapeutic possibilities. Purpose This study seeks to explore the therapeutic potential of the traditional Chinese medicine (TCM) compound new bitongling (NBTL) for RA, with an emphasis on its capacity to regulate gut microbiota and suppress angiogenesis via the vascular endothelial growth factor (VEGF) signaling pathway. Methods We utilized a collagen-induced arthritis (CIA) rat model to assess the impact of NBTL. The study employed 16S ribosomal DNA (16S rDNA) sequencing to analyze gut microbiota composition, machine learning techniques to identify characteristic microbial taxa, and transcriptomic analysis (GSVA) to assess the impact on the VEGF signaling pathway. The findings were further validated through analysis with deep neural network models and in vivo/in vitro experiments, including western blot, immunofluorescence, and miRNA analysis. Results NBTL treatment markedly diminished inflammation in RA rats, evidenced by the reduced expression of TNF-α, IL-17, IL-6, and ASC in synovial tissues. Histopathological analysis confirmed alleviation of joint damage. Five characteristic microbial taxa, including f_Mycoplasmataceae, s_Metamycoplasma_sualvi, and g_Prevotellaceae_Ga6A1_group, were identified and associated with NBTL's modulation of the VEGF pathway. Gene set variation analysis (GSVA) revealed significant downregulation of the VEGF signaling pathway following NBTL treatment. Subsequent experiments confirmed that NBTL inhibited VEGF and its receptors, VEGFR1 and VEGFR2, along with HIF-1α (hypoxia-inducible factor 1-alpha), thereby reducing angiogenesis. Additionally, NBTL upregulated miR-20a-5p and miR-223-3p, contributing to its anti-angiogenic effects. Conclusion NBTL exhibits significant therapeutic potential in RA by modulating gut microbiota and inhibiting the VEGF signaling pathway. These findings support NBTL's use as a promising candidate for RA treatment, emphasizing the need for further research on its mechanisms and clinical application.
Collapse
Affiliation(s)
- Yin Guan
- Department of Rheumatism Immunity Branch, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoqian Zhao
- Department of Ethics Committee, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Lu
- Department of Rheumatism Immunity Branch, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Zhang
- Department of Rheumatism Immunity Branch, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Lu
- Department of Rheumatism Immunity Branch, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Wang
- Department of Rheumatism Immunity Branch, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
Song H, Yu J, Yang Y, Zhou L, Liu X, Yu J, Huang Q, Wang S, Zhang X, Liu Y, Zhang D, Meng J, Han T, Li W, Niu X. Exploring molecular mechanism of Panlongqi Tablet (PLQT) against RA: Integrated network pharmacology, molecular docking and experiment validation. Int Immunopharmacol 2025; 144:113639. [PMID: 39616851 DOI: 10.1016/j.intimp.2024.113639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND AND PURPOSE Panlongqi Tablet (PLQT), a proprietary Chinese medicine composed of 29 herbs, has been included in the Chinese Medical Insurance List and has shown promising therapeutic effects on patients with rheumatoid arthritis (RA) in clinical practice. However, the molecular mechanisms of PLQT against RA have not been fully elucidated. This study aimed to further decipher the active ingredients and molecular mechanism of PLQT anti-RA. METHODS A Complete Freund's adjuvant (CFA)-induced rat model was established to evaluate the pharmacodynamic effects of PLQT against RA, the assessment included arthritis index, paw thickness, ankle diameter, morphological and histopathological analysis. Network analysis was used to elucidate the active ingredients and underlying mechanisms of PLQT in the treatment of RA, molecular docking was conducted to assess the binding of active ingredients to key targets. In vitro and in vivo experimental verification were employed to reveal the mechanism of PLQT against RA. RESULTS Experimentally, PLQT improved CFA-induced arthritis without evident side effects. Network analysis revealed that the active ingredients in PLQT were mainly flavonoids, biscoumarin derivatives, alklaloid and lignans. Integrated with molecular docking studies, the molecular mechanisms of PLQT against RA were enriched in inflammatory response, immune regulation, angiogenesis, osteoclast differentiation and autophagy. In vitro experiments confirmed that PLQT exerted anti-inflammatory and immune regulatory effects by targeting the inflammatory response of M1 macrophages and the biological functions of T lymphocytes. In addition, In vivo experiments verified that PLQT could further inhibit synovial angiogenesis to prevent RA. CONCLUSION This study integrated network pharmacology analysis, molecular docking and experimental validation to elucidate the active components of PLQT and its mechanisms in intervening the pathological progression of RA, providing a more comprehensive theoretical basis for the clinical application of PLQT in the treatment of RA.
Collapse
Affiliation(s)
- Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinya Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Jianguo Meng
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
6
|
Guo Y, Yang P, Wu Z, Zhang S, You F. Mechanisms of Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao (huang qi) and Angelica sinensis (Oliv.) Diels (dang gui) in Ameliorating Hypoxia and Angiogenesis to Delay Pulmonary Nodule Malignant Transformation. Integr Cancer Ther 2025; 24:15347354241311917. [PMID: 39882753 PMCID: PMC11780663 DOI: 10.1177/15347354241311917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Screening for pulmonary nodules (PN) using low-dose CT has proven effective in reducing lung cancer (LC) mortality. However, current treatments relying on follow-up and surgical excision fail to fully address clinical needs. Pathological angiogenesis plays a pivotal role in supplying oxygen necessary for the progression of PN to LC. The interplay between hypoxia and angiogenesis establishes a vicious cycle, rendering anti-angiogenesis therapy alone insufficient to prevent PN to LC transformation. In traditional Chinese medicine (TCM), PN is referred to as "Feiji," which is mainly attributed to Qi and blood deficiency, correspondingly, the most commonly prescribed medicines are Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao (huang qi) (AR) and Angelica sinensis (Oliv.) Diels (dang gui) (ARS). Modern pharmacological studies have demonstrated that AR and ARS possess immune-enhancing, anti-tumor, anti-inflammatory, and anti-angiogenic properties. However, the precise mechanisms through which AR and ARS exert anti-angiogenic effects to delay PN progression to LC remain inadequately understood. This review explores the critical roles of hypoxia and angiogenesis in the transition from PN to LC. It emphasizes that, compared to therapies targeting angiogenic growth factors alone, AR, ARS, and their compound-based prescriptions offer additional benefits. These include ameliorating hypoxia by restoring blood composition, enhancing vascular structure, accelerating circulation, promoting vascular normalization, and blocking or inhibiting various pro-angiogenic expressions and receptor interactions. Collectively, these actions inhibit angiogenesis and delay the PN-to-LC transformation. Finally, this review summarizes recent advancements in related research, identifies existing limitations and gaps in knowledge, and proposes potential strategies and recommendations to address these challenges.
Collapse
Affiliation(s)
- Ying Guo
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Yang
- Chengdu Fifth People’s Hospital, Chengdu, China
| | - Zihong Wu
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanyin Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu Integrated TCM & Western Medicine Hospital, Chengdu, China
| | - Fengming You
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Guo X, Zhang J, Feng Z, Ji J, Shen X, Hou X, Mei Z. The antiangiogenic effect of total saponins of Panax japonicus C.A. Meyer in rheumatoid arthritis is mediated by targeting the HIF-1α/VEGF/ANG-1 axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118422. [PMID: 38838922 DOI: 10.1016/j.jep.2024.118422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 05/19/2024] [Accepted: 06/03/2024] [Indexed: 06/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese herbal medicine Panax japonicus C.A. Meyer has a long history in clinical treatment of rheumatoid arthritis (RA). Total saponins of Panax japonicus C.A. Meyer (TSPJs) were extracted from the root of Panax japonicus C.A. Meyer, and its anti-rheumatism mechanism is still unclear. AIM OF THE STUDY To investigate whether TSPJs attenuated synovial angiogenesis in RA and explore the potential mechanisms. MATERIALS AND METHODS Potential TSPJs targets involving gene function were predicted by network pharmacology related databases. Bioinformatics analysis and molecular docking technology were used to predict the mechanism of TSPJs in the treatment of RA. The predicted results were validated by cell experiments and a collagen-induced arthritis (CIA) mouse model. RESULTS Bioinformatics analysis results showed that TSPJs may inhibit RA-related angiogenesis through the hypoxia-inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) pathways. In vitro, different doses of TSPJs showed a good inhibitory effect on the tube formation of EA.hy926 cells. The results of the cellular thermal shift assay indicated that TSPJs can bind to the HIF-1α, VEGFA, and angiopoietin-1 (ANG-1) proteins. In vivo, the administration of TSPJs alleviated the symptoms of CIA mice, including the arthritis index, hind paw thickness, and swollen joint count. The histological results demonstrated that TSPJs inhibited inflammation, angiogenesis, bone damage, and cartilage destruction. Furthermore, TSPJs decreased the number of vessels and the expression level of CD31. The mechanistic results revealed that TSPJs decreased the expression of HIF-1α, VEGFA, and ANG-1 in the serum or synovial tissues of CIA mice. CONCLUSION These results suggest that TSPJs effectively inhibit angiogenesis in RA, and the mechanism may be related to inhibiting the HIF-1α/VEGF/ANG-1 axis.
Collapse
Affiliation(s)
- Xiang Guo
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Science, China Three Gorges University, Yichang, Hubei, 443002, China; The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China.
| | - Jinkai Zhang
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Science, China Three Gorges University, Yichang, Hubei, 443002, China.
| | - Zhitao Feng
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Science, China Three Gorges University, Yichang, Hubei, 443002, China; Institute of Rheumatology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, 443003, China.
| | - Jinyu Ji
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Science, China Three Gorges University, Yichang, Hubei, 443002, China.
| | - Xiaolan Shen
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Science, China Three Gorges University, Yichang, Hubei, 443002, China.
| | - Xiaoqiang Hou
- Institute of Rheumatology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, 443003, China.
| | - Zhigang Mei
- The Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
8
|
Gao C, Song XD, Chen FH, Wei GL, Guo CY. The protective effect of natural medicines in rheumatoid arthritis via inhibit angiogenesis. Front Pharmacol 2024; 15:1380098. [PMID: 38881875 PMCID: PMC11176484 DOI: 10.3389/fphar.2024.1380098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Rheumatoid arthritis is a chronic immunological disease leading to the progressive bone and joint destruction. Angiogenesis, accompanied by synovial hyperplasia and inflammation underlies joint destruction. Delaying or even blocking synovial angiogenesis has emerged as an important target of RA treatment. Natural medicines has a long history of treating RA, and numerous reports have suggested that natural medicines have a strong inhibitory activity on synovial angiogenesis, thereby improving the progression of RA. Natural medicines could regulate the following signaling pathways: HIF/VEGF/ANG, PI3K/Akt pathway, MAPKs pathway, NF-κB pathway, PPARγ pathway, JAK2/STAT3 pathway, etc., thereby inhibiting angiogenesis. Tripterygium wilfordii Hook. f. (TwHF), sinomenine, and total glucoside of Paeonia lactiflora Pall. Are currently the most representative of all natural products worthy of development and utilization. In this paper, the main factors affecting angiogenesis were discussed and different types of natural medicines that inhibit angiogenesis were systematically summarized. Their specific anti-angiogenesis mechanisms are also reviewed which aiming to provide new perspective and options for the management of RA by targeting angiogenesis.
Collapse
Affiliation(s)
- Chang Gao
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Xiao-Di Song
- Gannan Medical University, Jiangxi, Ganzhou, China
| | - Fang-Hui Chen
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Gui-Lin Wei
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Chun-Yu Guo
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| |
Collapse
|
9
|
Jiang F, Wang MQ, Zhang MY, Gu SL, Xie YW, Huang Y, Zhou MY, Li FL, Yang YC, Zhang PP, Liu XS, Li R. CPD-002, a novel VEGFR2 inhibitor, relieves rheumatoid arthritis by reducing angiogenesis through the suppression of the VEGFR2/PI3K/AKT signaling pathway. Int Immunopharmacol 2024; 131:111850. [PMID: 38479157 DOI: 10.1016/j.intimp.2024.111850] [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: 12/07/2023] [Revised: 02/06/2024] [Accepted: 03/10/2024] [Indexed: 04/10/2024]
Abstract
Synovial angiogenesis is a key player in the development of rheumatoid arthritis (RA), and anti-angiogenic therapy is considered a promising approach for treating RA. CPD-002 has demonstrated efficacy in suppressing tumor angiogenesis as a VEGFR2 inhibitor, but its specific impacts on RA synovial angiogenesis and possible anti-RA effects need further study. We examined the influences of CPD-002 on the migration and invasion of human umbilical vein endothelial cells (HUVECs) and its impacts on HUVECs' tube formation and vessel sprouting ex vivo. The therapeutic potential of CPD-002 in adjuvant-induced arthritis (AIA) rats and its suppression of synovial angiogenesis were examined. The involvement of the VEGFR2/PI3K/AKT pathway was assessed both in HUVECs and AIA rat synovium. Here, CPD-002 inhibited the migration and invasion of VEGF-stimulated HUVECs, decreased their chemotactic response to RA fibroblast-like synoviocyte-released chemoattractants, and exhibited anti-angiogenic effects in vitro and ex vivo. CPD-002's targeting of VEGFR2 was confirmed with molecular docking and cellular thermal shift assays, supported by the abolishment of CPD-002's effects upon using VEGFR2 siRNA. CPD-002 relieved paw swelling, arthritis index, joint damage, and synovial angiogenesis, indicating its anti-arthritic and anti-angiogenic effects in AIA rats. Moreover, the anti-inflammatory effects in vivo and in vitro of CPD-002 contributed to its anti-angiogenic effects. Mechanistically, CPD-002 hindered the activation of VEGFR2/PI3K/AKT pathway in VEGF-induced HUVECs and AIA rat synovium, as evidenced by reduced p-VEGFR2, p-PI3K, and p-AKT protein levels alongside elevated PTEN protein levels. Totally, CPD-002 showed anti-rheumatoid effects via attenuating angiogenesis through the inhibition of the VEGFR2/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Fei Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Meng-Qing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Ya-Wen Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Fei-Long Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Yu-Chen Yang
- The First Clinical Medical College, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Pei-Pei Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China
| | - Xue-Song Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, PR China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230026, Anhui Province, PR China.
| |
Collapse
|
10
|
Fu X, Song Y, Feng X, Liu Z, Gao W, Song H, Zhang Q. Synergistic chemotherapy/PTT/oxygen enrichment by multifunctional liposomal polydopamine nanoparticles for rheumatoid arthritis treatment. Asian J Pharm Sci 2024; 19:100885. [PMID: 38434718 PMCID: PMC10906176 DOI: 10.1016/j.ajps.2024.100885] [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: 08/14/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 03/05/2024] Open
Abstract
Amultifunctional liposomal polydopamine nanoparticle (MPM@Lipo) was designed in this study, to combine chemotherapy, photothermal therapy (PTT) and oxygen enrichment to clear hyperproliferating inflammatory cells and improve the hypoxic microenvironment for rheumatoid arthritis (RA) treatment. MPM@Lipo significantly scavenged intracellular reactive oxygen species and relieved joint hypoxia, thus contributing to the repolarization of M1 macrophages into M2 phenotype. Furthermore, MPM@Lipo could accumulate at inflammatory joints, inhibit the production of inflammatory factors, and protect cartilage in vivo, effectively alleviating RA progression in a rat adjuvant-induced arthritis model. Moreover, upon laser irradiation, MPM@Lipo can elevate the temperature to not only significantly obliterate excessively proliferating inflammatory cells but also accelerate the production of methotrexate and oxygen, resulting in excellent RA treatment effects. Overall, the use of synergistic chemotherapy/PTT/oxygen enrichment therapy to treat RA is a powerful potential strategy.
Collapse
Affiliation(s)
- Xiaoling Fu
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Yutong Song
- First school of clinical medicine, Nanjing Medical University, Nanjing 211166, China
| | - Xianquan Feng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Zhihong Liu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Wenhao Gao
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Hongtao Song
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Qian Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
| |
Collapse
|
11
|
Hu W, Cai W, Wu Y, Ren C, Yu D, Li T, Shen T, Xu D, Yu J. Topical Application of Cell-Penetrating Peptide Modified Anti-VEGF Drug Alleviated Choroidal Neovascularization in Mice. Int J Nanomedicine 2024; 19:35-51. [PMID: 38187905 PMCID: PMC10771783 DOI: 10.2147/ijn.s428684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/19/2023] [Indexed: 01/09/2024] Open
Abstract
Background Age-related macular degeneration (AMD) stands as the foremost cause of irreversible central vision impairment, marked by choroidal neovascularization (CNV). The prevailing clinical approach to AMD treatment relies on intravitreal injections of anti-vascular endothelial growth factor (VEGF) drugs. However, this method is encumbered by diverse complications, prompting exploration of non-invasive alternatives such as ocular administration via eye drops for anti-VEGF therapy. Methods Two complexes, 5-FITC-CPP-Ranibizumab (5-FCR) and 5-FITC-CPP-Conbercept (5-FCC), were synthesized by incorporating the anti-VEGF drugs Ranibizumab (RBZ) or Conbercept (CBC) with cell-penetrating peptide (CPP). Circular dichroism spectrum (CD) facilitated complexes characterization. Eye drops was utilized to address laser-induced CNV in mice. Fluorescein fundus angiography (FFA) observe the CNV lesion, while FITC-dextran and IB4 dual fluorescent staining, along with hematoxylin-eosin (HE) staining, assessed in lesion size. Tissue immunofluorescence examined CD31 and VEGF expression in choroidal/retinal pigment epithelial (RPE) tissues. Biocompatibility and biosafety of 5-FCR and 5-FCC was evaluated through histological examination of various organs or cell experiments. Results Both 5-FCR and 5-FCC exhibited favorable biocompatibility and safety profiles. VEGF-induced migration of Human umbilical vein endothelial cells (HUVECs) significantly decreased post-5-FCR/5-FCC treatment. Additionally, both complexes suppressed VEGF-induced tube formation in HUVECs. FFA results revealed a significant improvement in retinal exudation in mice. Histological examination unveiled the lesion areas in the 5-FCR and 5-FCC groups showed a significant reduction compared to the control group. Similar outcomes were observed in histological sections of the RPE-choroid-sclera flat mounts. Conclusion In this study, utilizing the properties of CPP and two anti-VEGF drugs, we successfully synthesized two complexes, 5-FCR and 5-FCC, through a straightforward approach. Effectively delivering the anti-VEGF drugs to the target area in a non-invasive manner, suppressing the progression of laser-induced CNV. This offers a novel approach for the treatment of wet AMD.
Collapse
Affiliation(s)
- Weinan Hu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Wenting Cai
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Chengda Ren
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Donghui Yu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Tingting Li
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Tianyi Shen
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Ding Xu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| | - Jing Yu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
12
|
Liang H, Yin G, Shi G, Liu Z, Liu X, Li J. Echinacoside regulates PI3K/AKT/HIF-1α/VEGF cross signaling axis in proliferation and apoptosis of breast cancer. Anal Biochem 2024; 684:115360. [PMID: 37865269 DOI: 10.1016/j.ab.2023.115360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
CONTEXT Echinacoside (ECH) is a natural anti-cancer compound and is of great value in cancer treatment. However, the mechanism underlying this effect on breast cancer (BC) was unclear. OBJECTIVE To explore the mechanism of ECH treating BC by network pharmacology and experimental validation. MATERIALS & METHODS Several databases were searched to screen potential targets of ECH and obtain information on targets related to BC. STRING was applied to construct a Protein-protein interaction (PPI) network. DAVID was applied for Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Gene Expression Profiling Interactive Analysis (GEPIA) was searched for the relationship between the expression profile and overall survival of major targets in normal breast and BC tissues. Finally, the results of network pharmacology analysis were validated by experiments. RESULTS Seventeen targets of ECH overlapped with targets in BC. Ten hub targets were determined through PPI. By GO and KEGG analysis 15 entries and 25 pathways were obtained, in which phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), hypoxia inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) played greater roles. Validation of key targets in the GEPIA database showed that PIK3R1 and PIK3CD remained consistent with the results of the study. Experiments in vitro showed ECH inhibited proliferation, induced apoptosis and reduced mRNA levels and protein expression of PI3K, AKT, hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA) in MCF-7 cells. Furthermore, experiments in vivo revealed that ECH significantly reduced tumor growth, promoted apoptosis and decreased the related mRNA levels and protein expression, suggesting ECH works on BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. DISCUSSION & CONCLUSION In summary, ECH played an important role in anti-BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. Furthermore, ECH had multi-target and multi-pathway effects, which may be a promising natural compound for treating BC.
Collapse
Affiliation(s)
- Hongyi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guangxi Shi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Zhiyong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Xiaofei Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Jingwei Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
13
|
Qiao J, Guo S, Huang X, Zhang L, Li F, Fan Y. Expression of Angiopoietin-2 in Lung Tissue of Juvenile SD Rats with Lipopolysaccharide-Induced Acute Lung Injury and the Role of Ulinastatin. Arch Immunol Ther Exp (Warsz) 2023; 71:23. [PMID: 37882869 DOI: 10.1007/s00005-023-00688-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023]
Abstract
This study aimed to observe the expression of angiopoietin-2 (Ang-2) in the lung tissue of juvenile SD rats with lipopolysaccharide (LPS)-induced acute lung injury (ALI) and to clarify the role of ulinastatin (UTI). Ninety 18-21-day-old juvenile SD male rats were randomly divided into five groups (n = 18). ALI rat model was established by intraperitoneal injection of LPS (LPS 10 mg/kg), while the control group was given the same dose of normal saline. The UTI intervention group was given the injection of UTI (5000 U/mL) immediately after the injection of LPS, which was divided into UTI low-dose group (LPS + 5 ml/kg UTI), UTI medium-dose group (LPS + 10 ml/kg UTI), and UTI high-dose group (LPS + 20 ml/kg UTI).The respiratory status of each group of rats was observed, and six rats were randomly selected to be killed in each group at 6, 12, and 24 h, and the lung tissues were dissected and retained. The pathological changes of the lung tissues were observed by hematoxylin-eosin (HE) staining, the expression levels and locations of Ang-2 and vascular endothelial growth factor (VEGF) in lung tissue were observed by immunohistochemical staining, and the expressions of genes and proteins of Ang-2 and VEGF were detected by quantitative reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis. Three hours after intraperitoneal injection, rats in the model group developed shortness of breath and the developed respiratory distress progressed over time. The lung pathological changes in the model group were obvious compared with those in the control group, and gradually worsened with time, and the pathological changes of lung in the rats in the UTI intervention group were reduced compared with those in the model group. At different time points, the expressions of Ang-2 and VEGF in the lung tissue of rats in the model group were higher than those in the control group, and were lower in the UTI intervention group than those in the model group. The expressions of Ang-2 and VEGF protein were lower in the low-dose group of UTI group than those in the high-dose group of UTI group at different time points (P < 0.05), and the expressions of Ang-2 and VEGF protein in the low-dose group of UTI were significantly lower than those in the medium-dose group at 12 h and 24 h (P < 0.05). The expression of Ang-2 was increased in the lung tissue of juvenile SD rats with LPS-induced ALI, and was associated with the degree of lung injury. UTI might attenuate LPS-induced ALI by inhibiting the expression of Ang-2 in lung tissue, and the low dose was more obvious than the medium and high dose.
Collapse
Affiliation(s)
- Junying Qiao
- Department of Pediatric Critical Care Medicine, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Zhengzhou, 450052, Henan, China.
| | - Shanshan Guo
- Department of Pediatrics, The Third Clinical College of Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Xianjie Huang
- Department of Pediatric Critical Care Medicine, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Zhengzhou, 450052, Henan, China
| | - Luodan Zhang
- Department of Pediatric Critical Care Medicine, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Zhengzhou, 450052, Henan, China
| | - Fan Li
- Department of Pediatric Critical Care Medicine, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Zhengzhou, 450052, Henan, China
| | - Yazhen Fan
- Department of Pediatric Critical Care Medicine, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Zhengzhou, 450052, Henan, China
| |
Collapse
|
14
|
Li D, Zhao B, Zhuang P, Mei X. Development of nanozymes for promising alleviation of COVID-19-associated arthritis. Biomater Sci 2023; 11:5781-5796. [PMID: 37475700 DOI: 10.1039/d3bm00095h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has been identified as a culprit in the development of a variety of disorders, including arthritis. Although the emergence of arthritis following SARS-CoV-2 infection may not be immediately discernible, its underlying pathogenesis is likely to involve a complex interplay of infections, oxidative stress, immune responses, abnormal production of inflammatory factors, cellular destruction, etc. Fortunately, recent advancements in nanozymes with enzyme-like activities have shown potent antiviral effects and the ability to inhibit oxidative stress and cytokines and provide immunotherapeutic effects while also safeguarding diverse cell populations. These adaptable nanozymes have already exhibited efficacy in treating common types of arthritis, and their distinctive synergistic therapeutic effects offer great potential in the fight against arthritis associated with COVID-19. In this comprehensive review, we explore the potential of nanozymes in alleviating arthritis following SARS-CoV-2 infection by neutralizing the underlying factors associated with the disease. We also provide a detailed analysis of the common therapeutic pathways employed by these nanozymes and offer insights into how they can be further optimized to effectively address COVID-19-associated arthritis.
Collapse
Affiliation(s)
- Dan Li
- Department of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Baofeng Zhao
- Liaoning Provincial Key Laboratory of Medical Testing, Jinzhou Medical University, Jinzhou, 121001, China.
| | - Pengfei Zhuang
- Department of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China.
| | - Xifan Mei
- Liaoning Provincial Key Laboratory of Medical Testing, Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
15
|
Tai Y, Zheng L, Liao J, Wang Z, Zhang L. Roles of the HIF-1α pathway in the development and progression of keloids. Heliyon 2023; 9:e18651. [PMID: 37636362 PMCID: PMC10448433 DOI: 10.1016/j.heliyon.2023.e18651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Keloids, a pathological scar that is induced by the consequence of aberrant wound healing, is still a major global health concern for its unsatisfactory treatment outcomes. HIF-1α, a main regulator of hypoxia, mainly acts through some proteins or signaling pathways and plays important roles in a variety of biological processes. Accumulating evidence has shown that HIF-1α played a crucial role in the process of keloid formation. In this review, we attempted to summarize the current knowledge on the association between HIF-1α expression and the development and progression of keloids. Through a comprehensive analysis, the molecular mechanisms underlying HIF-1α in keloids were shown to be correlated to the proliferation of fibroblasts, angiogenesis, and collagen deposits. The affected proteins and the signaling pathways were multiple. For instance, HIF-1α was reported to promote keloids formation by enhancing angiogenesis, fibroblast proliferation, and collagen deposition through the activation of periostin PI3K/Akt, TGF-β/Smad and TLR4/MyD88/NF-κB pathway. However, the specific effects of HIF-1α on keloids keloid illnesses in clinical practice is are entirely unclear, and further studies in clinical trials are still warranted. Therefore, an in-depth understanding of the biological mechanisms of HIF-1α in keloid formation is significant to develop promising therapeutic targets for the treatment of keloids in clinical practice.
Collapse
Affiliation(s)
- Yuncheng Tai
- Department of Burn Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China
| | - Liying Zheng
- Postgraduate Department, First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Jiao Liao
- Department of Nephrology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China
| | - Zixiong Wang
- Department of Burn and Plastic Surgery, Xinjiang Military General Hospital, Urumqi, 830063, Xinjiang, China
| | - Lai Zhang
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, 318000, Zhejiang, China
| |
Collapse
|
16
|
Tong Y, Li X, Deng Q, Shi J, Feng Y, Bai L. Advances of the small molecule drugs regulating fibroblast-like synovial proliferation for rheumatoid arthritis. Front Pharmacol 2023; 14:1230293. [PMID: 37547337 PMCID: PMC10400780 DOI: 10.3389/fphar.2023.1230293] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
Rheumatoid arthritis (RA) is a type of chronic autoimmune and inflammatory disease. In the pathological process of RA, the alteration of fibroblast-like synoviocyte (FLS) and its related factors is the main influence in the clinic and fundamental research. In RA, FLS exhibits a uniquely aggressive phenotype, leading to synovial hyperplasia, destruction of the cartilage and bone, and a pro-inflammatory environment in the synovial tissue for perpetuation and progression. Evidently, it is a highly promising way to target the pathological function of FLS for new anti-RA drugs. Based on this, we summed up the pathological mechanism of RA-FLS and reviewed the recent progress of small molecule drugs, including the synthetic small molecule compounds and natural products targeting RA-FLS. In the end, there were some views for further action. Compared with MAPK and NF-κB signaling pathways, the JAK/STAT signaling pathway has great potential for research as targets. A small number of synthetic small molecule compounds have entered the clinic to treat RA and are often used in combination with other drugs. Meanwhile, most natural products are currently in the experimental stage, not the clinical trial stage, such as triptolide. There is an urgent need to unremittingly develop new agents for RA.
Collapse
Affiliation(s)
- Yitong Tong
- Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Xinyu Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qichuan Deng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Lv Y, Zhang J, Li C, Wang L, Lei L, Huang X. Network pharmacological analysis to reveal the mechanism governing the effect of Qin Xi Tong on osteoarthritis and rheumatoid arthritis. Clin Rheumatol 2023:10.1007/s10067-023-06625-5. [PMID: 37162694 DOI: 10.1007/s10067-023-06625-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 04/03/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
INTRODUCTION Qin Xi Tong (QXT), produced by water extracts of Caulis Sinomenii, is clinically effective in the therapy of rheumatoid arthritis (RA). It is also a complementary agent for osteoarthritis (OA). This study aimed to screen the candidate targets and identify the potential mechanisms of QXT against RA and OA. METHOD The active ingredients contained in QXT were queried from the TCMSP database. Their predicted targets were obtained through web-based databases, including TCMSP, BATMAN-TCM, CTD, and PharmMapper. The OA and RA targets were collected from the Genecards database and the GSE55235 dataset. Based on the DAVID database, GO and KEGG enrichment analyses of disease-drug common targets predicted potential signaling pathways for QXT. In addition, core targets were identified by mapping component-target-disease interaction networks with Cytoscape 3.9.1 and STRING. The Swissdock and Pymol tools further validate the predicted results. RESULTS A total of 161 genes were put forward as potential targets for treating RA and OA. These genes might be involved in joint inflammation, including the IL-17 signaling pathway, MAPK signaling pathway, and TNF signaling pathway. They also regulated the progression of joint injuries, such as apoptosis, Th17 cell differentiation, and osteoclast differentiation. In addition, we identified 12 core targets of QXT. Molecular docking results showed that QXT has a high affinity with these core targets. CONCLUSIONS This study reveals the mechanism governing the effect of QXT on RA and OA, predicts the direct target, and provides new ideas for clinical treatment. Key Points • Our study reveals the underlying mechanism of QXT in the treatment of RA and OA. • Further research into the effects of compounds in QXT alone would be of interest.
Collapse
Affiliation(s)
- Yanyan Lv
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Jie Zhang
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Chao Li
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Li Wang
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Lei Lei
- School of Life Sciences and Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, China
| | - Xiaoqiang Huang
- Department of Orthopedics, Xi'an No.5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China.
| |
Collapse
|
18
|
Network pharmacology and experimental validation to identify the potential mechanism of Hedyotis diffusa Willd against rheumatoid arthritis. Sci Rep 2023; 13:1425. [PMID: 36697436 PMCID: PMC9877023 DOI: 10.1038/s41598-022-25579-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 12/01/2022] [Indexed: 01/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, autoimmune disease that may lead to joint damage, deformity, and disability, if not treated effectively. Hedyotis diffusa Willd (HDW) and its main components have been widely used to treat a variety of tumors and inflammatory diseases. The present study utilized a network pharmacology approach, microarray data analysis and molecular docking to predict the key active ingredients and mechanisms of HDW against RA. Eleven active ingredients in HDW and 180 potential anti-RA targets were identified. The ingredients-targets-RA network showed that stigmasterol, beta-sitosterol, quercetin, kaempferol, and 2-methoxy-3-methyl-9,10-anthraquinone were key components for RA treatment. KEGG pathway results revealed that the 180 potential targets were inflammatory-related pathways with predominant enrichment of the AGE-RAGE, TNF, IL17, and PI3K-Akt signaling pathways. Screened through the PPI network and with Cytoscape software, RELA, TNF, IL6, TP53, MAPK1, AKT1, IL10, and ESR1 were identified as the hub targets in the HDW for RA treatment. Molecular docking was used to identify the binding of 5 key components and the 8 related-RA hub targets. Moreover, the results of network pharmacology were verified by vitro experiments. HDW inhibits cell proliferation in MH7A cells in a dose and time-dependent manner. RT-qPCR and WB results suggest that HDW may affect hub targets through PI3K/AKT signaling pathway, thereby exerting anti-RA effect. This study provides evidence for a clinical effect of HDW on RA and a research basis for further investigation into the active ingredients and mechanisms of HDW against RA.
Collapse
|
19
|
Zhao L, Tang X, Huang R, Liu Q, Liao L, Hu Y, He K, Zhang X, Guo J, Chen S, Yang S. Acute hypoxia promotes the liver angiogenesis of largemouth bass (Micropterus salmoides) by HIF - Dependent pathway. FISH & SHELLFISH IMMUNOLOGY 2022; 131:264-273. [PMID: 35940542 DOI: 10.1016/j.fsi.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 06/15/2023]
Abstract
A 24-h hypoxia exposure experiment was conducted to determine how hypoxia exposure induce liver angiogenesis in largemouth bass. Nitrogen (N2) was pumped into water to exclude dissolved oxygen into 1.2 ± 0.2 mg/L, and liver tissues were sampled during hypoxia exposure of 0 h, 4 h, 8 h, 12 h, 24 h and re-oxygenation for 12 h. Firstly, the results showed that hypoxia exposure promoted the angiogenesis occurrence by immunohistochemical analysis of vascular endothelial growth factor receptor 2 (VEGFR2). Secondly, the concentration of vasodilation factor increased and it's activity was elevated during 8 h exposure, such as nitric oxide (NO) and nitric oxide synthase (NOS) (p < 0.05). Thirdly, hypoxia exposure promoted angiogenesis through up-regulation the expression of matrix metalloproteinase 2 (MMP-2), jagged, protein kinase B (AKT), phosphoinositide-3-kinase (PI3K), mitogen-activated protein kinase (MAPK) at 4 h; contrarily, the expression of inhibiting angiogenesis genes presented up-regulated at 8 h (p < 0.05), such as matrix metalloproteinase inhibitor-2 (TIMP-2), matrix metalloproteinase inhibitor-3 (TIMP-3). Finally, the genes and proteins that regulate angiogenesis presented obvious chronological order. Parts of them promoted the budding and extension of blood vessels were up-regulated during 4 h-8 h (p < 0.05), such as vascular endothelial growth factor a (VEGFA), VEGFR2, monocarboxylic acid transporter 1 (MCT1), CD147, prolyl hydroxylase (PHD), nuclear factor kappa-B (NF-κB); other part of them promoted blood vessel maturation were highly expressed during 12 h-24 h (p < 0.05), such as angiogenin-1 (Ang-1) and angiogenin-2 (Ang-2). In short, acute hypoxia can promote the liver angiogenesis of largemouth bass by HIF - dependent pathway.
Collapse
Affiliation(s)
- Liulan Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Xiaohong Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Fish Resources and Environment in the Lpper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu, Sichuan, 610011, China.
| | - Rui Huang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Qiao Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Lei Liao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Yifan Hu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Kuo He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Xin Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Jiazhong Guo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Shiyi Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Song Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
20
|
Bu Y, Wu H, Deng R, Wang Y. Geniposide restricts angiogenesis in experimentary arthritis via inhibiting Dnmt1-mediated PTEN hypermethylation. Int Immunopharmacol 2022; 111:109087. [PMID: 35908504 DOI: 10.1016/j.intimp.2022.109087] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/01/2022]
Abstract
Neovascularization in rheumatoid arthritis (RA) is a key bridge between malignant proliferative synovial tissue and pannus. In view of previous studies on the efficacy of Geniposide (GE) in experimentary arthritis, the purpose of this study was to investigate the possible mechanism of GE inhibiting angiogenesis by regulating the gene of phosphate and tension homology deleted on chromosome ten (PTEN). In this study, human umbilical vein endothelial cells (HUVEC) and adjuvant arthritis (AA) rat models were performed to research in vitro and in vivo. The results showed that GE treatment significantly reduced synovitis and angiogenesis in AA rats, which may be associated with the increased expression of PTEN with GE treatment. Meanwhile, the hypermethylation of PTEN accompanied by the over-expression of DNA methyltransferases (Dnmts) was demonstrated in TNF-α-induced HUVEC and AA rats. Knockdown of Dnmt1 by Dnmt1- siRNA significantly inhibited the tube formation of HUVEC in vitro. GE significantly restricted the angiogenesis of HUVEC by inhibiting DNA methylation, which was attributed to the down-regulation of Dnmt1 rather than Dnmt3a and Dnmt3b. The anti-angiogenesis effect of GE was further verified in AA model by the inhibition of Dnmt1. These results indicate that GE exhibited anti-angiogenesis effects in experimentary arthritis by inhibiting Dnmt1-mediated PTEN gene hypermethylation, which may brings new insights for the prevention and research of RA.
Collapse
Affiliation(s)
- Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China.
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
21
|
Mao Y, Meng L, Liu H, Lu Y, Yang K, Ouyang G, Ban Y, Chen S. Therapeutic potential of traditional Chinese medicine for vascular endothelial growth factor. J Zhejiang Univ Sci B 2022; 23:353-364. [PMID: 35557037 DOI: 10.1631/jzus.b2101055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vascular endothelial growth factor (VEGF) is the main regulator of physiological angiogenesis during embryonic development, bone growth, and reproductive function, and it also participates in a series of pathological changes. Traditional Chinese medicine (TCM), with a history of more than 2000 years, has been widely used in clinical practice, while the exploration of its mechanisms has only begun. This review summarizes the research of recent years on the influence of TCM on VEGF. It is found that many Chinese medicines and recipes have a regulatory effect on VEGF, indicating that Chinese medicine has broad prospects as a complementary and alternative therapy, providing new treatment ideas for clinical applications and the theoretical basis for research on the mechanisms of TCM.
Collapse
Affiliation(s)
- Yijia Mao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Lingkai Meng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Huayi Liu
- Department of Digestive Diseases, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China.
| | - Yuting Lu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Kuo Yang
- Department of Digestive Diseases, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Guangze Ouyang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Yanran Ban
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Shuang Chen
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| |
Collapse
|