1
|
Fan B, Liu Q, Yang Y, Wu W, Wei Q, Yang J, Hu C, Sun X, Cao P. Soufeng sanjie formula alleviates osteoarthritis by inhibiting macrophage M1 polarization and modulating intestinal metabolites. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119147. [PMID: 39592076 DOI: 10.1016/j.jep.2024.119147] [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: 06/04/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Osteoarthritis (OA) is defined as "bone bi" disease based on clinical symptoms in Chinese medicine. Soufeng sanjie formula (SF) is a traditional formula for treating "bone bi" disease, which consists of Scolopendra (dried body of Scolopendra subspinipes mutilans L. Koch) (0.5 g), Scorpions (dried body of Buthus martensii Karsch) (0.5 g), Astragali radix (dried root of Astragalus membranaceus (Fisch.) Bge) (20 g) and Black soybean seed coats (seed coats of Glycine max (L.) Merr) (30 g), and it can be used to treat rheumatoid arthritis. Nonetheless, the potential of SF to postpone the advancement of OA and its underlying mechanisms remain unexplored. AIM OF THE STUDY This study investigated whether SF could alleviate OA and the underlying mechanisms. MATERIALS AND METHODS Anterior cruciate ligament transection (ACLT) was performed to establish an OA mice model. Mechanical pain and cold pain were assessed to evaluate changes in pain sensitivity in OA mice. Micro-CT was used to observe the microstructure and quantify the bone morphological parameters of knee joints. Safranin O-fast green staining was used to evaluate cartilage damage, and Osteoarthritis Research Society International (OARSI) scores were calculated. Immunohistochemistry was used to assess the expression of inflammatory factors in the synovium of OA mice following SF administration. Immunofluorescence analyzed the fraction of CD80 and iNOS positive regions in the synovium of knee joints. The effect of SF on macrophage M1 polarization was investigated using flow cytometry, western blot and quantitative PCR (qPCR) in vitro. Untargeted metabolomics was used to identify the differential metabolites associated with OA. RESULTS SF-treatment markedly reduced the cartilage damage, lowered the OARSI score and downregulated the pain sensitivity in the OA mice. Secondly, SF decreased the expression of IL-6, IL-1β, and TNF-α in the OA synovium. SF also reduced the percentage of CD80 and iNOS in the synovium of the knee joint after ACLT surgery by immunofluorescence. Thirdly, SF inhibited the protein expression of iNOS and COX-2, decreased the percentage of CD80, and reduced the mRNA levels of IL-6, IL-1β, and TNF-α in BMDM cells. Furthermore, SF inhibited the macrophage M1 polarization-related AKT/NF-κB signaling pathway. Finally, untargeted metabolomics showed that SF effectively reduced the levels of intestinal metabolite 18-hydroxyoleic acid in OA mice. CONCLUSION Our results suggested that SF reduced pain symptoms and joint inflammation in mice with OA. Furthermore, SF inhibited synovial macrophage M1 polarization and modified the levels of the pro-inflammatory intestinal metabolite 18-hydroxyoleic acid in OA mice. Therefore, SF may be act as a potential Chinese medicine for the treatment of OA.
Collapse
Affiliation(s)
- Bo Fan
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Qingyu Liu
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Yan Yang
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Wenhui Wu
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Qingyun Wei
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China; Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China
| | - Jie Yang
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Chunping Hu
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Xiaoyan Sun
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China.
| | - Peng Cao
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China; Shandong Academy of Chinese Medicine, Jinan, 250014, China; Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
2
|
Sun H, Qu Y, Lei X, Xu Q, Li S, Shi Z, Xiao H, Zhang C, Yang Z. Therapeutic Potential of Bee and Wasp Venom in Anti-Arthritic Treatment: A Review. Toxins (Basel) 2024; 16:452. [PMID: 39591207 PMCID: PMC11598298 DOI: 10.3390/toxins16110452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/28/2024] [Accepted: 10/13/2024] [Indexed: 11/28/2024] Open
Abstract
Arthritis has a high global prevalence. During the early ancient human era, bee (Apis) venom therapy was employed in Egypt, Greece, and China to alleviate ailments such as arthritis and neuralgia. In addition, bee venom has long been used as a traditional medicine for immune-related diseases in Korea. Wasp (Vespa) venom is a folk medicine of the Jingpo people in Yunnan, China, and has been widely used to treat rheumatoid arthritis. In spite of this, the underlying mechanisms of bee and wasp venoms for the treatment of arthritis are yet to be fully understood. In recent years, researchers have investigated the potential anti-arthritic properties of bee and wasp venoms. Studies have shown that both bee and wasp venom can improve swelling, pain, and inflammation caused by arthritis. The difference is that bee venom reduces arthritis damage to bone and cartilage by inhibiting the IRAK2/TAK1/NF-κB signaling pathway, NF-κB signaling pathway, and JAK/STAT signaling pathway, as well as decreasing osteoclastogenesis by inhibiting the RANKL/RANK signaling pathway. Wasp venom, on the other hand, regulates synovial cell apoptosis via the Bax/Bcl-2 signaling pathway, inhibits the JAK/STAT signaling pathway to reduce inflammation production, and also ameliorates joint inflammation by regulating redox balance and iron death in synovial cells. This review provides a detailed overview of the various types of arthritis and their current therapeutic approaches; additionally, it comprehensively analyzes the therapeutic properties of bee venom, wasp venom, or venom components used as anti-arthritic drugs and explores their mechanisms of action in anti-arthritic therapy.
Collapse
Affiliation(s)
- Hongmei Sun
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Yunxia Qu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Xiaojing Lei
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Qingzhu Xu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Siming Li
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Zhengmei Shi
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
- National-Local Joint Engineering Research Center of Entomoceutics, Dali 671000, China
| | - Zhibin Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali 671000, China; (H.S.); (Y.Q.); (X.L.); (Q.X.); (S.L.); (Z.S.); (H.X.)
- National-Local Joint Engineering Research Center of Entomoceutics, Dali 671000, China
| |
Collapse
|
3
|
Kimariyo PF, Kurati SP, Bhargavi SNVD, Gordon A, Kayabu D, Muthyala MKK. Synthesized pyrrole ester ameliorates adjuvant‑induced arthritis in Wistar rats by alleviating inflammation and downregulating the pro‑inflammatory cytokines. Inflammopharmacology 2024; 32:2361-2375. [PMID: 38683276 DOI: 10.1007/s10787-024-01470-7] [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: 07/05/2023] [Accepted: 03/27/2024] [Indexed: 05/01/2024]
Abstract
Piperine is an amide alkaloid responsible for producing the pungent smell that comes from black pepper. Piperine has been explained to exhibit significant properties such as anti-rheumatic, anti-inflammatory, and antihypertensive effects. The aim of the study was to synthesize pyrrole ester from piperine and evaluate its anti-arthritis effects in adjuvant-induced arthritis female Wistar rats. In this study, pyrrole ester (AU-5) was designed, synthesized and evaluated for ant-arthritic activity in adjuvant-induced arthritis Wistar rats. The synthesized pyrrole ester (AU-5) was administered in three selected doses (20, 10 and 5 mg/kg) to the arthritic-induced model. The administered ester significantly inhibited the increase in arthritis index, paw and ankle joint swelling compared to the arthritic control group. Similarly, the treated rats exhibited a remarkable increase in body weight increase, improved haematological, biochemical, histopathological and radiological parameters. Moreover, the excess production of rheumatoid factor (RF), C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) was noticeably attenuated in all AU-5-treated rats. However, the spleen index, tumour necrosis factor (TNF-α) and interleukin-6 (IL-6) were distinctly lowered compared to arthritic control rats. Moreover, AU-5 showed promising liver protection by lowering the level of liver function markers Serum glutamic pyruvic transaminase (SGPT), Serum glutamic-oxaloacetic transaminase (SGOT) and alkaline phosphatase (ALP) in serum. Henceforth, it might be concluded that AU-5 has an anti-arthritic effect which can be credited to the down regulation of inflammatory markers and the pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Patrick Francis Kimariyo
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India
- Science and Laboratory Technology Department, Dar es Salaam Institute of Technology (DIT), Dar es Salaam, Tanzania
| | - Sony Priya Kurati
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India
| | | | - Andrew Gordon
- Science Laboratory Technology Department, Accra Technical University, Accra, Ghana
| | - Dickson Kayabu
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India
| | | |
Collapse
|
4
|
Wang X, Sun B, Wang Y, Gao P, Song J, Chang W, Xiao Z, Xi Y, Li Z, An F, Yan C. Research progress of targeted therapy regulating Th17/Treg balance in bone immune diseases. Front Immunol 2024; 15:1333993. [PMID: 38352872 PMCID: PMC10861655 DOI: 10.3389/fimmu.2024.1333993] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
Rheumatoid arthritis (RA) and postmenopausal osteoporosis (PMOP) are common bone-immune diseases. The imbalance between helper (Th17) and regulatory T cells (Tregs) produced during differentiation of CD4+ T cells plays a key regulatory role in bone remodelling disorders in RA and PMOP. However, the specific regulatory mechanism of this imbalance in bone remodelling in RA and PMOP has not been clarified. Identifying the regulatory mechanism underlying the Th17/Treg imbalance in RA and PMOP during bone remodelling represents a key factor in the research and development of new drugs for bone immune diseases. In this review, the potential roles of Th17, Treg, and Th17/Treg imbalance in regulating bone remodelling in RA and PMOP have been summarised, and the potential mechanisms by which probiotics, traditional Chinese medicine compounds, and monomers maintain bone remodelling by regulating the Th17/Treg balance are expounded. The maintenance of Th17/Treg balance could be considered as an therapeutic alternative for the treatment of RA and PMOP. This study also summarizes the advantages and disadvantages of conventional treatments and the quality of life and rehabilitation of patients with RA and PMOP. The findings presented her will provide a better understanding of the close relationship between bone immunity and bone remodelling in chronic bone diseases and new ideas for future research, prevention, and treatment of bone immune diseases.
Collapse
Affiliation(s)
- Xiaxia Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Bai Sun
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yongbin Xi
- Orthopaedics Department, The No.2 People's Hospital of Lanzhou, Lanzhou, Gansu, China
| | - Zhonghong Li
- Pathological Research Centre, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Fangyu An
- Teaching Experiment Training Centre, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Zhang Y, Ma J, Bao X, Hu M, Wei X. The role of retinoic acid receptor-related orphan receptors in skeletal diseases. Front Endocrinol (Lausanne) 2023; 14:1302736. [PMID: 38027103 PMCID: PMC10664752 DOI: 10.3389/fendo.2023.1302736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Bone homeostasis, depending on the balance between bone formation and bone resorption, is responsible for maintaining the proper structure and function of the skeletal system. As an important group of transcription factors, retinoic acid receptor-related orphan receptors (RORs) have been reported to play important roles in bone homeostasis by regulating the transcription of target genes in skeletal cells. On the other hand, the dysregulation of RORs often leads to various skeletal diseases such as osteoporosis, rheumatoid arthritis (RA), and osteoarthritis (OA). Herein, we summarized the roles and mechanisms of RORs in skeletal diseases, aiming to provide evidence for potential therapeutic strategies.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Orthodontics, Hospital of Stomatology Jilin University, Changchun, Jilin, China
| | - Jun Ma
- Department of Oral Anatomy and Physiology, Hospital of Stomatology Jilin University, Changchun, Jilin, China
| | - Xingfu Bao
- Department of Orthodontics, Hospital of Stomatology Jilin University, Changchun, Jilin, China
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology Jilin University, Changchun, Jilin, China
| | - Xiaoxi Wei
- Department of Orthodontics, Hospital of Stomatology Jilin University, Changchun, Jilin, China
| |
Collapse
|
6
|
Li W, Yu L, Li W, Ge G, Ma Y, Xiao L, Qiao Y, Huang W, Huang W, Wei M, Wang Z, Bai J, Geng D. Prevention and treatment of inflammatory arthritis with traditional Chinese medicine: Underlying mechanisms based on cell and molecular targets. Ageing Res Rev 2023; 89:101981. [PMID: 37302756 DOI: 10.1016/j.arr.2023.101981] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/25/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
Inflammatory arthritis, primarily including rheumatoid arthritis, osteoarthritis and ankylosing spondylitis, is a group of chronic inflammatory diseases, whose general feature is joint dysfunction with chronic pain and eventually causes disability in older people. To date, both Western medicine and traditional Chinese medicine (TCM) have developed a variety of therapeutic methods for inflammatory arthritis and achieved excellent results. But there is still a long way to totally cure these diseases. TCM has been used to treat various joint diseases for thousands of years in Asia. In this review, we summarize clinical efficacies of TCM in inflammatory arthritis treatment after reviewing the results demonstrated in meta-analyses, systematic reviews, and clinical trials. We pioneered taking inflammatory arthritis-related cell targets of TCM as the entry point and further elaborated the molecular targets inside the cells of TCM, especially the signaling pathways. In addition, we also briefly discussed the relationship between gut microbiota and TCM and described the role of drug delivery systems for using TCM more accurately and safely. We provide updated and comprehensive insights into the clinical application of TCM for inflammatory arthritis treatment. We hope this review can guide and inspire researchers to further explore mechanisms of the anti-arthritis activity of TCM and make a great leap forward in comprehending the science of TCM.
Collapse
Affiliation(s)
- Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Yong Ma
- Department of Integrated Chinese and Western Medicine, School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, Anhui, China
| | - Wenli Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230031, Anhui, China
| | - Minggang Wei
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhirong Wang
- Translational Medical Innovation Center, Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, Anhui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Orthopedic Institute, Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
7
|
Chen H, Zhang N, Li C, Zhang H. Effects of Astragalus membranaceus on systemic lupus erythematosus in a mouse model of pregnancy. Immun Inflamm Dis 2022; 10:e624. [PMID: 35634952 PMCID: PMC9092001 DOI: 10.1002/iid3.624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/16/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This study used astragalus membranaceus (AM) to treat systemic lupus erythematosus (SLE) model mice during pregnancy, aiming to explore the role of AM in Helper T cell 17 (Th17) differentiation and SLE during pregnancy. METHODS We used lipopolysaccharide to constructed the SLE mouse model. AM decoction given by intragastric administration lasted from the eighth week of the mouse age until the mouse was killed. We estimated the messenger RNA levels of IL-17a and Rorc, counted the Th17 cell number and examined the levels of cytokines including interleukin (IL)-12, tumor necrosis factor α, interferon gamma, IL-17A in mouse serum. Periodic acid-Schiff staining and renal glomerular/tubulointerstitial (TI) score were used to evaluate the progression of lupus nephritis (LN). RESULTS AM treatment improved the conception rate and increased the number and average weight of fetuses in SLE mice. It significantly decreased the urinary albumin/creatinine ratios and reduced the glomerular scores and TI scores in the pregnant SLE mice. AM gavage significantly decreased the weight of spleen, mesenteric lymph node, total splenocytes and T cells, and the expression of proinflammatory factors. Furthermore, AM treatment reduced the ratio of Th17 cells and the expression levels of RORγt and IL-17A. CONCLUSION AM significantly improved pregnancy outcomes and inhibited lupus nephritis during pregnancy in SLE mice.
Collapse
Affiliation(s)
- Hong‐Qing Chen
- Department of ObstetricsHengshui Fourth People's HospitalHengshuiHebeiChina
| | - Na Zhang
- Department of Clinical PharmacyThe Fourth Hospital of ShijiazhuangShijiazhuangHebeiChina
| | - Cai‐Xia Li
- The Fourth Hospital of ShijiazhuangShijiazhuangHebeiChina
| | - Hong‐Xia Zhang
- Department of PharmacyThe Fourth Hospital of ShijiazhuangShijiazhuangHebeiChina
| |
Collapse
|
8
|
Cao L, Xu E, Zheng R, Zhangchen Z, Zhong R, Huang F, Ye J, Sun H, Fan Y, Xie S, Chen Y, Xu Y, Cao J, Cao W, Liu C. Traditional Chinese medicine Lingguizhugan decoction ameliorate HFD-induced hepatic-lipid deposition in mice by inhibiting STING-mediated inflammation in macrophages. Chin Med 2022; 17:7. [PMID: 34983596 PMCID: PMC8728979 DOI: 10.1186/s13020-021-00559-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/18/2021] [Indexed: 11/25/2022] Open
Abstract
Background Stimulator of IFN genes (STING) is highly expressed in the livers of non-alcoholic fatty liver disease (NAFLD) patients and high fat diet (HFD) induced NAFLD mice model. The STING signaling-mediated inflammation has been shown to play a critical role in metabolic disorders. Lingguizhugan decoction (LGZG), a Traditional Chinese herbal decoction, has been applied to treat metabolic disorders for many years. However, whether LGZG can alleviate the progression of NAFLD through inhibiting inflammation remains unclear. This study was to determine the role of STING-mediated inflammation in the HFD-induced hepatic-lipid deposition treated with LGZG. Methods The anti-inflammatory and anti-steatotic effects of LGZG in vivo were detected by H&E staining, immunofluorescence and immuno-chemistry. Mice bone-marrow-derived macrophages (BMDMs) and primary liver macrophages were treated with STING-specific agonist (DMXAA), LGZG and its critical components respectively. The treated culture supernatant of BMDMs and primary liver macrophages from each group was co-cultured with palmitic acid-treated mouse primary hepatocytes or mouse liver cell line AML-12 respectively to detect whether the activation of STING-mediated pathway is involved in the anti-steatotic effect of LGZG. The hepatocyte lipid deposition in vivo and in vitro were detected by oil red staining. Mitochondrial DNA release of mouse liver extracts were detected by real time PCR. The expression of proteins and inflammatory cytokines related to STING-TBK1-NF-κB pathway was detected by western blotting and ELISA. Results LGZG significantly ameliorated HFD induced hepatic steatosis, oxidative stress, hepatic mitochondrial damage and mitochondrial DNA release, which was correlated with reduction of the expression level of STING as well as the infiltration of STING-positive macrophages in the livers of HFD fed mice. The critical components of LGZG directly inhibited the activation of STING-TBK1-NF-κB pathway in liver macrophages induced by DMXAA, LPS, thereby reducing the release of IFNβ and TNFα. Co-incubating the culture supernatant of LGZG treated liver macrophages and PA-stimulated hepatocytes significantly inhibited the PA-induced lipid deposition. Conclusion This study demonstrates that LGZG can ameliorate HFD-induced hepatic-lipid deposition through inhibiting STING-TBK1-NF-κB pathway in liver macrophages, which provides novel insight for elucidating the molecular mechanism of LGZG alleviating HFD induced hepatic steatosis.
Collapse
Affiliation(s)
- Lin Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.
| | - Erjin Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Rendong Zheng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Zhili Zhangchen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Rongling Zhong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Fei Huang
- Suzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No. 18 Yangsu Road, Gusu District, Suzhou, 215002, China
| | - Juan Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Hongping Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Yaofu Fan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Shaofeng Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Yu Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Yijiao Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Jing Cao
- Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Qinhuai District, Nanjing, 210029, China
| | - Wen Cao
- The Affiliated Jiangning Hospital of Nanjing Medical University, No. 169, Dongshan street, Hushan Road, Jiangning District, Nanjing, 211100, China.
| | - Chao Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.
| |
Collapse
|
9
|
Systematic review of robust experimental models of rheumatoid arthritis for basic research. DIGITAL CHINESE MEDICINE 2021. [DOI: 10.1016/j.dcmed.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
10
|
BTN2A2 protein negatively regulates T cells to ameliorate collagen-induced arthritis in mice. Sci Rep 2021; 11:19375. [PMID: 34588505 PMCID: PMC8481265 DOI: 10.1038/s41598-021-98443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/06/2021] [Indexed: 12/05/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by persistent inflammatory responses in target tissues and organs, resulting in the destruction of joints. Collagen type II (CII)-induced arthritis (CIA) is the most used animal model for human RA. Although BTN2A2 protein has been previously shown to inhibit T cell functions in vitro, its effect on autoimmune arthritis has not been reported. In this study, we investigate the ability of a recombinant BTN2A2-IgG2a Fc (BTN2A2-Ig) fusion protein to treat CIA. We show here that administration of BTN2A2-Ig attenuates established CIA, as compared with control Ig protein treatment. This is associated with reduced activation, proliferation and Th1/Th17 cytokine production of T cells in BTN2A2-Ig-treated CIA mice. BTN2A2-Ig also inhibits CII-specific T cell proliferation and Th1/Th17 cytokine production. Although the percentage of effector T cells is decreased in BTN2A2-Ig-treated CIA mice, the proportions of naive T cells and regulatory T cells is increased. Furthermore, BTN2A2-Ig reduces the percentage of proinflammatory M1 macrophages but increases the percentage of anti-inflammatory M2 macrophages in the CIA mice. Our results suggest that BTN2A2-Ig protein has the potential to be used in the treatment of collagen-induced arthritis models.
Collapse
|