1
|
Sawan S, Kumari A, Majie A, Ghosh A, Karmakar V, Kumari N, Ghosh S, Gorain B. siRNA-based nanotherapeutic approaches for targeted delivery in rheumatoid arthritis. BIOMATERIALS ADVANCES 2025; 168:214120. [PMID: 39577366 DOI: 10.1016/j.bioadv.2024.214120] [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: 07/03/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Rheumatoid arthritis (RA), characterized as a systemic autoimmune ailment, predominantly results in substantial joint and tissue damage, affecting millions of individuals globally. Modern treatment modalities are being explored as the traditional RA therapy with non-specific immunosuppressive drugs showcased potential side effects and variable responses. Research potential with small interfering RNA (siRNA) depicted potential in the treatment of RA. These siRNA-based therapies could include genes encoding pro-inflammatory cytokines like TNF-α, IL-1, and IL-6, as well as other molecular targets such as RANK, p38 MAPK, TGF-β, Wnt/Fz complex, and HIF. By downregulating the expression of these genes, siRNA-based nanoformulations can attenuate inflammation, inhibit immune system dysregulation, and prevent tissue damage associated with RA. Strategies of delivering siRNA molecules through nanocarriers could be targeted to treat RA effectively, where specific genes associated with this autoimmune disease pathology can be selectively silenced. Additionally, simultaneous targeting of multiple molecular pathways may offer synergistic therapeutic benefits, potentially leading to more effective and safer therapeutic strategies for RA patients. This review critically highlights the in-depth pathology of RA, RNA interference-mediated molecular targets, and nanocarrier-based siRNA delivery strategies, along with the challenges and opportunities to harbor future solutions.
Collapse
Affiliation(s)
- Sweta Sawan
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankita Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Nimmy Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Santanu Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India.
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India.
| |
Collapse
|
2
|
Song H, Yu J, Yang Y, Zhou L, Liu X, Yu J, Huang Q, Wang S, Zhang X, Liu Y, Zhang D, Meng J, Han T, Li W, Niu X. Exploring molecular mechanism of Panlongqi Tablet (PLQT) against RA: Integrated network pharmacology, molecular docking and experiment validation. Int Immunopharmacol 2025; 144:113639. [PMID: 39616851 DOI: 10.1016/j.intimp.2024.113639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND AND PURPOSE Panlongqi Tablet (PLQT), a proprietary Chinese medicine composed of 29 herbs, has been included in the Chinese Medical Insurance List and has shown promising therapeutic effects on patients with rheumatoid arthritis (RA) in clinical practice. However, the molecular mechanisms of PLQT against RA have not been fully elucidated. This study aimed to further decipher the active ingredients and molecular mechanism of PLQT anti-RA. METHODS A Complete Freund's adjuvant (CFA)-induced rat model was established to evaluate the pharmacodynamic effects of PLQT against RA, the assessment included arthritis index, paw thickness, ankle diameter, morphological and histopathological analysis. Network analysis was used to elucidate the active ingredients and underlying mechanisms of PLQT in the treatment of RA, molecular docking was conducted to assess the binding of active ingredients to key targets. In vitro and in vivo experimental verification were employed to reveal the mechanism of PLQT against RA. RESULTS Experimentally, PLQT improved CFA-induced arthritis without evident side effects. Network analysis revealed that the active ingredients in PLQT were mainly flavonoids, biscoumarin derivatives, alklaloid and lignans. Integrated with molecular docking studies, the molecular mechanisms of PLQT against RA were enriched in inflammatory response, immune regulation, angiogenesis, osteoclast differentiation and autophagy. In vitro experiments confirmed that PLQT exerted anti-inflammatory and immune regulatory effects by targeting the inflammatory response of M1 macrophages and the biological functions of T lymphocytes. In addition, In vivo experiments verified that PLQT could further inhibit synovial angiogenesis to prevent RA. CONCLUSION This study integrated network pharmacology analysis, molecular docking and experimental validation to elucidate the active components of PLQT and its mechanisms in intervening the pathological progression of RA, providing a more comprehensive theoretical basis for the clinical application of PLQT in the treatment of RA.
Collapse
Affiliation(s)
- Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinya Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Jianguo Meng
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
3
|
Xu D, Bai C, Hu R, Li X, Guo F, Zhang D, Shi B. Exploring the Changes in IL-6 and Related Cytokines in Angiogenesis after Tibial Transverse Transplantation in Diabetic Foot Ulcers. Orthop Surg 2024; 16:2181-2190. [PMID: 39223795 PMCID: PMC11572566 DOI: 10.1111/os.14221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE The transverse tibial transfer technique is employed primarily to treat diabetic foot ulcers (DFUs), aiming to enhance leg circulation and promote new blood vessel growth. This technique is also beneficial for various conditions associated with poor blood flow in the lower extremities. However, there is no clear molecular mechanism to explain the relationship between the transverse tibial transfer technique and angiogenesis in patients with diabetic foot. This study aims to preliminarily explore the change of IL-6 and related cytokines in promoting angiogenesis during transverse tibial transplantation, providing a direction for future research. METHODS We retrospectively assessed a study from April 2022 to November 2023 on 76 patients with severe DFUs at Wagner stages 3-4. Flow cytometry was used to detect the levels of 12 cytokines in serum before the operation and 3, 7, 14, 21, and 35 days after the operation. Ankle-brachial index (ABI), transcutaneous oxygen tension (TcPO2), and glycosylated hemoglobin (Hba1c) were recorded at admission and discharge. We examined the variations in cytokine levels, wound healing duration, amputation rates, infection incidence, and other key outcomes. RESULTS In our investigation, a total of 76 individuals participated, comprising 49 males and 27 females. These subjects had an average age of 64.7 years, with a standard deviation of 13 years. The mean ulcer healing time was 74 ± 31 days, amputation occurred in 3 patients, pin tract infection occurred in one patient (1.3%), and incision infection occurred in one patient (1.3%). By day 35 following the surgery, both the ABI and TcPO2 values showed a significant increase from their preoperative levels. HbA1c significantly improved compared with presurgery (p < 0.001), IL-6 levels were significantly increased compared with presurgery (p < 0.05), and then decreased. CONCLUSION The transverse tibial transfer (TTT) technique is safe and efficient for managing DFUs. The wound healing time in patients who smoke or consume alcohol is statistically significant compared with that of nonsmoking and nondrinking patients. IL-6 exhibited substantial changes at various postoperative time points. Future research could investigate the role of IL-6 in tibial transverse translation.
Collapse
Affiliation(s)
- Daofei Xu
- Chengdu Medical CollegeChengduChina
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| | - Chunxia Bai
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| | - Rong Hu
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| | - Xiaoya Li
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| | - Fudie Guo
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| | - Dingwei Zhang
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| | - Bo Shi
- Department of OrthopaedicsMianyang Central HospitalMianyangChina
| |
Collapse
|
4
|
Zhu R, Liao HY, Huang YC, Shen HL. Application of Injectable Hydrogels as Delivery Systems in Osteoarthritis and Rheumatoid Arthritis. Br J Hosp Med (Lond) 2024; 85:1-41. [PMID: 39212571 DOI: 10.12968/hmed.2024.0347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Osteoarthritis and rheumatoid arthritis, though etiologically distinct, are both inflammatory joint diseases that cause progressive joint injury, chronic pain, and loss of function. Therefore, long-term treatment with a focus on relieving symptoms is needed. At present, the primary treatment for arthritis is drug therapy, both oral and intravenous. Although significant progress has been achieved for these treatment methods in alleviating symptoms, certain prominent drawbacks such as the substantial side effects and limited absorption of medications call for an urgent need for improved drug delivery methods. Injected hydrogels can be used as a delivery system to deliver drugs to the joint cavity in a controlled manner and continuously release them, thereby enhancing drug retention in the joint cavity to improve therapeutic effectiveness, which is attributed to the desirable attributes of the delivery system such as low immunogenicity, good biodegradability and biocompatibility. This review summarizes the types of injectable hydrogels and analyzes their applications as delivery systems in arthritis treatment. We also explored how hydrogels counteract inflammation, bone and cartilage degradation, and oxidative stress, while promoting joint cartilage regeneration in the treatment of osteoarthritis (OA) and rheumatoid arthritis (RA). This review also highlights new approaches to developing injectable hydrogels as delivery systems for OA and RA.
Collapse
Affiliation(s)
- Rong Zhu
- Department of Rheumatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yi-Chen Huang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Hai-Li Shen
- Department of Rheumatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Chand Dakal T, Choudhary K, Tiwari I, Yadav V, Kumar Maurya P, Kumar Sharma N. Unraveling the Triad: Hypoxia, Oxidative Stress and Inflammation in Neurodegenerative Disorders. Neuroscience 2024; 552:126-141. [PMID: 38936458 DOI: 10.1016/j.neuroscience.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
The mammalian brain's complete dependence on oxygen for ATP production makes it highly susceptible to hypoxia, at high altitudes or in clinical scenarios including anemia or pulmonary disease. Hypoxia plays a crucial role in the development of various brain disorders, such as Alzheimer's, Parkinson's, and other age-related neurodegenerative diseases. On the other hand, a decrease in environmental oxygen levels, such as prolonged stays at high elevations, may have beneficial impacts on the process of ageing and the likelihood of death. Additionally, the utilization of controlled hypoxia exposure could potentially serve as a therapeutic approach for age-related brain diseases. Recent findings indicate that the involvement of HIF-1α and the NLRP3 inflammasome is of significant importance in the development of Alzheimer's disease. HIF-1α serves as a pivotal controller of various cellular reactions to oxygen deprivation, exerting influence on a multitude of physiological mechanisms such as energy metabolism and inflammatory responses. The NLRP3 plays a crucial role in the innate immune system by coordinating the initiation of inflammatory reactions through the assembly of the inflammasome complex. This review examines the information pertaining to the contrasting effects of hypoxia on the brain, highlighting both its positive and deleterious effects and molecular pathways that are involved in mediating these different effects. This study explores potential strategies for therapeutic intervention that focus on restoring cellular balance and reducing neuroinflammation, which are critical aspects in addressing this severe neurodegenerative condition and addresses crucial inquiries that warrant further future investigations.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Kanika Choudhary
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Isha Tiwari
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India
| | - Vikas Yadav
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India.
| |
Collapse
|
6
|
Ali M, Benfante V, Di Raimondo D, Laudicella R, Tuttolomondo A, Comelli A. A Review of Advances in Molecular Imaging of Rheumatoid Arthritis: From In Vitro to Clinic Applications Using Radiolabeled Targeting Vectors with Technetium-99m. Life (Basel) 2024; 14:751. [PMID: 38929734 PMCID: PMC11204982 DOI: 10.3390/life14060751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disorder caused by inflammation of cartilaginous diarthrodial joints that destroys joints and cartilage, resulting in synovitis and pannus formation. Timely detection and effective management of RA are pivotal for mitigating inflammatory arthritis consequences, potentially influencing disease progression. Nuclear medicine using radiolabeled targeted vectors presents a promising avenue for RA diagnosis and response to treatment assessment. Radiopharmaceutical such as technetium-99m (99mTc), combined with single photon emission computed tomography (SPECT) combined with CT (SPECT/CT), introduces a more refined diagnostic approach, enhancing accuracy through precise anatomical localization, representing a notable advancement in hybrid molecular imaging for RA evaluation. This comprehensive review discusses existing research, encompassing in vitro, in vivo, and clinical studies to explore the application of 99mTc radiolabeled targeting vectors with SPECT imaging for RA diagnosis. The purpose of this review is to highlight the potential of this strategy to enhance patient outcomes by improving the early detection and management of RA.
Collapse
Affiliation(s)
- Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (A.C.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (A.C.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Messina University, 98124 Messina, Italy;
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (A.C.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| |
Collapse
|
7
|
Fan D, Geng Q, Wang B, Wang X, Xia Y, Yang L, Zhang Q, Deng T, Xu Y, Zhao H, Liu B, Lu C, Gu X, Xiao C. Hypoxia-induced ALKBH5 aggravates synovial aggression and inflammation in rheumatoid arthritis by regulating the m6A modification of CH25H. Clin Immunol 2024; 261:109929. [PMID: 38331303 DOI: 10.1016/j.clim.2024.109929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/20/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Previous studies have shown that epigenetic factors are involved in the occurrence and development of rheumatoid arthritis (RA). However, the role of N6-methyladenosine (m6A) methylation in RA has not been determined. The aim of this study was to investigate the role and regulatory mechanisms of hypoxia-induced expression of the m6A demethylase alkB homolog 5 (ALKBH5) in RA fibroblast-like synoviocytes (FLSs). Synovial tissues were collected from RA and osteoarthritis (OA) patients, and RA FLSs were obtained. ALKBH5 expression in RA FLSs and collagen-induced arthritis (CIA) model rats was determined using quantitative reverse transcription-PCR (qRT-PCR), western blotting and immunohistochemistry (IHC). Using ALKBH5 overexpression and knockdown, we determined the role of ALKBH5 in RA FLS aggression and inflammation. The role of ALKBH5 in RA FLS regulation was explored using m6A-methylated RNA sequencing and methylated RNA immunoprecipitation coupled with quantitative real-time PCR. The expression of ALKBH5 was increased in RA synovial tissues, CIA model rats and RA FLSs, and a hypoxic environment increased the expression of ALKBH5 in FLSs. Increased expression of ALKBH5 promoted the proliferation and migration of RA-FLSs and inflammation. Conversely, decreased ALKBH5 expression inhibited the migration of RA-FLSs and inflammation. Mechanistically, hypoxia-induced ALKBH5 expression promoted FLS aggression and inflammation by regulating CH25H mRNA stability. Our study elucidated the functional roles of ALKBH5 and mRNA m6A methylation in RA and revealed that the HIF1α/2α-ALKBH5-CH25H pathway may be key for FLS aggression and inflammation. This study provides a novel approach for the treatment of RA by targeting the HIF1α/2α-ALKBH5-CH25H pathway.
Collapse
Affiliation(s)
- Danping Fan
- China-Japan Friendship Hospital (Department of Emergency, Institute of Clinical Medical Sciences), Beijing 100029, China; Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qishun Geng
- China-Japan Friendship Hospital (Department of Emergency, Institute of Clinical Medical Sciences), Beijing 100029, China; China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Bailiang Wang
- Department of Orthopaedic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xing Wang
- China-Japan Friendship Hospital (Department of Emergency, Institute of Clinical Medical Sciences), Beijing 100029, China; China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ya Xia
- China-Japan Friendship Hospital (Department of Emergency, Institute of Clinical Medical Sciences), Beijing 100029, China; China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liwen Yang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qian Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Tingting Deng
- China-Japan Friendship Hospital (Department of Emergency, Institute of Clinical Medical Sciences), Beijing 100029, China
| | - Yuan Xu
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Hongyan Zhao
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Xiaofeng Gu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Cheng Xiao
- China-Japan Friendship Hospital (Department of Emergency, Institute of Clinical Medical Sciences), Beijing 100029, China.
| |
Collapse
|
8
|
Zhang YJ, Chen LF, Li X, Chen JH, Tan ZK. Tetramethylpyrazine alleviates hypoxia-induced proliferation, migration, and inflammatory response of fibroblast-like synoviocytes via inhibiting the HIF-1α- circCDC42BPB pathway. Adv Rheumatol 2024; 64:19. [PMID: 38449057 DOI: 10.1186/s42358-024-00355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/02/2024] [Indexed: 03/08/2024] Open
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic inflammatory joint disease, which might trigger cartilage, bone damage, and disability. Recent studies have suggested that Tetramethylpyrazine (TMP), an alkaloid monomer isolated from the rhizome of the traditional herbal medicine Ligusticum wallichii Franch, exerts a broad spectrum of pharmacological properties, containing anti-inflammatory. This study aimed to analyze the role and underlying mechanism of TMP in RA. METHODS Under Hypoxia condition, RA-Fibroblast-like synoviocyte (FLS) were treated with TMP at different doses. Cell viability, proliferation, cell cycle progression, and migration were detected using Cell Counting Kit-8 (CCK-8) assay, 5-ethynyl-2'-deoxyuridine (EdU) assay, flow cytometry assay, wound healing assay, and transwell assay. Cyclin D1, Proliferating cell nuclear antigen (PCNA), Matrix metalloproteinase-2 (MMP2), MMP9, and hypoxia-inducible factor-1α (HIF-1α) protein levels were measured using western blot assay. Interleukin-6 (IL-6) and IL-8 were evaluated using ELISA. Circular RNA (circRNA) hsa_circ_0005178 (circCDC42BPB), CDC42BPB, and HIF-1α expression were determined using real-time quantitative polymerase chain reaction (RT-qPCR). Binding between HIF-1α and CDC42BPB promoter was predicted by JASPAR and verified using dual-luciferase reporter and Chromatin immunoprecipitation (ChIP) assays. RESULTS TMP might hinder FLS proliferation, cycle progression, migration, and inflammatory response under hypoxic conditions. CircCDC42BPB expression was increased in RA patients and RA-FLSs treated with hypoxia, while its level was obviously reduced in RA-FLSs treated with hypoxia and TMP. TMP might abolish hypoxia-induced circCDC42BPB expression. Upregulation of circCDC42BPB might partially overturn the repression of TMP on hypoxia-caused RA-FLS damage. TMP might regulate circCDC42BPB level via HIF-1α in RA-FLSs under hypoxic conditions. CONCLUSION TMP might block RA-FLS injury partly via regulating the HIF-1α- circCDC42BPB pathway, providing a promising therapeutic target for RA.
Collapse
Affiliation(s)
- Yu-Jing Zhang
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, Hubei, 430070, China
| | - Li-Feng Chen
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, Hubei, 430070, China.
| | - Xu Li
- Department of Cardiology, Guiqian International General Hospital, No. 1 Dongfeng Avenue, Wudang District, Guiyang, Guizhou, 550018, China
| | - Jian-Hua Chen
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, Hubei, 430070, China
| | - Zhang-Kui Tan
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, Hubei, 430070, China
| |
Collapse
|
9
|
Knowles HJ, Vasilyeva A, Sheth M, Pattinson O, May J, Rumney RMH, Hulley PA, Richards DB, Carugo D, Evans ND, Stride E. Use of oxygen-loaded nanobubbles to improve tissue oxygenation: Bone-relevant mechanisms of action and effects on osteoclast differentiation. Biomaterials 2024; 305:122448. [PMID: 38218121 DOI: 10.1016/j.biomaterials.2023.122448] [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: 07/31/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/15/2024]
Abstract
Gas-loaded nanobubbles have potential as a method of oxygen delivery to increase tumour oxygenation and therapeutically alleviate tumour hypoxia. However, the mechanism(s) whereby oxygen-loaded nanobubbles increase tumour oxygenation are unknown; with their calculated oxygen-carrying capacity being insufficient to explain this effect. Intra-tumoural hypoxia is a prime therapeutic target, at least partly due to hypoxia-dependent stimulation of the formation and function of bone-resorbing osteoclasts which establish metastatic cells in bone. This study aims to investigate potential mechanism(s) of oxygen delivery and in particular the possible use of oxygen-loaded nanobubbles in preventing bone metastasis via effects on osteoclasts. Lecithin-based nanobubbles preferentially interacted with phagocytic cells (monocytes, osteoclasts) via a combination of lipid transfer, clathrin-dependent endocytosis and phagocytosis. This interaction caused general suppression of osteoclast differentiation via inhibition of cell fusion. Additionally, repeat exposure to oxygen-loaded nanobubbles inhibited osteoclast formation to a greater extent than nitrogen-loaded nanobubbles. This gas-dependent effect was driven by differential effects on the fusion of mononuclear precursor cells to form pre-osteoclasts, partly due to elevated potentiation of RANKL-induced ROS by nitrogen-loaded nanobubbles. Our findings suggest that oxygen-loaded nanobubbles could represent a promising therapeutic strategy for cancer therapy; reducing osteoclast formation and therefore bone metastasis via preferential interaction with monocytes/macrophages within the tumour and bone microenvironment, in addition to known effects of directly improving tumour oxygenation.
Collapse
Affiliation(s)
- Helen J Knowles
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Alexandra Vasilyeva
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK; Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Mihir Sheth
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK; Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Oliver Pattinson
- Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Jonathan May
- Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Robin M H Rumney
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| | - Philippa A Hulley
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Duncan B Richards
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Dario Carugo
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Nicholas D Evans
- Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Eleanor Stride
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK; Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Xu XX, Shao H, Wang QX, Wang ZY. Network Pharmacology and Experimental Validation Explore the Pharmacological Mechanisms of Herb Pair for Treating Rheumatoid Arthritis. Comb Chem High Throughput Screen 2024; 27:1808-1822. [PMID: 38213142 DOI: 10.2174/0113862073263839231129163200] [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: 06/23/2023] [Revised: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE This study aimed to elucidate the multitarget mechanism of the Mori Ramulus - Taxilli Herba (MT) herb pair in treating rheumatoid arthritis (RA). METHODS The targets of the herb pair and RA were predicted from databases and screened through cross-analysis. The core targets were obtained using protein-protein interaction (PPI) network analysis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. Finally, animal experiments were conducted to validate the anti-RA effect and mechanism of this herb pair. RESULTS This approach successfully identified 9 active compounds of MT that interacted with 6 core targets (AKT1, TNF, IL6, TP53, VEGFA, and IL1β). Pathway and functional enrichment analyses revealed that MT had significant effects on the TNF and IL-17 signaling pathways. The consistency of interactions between active components and targets in these pathways was confirmed through molecular docking. Moreover, the potential therapeutic effect of MT was verified in vivo, demonstrating its ability to effectively relieve inflammation by regulating these targeted genes and pathways. CONCLUSION The present work suggests that the therapeutic effect of MT herb pair on RA may be attributed to its ability to regulate the TNF signaling pathway and IL-17 signaling pathway.
Collapse
Affiliation(s)
- Xi-Xi Xu
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Qiao-Xue Wang
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Zi-Yuan Wang
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211100, P. R. China
| |
Collapse
|
11
|
Cai L, Meng B, Jiang F, Shu WH, Wang XH, Wang MQ, Wu XJ, Hu MW, Yang YC, Ran X, Li R. Novel HIF-1α Inhibitor AMSP-30m Mitigates the Pathogenic Cellular Behaviors of Hypoxia-Stimulated Fibroblast-Like Synoviocytes and Alleviates Collagen-Induced Arthritis in Rats via Inhibiting Sonic Hedgehog Pathway. Inflammation 2023; 46:2289-2305. [PMID: 37480451 DOI: 10.1007/s10753-023-01878-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/24/2023]
Abstract
Synovial hypoxia-inducible factor 1α (HIF-1α) is a prospective therapeutic target for rheumatoid arthritis (RA). AMSP-30 m, a novel HIF-1α inhibitor, was reported to have notable anti-arthritic effects in rats with adjuvant-induced arthritis. However, its roles in inhibiting the pathogenic behaviors of fibroblast-like synoviocytes (FLS) and the involved mechanisms remain unknown. Here, AMSP-30 m inhibited proliferation and induced apoptosis in hypoxia-induced RA FLS (MH7A cell line), as evidenced by decreased cell viability, reduced Ki67-positive cells, G0/G1 phase arrest, lowered C-myc and Cyclin D1 protein levels, emergence of apoptotic nuclear fragmentation, raised apoptosis rates, and activation of caspase 3. Furthermore, AMSP-30 m prevented hypoxia-induced increases in pro-inflammatory factor production, MMP-2 activity, migration index, migrated/invasive cells, and actin cytoskeletal rearrangement. In vivo, AMSP-30 m alleviated the severity of rat collagen-induced arthritis (CIA). Mechanically, AMSP-30 m reduced HIF-1α expression and blocked sonic hedgehog (Shh) pathway activation in hypoxia-induced MH7A cells and CIA rat synovium, as shown by declines in pathway-related proteins (Shh, Smo, and Gli-1). Particularly, the combination of Shh pathway inhibitor cyclopamine enhanced AMSP-30 m's inhibitory effects on the pathogenic behaviors of hypoxia-stimulated MH7A cells, whereas the combination of Shh pathway activator SAG canceled AMSP-30 m's therapeutic effects in vitro and in CIA rats, implying a close involvement of Shh pathway inhibition in its anti-arthritic effects. We likewise confirmed AMSP-30 m's anti-proliferative role in hypoxia-induced primary CIA FLS. Totally, AMSP-30 m suppressed hypoxia-induced proliferation, inflammation, migration, and invasion of MH7A cells and ameliorated the severity of rat CIA via inhibiting Shh signaling.
Collapse
Affiliation(s)
- Li Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China
| | - Bo Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China
| | - Fei Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China
| | - Wen-Hao Shu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China
| | - Xiao-Hua Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China
| | - Meng-Qing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China
| | - Xin-Jie Wu
- The First Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China
| | - Ming-Wang Hu
- The Second Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China
| | - Yu-Chen Yang
- The First Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China
| | - Xiang Ran
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui Province, People's Republic of China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230026, Anhui Province, People's Republic of China.
| |
Collapse
|
12
|
Xu Q, Kong H, Ren S, Meng F, Liu R, Jin H, Zhang J. Coix seed oil alleviates synovial angiogenesis through suppressing HIF-1α/VEGF-A signaling pathways via SIRT1 in collagen-induced arthritis rats. Chin Med 2023; 18:119. [PMID: 37715217 PMCID: PMC10504826 DOI: 10.1186/s13020-023-00833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by symmetric arthritis. Coix Seed Oil (CSO) has been shown to reduce inflammation in collagen induced arthritis (CIA) rats. However, the effect of CSO on synovial angiogenesis in RA is unknown. In this study, we aimed to explore whether CSO could inhibit RA synovial angiogenesis and elucidate the underlying mechanisms. METHODS CIA rat models were established and subjected to different doses of CSO treatments for four weeks in vivo. Arthritis index, paw swelling, and weight were recorded to assess clinical symptoms. Hematoxylin and Eosin staining, Safarnin O fast green staining, Micro-CT, Immunohistochemical, and Immunofluorescence (IF) staining were performed to examined changes in synovial and joint tissues. The serum HIF-1α and VEGF-A levels were evaluated through enzyme-linked immunosorbent assay. Fibroblast-like synoviocytes (FLS) of rats was stimulated with tumor necrosis factor-α (TNF-α) for developing inflammatory model in vitro. Optimal concentrations of CSO and TNF-α for stimulation were measured through Cell Counting Kit-8 test. Wound healing and Transwell migration experiments were employed to determine FLS migratory ability. IF staining was performed to assess HIF-1α nuclear translocation in FLS. Protein levels of SIRT1, HIF-1α, VEGF-A, and CD31 were assessed through Western blot. The isolated aortic rings were induced with recombinant rat VEGF-A 165 (VEGF-A165) to observe the CSO inhibitory impact on angiogenesis ex vivo. RESULTS CSO attenuated the progression of arthritis in CIA rats, mitigated histopathological deterioration in synovial and joint tissues, significantly inhibited immature vessels labeled with CD31+/αSMA-, and reduced the micro-vessels in VEGF-A165 induced aortic rings. Moreover, it upregulated SIRT1 protein levels in CIA rats and TNF-α induced FLS, but decreased HIF-1α and VEGF-A protein levels. Furthermore, CSO inhibited the migration ability and HIF-1α nuclear translocation of TNF-α induced FLS. Finally, suppressing SIRT1 levels in TNF-α induced FLS enhanced their migration ability, HIF-1α nuclear translocation, and the protein levels of HIF-1α, VEGF-A, and CD31, whereas the inhibitory effect of CSO on TNF-α induced FLS was severely constrained. CONCLUSIONS This study indicates that CSO can alleviate synovial angiogenesis through suppressing HIF-1α/VEGF-A signaling pathways via SIRT1 in CIA rats.
Collapse
Affiliation(s)
- Qiangqiang Xu
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Hongxi Kong
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Shuang Ren
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Fanyan Meng
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Ruoshi Liu
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Hongxin Jin
- Guangzhou University of Traditional Chinese Medicine, Guangdong, 510006, China
| | - Jie Zhang
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China.
| |
Collapse
|
13
|
Chen Z, Xu J, Sui J, Dai H. Expressions of Peptidoglycan Recognition Protein 1, Neuron Towards Axon Guidance Factor-1 and miR-142-3p and Their Correlations in Patients with Rheumatoid Arthritis. Int J Gen Med 2023; 16:3457-3464. [PMID: 37601805 PMCID: PMC10439792 DOI: 10.2147/ijgm.s418396] [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: 05/09/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Purpose We aimed to explore the expressions of peptidoglycan recognition protein 1 (PGLYRP-1), neuron towards axon guidance factor-1 (Netrin-1) and miR-142-3p and their correlations in patients with rheumatoid arthritis. Patients and Methods Sixty patients with rheumatoid arthritis treated from January 2022 to January 2023 were enrolled as a rheumatoid group, 30 patients with osteoarthritis were selected as an osteoarthritis group, and 30 healthy volunteers were recruited as a control group. The enzyme-linked immunosorbent assay, Western blotting and reverse transcriptase-polymerase chain reaction were employed to measure the expressions of PGLYRP-1, Netrin-1 and miR-142-3p, respectively. The correlations among PGLYRP-1, Netrin-1 and miR-142-3p expressions in patients with rheumatoid arthritis were analyzed. Results In patients with rheumatoid arthritis, PGLYRP-1 expression was negatively correlated with Netrin-1 expression (r=-0.570, P=0.001) but positively correlated with miR-142-3p expression (r=0.599, P=0.001), and a negative correlation was found between Netrin-1 and miR-142-3p expressions (r=-0.468, P=0.001). The combined detection of PGLYRP-1, Netrin-1 and miR-142-3p was more sensitive and less specific for predicting the prognosis of patients with rheumatoid arthritis than the measurement of a single marker (P<0.05). Conclusion The combined measurement of PGLYRP-1, Netrin-1 and miR-142-3p has a predictive value for the prognosis of patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- Zhijian Chen
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Juanjuan Xu
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Jingzhe Sui
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Hai Dai
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| |
Collapse
|
14
|
Sakyi SA, Buckman TA, Yeboah‐Mensah K, Senu E, Effah A, Antwi‐Berko D, Dey D, Antwi MH, Yorke J, Boateng AO, Addei AM, Tanko MM, Boateng R. Receptors expressions on peripheral lymphocytes and CD4 + CD183 + as a diagnostics biomarker for rheumatoid arthritis: A case-control study in Ghana. Immun Inflamm Dis 2023; 11:e976. [PMID: 37647423 PMCID: PMC10465995 DOI: 10.1002/iid3.976] [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: 02/26/2023] [Revised: 05/15/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND T cell receptors play important roles in the development and progression of rheumatoid arthritis (RA). Their involvement has been reported in inflammatory autoimmune diseases. However, their role in predicting RA is still under exploration. This study evaluated the expression of CD183 (CXCR3) receptors on T-cells and other relevant biomarkers for detecting RA and determine their relationship with disease activity. METHODS This unmatched case-control study included 48 newly diagnosed RA patients and 30 apparent healthy controls from the orthopedic units of Komfo Anokye Teaching Hospital (KATH), Kumasi and Korle-Bu Teaching Hospital (KBTH), Accra, Ghana. Sociodemographic data was obtained, and blood samples were also collected and processed for flow cytometric analysis. Statistical analyses were done using SPSS version 26.0 and R programming language. p < .05 was considered statistically significant. RESULTS This study found a significant difference in age group (p < .0001), marital status (p = .0210), occupation (p = .0140), educational level (p = .0210) and religion (p = .0100) between RA patients and healthy controls. Moreover, hemoglobin level (p = .0010), waist circumference (p < .0001) and hip circumference (p = .0040) were significantly different between RA patients and controls. RA patients had significantly lower levels of CD4+ CD183+ compared with the control group (p < .001), and was positively correlated with DAS score (r = .0397, p = .789). In Receiver Operator Characteristics analysis, CD4+ CD183+ could significantly detect RA with a high area under the curve (AUC = 0.687, p = .018). At a cut-off of 0.082, CD4+ CD183+ was the best receptor biomarker for detecting RA with a sensitivity of 90.0%, specificity of 25.9%, a positive predictive value of 69.2%, and a negative predictive value of 58.3%. CONCLUSION CD4+ CD183+ best predict RA and is positively correlated with disease activity. CD4+ CD183+ could serve as diagnostics and disease-monitoring biomarker for RA; however, it demonstrates low specificity. Future studies should be directed on CD4+ CD183+ and other biomarkers to augment their diagnostics performances and routine management in RA.
Collapse
Affiliation(s)
- Samuel Asamoah Sakyi
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiAshanti RegionGhana
| | - Tonnies Abeku Buckman
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiAshanti RegionGhana
- Department of Medical Laboratory ScienceUniversity of Energy and Natural ResourcesSunyaniGhana
- Department of Medical Laboratory SciencesKAAF University CollegeAccraGhana
| | - Kwame Yeboah‐Mensah
- Department of Medicine, School of Medicine and Dentistry, Komfo Anokye Teaching HospitalKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Ebenezer Senu
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiAshanti RegionGhana
| | - Alfred Effah
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiAshanti RegionGhana
| | - Daniel Antwi‐Berko
- Neurochemistry Laboratory, Department of Clinical ChemistryVU University Medical Center (VUmc)AmsterdamThe Netherlands
| | - Dzifa Dey
- Department of Medicine and Therapeutics, Korle‐Bu Teaching HospitalUniversity of Ghana Medical SchoolAccraGhana
| | - Maxwell H. Antwi
- Department of Medical Laboratory SciencesKoforidua Technical UniversityKoforiduaGhana
| | - Joseph Yorke
- Department of Surgery, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
- Directorate of SurgeryKomfo Anokye Teaching HospitalKumasiGhana
| | - Andy O. Boateng
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiAshanti RegionGhana
| | - Akwasi M. Addei
- Department of Biological SciencesKwame Nkrumah University of Science and TechnologyKumasiAshanti RegionGhana
| | - Muniru M. Tanko
- Department of Immunology and ImmunodiagnosticsUniversity for Development StudiesTamaleNorthern RegionGhana
| | - Richard Boateng
- Department of Clinical MicrobiologyKomfo Anokye Teaching HospitalKumasiAshanti RegionGhana
| |
Collapse
|
15
|
Häupl T, Sörensen T, Smiljanovic B, Darcy M, Scheder-Bieschin J, Steckhan N, Hartmann AM, Koppold DA, Stuhlmüller B, Skriner K, Walewska BM, Hoppe B, Bonin M, Burmester GR, Schendel P, Feist E, Liere K, Meixner M, Kessler C, Grützkau A, Michalsen A. Intestinal Microbiota Reduction Followed by Fasting Discloses Microbial Triggering of Inflammation in Rheumatoid Arthritis. J Clin Med 2023; 12:4359. [PMID: 37445394 DOI: 10.3390/jcm12134359] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/03/2023] [Accepted: 06/09/2023] [Indexed: 07/15/2023] Open
Abstract
Rheumatoid arthritis (RA) synovitis is dominated by monocytes/macrophages with inflammatory patterns resembling microbial stimulation. In search of triggers, we reduced the intestinal microbiome in 20 RA patients (open label study DRKS00014097) by bowel cleansing and 7-day fasting (≤250 kcal/day) and performed immune monitoring and microbiome sequencing. Patients with metabolic syndrome (n = 10) served as a non-inflammatory control group. Scores of disease activity (DAS28/SDAI) declined within a few days and were improved in 19 of 20 RA patients after breaking the fast (median ∆DAS28 = -1.23; ∆SDAI = -43%) or even achieved remission (DAS28 < 2.6/n = 6; SDAI < 3.3/n = 3). Cytometric profiling with 46 different surface markers revealed the most pronounced phenomenon in RA to be an initially increased monocyte turnover, which improved within a few days after microbiota reduction and fasting. Serum levels of IL-6 and zonulin, an indicator of mucosal barrier disruption, decreased significantly. Endogenous cortisol levels increased during fasting but were insufficient to explain the marked improvement. Sequencing of the intestinal microbiota indicated that fasting reduced potentially arthritogenic bacteria and changed the microbial composition to species with broader metabolic capabilities. More eukaryotic, predominantly fungal colonizers were observed in RA, suggesting possible involvement. This study demonstrates a direct link between the intestinal microbiota and RA-specific inflammation that could be etiologically relevant and would support targeted nutritional interventions against gut dysbiosis as a causal therapeutic approach.
Collapse
Affiliation(s)
- Thomas Häupl
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Department of Rheumatology, Helios Fachklinik Vogelsang-Gommern GmbH, 39245 Gommern, Germany
| | - Till Sörensen
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Biljana Smiljanovic
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Marine Darcy
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Justus Scheder-Bieschin
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Nico Steckhan
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Anika M Hartmann
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Daniela A Koppold
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Department of Internal and Integrative Medicine, Immanuel Hospital Berlin, 14109 Berlin, Germany
- Department of Pediatrics, Division of Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin and Berlin Institute of Health, 10117 Berlin, Germany
| | - Bruno Stuhlmüller
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Karl Skriner
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Barbara M Walewska
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Berthold Hoppe
- Institute of Laboratory Medicine, Unfallkrankenhaus Berlin, 12683 Berlin, Germany
| | - Marc Bonin
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Pascal Schendel
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Eugen Feist
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Department of Rheumatology, Helios Fachklinik Vogelsang-Gommern GmbH, 39245 Gommern, Germany
| | - Karsten Liere
- Amedes Genetics, 10117 Berlin, Germany
- Services in Molecular Biology GmbH, 10115 Rüdersdorf, Germany
| | - Martin Meixner
- Amedes Genetics, 10117 Berlin, Germany
- Services in Molecular Biology GmbH, 10115 Rüdersdorf, Germany
| | - Christian Kessler
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Department of Internal and Integrative Medicine, Immanuel Hospital Berlin, 14109 Berlin, Germany
| | | | - Andreas Michalsen
- Institute of Social Medicine, Epidemiology and Health Economics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, 10117 Berlin, Germany
- Department of Internal and Integrative Medicine, Immanuel Hospital Berlin, 14109 Berlin, Germany
| |
Collapse
|
16
|
Phua TJ. Understanding human aging and the fundamental cell signaling link in age-related diseases: the middle-aging hypovascularity hypoxia hypothesis. FRONTIERS IN AGING 2023; 4:1196648. [PMID: 37384143 PMCID: PMC10293850 DOI: 10.3389/fragi.2023.1196648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 06/30/2023]
Abstract
Aging-related hypoxia, oxidative stress, and inflammation pathophysiology are closely associated with human age-related carcinogenesis and chronic diseases. However, the connection between hypoxia and hormonal cell signaling pathways is unclear, but such human age-related comorbid diseases do coincide with the middle-aging period of declining sex hormonal signaling. This scoping review evaluates the relevant interdisciplinary evidence to assess the systems biology of function, regulation, and homeostasis in order to discern and decipher the etiology of the connection between hypoxia and hormonal signaling in human age-related comorbid diseases. The hypothesis charts the accumulating evidence to support the development of a hypoxic milieu and oxidative stress-inflammation pathophysiology in middle-aged individuals, as well as the induction of amyloidosis, autophagy, and epithelial-to-mesenchymal transition in aging-related degeneration. Taken together, this new approach and strategy can provide the clarity of concepts and patterns to determine the causes of declining vascularity hemodynamics (blood flow) and physiological oxygenation perfusion (oxygen bioavailability) in relation to oxygen homeostasis and vascularity that cause hypoxia (hypovascularity hypoxia). The middle-aging hypovascularity hypoxia hypothesis could provide the mechanistic interface connecting the endocrine, nitric oxide, and oxygen homeostasis signaling that is closely linked to the progressive conditions of degenerative hypertrophy, atrophy, fibrosis, and neoplasm. An in-depth understanding of these intrinsic biological processes of the developing middle-aged hypoxia could provide potential new strategies for time-dependent therapies in maintaining healthspan for healthy lifestyle aging, medical cost savings, and health system sustainability.
Collapse
Affiliation(s)
- Teow J. Phua
- Molecular Medicine, NSW Health Pathology, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
17
|
Xu C, Jiang Y, Wang H, Zhang Y, Ye Y, Qin H, Gao J, Dan Q, Du L, Liu L, Peng F, Li Y, Tu Y. Arthritic Microenvironment Actuated Nanomotors for Active Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204881. [PMID: 36373692 PMCID: PMC9896045 DOI: 10.1002/advs.202204881] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/05/2022] [Indexed: 05/20/2023]
Abstract
Increasing O2 demand and excessive ROS production are the main features of arthritic microenvironment in rheumatoid arthritis (RA) joints and further play pivotal roles in inflammation exacerbation. In this work, a system of in situ regulation of arthritic microenvironment based on nanomotor strategy is proposed for active RA therapy. The synthesized MnO2 -motors enable catalytic regulation of RA microenvironment by consuming the overproduced H2 O2 and generating O2 synergistically. The generated O2 under H2 O2 -rich conditions functions as inflammation detector, propellant for enhanced diffusion, as well as ameliorator for the hypoxic synovial microenvironment. Owing to O2 generation and inflammation scavenging, the MnO2 -motors block the re-polarization of pro-inflammatory macrophages, which results in significantly decreased secretion of multiple pro-inflammatory cytokines both in vitro and in vivo. In addition, intra-articular administration of MnO2 -motors to collagen-induced arthritis rats (CIA rats) effectively alleviates hypoxia, synovial inflammation, bone erosion, and cartilage degradation in joints. Therefore, the proposed arthritic regulation strategy shows great potential to seamlessly integrate basic research of RA with clinical translation.
Collapse
Affiliation(s)
- Cong Xu
- Department of Medicine UltrasonicsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Yuejun Jiang
- Department of Medicine UltrasonicsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Hong Wang
- Department of Medicine UltrasonicsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Yuxin Zhang
- Department of UltrasoundFirst Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510120China
| | - Yicheng Ye
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Hanfeng Qin
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Junbin Gao
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Qing Dan
- Department of Medicine UltrasonicsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Lingli Du
- Department of Medicine UltrasonicsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Lu Liu
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Fei Peng
- School of Materials Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Yingjia Li
- Department of Medicine UltrasonicsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yingfeng Tu
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
18
|
Hypoxia and TNF-α Synergistically Induce Expression of IL-6 and IL-8 in Human Fibroblast-like Synoviocytes via Enhancing TAK1/NF-κB/HIF-1α Signaling. Inflammation 2023; 46:912-924. [PMID: 36607540 DOI: 10.1007/s10753-022-01779-x] [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: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
Hypoxia and increased levels of inflammatory cytokines in the joints are characteristics of rheumatoid arthritis (RA). However, the effects of hypoxia and tumor necrosis factor-α (TNF-α) on interleukin (IL)-6 and IL-8 production on fibroblast-like synoviocytes (FLSs) remain to be clarified. This study aimed to explore how hypoxia and TNF-α affect the expression of IL-6 and IL-8 in human FLSs isolated from RA patients. Hypoxia or TNF-α treatment alone significantly increased the expression and promoter activity of IL-6, IL-8, and hypoxia-inducible factor-1α (HIF-1α). Treatment of hypoxic FLSs with TNF-α further significantly elevated the expression of these cytokines and enhanced promoter activity of HIF-1α, which was abrogated by treatment with the HIF-1α inhibitor YC-1. Similarly, TNF-α alone elevated the phosphorylation and promoter activity of nuclear factor-κBp65 (NF-κBp65) in the FLSs. These effects were further enhanced by the combined treatment of hypoxia and TNFα but were attenuated by the NF-κB inhibitor BAY11-7082. NF-κB-p65 inhibition decreased the effect of TNF-α on HIF-1α upregulation in the FLSs in response to hypoxia. The combination of hypoxia and TNF-α also significantly upregulated transforming growth factor-β-activated kinase 1 (TAK1) expression, and silencing TAK1 dramatically decreased NF-κB-p65, HIF-1α, IL-6, and IL-8 expression under the same conditions. Our results indicate that hypoxia and TNF-α synergistically increase IL-6 and IL-8 expression in human FLSs via enhancing TAK1/NF-κB/HIF-1α signaling.
Collapse
|
19
|
Wang D, Liu Y, Diao S, Shan L, Zhou J. Long Non-Coding RNAs Within Macrophage-Derived Exosomes Promote BMSC Osteogenesis in a Bone Fracture Rat Model. Int J Nanomedicine 2023; 18:1063-1083. [PMID: 36879890 PMCID: PMC9985426 DOI: 10.2147/ijn.s398446] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
Purpose To investigate the effect of macrophage exosomal long non-coding (lnc)RNAs on bone mesenchymal stem cell (BMSC) osteogenesis and the associated mechanism. Methods Rat BMSCs and spleen macrophages were co-cultured with serum derived from the fracture microenvironment of rat tibia. BMSC osteogenesis was evaluated using Alizarin red staining and the expression of BMP-2, RUNX2, OPN, and OC mRNA. BMSC osteogenesis was evaluated after co-culture with macrophages stimulated using hypoxic conditions or colony-stimulating factor (CSF). The uptake of macrophage-derived exosomes by BMSCs was evaluated using the exosome uptake assay. High-throughput sequencing and bioinformatics analyses were performed to identify key lncRNAs in the macrophage exosomes. The effect of lncRNA expression levels on BMSC osteogenesis was also assessed using a lncRNA overexpression plasmid and siRNA technology. M1 and M2 macrophages were distinguished using flow cytometry and the key exosomal lncRNA was detected by in situ hybridization. Results In the fracture microenvironment, macrophages (stimulated using either hypoxia or CSF) significantly increased the osteogenic ability of BMSCs. We showed that BMSCs assimilated macrophage-derived vesicles and that the inhibition of exosomal secretion significantly attenuated the macrophage-mediated induction of BMSC osteogenesis. The hypoxia condition led to the up-regulation of 310 lncRNAs and the down-regulation of 575 lncRNAs in macrophage exosomes, while CSF stimulation caused the up-regulation of 557 lncRNAs and the down-regulation of 407 lncRNAs. In total, 108 lncRNAs were co-up-regulated and 326 lncRNAs were co-down-regulated under both conditions. We eventually identified LOC103691165 as a key lncRNA that promoted BMSC osteogenesis and was expressed at similar levels in both M1 and M2 macrophages. Conclusion In the fracture microenvironment, M1 and M2 macrophages promoted BMSC osteogenesis by secreting exosomes containing LOC103691165.
Collapse
Affiliation(s)
- Dong Wang
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Yang Liu
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Shuo Diao
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Lei Shan
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Junlin Zhou
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| |
Collapse
|
20
|
Hong Z, Tie Q, Zhang L. Targeted inhibition of the GRK2/HIF-1α pathway is an effective strategy to alleviate synovial hypoxia and inflammation. Int Immunopharmacol 2022; 113:109271. [PMID: 36461590 DOI: 10.1016/j.intimp.2022.109271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
G-protein coupled receptor (GPCR) kinases (GRKs) and hypoxia-inducible factor-1α (HIF-1α) play key roles in rheumatoid arthritis (RA). Several studies have demonstrated that HIF-1α expression is positively regulated by GRK2, suggesting its posttranscriptional effects on HIF-1α. In this study, we review the role of HIF-1α and GRK2 in RA pathophysiology, focusing on their proinflammatory roles in immune cells and fibroblast-like synoviocytes (FLS).We then introduce several drugs that inhibit GRK2 and HIF-1α, and briefly outline their molecular mechanisms. We conclude by presenting gaps in knowledge and our prospects for the pharmacological potential of targeting these proteins and the relevant downstream signaling pathways.Future research is warranted and paramount for untangling these novel and promising roles for GRK2 and HIF-1α in RA.
Collapse
Affiliation(s)
- Zhongyang Hong
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China.
| | - Qingsong Tie
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
21
|
Liu X, Wang Z, Qian H, Tao W, Zhang Y, Hu C, Mao W, Guo Q. Natural medicines of targeted rheumatoid arthritis and its action mechanism. Front Immunol 2022; 13:945129. [PMID: 35979373 PMCID: PMC9376257 DOI: 10.3389/fimmu.2022.945129] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease involving joints, with clinical manifestations of joint inflammation, bone damage and cartilage destruction, joint dysfunction and deformity, and extra-articular organ damage. As an important source of new drug molecules, natural medicines have many advantages, such as a wide range of biological effects and small toxic and side effects. They have become a hot spot for the vast number of researchers to study various diseases and develop therapeutic drugs. In recent years, the research of natural medicines in the treatment of RA has made remarkable achievements. These natural medicines mainly include flavonoids, polyphenols, alkaloids, glycosides and terpenes. Among them, resveratrol, icariin, epigallocatechin-3-gallate, ginsenoside, sinomenine, paeoniflorin, triptolide and paeoniflorin are star natural medicines for the treatment of RA. Its mechanism of treating RA mainly involves these aspects: anti-inflammation, anti-oxidation, immune regulation, pro-apoptosis, inhibition of angiogenesis, inhibition of osteoclastogenesis, inhibition of fibroblast-like synovial cell proliferation, migration and invasion. This review summarizes natural medicines with potential therapeutic effects on RA and briefly discusses their mechanisms of action against RA.
Collapse
Affiliation(s)
- Xueling Liu
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhiguo Wang
- Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Hua Qian
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang City, China
| | - Wenhua Tao
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang City, China
| | - Ying Zhang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chunyan Hu
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Weiwei Mao
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qi Guo
- School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Qi Guo,
| |
Collapse
|
22
|
Zeng H, Zhou K, Ye Z. Biology of interleukin‑37 and its role in autoimmune diseases (Review). Exp Ther Med 2022; 24:495. [PMID: 35837057 PMCID: PMC9257848 DOI: 10.3892/etm.2022.11422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 05/10/2022] [Indexed: 11/06/2022] Open
Abstract
Autoimmune diseases (AIDs) are characterized by dysfunction and tissue destruction, and recent studies have shown that interleukin (IL)-37 expression is dysregulated in AIDs. Among cytokines of the IL-1 family, most are pro-inflammatory agents, and as an anti-inflammatory cytokine, IL-37 may have the potential to alleviate excessive inflammation and can be used as a ligand or transcription factor that is involved in regulating innate and adaptive immunity. IL-37 plays important roles in the development of AIDs. This review summarizes the biological characteristics and functions of IL-37 and discusses the potential of IL-37 as a therapeutic target for effective cytokine therapy and as a biomarker in AIDs.
Collapse
Affiliation(s)
- Huiqiong Zeng
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong 518040, P.R. China
| | - Kaixia Zhou
- School of Biomedical Sciences, CUHK‑GIBH CAS Joint Laboratory on Stem Cell and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, P.R. China
| | - Zhizhong Ye
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong 518040, P.R. China
| |
Collapse
|