1
|
Xie Y, Fan Y, Liu X, Li Z, Liu S. 4D-DIA-based proteomics analysis reveals the protective effects of Pidanjiangtang granules in IGT rat model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119012. [PMID: 39481621 DOI: 10.1016/j.jep.2024.119012] [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: 09/09/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Pidanjiangtang (PDJT) formula was founded on the "Pidan" theory from the "Nei Jing." PDJT is considered to eliminate the accumulation of pathological products, remove heat sources, and prevent damage to organs such as the liver and islets. It is widely used in clinical practice to treat impaired glucose tolerance (IGT). However, the bioactive ingredients and underlying mechanisms are still unclear and need further investigation. OBJECTIVE This study aimed to determine the therapeutic effect of PDJT on IGT rats and explore the mechanism of PDJT intervention on IGT by four-dimensional independent data acquisition (4D-DIA) proteomics analysis. MATERIALS AND METHODS The IGT model was established by a high-fat diet combined with Streptozotocin (STZ) injection. The IGT rats were treated with low, medium, and high doses of PDJT orally for 42 days and compared with the Metformin positive control group. The therapeutic effects of PDJT on IGT rats were evaluated using the oral glucose tolerance test (OGTT), serum lipoprotein detection, insulin detection, liver histopathology, and hepatic steatosis assessment. 4D-DIA proteomics analysis was used to explore the differential proteins (DEPs) and potential pathways of PDJT. Finally, Western blotting and ELISA techniques were used to verify DEPs and major targets. RESULTS PDJT can enhance glucose metabolism, restore islet β cell function, regulate lipoprotein metabolism, reduce hepatic steatosis, and consequently slow down the progression of IGT. In the proteomic analysis, a total of 355 DEPs were identified, and critical proteins were validated. The results indicated that the JAK2/STAT1 signaling pathway plays a pivotal role in the effects of PDJT. IκB-ζ may be a potential target for PDJT in regulating the inflammatory response of IGT. CONCLUSION PDJT is an effective formula for improving IGT, with its potential mechanism linked to the JAK2/STAT1/IκB-ζ signaling pathway. This study offers a novel approach to investigating the mechanisms of TCM formula through proteomics and offers new insight into exploring TCM treatment for IGT.
Collapse
Affiliation(s)
- Yu Xie
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Fan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Liu
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zirong Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Shangjian Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Zhang Y, Lian X, Xu H, Zhu S, Zhang H, Ni Z, Fu T, Liu S, Tao L, Zhou Y, Zhu F. OrgXenomics: an integrated proteomic knowledge base for patient-derived organoid and xenograft. Nucleic Acids Res 2025; 53:D504-D515. [PMID: 39373514 PMCID: PMC11701540 DOI: 10.1093/nar/gkae861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Patient-derived models (PDMs, particularly organoids and xenografts) are irreplaceable tools for precision medicine, from target development to lead identification, then to preclinical evaluation, and finally to clinical decision-making. So far, PDM-based proteomics has emerged to be one of the cutting-edge directions and massive data have been accumulated. However, such PDM-based proteomic data have not been provided by any of the available databases, and proteomics profiles of all proteins in proteomic study are also completely absent from existing databases. Herein, an integrated database named 'OrgXenomics' was thus developed to provide the proteomic data for PDMs, which was unique in (a) explicitly describing the establishment detail for a wide array of models, (b) systematically providing the proteomic profiles (expression/function/interaction) for all proteins in studied proteomic analysis and (c) comprehensively giving the raw data for diverse organoid/xenograft-based proteomic studies of various diseases. Our OrgXenomics was expected to server as one good complement to existing proteomic databases, and had great implication for the practice of precision medicine, which could be accessed at: https://idrblab.org/orgxenomics/.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Xichen Lian
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hangwei Xu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Sisi Zhu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Zhang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
3
|
Lyu J, Zhang H, Wang C, Pan M. New insight in treating autoimmune diseases by targeting autophagy. Autoimmunity 2024; 57:2351872. [PMID: 38739691 DOI: 10.1080/08916934.2024.2351872] [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: 01/22/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Autophagy is a highly conserved biological process in eukaryotes, which degrades cellular misfolded proteins, damaged organelles and invasive pathogens in the lysosome-dependent manner. Autoimmune diseases caused by genetic elements, environments and aberrant immune responses severely impact patients' living quality and even threaten life. Recently, numerous studies have reported autophagy can regulate immune responses, and play an important role in autoimmune diseases. In this review, we summarised the features of autophagy and autophagy-related genes, enumerated some autophagy-related genes involved in autoimmune diseases, and further overviewed how to treat autoimmune diseases through targeting autophagy. Finally, we outlooked the prospect of relieving and curing autoimmune diseases by targeting autophagy pathway.
Collapse
Affiliation(s)
- Jiao Lyu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hongqian Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chaoyang Wang
- The Key Medical Laboratory for Chemical Poison Detection of Henan Province, The Third People's Hospital of Henan Province, Zhengzhou, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China
| | - Mingyu Pan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Cheng X, Wan J, Sun D, Zhan Y, Yu J, Li Y, Xiong Y, Liu W. Proteomic Insights into the Effects of Jianweixiaoshi Tablets on Functional Dyspepsia with Spleen Deficiency in Rats. Drug Des Devel Ther 2024; 18:5129-5148. [PMID: 39554757 PMCID: PMC11568854 DOI: 10.2147/dddt.s477034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024] Open
Abstract
Background Jianweixiaoshi tablets (JWXS) is widely used in traditional Chinese medicine for treating functional dyspepsia with spleen deficiency (SD-FD) in China. However, the molecular mechanisms underlying the therapeutic effects of JWXS remain incompletely understood. Methods Functional dyspepsia was induced in rats with spleen deficiency by iodoacetamide in combination with the modified multiple platform method. The SD-FD rats were administered JWXS at both low and high doses, as well as domperidone. We conducted a comprehensive evaluation of the treatment effects of JWXS, including body weight, gastrointestinal motility, immune organ index, biochemical analysis, gastrointestinal hormones, and hematological studies. Quantitative proteomic analysis based on data-independent acquisition (DIA) was used to determine the changes in protein profiles of gastric and duodenal tissues in SD-FD rats and JWXS intervention rats. Results The results showed that JWXS effectively alleviated gastrointestinal motility disorders in SD-FD rats, as indicated by accelerated gastric emptying and intestinal propulsion, increased levels of gastrin, motilin, and ghrelin, and reduced levels of cholecystokinin-octapeptide, vasoactive intestinal peptide, and somatostatin. Additionally, JWXS increased the spleen and thymus index, increased %lymphocyte in blood, reduced white blood cell count and %neutrophil, and improved immune function. Through quantitative proteomic analysis of gastric tissues, we identified 333 differentially expressed proteins in the JWXS treatment group and the model group. Notably, the mechanism by which JWXS accelerated gastric emptying may be related to PLC-γ and SERCA2 in the calcium signaling pathway. Furthermore, JWXS treatment altered the expression of 732 proteins in rat duodenal samples. The differentially expressed proteins were enriched in immune-related functions and pathways, including antigen processing and presentation, as well as the intestinal immune network for IgA production. Conclusion In conclusion, JWXS exhibits a multi-faceted impact on various pathways, demonstrating its efficacy in treating SD-FD. These findings provide a foundation for the clinical application of JWXS in managing SD-FD.
Collapse
Affiliation(s)
- Xiaoying Cheng
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Jianhua Wan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Denglong Sun
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Yang Zhan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Jingting Yu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Yingmeng Li
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Yanxia Xiong
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| | - Wenjun Liu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, People’s Republic of China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330103, People’s Republic of China
| |
Collapse
|
5
|
Lv Y, Xu Y, Liu S, Zeng X, Yang B. Biochanin A Attenuates Psoriasiform Inflammation by Regulating Nrf2/HO-1 Pathway Activation and Attenuating Inflammatory Signalling. Cell Biochem Biophys 2024:10.1007/s12013-024-01595-0. [PMID: 39499389 DOI: 10.1007/s12013-024-01595-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 11/07/2024]
Abstract
Psoriasis is a long-term inflammatory skin condition marked by an overabundance of keratinocytes and the release of pro-inflammatory cytokines in the outer layer of skin. For the comprehensive management of intermediate to advanced psoriasis, innovative biological treatments have been developed. Products for the superficial therapy of mild to moderate psoriasis are still necessary, though. Trifolium pratense contains the isoflavone biochanin A (BCA), which exhibits antiviral, antioxidant, anti-carcinogenic, and anti-inflammatory properties, and helps protect the integrity and function of the endothelium. Although investigations have not shown that BCA is effective in treating psoriasis, it has been shown to slow down the breakdown of the skin barrier by regulating keratinocyte growth. We sought to clarify the basic mechanisms behind BCA's impact on psoriasis in vitro and in vivo using experimental research via regulating Nrf2/HO-1 signaling pathway. By subjecting human primary keratinocytes to psoriasis-related cytokines, psoriasis-like keratinocytes were produced. The CCK8 test was used in this investigation to assess cell viability. BCA reduced keratinocyte growth and inflammatory cascade stimulation produced by TNF-α and IL-6, according to in vitro investigations conducted on HaCaT cells. The in vivo findings showed that six days of BCA therapy significantly decreased the skin, hematological indicators, levels of NO, TBARS, histopathological, and pro-inflammatory factors of COX-2, iNOS, NF-κB pathway. It additionally influenced the protein content of pro-inflammatory cytokines such as IL-17, IL-23, IL-1β in the epidermis along with IL-6, TNF-α among the epidermis and serum. In addition, in contrast to the IMQ group, BCA improved the skin's level of Nrf2/HO-1 protein, anti-inflammatory cytokine IL-10, and antioxidant indicators like SOD, CAT, GST, GSH, GR, and Vit-C. Ultimately, our research shows that BCA was effective in treating psoriasis in pre-clinical animal models by activating the Nrf2/HO-1 pathway, leading to an increase in antioxidant and anti-inflammatory markers.
Collapse
Affiliation(s)
- Yaping Lv
- Department of Dermatology and Venereology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, Shanxi, 030032, China
| | - Yingsheng Xu
- Department of Clinical Nutrition, Ezhou Central Hospital, Ezhou, Hubei, 436000, China
| | - Songchun Liu
- Department of Clinical Nutrition, Ezhou Central Hospital, Ezhou, Hubei, 436000, China
| | - Xianjing Zeng
- General Practice Medicine, Affiliated Hospital of Jinggangshan University, Ji 'an, Jiangxi, 343000, China
| | - Bin Yang
- Department of Dermatology, Affiliated Hospital of Jinggangshan University, Ji 'an, Jiangxi, 343000, China, Jinggangshan University, Ji 'an, Jiangxi, 343009, China.
| |
Collapse
|
6
|
Chen H, Yang G, Xu DE, Du YT, Zhu C, Hu H, Luo L, Feng L, Huang W, Sun YY, Ma QH. Autophagy in Oligodendrocyte Lineage Cells Controls Oligodendrocyte Numbers and Myelin Integrity in an Age-dependent Manner. Neurosci Bull 2024:10.1007/s12264-024-01292-1. [PMID: 39283565 DOI: 10.1007/s12264-024-01292-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/10/2024] [Indexed: 12/08/2024] Open
Abstract
Oligodendrocyte lineage cells, including oligodendrocyte precursor cells (OPCs) and oligodendrocytes (OLs), are essential in establishing and maintaining brain circuits. Autophagy is a conserved process that keeps the quality of organelles and proteostasis. The role of autophagy in oligodendrocyte lineage cells remains unclear. The present study shows that autophagy is required to maintain the number of OPCs/OLs and myelin integrity during brain aging. Inactivation of autophagy in oligodendrocyte lineage cells increases the number of OPCs/OLs in the developing brain while exaggerating the loss of OPCs/OLs with brain aging. Inactivation of autophagy in oligodendrocyte lineage cells impairs the turnover of myelin basic protein (MBP). It causes MBP to accumulate in the cytoplasm as multimeric aggregates and fails to be incorporated into integral myelin, which is associated with attenuated endocytic recycling. Inactivation of autophagy in oligodendrocyte lineage cells impairs myelin integrity and causes demyelination. Thus, this study shows autophagy is required to maintain myelin quality during aging by controlling the turnover of myelin components.
Collapse
Affiliation(s)
- Hong Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Gang Yang
- Lab Center, Medical College of Soochow University, Suzhou, 215021, China
| | - De-En Xu
- The Wuxi No.2 People Hospital, Wuxi, 214002, China
| | - Yu-Tong Du
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Chao Zhu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Hua Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Li Luo
- School of Physical Education and Sports Science, Soochow University, Suzhou, 215021, China
| | - Lei Feng
- Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421, Homburg, Germany
| | - Yan-Yun Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
7
|
Jiang T, Sun L, Wang Y, Zhang F, Guo J, Sun L, Jiang Y, Xue J, Duan J, Liu C. Podophyllotoxin via SIRT1/PPAR /NF-κB axis induced cardiac injury in rats based on the toxicological evidence chain (TEC) concept. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155655. [PMID: 38838636 DOI: 10.1016/j.phymed.2024.155655] [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: 09/14/2023] [Revised: 02/28/2024] [Accepted: 04/17/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND The study of cardiotoxicity of drugs has become an important part of clinical safety evaluation of drugs. It is commonly known that podophyllotoxin (PPT) and its many derivatives and congeners are broad-spectrum pharmacologically active substances. Clinical cardiotoxicity of PPT and its derivatives has been raised, basic research on the mechanism of cardiotoxicity remains insufficient. PURPOSE In present study, our group's innovative concept of toxicological evidence chain (TEC) was applied to reveal the cardiac toxicity mechanism of PPT by targeted metabolomics, TMT-based quantitative proteomics and western blot. METHODS The injury phenotype evidence (IPE) acquired from the toxicity manifestations, such as weight and behavior observation of Sprague-Dawley rat. The damage to rat hearts were assessed through histopathological examination and myocardial enzymes levels, which were defined as Adverse Outcomes Evidence (AOE). The damage to rat hearts was assessed through histopathological examination and myocardial enzyme levels, which were defined as evidence of adverse outcomes.Overall measurements of targeted metabolomics based on energy metabolism and TMT-based quantitative proteomics were obtained after exposure to PPT to acquire the Toxic Event Evidence (TEE). The mechanism of cardiac toxicity was speculated based on the integrated analysis of targeted metabolomics and TMT-based quantitative proteomics, which was verified by western blot. RESULTS The results indicated that exposure to PPT could result in significant elevation of myocardial enzymes and pathological alterations in rat hearts. In addition, we found that PPT caused disorders in cardiac energy metabolism, characterized by a decrease in energy metabolism fuels. TMT-based quantitative proteomics revealed that the PPAR (Peroxisome proliferators-activated receptor) signaling pathway needs further study. It is worth noting that PPT may suppress the expression of SIRT1, subsequently inhibiting AMPK, decreasing the expression of PGC-1α, PPARα and PPARγ. This results in disorders of glucose oxidation, glycolysis and ketone body metabolism. Additionally, the increase in the expression of p-IKK and p-IκBα, leads to the nuclear translocation of NF-κB p65 from the cytosol, thus triggering inflammation. CONCLUSION This study comprehensively evaluated cardiac toxicity of PPT and initially revealed the mechanism of cardiotoxicity,suggesting that PPT induced disorders of energy metabolism and inflammation via SIRT1/PPAR/NF-κB axis, potentially contributing to cardiac injury.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Lu Sun
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong 030600, China
| | - Yuming Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fangfang Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jia Guo
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Lingyun Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Yalin Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Juan Xue
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Jiajia Duan
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
8
|
Wu X, Hu S, Jia N, Zhang C, Liu C, Song J, Kuai L, Jiang W, Li B, Chen Q. Accurate network pharmacology and novel ingredients formula of herbal targeting estrogen signaling for psoriasis intervention. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118099. [PMID: 38554853 DOI: 10.1016/j.jep.2024.118099] [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: 12/30/2023] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a common chronic inflammatory skin disease, psoriasis is incompletely understood and brings a lot of distress to patients. The estrogen signaling pathway has been implicated in its pathogenesis, making it a potential therapeutic target. Si Cao Formula (SCF) has demonstrated promise in treating psoriasis clinically. However, its molecular mechanisms concerning psoriasis remain largely unexplored. AIM OF THE STUDY To elucidate the underlying mechanisms of the action of SCF on psoriasis. MATERIALS AND METHODS Active ingredients were identified by LC-MS/MS. After the treatment with SCF, the exploration of differentially expressed proteins (DEPs) were conducted using tandem mass tag (TMT)-based quantitative proteomics analysis. By GO/KEGG, WikiPathways and network pharmacology, core signaling pathway and protein targets were explored. Consequently, major signaling pathway and protein targets were validated by RT-qPCR, immunoblotting and immunofluorescence. Based on Lipinski's Rule of Five rules and molecular docking, 8 active compounds were identified that acted on the core targets. RESULTS 41 compounds of SCF and 848 specific targets of these compounds were identified. There were 570 DEPs between IMQ (Imiquimod) and IMQ + SCF group, including 279 up-regulated and 304 down-regulated proteins. GO/KEGG, WikiPathways and network pharmacology revealed estrogen signaling pathway as the paramount pathways, through which SCF functioned on psoriasis. We further show novel ingredients formula of SCF contributes to estrogen signaling intervention, including liquiritin, parvisoflavone B, glycycoumarin, 8-prenylluteone, licochalcone A, licochalcone B, oxymatrine, and 13-Hydroxylupanine, where targeting MAP2K1, ILK, HDAC1 and PRKACA, respectively. Molecular docking proves that they have good binding properties. CONCLUSION Our results provide an in-depth view of psoriasis pathogenesis and herbal intervention, which expands our understanding of the systemic pharmacology to reveal the multiple ingredients and multiple targets of SCF and focus on one pathway (estrogen signaling pathway) may be a novel therapeutic strategy for psoriasis treatment of herbal medicine.
Collapse
Affiliation(s)
- Xinxin Wu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Sheng Hu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Ning Jia
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Caiyun Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Changya Liu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jiankun Song
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Le Kuai
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Wencheng Jiang
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| | - Bin Li
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| | - Qilong Chen
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| |
Collapse
|
9
|
Wu X, Zheng Q, Shen F, Song J, Luo Y, Fei X, Jiang W, Xie S, Ma X, Kuai L, Wang R, Ding X, Li M, Luo Y, Li B. The therapeutic efficacy and mechanism action of Si Cao formula in the treatment of psoriasis: A pilot clinical investigation and animal validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117662. [PMID: 38160866 DOI: 10.1016/j.jep.2023.117662] [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: 06/26/2023] [Revised: 12/17/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Psoriasis is a chronic inflammation and relapsing disease that affected approximately 100 million individuals worldwide. In previous clinical study, it was observed that the topical application of Si Cao Formula (SCF) ameliorated psoriasis skin lesions and reduced the recurrence rate of patients over a period of three months. However, the precise mechanism remains unclear. AIM OF THE STUDY The objective of this study was to assess the effectiveness and safety of SCF in patients diagnosed with psoriasis and explore the molecular mechanisms that contribute to SCF's therapeutic efficacy in psoriasis treatment. MATERIALS AND METHODS A randomized, controlled, and pilot clinical study was performed. This study assessed 30 individuals diagnosed with mild to moderate plaque psoriasis. 15 of them underwent local SCF treatment, the others received calcipotriol intervention. The outcome measure focused on Psoriasis Area and Severity Index (PASI), Dermatology Life Quality Index (DLQI), and recurrence rate. In addition, IMQ-induced psoriasis-like mice model were used to assess the impact of SCF on ameliorating epidermal hyperplasia, suppressing angiogenesis, and modulating immune response. Furthermore, we performed bioinformatics analysis on transcriptome data obtained from skin lesions of mice model. This analysis allowed us to identify the targets and signaling pathways associated with the action of SCF. Subsequently, we conducted experimental validation to confirm the core targets. RESULTS Our clinical pilot study demonstrated that SCF could ameliorate skin lesions in psoriasis patients with comparable efficacy of calcipotriol in drop of PASI and DLQI scores. SCF exhibited a significantly reduced recurrence rate within 12 weeks (33.3%). Liquid Chromatography Mass Spectrometry (LC-MS) identified 41 active constituents of SCF (26 cations and 15 anions). Animal experiments showed SCF ameliorates the skin lesions of IMQ-induced psoriasis like mice model and suppresses epidermal hyperkeratosis and angiogenesis. There were 845 up-regulated and 764 down-regulated DEGs between IMQ and IMQ + SCF groups. GO analysis revealed that DEGs were linked to keratinization, keratinocyte differentiation, organic acid transport epidermal cell differentiation, and carboxylic acid transport interferon-gamma production. KEGG pathway analysis showed that SCF may play a vital part through IL-17 and JAK/STAT signaling pathway. In addition, SCF could reduce the number of positive cells expressing PCNA, CD31, pSTAT3, CD3, and F4/80 within the epidermis of psoriatic lesions, as well as the expression of Il-17a and Stat3 in IMQ-induced psoriasis mice. CONCLUSIONS Our research suggests that SCF serves as a reliable and efficient local approach for preventing and treating psoriasis. The discovery of plausible molecular mechanisms and therapeutic targets associated with SCF may support its broad implementation in clinical settings.
Collapse
Affiliation(s)
- Xinxin Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Qi Zheng
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Fang Shen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jiankun Song
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Yue Luo
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xiaoya Fei
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Wencheng Jiang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Shaoqiong Xie
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xin Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ruiping Wang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xiaojie Ding
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Miao Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
10
|
Niu B, Zhao M, Gao X, Xu J, Yu L. TMT-based quantitative proteomics analysis of neuroprotective effects of Forsythoside A on the MPTP-induced Parkinson's disease mouse model. Exp Neurol 2024; 373:114642. [PMID: 38056584 DOI: 10.1016/j.expneurol.2023.114642] [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: 09/29/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder characteristized by the presence of dyskinesia and the progressive loss of dopaminergic neurons. Although certain drugs can mitigate the symptoms of PD, they are unable to delay the disease progression, and their prolonged use may result in complications. Therefore, there exists an urgent necessity to identify potential agents that can effectively delay PD progression with fewer side effects. Recent research has unveiled that several traditional Chinese medicines (TCM) exhibit neuroprotective properties in various models pertinent to PD. Forsythoside A (FSA), the primary bioactive compound derived from TCM Lianqiao, has undergone extensive research in animal models of Alzheimer's disease and cerebral ischemia. However, the investigation into the impact of FSA on PD is limited in existing research. In this study, we aimed to evaluate the neuroprotective effects of FSA on MPTP-induced PD mouse model. FSA demonstrated significant improvements in the behavioral and neuropathological changes triggered by MPTP in mice. Furthermore, it exerted a suppressive effect on the activations of astrocyte and microglia. Meanwhile, Tandem mass tag (TMT)-based quantitative proteomics of striatal tissue and bioinformatics analysis were performed to elucidate the underlying mechanisms of FSA on PD mouse model. Proteomics demonstrated a total of 68 differentially expressed proteins (DEPs) were identified between HFSA and MPTP groups including 26 upregulated and 42 downregulated. Systematic bioinformatics analysis of the 68 DEPs illustrated that they were predominantly related to estrogen signaling pathway and calcium signaling pathway. The related DEPs (PLCβ4, Grm2, HPAC and Cox4i1) expression levels were verified by Western blot. FSA effectively restored the altered expression of the four DEPs induced by MPTP. Summarily, FSA exerted remarkable neuroprotective effects in MPTP-induced mice. Further, our research may provide proteomics insights that contribute to the further exploration of FSA as a potential treatment for PD.
Collapse
Affiliation(s)
- Bo Niu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, China.
| | - Minhong Zhao
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, China.
| | - Xiu'an Gao
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, China.
| | - Jiangping Xu
- School of Pharmaceutical Sciences, Southern Medical University, Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China.
| | - Linzhong Yu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
11
|
Zhang B, Yan G, Li F, Tang Y, Xu G, Zhang Y, Ze K. Qingxiong Ointment and its Active Ingredient, Shikonin Treat Psoriasis through HIF-1 Signaling Pathway. Curr Pharm Des 2024; 30:1927-1938. [PMID: 38835124 DOI: 10.2174/0113816128287142240529120346] [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: 01/10/2024] [Revised: 04/21/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Psoriasis is a common chronic inflammatory skin disorder. Qingxiong ointment (QX) is a natural medicinal combination frequently employed in clinical treatment of psoriasis. However, the active ingredients of QX and its precise mechanisms of improving psoriasis remain unclear. This study elucidated the effects of QX on an Imiquimod (IMQ)-induced mouse model of psoriasis while also exploring the regulation of the active ingredient of QX, shikonin, on the HIF-1 signaling pathway in HaCaT cells. METHODS A mouse model of psoriasis was established through topical application of IMQ, and the local therapeutic effect of QX was evaluated using dorsal skin tissue with mouse psoriatic lesion and Psoriasis Area Severity Index (PASI) scores, hematoxylin-eosin (HE) staining, and immunohistochemical staining. Elisa and qPCR were employed to identify changes in the expression of inflammation-related factors in the mouse dorsal skin. Immunofluorescence was used to assess changes in the expression of T cell subsets before and after treatment with various doses of QX. HPLC was used to analyze the content of shikonin, and network pharmacology was employed to analyze the main targets of shikonin. Immunofluorescence was used to identify the effects of shikonin on the HIF-1 signaling pathway in IL6-induced psoriasis HaCaT cells. Finally, qPCR was used to identify the differential expression of the HIF-1 signaling pathway in skin tissues. RESULTS QX significantly reduces PASI scores on the backs of IMQ-induced psoriasis mice. HE staining reveals alleviated epidermal thickness in the QX group. Immunohistochemical analysis shows a significant reduction in ICAM, KI67, and IL17 expression levels in the QX group. Immunofluorescence results indicate that QX can notably decrease the proportions of CD4+ T cells, γδ T cells, and CD8+ T cells while increasing the proportion of Treg cells. Network pharmacology analysis demonstrates that the main targets of shikonin are concentrated in the HIF-1 signaling pathway. Molecular docking results show favorable binding affinity between shikonin and key genes of the HIF-1 signaling pathway. Immunofluorescence results reveal that shikonin significantly reduces p-STAT3, SLC2A1, HIF1α, and NOS2 expression levels. qPCR results show significant downregulation of the HIF-1 signaling pathway at cellular and tissue levels. CONCLUSION Our study revealed that QX can significantly reduce the dorsal inflammatory response in the IMQ-induced psoriasis mouse model. Furthermore, we discovered that its main component, shikonin, exerts its therapeutic effect by diminishing the HIF-1 signaling pathway in HaCaT cells.
Collapse
Affiliation(s)
- Bin Zhang
- Derpartment of Surgery VIII (Dermatology and Sores), Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Ge Yan
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Fei Li
- Bozhou City Food and Drug Inspection Center, Bozhou 236800, China
| | - Ye Tang
- Derpartment of Surgery VIII (Dermatology and Sores), Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Guangyao Xu
- Derpartment of Surgery VIII (Dermatology and Sores), Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Yanan Zhang
- Derpartment of Surgery VIII (Dermatology and Sores), Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Kan Ze
- Derpartment of Surgery VIII (Dermatology and Sores), Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| |
Collapse
|
12
|
Xiao T, Yu X, Yang L, Duan X. Palmatine treats urticaria by reducing inflammation and increasing autophagy. Front Immunol 2023; 14:1268467. [PMID: 38035098 PMCID: PMC10682667 DOI: 10.3389/fimmu.2023.1268467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Chronic spontaneous urticaria (CSU) is mainly manifested as wheals and erythema on the skin accompanied by itching, which will cause emotional anxiety and seriously affect the quality of life in patients. Palmatine (PAL) is a main chemical component of Yajieshaba, which has been found to effectively alleviate the symptoms of food allergy. However, its role and mechanism in CSU remain unclear. The present study aimed to investigate the protective effect of PAL on CSU rats. Methods We replicated the CSU rat model by intraperitoneal injection of ovalbumin (OVA) in rats on days 0, 2, 4, and 14, with a double dose given on the last challenge. PAL, loratadine and saline were given by gavage from day 5 to day 14. We observed the skin pathologic changes, mast cell degranulation, immune factor levels, inflammatory response and autophagy-related protein expression in CSU rats. Results We found PAL treatment to be effective in alleviating CSU-like skin lesions and reducing itching and mast cell degranulation in rats. Compared with the OVA group, the levels of immune and inflammatory factors were significantly reduced, neutrophil recruitment was alleviated, suggesting a reduced inflammatory response. The autophagy results showed that PAL further increased the expression of LC3, Beclin-1 and p-LKB1, p-AMPK, Atg5, Atg12 and Atg5-Atg12, while P62 and p-p70S6K1 expression decreased. They collectively suggested that autophagic flux was activated after PAL treatment. However, there was an increase in the expression of LC3I, probably due to the fact that PAL induced its accumulation in order to provide substrate for the generation of more LC3II. Discussion Overall, PAL had a protective effect on CSU in normal rats, activated the expression of autophagy and improved the inflammatory response.
Collapse
Affiliation(s)
| | | | | | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
13
|
Chen H, Su Z, Pan X, Zheng X, Li H, Ye Z, Tang B, Lu Y, Zheng G, Lu C. Phytochemicals: Targeting autophagy to treat psoriasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155041. [PMID: 37678054 DOI: 10.1016/j.phymed.2023.155041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Psoriasis is an immune-mediated chronic inflammatory skin disease characterized by well-defined erythema and white scales, which affects approximately 2% of the worldwide population and causes long-term distress to patients. Therefore, development of safe and effective therapeutic drugs is imminent. Autophagy, an evolutionarily conserved catabolic process, degrades intracellular constituents to maintain cellular energy homeostasis. Numerous studies have revealed that autophagy is closely related to immune function, such as removal of intracellular bacteria, inflammatory cytokine secretion, antigen presentation, and lymphocyte development. Phytochemicals derived from natural plants are often used to treat psoriasis due to their unique therapeutic properties and favorable safety. So far, a mass of phytochemicals have been proven to be able to activate autophagy and thus alleviate psoriasis. This review aimed to provide directions for finding phytochemicals that target autophagy to treat psoriasis. METHODS The relevant literatures were collected from classical TCM books and a variety of databases (PubMed, Google Scholar, ScienceDirect, Springer Link, Web of Science and China National Knowledge Infrastructure) till December 2022. Search terms were "Phytochemical", "Psoriasis" and "Autophagy". The retrieved data followed PRISMA criteria (preferred reporting items for systematic review). RESULTS Phytochemicals treat psoriasis mainly through regulating immune cell function, inhibiting excessive inflammatory response, and reducing oxidative stress. While the role and mechanism of autophagy in the pathogenesis of psoriasis have been confirmed in human trials, most of the evidence for phytochemicals that target autophagy to treat psoriasis comes from animal studies. The research focusing on the role of phytochemical-mediated autophagy in the prevention and treatment of psoriasis is limited, and the definite relationship between phytochemical-regulated autophagy and treatment of psoriasis still deserves further experimental confirmation. CONCLUSIONS Phytochemicals with autophagic activities will provide new insights into the therapeutic intervention for psoriasis.
Collapse
Affiliation(s)
- Haiming Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zuqing Su
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xin Pan
- Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xuwei Zheng
- Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Hongxia Li
- Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zeting Ye
- Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bin Tang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yue Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Guangjuan Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Chuanjian Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
14
|
Gao C, Ni B, Lu X, Guo C, Wei G. An integrated investigation of 16S rRNA gene sequencing and proteomics to elucidate the mechanism of Corydalis bungeana Turcz. on dextran sulfate sodium-induced colitis. Biomed Pharmacother 2023; 167:115550. [PMID: 37741254 DOI: 10.1016/j.biopha.2023.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Corydalis bungeana Turcz. (CBT) is frequently used to treat inflammatory illnesses, the mechanisms underlying its use to ulcerative colitis (UC) remain unclear. A dextran sulfate sodium (DSS)-induced UC mice model was established. The disease activity index (DAI), colonic length, histological inspection by hematoxylin-eosin staining, the cytokines levels in the colon, proteomics and intestinal flora in mice were investigated to evaluate the effect of CBT. The results showed that CBT can significantly reduce the DAI, increase the length of colon, improve the pathological injury of colon tissue, decrease the level of TNF-α, IL-6, IL-1β and increase the level of IL-10 in UC mice. Gut microbe sequencing showed that CBT could enhance the abundance of the intestinal microbiome, decrease possibly harmful bacteria and promote potentially helpful microbes. Proteomics investigation showed that 20 overlapping differentially expressed proteins (DEPs) were discovered in the control, model, and CBT administration groups. The DEPs in the CBT administration group were connected to biological procedures mainly involving detoxification. Extracellular matrix (ECM) receptor-associated proteins such as Col6a1 and CD36 may be important targets for CBT treatment of UC. Overall, this integrated methodology identified a comprehensive multi-omics network, composed of a certain set of gut microbiota and proteins, which may be potential targets for CBT treatment with UC.
Collapse
Affiliation(s)
- Chang Gao
- Department of pharmacy, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi, China; Ganzhou Key Laboratory of Immunotherapeutic Drugs Developing for Childhood Leukemia, Ganzhou 341000, Jiangxi, China; Ganzhou Key Laboratory of Antitumor Drugs Developing of Natural Drugs, Ganzhou 341000, Jiangxi, China.
| | - Bin Ni
- Department of pharmacy, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi, China
| | - Xiaolu Lu
- Gannan Medical University, Ganzhou 341000, Jiangxi, China
| | - Chunyu Guo
- Department of pharmacy, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi, China
| | - Guilin Wei
- Department of pharmacy, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi, China.
| |
Collapse
|
15
|
Dai Y, Sun L, Han S, Xu S, Wang L, Ding Y. Proteomic Study on the Reproductive Toxicity of Tripterygium Glycosides in Rats. Front Pharmacol 2022; 13:888968. [PMID: 35668950 PMCID: PMC9163711 DOI: 10.3389/fphar.2022.888968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
Tripterygium glycoside tablet (TGT) is a common clinically used and effective non-steroidal immunosuppressant. However, its reproductive toxicity limits its application in pediatric immune diseases, warranting the study of the molecular mechanism behind its reproductive toxicity. In the present study, 4-week-old male Sprague Dawley (SD) rats were provided TGT through continuous gavage with a clinically equivalent dose of 12 mg/kg for 12 weeks. The reproductive toxicity of TGT was recorded, and its toxicity mechanism was verified through experimental validation and proteomics analyses. Our results demonstrated that TGT could significantly reduce the testosterone level in the serum as well as the concentration and survival rate of sperms. Pathological sections of the testis revealed that TGT could reduce spermatocytes at different levels and make the convoluted meridians vacuolated. Based on tandem mass tag (TMT)-labeled quantitative rats testicular tissue proteomics, 34 differential proteins were screened, involving protein digestion and absorption, PPAR signaling pathway, PI3K-Akt, and other pathways, among which PI3K-Akt plays an important role in the study of reproductive injury. Western blotting results revealed that TGT could significantly downregulate the Col1A1, Col1A2, p-PI3K, and p-Akt expressions and inhibit the expression of proteins related to the PI3K-Akt signaling pathway. In summary, the clinically equivalent dose of TGT induced reproductive toxicity of 4-week-old male SD rats, possibly in relation to the inhibition of the PI3K-Akt pathway expression.
Collapse
Affiliation(s)
- Yanlin Dai
- Pediatric Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Lihui Sun
- Pediatric Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Shanshan Han
- Pediatric Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Shanshan Xu
- Pediatric Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Long Wang
- Pediatric Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ying Ding
- Pediatric Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|