1
|
Liu Y, Kong X, Sun Q, Cui T, Xu S, Ding C. Identification and validation of the common pathogenesis and hub biomarkers in Papillary thyroid carcinoma complicated by rheumatoid arthritis. PLoS One 2025; 20:e0317369. [PMID: 40063597 PMCID: PMC11892850 DOI: 10.1371/journal.pone.0317369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/19/2024] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Papillary thyroid carcinoma coexisting with rheumatoid arthritis is frequently observed in clinical patients, yet its pathogenesis has not been fully elucidated. This investigation sought to further explore the molecular underpinnings of these two diseases. METHODS Gene expression profiles for thyroid papillary carcinoma and rheumatoid arthritis patients were obtained from the Comprehensive Gene Expression Database (GEO). Following the discovery of shared differentially expressed genes (DEGs) between these two conditions, three separate analyses were conducted. These included functional annotation, the establishment of a protein‒protein interaction (PPI) network and module, and the identification of hub genes via coexpression analysis. The final step involved the validation of target genes via clinical specimens. RESULTS This study analyzed datasets from four GEO databases and identified 64 common DEGs. Functional enrichment analysis revealed that these genes are predominantly associated with pathways related to immunity and signal transduction. Protein‒protein interaction (PPI) network analysis revealed complex interactions among these differentially expressed genes and highlighted several genes that may play pivotal roles in shared pathological mechanisms, namely, CCR5, CD4, IL6, CXCL13, FOXM1, CXCL9, and CXCL10. CONCLUSION Our study highlights the shared pathogenesis between papillary thyroid cancer and rheumatoid arthritis. Shared pathways and crucial genes could offer novel perspectives for subsequent investigations into the mechanisms of these diseases.
Collapse
MESH Headings
- Humans
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/complications
- Thyroid Cancer, Papillary/pathology
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/complications
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Protein Interaction Maps/genetics
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/complications
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Databases, Genetic
- Transcriptome
Collapse
Affiliation(s)
- Yingming Liu
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangjun Kong
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qianshu Sun
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianxing Cui
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shengnan Xu
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chao Ding
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
2
|
Liu W, Tao Z, Liang R, Hu X. Castleman disease complicated by rheumatoid arthritis and postoperative chylous leakage: A case report. Medicine (Baltimore) 2025; 104:e41559. [PMID: 39960936 PMCID: PMC11835113 DOI: 10.1097/md.0000000000041559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025] Open
Abstract
RATIONALE Castleman disease (CD) is a rare disorder characterized by nonspecific clinical presentations and imaging findings, making it prone to misdiagnosis and missed diagnosis. This report details the diagnosis and treatment process of a patient with CD complicated by rheumatoid arthritis (RA) who developed chylous leakage postoperatively, highlighting the potential risks of infection and nutritional depletion, aiming to enhance the understanding of CD. PATIENT CONCERNS A patient with CD complicated by RA developed chylous leakage postoperatively, which posed risks of infection and nutritional depletion. DIAGNOSES The patient underwent a left axillary lymph node excisional biopsy on November 13, 2019, and was diagnosed with HV-CD based on pathology. INTERVENTIONS No further axillary lymph node dissection or adjuvant therapy was performed. Postoperatively, the patient developed significant chylous leakage from the biopsy incision, which resolved after 2 weeks of drainage, dietary modifications, and anti-infective treatment. OUTCOMES Follow-up showed no disease progression, and the chylous leakage resolved with appropriate management. LESSONS A history of RA may be associated with the onset of CD. HV-CD generally has a favorable prognosis, and chylous leakage post-axillary lymph node biopsy, though rare, can be effectively managed with appropriate drainage, dietary control, and infection prevention.
Collapse
Affiliation(s)
- Wei Liu
- Department of Breast, Jinan University Affiliated Guangzhou Red Cross Hospital: Guangzhou Red Cross Hospital, Guangzhou, Guangdong, China
| | - Zhuoyan Tao
- Department of Breast Surgery, Shenzhen Futian District Maternal and Child Health Care Hospital, Shenzhen, Guangdong, China
| | - Rong Liang
- Department of Breast, Jinan University Affiliated Guangzhou Red Cross Hospital: Guangzhou Red Cross Hospital, Guangzhou, Guangdong, China
| | - Xinpeng Hu
- Department of Breast Surgery, The Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Wijesinghe SN, Ditchfield C, Flynn S, Agrawal J, Davis ET, Dajas-Bailador F, Chapman V, Jones SW. Immunomodulation and fibroblast dynamics driving nociceptive joint pain within inflammatory synovium: Unravelling mechanisms for therapeutic advancements in osteoarthritis. Osteoarthritis Cartilage 2024; 32:1358-1370. [PMID: 38960140 DOI: 10.1016/j.joca.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/21/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE Synovitis is a widely accepted sign of osteoarthritis (OA), characterised by tissue hyperplasia, where increased infiltration of immune cells and proliferation of resident fibroblasts adopt a pro-inflammatory phenotype, and increased the production of pro-inflammatory mediators that are capable of sensitising and activating sensory nociceptors, which innervate the joint tissues. As such, it is important to understand the cellular composition of synovium and their involvement in pain sensitisation to better inform the development of effective analgesics. METHODS Studies investigating pain sensitisation in OA with a focus on immune cells and fibroblasts were identified using PubMed, Web of Science and SCOPUS. RESULTS In this review, we comprehensively assess the evidence that cellular crosstalk between resident immune cells or synovial fibroblasts with joint nociceptors in inflamed OA synovium contributes to peripheral pain sensitisation. Moreover, we explore whether the elucidation of common mechanisms identified in similar joint conditions may inform the development of more effective analgesics specifically targeting OA joint pain. CONCLUSION The concept of local environment and cellular crosstalk within the inflammatory synovium as a driver of nociceptive joint pain presents a compelling opportunity for future research and therapeutic advancements.
Collapse
Affiliation(s)
- Susanne N Wijesinghe
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Caitlin Ditchfield
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Sariah Flynn
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Jyoti Agrawal
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | - Victoria Chapman
- Pain Centre Versus Arthritis, NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
4
|
Taylor PC, Feist E, Pope JE, Nash P, Sibilia J, Caporali R, Balsa A. What have we learnt from the inhibition of IL-6 in RA and what are the clinical opportunities for patient outcomes? Ther Adv Musculoskelet Dis 2024; 16:1759720X241283340. [PMID: 39444594 PMCID: PMC11497505 DOI: 10.1177/1759720x241283340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterised by persistent inflammation of the synovial joints as well as other tissues and organs. Left untreated, it can lead to joint damage, disability and even increased mortality. The disease is driven by inflammatory cytokines that contribute to the chronic inflammation seen in RA. Interleukin-6 (IL-6) is a key pathological cytokine and a target for treatments aiming to alleviate local and systemic inflammation. Despite advances in understanding RA and the introduction of new treatments, achieving sustained remission remains challenging. This review explores the role of IL-6 in RA pathogenesis, its potential as a treatment target and the significance of personalised medicine in RA management. IL-6 has a dual signalling mechanism, classical and trans-signalling, which influences various intracellular pathways. While several targeted therapies have emerged, no single mechanism-based therapy is universally effective due to the diversity and complexity of the disease. Different approaches to targeting IL-6 have been tested, including biologic blockade of receptors or ligands, and inhibition of IL-6 signalling. IL-6 receptor inhibitors have been validated as RA therapeutics, either alone or in combination with other treatments. Tocilizumab, the first approved IL-6 inhibitor, blocks both soluble and membrane-bound IL-6 receptors, reducing the inflammatory cascade. Clinical trials confirm the efficacy and safety of tocilizumab and its role as a treatment option for patients unresponsive to conventional therapies. The benefits of IL-6 inhibition extend beyond reduced joint inflammation to the amelioration of comorbidities like anaemia, cardiovascular disease, depression and osteoporosis. Tailoring treatment to patients' profiles and comorbidities is essential for optimal outcomes. A 'treat-to-profile' approach, focusing on a holistic view of the patient, could improve personalised medicine strategies. Biosimilars - lower-cost alternatives to biologics - further enhance the accessibility and cost-effectiveness of treatment. IL-6 inhibitors present a valuable treatment option for RA management, particularly for patients with specific comorbidities.
Collapse
Affiliation(s)
- Peter C. Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Oxford OX3 7LD, UK
| | - Eugen Feist
- Department of Rheumatology, Helios Clinic Vogelsang-Gommern, Cooperation Partner of the Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Janet E. Pope
- Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Peter Nash
- School of Medicine, Griffith University, Brisbane, QLD, Australia
| | - Jean Sibilia
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Roberto Caporali
- Department of Rheumatology and Medical Sciences, ASST Gaetano Pini-CTO, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alejandro Balsa
- Rheumatology Department, Hospital Universitario La Paz, Madrid, Spain
- Inmuno-Rheumatology Research Group, Hospital La Paz, Institute for Health Research – IdiPAZ, Madrid, Spain
| |
Collapse
|
5
|
Cascetta G, Colombo G, Eremita G, Garcia JGN, Lenti MV, Di Sabatino A, Travelli C. Pro- and anti-inflammatory cytokines: the hidden keys to autoimmune gastritis therapy. Front Pharmacol 2024; 15:1450558. [PMID: 39193325 PMCID: PMC11347309 DOI: 10.3389/fphar.2024.1450558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Autoimmune gastritis (AIG) is an autoimmune disorder characterized by the destruction of gastric parietal cells and atrophy of the oxyntic mucosa which induces intrinsic factor deficiency and hypo-achlorhydria. AIG predominantly affects the antral mucosa with AIG patients experiencing increased inflammation and a predisposition toward the development of gastric adenocarcinoma and type I neuroendocrine tumors. The exact pathogenesis of this autoimmune disorder is incompletely understood although dysregulated immunological mechanisms appear to major contributors. This review of autoimmune gastritis, an unmet medical need, summarizes current knowledge on pro- and anti-inflammatory cytokines and strategies for the discovery of novel biomarkers and potential pharmacological targets.
Collapse
Affiliation(s)
- Greta Cascetta
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Giorgia Colombo
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Gianmarco Eremita
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Joe G. N. Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL, United States
| | - Marco Vincenzo Lenti
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
6
|
Iqbal U, Malik A, Sial NT, Uttra AM, Rehman MFU, Mehmood MH. Molecular insights of Eucalyptol (1,8-Cineole) as an anti-arthritic agent: in vivo and in silico analysis of IL-17, IL-10, NF-κB, 5-LOX and COX-2. Inflammopharmacology 2024; 32:1941-1959. [PMID: 38649658 DOI: 10.1007/s10787-024-01465-4] [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: 01/25/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024]
Abstract
The monoterpene oxide, Eucalyptol (1,8-Cineole), a primary component of eucalyptus oil, has been evaluated pharmacologically for anti-inflammatory and analgesic activity. Current research aimed to evaluate Eucalyptol's anti-arthritic potential in a Complete Freund's adjuvant induced arthritis that resembles human rheumatoid arthritis. Polyarthritis developed after 0.1 mL CFA injection into the left hind footpad in rats. Oral administration of Eucalyptol at various doses (100, 200 and 400 mg/kg) significantly reduced paw edema, body weight loss, 5-LOX, PGE2 and Anti-CCP levels. Real-time PCR investigation showed significant downregulation of COX-2, TNF-α, NF-κB, IL-17, IL-6, IL-1β and upregulation of IL-4 and IL-10 in Eucalyptol treated groups. Hemoglobin and RBCs counts significantly increased post-treatment with Eucalyptol while ESR, CRP, WBCs and platelets count significantly decreased. Eucalyptol significantly increased Superoxide Dismutase, Catalase and Glutathione levels compared to CFA-induced arthritic control however, MDA significantly decreased post-treatment. Further, radiographic and histopathological examination of the ankle joints of rodents administered Eucalyptol revealed an improvement in the structure of the joints. Piroxicam was taken as standard. Furthermore, molecular docking findings supported the anti-arthritic efficacy of Eucalyptol exhibited high binding interaction against IL-17, TNF-α, IL-4, IL-10, iNOS NF-κB, 5-LOX, and COX-2. Eucalyptol has reduced the severity of CFA induced arthritis by promoting anti-inflammatory cytokines for example IL-4, IL-10 and by inhibiting pro-inflammatory cytokines such as 5-LOX, COX-2, IL-17, NF-κB, TNF-α, IL-6 and IL-1β. Therefore, Eucalyptol might be as a potential therapeutic agent because of its pronounced anti-oxidant and anti-arthritic activity.
Collapse
Affiliation(s)
- Urooj Iqbal
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Abdul Malik
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan.
| | - Nabeela Tabassum Sial
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
- Institute of Pharmacy, Lahore College for Women University, Lahore, Pakistan
| | - Ambreen Malik Uttra
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | | | - Malik Hassan Mehmood
- Department of Pharmaceutical Sciences, Government College University, Lahore, Pakistan
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| |
Collapse
|
7
|
Chu YR, Xu YC, Ma LL, Wang JX, Zong HX, Tong WQ, Wang XL, Zhao X, Xu SQ. Skeletal muscle index together with body mass index is associated with secondary osteoporosis in patients with rheumatoid arthritis. Eur J Med Res 2024; 29:61. [PMID: 38245751 PMCID: PMC10799370 DOI: 10.1186/s40001-024-01665-2] [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: 05/31/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
OBJECTIVE The objective of this study was to explore the associations of body mass index (BMI), fat mass index (FMI), skeletal mass index (SMI) and secondary osteoporosis (OP) in patients with rheumatoid arthritis (RA). METHODS The bone mineral density (BMD) at sites of the femur neck (Neck), total hip (Hip) and lumbar vertebrae 1-4 (L1-4) was measured by dual-energy X-ray absorptiometry. The skeletal muscle index, body fat percentage and mineral content were measured by biological electrical impedance for calculating BMI, FMI and SMI. RESULTS A total of 433 patient with RA and 158 healthy controls were enrolled. The BMDs at each site of the RA patients were lower compared with those of the healthy controls (p < 0.0001), and the prevalence of OP (36.1%, 160/443) and sarcopenia (65.2%, 288/443) in the RA patients were higher than those in the controls (12.7%, 20/158, p < 0.0001; 9.0%, 14/156, p < 0.0001). Significant differences in the BMD, FMI, SMI, mineral content, body fat percentage and skeletal muscle mass were found among the RA patients in the different BMI groups (p < 0.05). In RA patients with BMI < 18.5 kg/m2, the prevalence of OP in the RA patients with sarcopenia was similar to that in those without sarcopenia (44.4% vs. 66. 7%, χ2 = 0. 574, p = 0.449). In the RA patients with a normal BMI or who were overweight or obese, prevalence of OP in the RA patients with sarcopenia was significantly higher than that in the RA patients without sarcopenia (42.8% vs. 21.7%, χ2 = 10.951, p = 0.001; 61.1% vs. 13.0%, χ2 = 26.270, p < 0.0001). In the RA patients without sarcopenia, the prevalence of OP in the RA patients in the different BMI groups was different (p = 0.039). In the RA patients with sarcopenia, there was no significant difference in the prevalence of OP among the RA patients in the different BMI groups (p = 0. 128). The linear correlation analysis showed that the SMI in RA patients was positively correlated with the BMD of each site measured and BMI and FMI (p < 0.0001). However, there was a negative linear correlation between SMI and disease duration (p = 0.048). The logistic regression analysis found that SMI (OR = 0.569, p = 0.002, 95% CI 0.399-0.810), BMI (OR = 0.884, p = 0.01, 95% CI 0.805-0.971) and gender (1 = female, 2 = male) (OR = 0.097, p < 0.0001, 95% CI 0.040-0.236) were protective factors for OP in RA, while age (OR = 1.098, p < 0.0001, 95% CI 1.071-1.125) was the risk factor. CONCLUSION BMI and SMI are associated with the occurrence of OP in RA patients, and both SMI and BMI are important protective factors for OP secondary to RA.
Collapse
Affiliation(s)
- Yi-Ran Chu
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Yue-Chen Xu
- Department of Radiotherapy, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Ling-Li Ma
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Jian-Xiong Wang
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - He-Xiang Zong
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Wan-Qiu Tong
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Xi-le Wang
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Xu Zhao
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Sheng-Qian Xu
- Department of Rheumatology & Immunology, the First Affiliated Hospital of Anhui Medical University, No. 218, Ji-Xi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
8
|
Giannandrea D, Parolini M, Citro V, De Felice B, Pezzotta A, Abazari N, Platonova N, Sugni M, Chiu M, Villa A, Lesma E, Chiaramonte R, Casati L. Nanoplastic impact on bone microenvironment: A snapshot from murine bone cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 462:132717. [PMID: 37820528 DOI: 10.1016/j.jhazmat.2023.132717] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023]
Abstract
Our world is made of plastic. Plastic waste deeply affects our health entering the food chain. The degradation and/or fragmentation of plastics due to weathering processes result in the generation of nanoplastics (NPs). Only a few studies tested NPs effects on human health. NPs toxic actions are, in part, mediated by oxidative stress (OS) that, among its effects, affects bone remodeling. This study aimed to assess if NPs influence skeleton remodeling through OS. Murine bone cell cultures (MC3T3-E1 preosteoblasts, MLOY-4 osteocyte-like cells, and RAW264.7 pre-osteoclasts) were used to test the NPs detrimental effects on bone cells. NPs affect cell viability and induce ROS production and apoptosis (by caspase 3/7 activation) in pre-osteoblasts, osteocytes, and pre-osteoclasts. NPs impair the migration capability of pre-osteoblasts and potentiate the osteoclastogenesis of preosteoclasts. NPs affected the expression of genes related to inflammatory and osteoblastogenic pathways in pre-osteoblasts and osteocytes, related to the osteoclastogenic commitment of pre-osteoclasts. A better understanding of the impact of NPs on bone cell activities resulting in vivo in impaired bone turnover could give more information on the possible toxicity consequence of NPs on bone mass and the subsequent public health problems, such as bone disease.
Collapse
Affiliation(s)
| | - Marco Parolini
- Department of Environmental Science and Policy, University of Milan, Italy
| | | | - Beatrice De Felice
- Department of Environmental Science and Policy, University of Milan, Italy
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | | | | | - Michela Sugni
- Department of Environmental Science and Policy, University of Milan, Italy
| | - Martina Chiu
- Department of Medicine and Surgery, University of Parma, Italy
| | | | - Elena Lesma
- Department of Health Sciences, University of Milan, Italy
| | | | - Lavinia Casati
- Department of Health Sciences, University of Milan, Italy.
| |
Collapse
|
9
|
M. S. Barron A, Fabre T, De S. Distinct fibroblast functions associated with fibrotic and immune-mediated inflammatory diseases and their implications for therapeutic development. F1000Res 2024; 13:54. [PMID: 38681509 PMCID: PMC11053351 DOI: 10.12688/f1000research.143472.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 05/01/2024] Open
Abstract
Fibroblasts are ubiquitous cells that can adopt many functional states. As tissue-resident sentinels, they respond to acute damage signals and shape the earliest events in fibrotic and immune-mediated inflammatory diseases. Upon sensing an insult, fibroblasts produce chemokines and growth factors to organize and support the response. Depending on the size and composition of the resulting infiltrate, these activated fibroblasts may also begin to contract or relax thus changing local stiffness within the tissue. These early events likely contribute to the divergent clinical manifestations of fibrotic and immune-mediated inflammatory diseases. Further, distinct changes to the cellular composition and signaling dialogue in these diseases drive progressive fibroblasts specialization. In fibrotic diseases, fibroblasts support the survival, activation and differentiation of myeloid cells, granulocytes and innate lymphocytes, and produce most of the pathogenic extracellular matrix proteins. Whereas, in immune-mediated inflammatory diseases, sequential accumulation of dendritic cells, T cells and B cells programs fibroblasts to support local, destructive adaptive immune responses. Fibroblast specialization has clear implications for the development of effective induction and maintenance therapies for patients with these clinically distinct diseases.
Collapse
Affiliation(s)
- Alexander M. S. Barron
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Thomas Fabre
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Saurav De
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
10
|
Negishi Y, Adili A, de Vega S, Momoeda M, Kaneko H, Cilek MZ, Yoshinaga C, Takafuji K, Otsuka Y, Shimoda M, Negishi-Koga T, Ishijima M, Okada Y. IL-6 Reduces Spheroid Sizes of Osteophytic Cells Derived from Osteoarthritis Knee Joint via Induction of Apoptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:135-149. [PMID: 37918800 DOI: 10.1016/j.ajpath.2023.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/01/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023]
Abstract
Osteophytes in osteoarthritis (OA) joints contribute to restriction of joint movement, joint pain, and OA progression, but little is known about osteophyte regulators. Examination of gene expression related to cartilage extracellular matrix, endochondral ossification, and growth factor signaling in articular cartilage and osteophytes obtained from OA knee joints showed that several genes such as COL1A1, VCAN, BGLAP, BMP8B, RUNX2, and SOST were overexpressed in osteophytes compared with articular cartilage. Ratios of mesenchymal stem/progenitor cells, which were characterized by co-expression of CD105 and CD166, were significantly higher in osteophytic cells than articular cells. A three-dimensional culture method for cartilage and osteophyte cells was developed by modification of cultures of self-assembled spheroid cell organoids (spheroids). These spheroids cultured in the media for mesenchymal stem cells containing transforming growth factor-β3 showed characteristic morphologies and gene expression profiles of articular cartilage and osteophytes, respectively. The effects of IL-1β, tumor necrosis factor-α, and IL-6 on the spheroids of articular and osteophytic cells were studied. To the best of our knowledge, they provide the first evidence that IL-6 suppresses the spheroid size of osteophytic cells by inducing apoptosis and reducing extracellular matrix molecules. These data show that IL-6 is the suppressor of osteophyte growth and suggest that IL-6 expression and/or activity are implicated in the regulation of osteophyte formation in pathologic joints.
Collapse
Affiliation(s)
- Yoshifumi Negishi
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Arepati Adili
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University, Tokyo, Japan
| | - Susana de Vega
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Momoeda
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Haruka Kaneko
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mehmet Z Cilek
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University, Tokyo, Japan
| | - Chiho Yoshinaga
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuaki Takafuji
- Research Institute, Suntory Global Innovation Center, Ltd., Kyoto, Japan
| | - Yuta Otsuka
- Institute for Science of Life, Suntory Wellness, Ltd., Kyoto, Japan
| | - Masayuki Shimoda
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Takako Negishi-Koga
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Community Medicine and Research for Bone and Joint Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Muneaki Ishijima
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University, Tokyo, Japan; Department of Community Medicine and Research for Bone and Joint Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
11
|
Battaglia F, Torrini F, Palladino P, Scarano S, Minunni M. Serotonin: A new super effective functional monomer for molecular imprinting. The case of TNF-α detection in real matrix by Surface Plasmon Resonance. Biosens Bioelectron 2023; 242:115713. [PMID: 37801835 DOI: 10.1016/j.bios.2023.115713] [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: 07/03/2023] [Revised: 09/07/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023]
Abstract
Molecular imprinting and related technologies are becoming increasingly appreciated in bioanalysis and diagnostic applications. Among the imprinted polymers, we have already demonstrated that the endogenous neurotransmitters (NTs) dopamine (DA) and norepinephrine (NE) can be efficiently used as natural and sustainable monomers to straightforwardly design and synthesize a new generation of green and "soft" Molecularly Imprinted BioPolymers (MIBPs). Here, we demonstrated for the first time the ability of a further NT, i.e., serotonin (SE), in forming adhesive imprinted nanofilms coupled to label-free optical biosensing. Its imprinting efficiency is compared with those obtained with PDA and PNE. As a model study, tumor necrosis factor-alpha (TNF-α) was selected as a biomolecular target of interest in clinical diagnostics. The biomimetic receptor was coupled to Surface Plasmon Resonance (SPR), and TNF-α detection was performed in label-free and real-time manner both in buffer and biological matrices, i.e. synovial fluid and human serum. The results indicate that, under the same imprinting and binding conditions, the analytical performances of PSE are impressively superior to those of PDA and PNE. The PSE-based MIBP was able to detect TNF-α in human matrices with a good sensitivity, selectivity, and repeatability.
Collapse
Affiliation(s)
- Federica Battaglia
- Department of Chemistry "Ugo Schiff', University of Florence, 50019, Sesto Fiorentino, FI, Italy.
| | - Francesca Torrini
- Department of Chemistry and Applied Biosciences, ETH Zurich, Ramistrasse 101, 8092, Zurich, Switzerland.
| | - Pasquale Palladino
- Department of Chemistry "Ugo Schiff', University of Florence, 50019, Sesto Fiorentino, FI, Italy.
| | - Simona Scarano
- Department of Chemistry "Ugo Schiff', University of Florence, 50019, Sesto Fiorentino, FI, Italy.
| | - Maria Minunni
- Department of Chemistry "Ugo Schiff', University of Florence, 50019, Sesto Fiorentino, FI, Italy.
| |
Collapse
|
12
|
Zeng M, Issotina Zibrila A, Li X, Liu X, Wang X, Zeng Z, Wang Z, He Y, Meng L, Liu J. Pyridostigmine ameliorates pristane-induced arthritis symptoms in Dark Agouti rats. Scand J Rheumatol 2023; 52:627-636. [PMID: 37339380 DOI: 10.1080/03009742.2023.2196783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/27/2023] [Indexed: 06/22/2023]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a chronic inflammatory disorder. Pyridostigmine (PYR), an acetylcholinesterase (AChE) inhibitor, has been shown to reduce inflammation and oxidative stress in several animal models for inflammation-associated conditions. The present study aimed to investigate the effects of PYR on pristane-induced (PIA) in Dark Agouti (DA) rats. METHOD DA rats were intradermally infused with pristane to establish the PIA model, which was treated with PYR (10 mg/kg/day) for 27 days. The effects of PYR on synovial inflammation, oxidative stress, and gut microbiota were evaluated by determining arthritis scores, H&E staining, quantitative polymerase chain reaction, and biochemical assays, as well as 16S rDNA sequencing. RESULTS Pristane induced arthritis, with swollen paws and body weight loss, increased arthritis scores, synovium hyperplasia, and bone or cartilage erosion. The expression of pro-inflammatory cytokines in synovium was higher in the PIA group than in the control group. PIA rats also displayed elevated levels of malondialdehyde, nitric oxide, superoxide dismutase, and catalase in plasma. Moreover, sequencing results showed that the richness, diversity, and composition of the gut microbiota dramatically changed in PIA rats. PYR abolished pristane-induced inflammation and oxidative stress, and corrected the gut microbiota dysbiosis. CONCLUSION The results of this study support the protective role of PYR in PIA in DA rats, associated with the attenuation of inflammation and correction of gut microbiota dysbiosis. These findings open new perspectives for pharmacological interventions in animal models of RA.
Collapse
Affiliation(s)
- M Zeng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, PR China
| | - A Issotina Zibrila
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, PR China
| | - X Li
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, PR China
| | - X Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, PR China
| | - X Wang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, PR China
| | - Z Zeng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, PR China
| | - Z Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Y He
- Department of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, PR China
| | - L Meng
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, PR China
| | - J Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, PR China
| |
Collapse
|
13
|
Lee ES, Ko H, Kim CH, Kim HC, Choi SK, Jeong SW, Lee SG, Lee SJ, Na HK, Park JH, Shin JM. Disease-microenvironment modulation by bare- or engineered-exosome for rheumatoid arthritis treatment. Biomater Res 2023; 27:81. [PMID: 37635253 PMCID: PMC10464174 DOI: 10.1186/s40824-023-00418-2] [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: 05/22/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Exosomes are extracellular vesicles secreted by eukaryotic cells and have been extensively studied for their surface markers and internal cargo with unique functions. A deeper understanding of exosomes has allowed their application in various research areas, particularly in diagnostics and therapy. MAIN BODY Exosomes have great potential as biomarkers and delivery vehicles for encapsulating therapeutic cargo. However, the limitations of bare exosomes, such as rapid phagocytic clearance and non-specific biodistribution after injection, pose significant challenges to their application as drug delivery systems. This review focuses on exosome-based drug delivery for treating rheumatoid arthritis, emphasizing pre/post-engineering approaches to overcome these challenges. CONCLUSION This review will serve as an essential resource for future studies to develop novel exosome-based therapeutic approaches for rheumatoid arthritis. Overall, the review highlights the potential of exosomes as a promising therapeutic approach for rheumatoid arthritis treatment.
Collapse
Affiliation(s)
- Eun Sook Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Hyewon Ko
- Bionanotechnology Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyun-Chul Kim
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Seong-Kyoon Choi
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Sang Won Jeong
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Se-Guen Lee
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Sung-Jun Lee
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea
| | - Hee-Kyung Na
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung Min Shin
- Division of Biotechnology, Convergence Research Institute, DGIST, 333 Techno Jungang-Daero, Daegu, 42988, Republic of Korea.
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
14
|
Philippon EML, van Rooijen LJE, Khodadust F, van Hamburg JP, van der Laken CJ, Tas SW. A novel 3D spheroid model of rheumatoid arthritis synovial tissue incorporating fibroblasts, endothelial cells, and macrophages. Front Immunol 2023; 14:1188835. [PMID: 37545512 PMCID: PMC10402919 DOI: 10.3389/fimmu.2023.1188835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Objective Rheumatoid Arthritis (RA) is a progressive and systemic autoimmune disorder associated with chronic and destructive joint inflammation. The hallmarks of joint synovial inflammation are cellular proliferation, extensive neoangiogenesis and infiltration of immune cells, including macrophages. In vitro approaches simulating RA synovial tissue are crucial in preclinical and translational research to evaluate novel diagnostic and/or therapeutic markers. Two-dimensional (2D) settings present very limited in vivo physiological proximity as they cannot recapitulate cell-cell and cell-matrix interactions occurring in the three-dimensional (3D) tissue compartment. Here, we present the engineering of a spheroid-based model of RA synovial tissue which mimics 3D interactions between cells and pro-inflammatory mediators present in the inflamed synovium. Methods Spheroids were generated by culturing RA fibroblast-like-synoviocytes (RAFLS), human umbilical vein endothelial cells (ECs) and monocyte-derived macrophages in a collagen-based 3D scaffold. The spheroids were cultured in the presence or absence of vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (bFGF) or RA synovial fluid (SF). Spheroid expansion and cell migration were quantified for all conditions using confocal microscopy and digital image analysis. Results A novel approach using machine learning was developed to quantify spheroid outgrowth and used to reexamine the existing spheroid-based model of RA synovial angiogenesis consisting of ECs and RAFLS. A 2-fold increase in the spheroid outgrowth ratio was demonstrated upon VEGF/bFGF stimulation (p<0.05). The addition of macrophages within the spheroid structure (3.75x104 RAFLS, 7.5x104 ECs and 3.0x104 macrophages) resulted in good incorporation of the new cell type. The addition of VEGF/bFGF significantly induced spheroid outgrowth (p<0.05) in the new system. SF stimulation enhanced containment of macrophages within the spheroids. Conclusion We present a novel spheroid based model consisting of RAFLS, ECs and macrophages that reflects the RA synovial tissue microenvironment. This model may be used to dissect the role of specific cell types in inflammatory responses in RA, to study specific signaling pathways involved in the disease pathogenesis and examine the effects of novel diagnostic (molecular imaging) and therapeutic compounds, including small molecule inhibitors and biologics.
Collapse
Affiliation(s)
- Eva M. L. Philippon
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Lisanne J. E. van Rooijen
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Fatemeh Khodadust
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Conny J. van der Laken
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
15
|
Nada H, Sivaraman A, Lu Q, Min K, Kim S, Goo JI, Choi Y, Lee K. Perspective for Discovery of Small Molecule IL-6 Inhibitors through Study of Structure–Activity Relationships and Molecular Docking. J Med Chem 2023; 66:4417-4433. [PMID: 36971365 DOI: 10.1021/acs.jmedchem.2c01957] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Interleukin-6 (IL-6) is a proinflammatory cytokine that plays a key role in the pathogenesis and physiology of inflammatory and autoimmune diseases, such as coronary heart disease, cancer, Alzheimer's disease, asthma, rheumatoid arthritis, and most recently COVID-19. IL-6 and its signaling pathway are promising targets in the treatment of inflammatory and autoimmune diseases. Although, anti-IL-6 monoclonal antibodies are currently being used in clinics, huge unmet medical needs remain because of the high cost, administration-related toxicity, lack of opportunity for oral dosing, and potential immunogenicity of monoclonal antibody therapy. Furthermore, nonresponse or loss of response to monoclonal antibody therapy has been reported, which increases the importance of optimizing drug therapy with small molecule drugs. This work aims to provide a perspective for the discovery of novel small molecule IL-6 inhibitors by the analysis of the structure-activity relationships and computational studies for protein-protein inhibitors targeting the IL-6/IL-6 receptor/gp130 complex.
Collapse
|
16
|
Liu K, Sun L, Li S, Xu H. Combined application of multiple biomarkers for early auxiliary diagnosis of silicosis. Toxicol Ind Health 2023; 39:138-145. [PMID: 36734071 DOI: 10.1177/07482337231154636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Silicosis is an important industrial health problem for those workers exposed to silica. The present study aimed to investigate the sensitivity and specificity of combined detection of biomarkers in early auxiliary diagnosis of silicosis, the risk factors of silicosis were also studied. The study sample comprised 65 workers who had clinical silicosis and 70 matched control subjects who were exposed to silica but did not have clinical silicosis. The levels of superoxide dismutase, malondialdehyde, interleukin 6 (IL-6), tumor necrosis factor-alpha, and cholinesterases in the serum of 135 subjects were measured. After completing the biochemical assays, a logistic regression model based on the above biochemical determination results was established, and the receiver operating characteristic curve was used for judging the discrimination ability of different statistical indexes. The expression levels of MDA, IL-6, and TNF-alpha in serum samples of patients with stage I silicosis and MDA and IL-6 in serum samples of patients with stage II silicosis were all significantly higher. Results from logistic regression analysis showed that ChEs were protective factors for silicosis, while age, chronic respiratory symptoms, IL-6, and MDA were risk factors. The areas under the ROC curve (AUC) were 0.86 (IL-6), 0.81 (MDA), and 0.65 (TNF-alpha or ChEs). AUC-ROC = 0.90 (95%CI:0.84-0.95). The diagnostic efficiency of IL-6 combined with MDA and TNF-alpha was better than that of any single biomarker.
Collapse
Affiliation(s)
- Keliang Liu
- School of Health Management, 530224Xinxiang Medical University SanQuan Medical College, Xinxiang, China
| | - Linqing Sun
- School of Health Management, 530224Xinxiang Medical University SanQuan Medical College, Xinxiang, China
| | - Sirui Li
- School of Health Management, 530224Xinxiang Medical University SanQuan Medical College, Xinxiang, China
| | - Haiming Xu
- School of Public Health and Management, 105002Ningxia Medical University, Yinchuan, China.,Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| |
Collapse
|
17
|
Sekelova T, Danisovic L, Cehakova M. Rejuvenation of Senescent Mesenchymal Stem Cells to Prevent Age-Related Changes in Synovial Joints. Cell Transplant 2023; 32:9636897231200065. [PMID: 37766590 PMCID: PMC10540599 DOI: 10.1177/09636897231200065] [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: 04/05/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Mesenchymal/medicinal stem/signaling cells (MSCs), well known for regenerative potential, have been involved in hundreds of clinical trials. Even if equipped with reparative properties, aging significantly decreases their biological activity, representing a major challenge for MSC-based therapies. Age-related joint diseases, such as osteoarthritis, are associated with the accumulation of senescent cells, including synovial MSCs. An impaired ability of MSCs to self-renew and differentiate is one of the main contributors to the human aging process. Moreover, senescent MSCs (sMSCs) are characterized by the senescence-messaging secretome (SMS), which is typically manifested by the release of molecules with an adverse effect. Many factors, from genetic and metabolic pathways to environmental stressors, participate in the regulation of the senescent phenotype of MSCs. To better understand cellular senescence in MSCs, this review discusses the characteristics of sMSCs, their role in cartilage and synovial joint aging, and current rejuvenation approaches to delay/reverse age-related pathological changes, providing evidence from in vivo experiments as well.
Collapse
Affiliation(s)
- Tatiana Sekelova
- National Institute of Rheumatic Diseases, Piestany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Piestany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Michaela Cehakova
- National Institute of Rheumatic Diseases, Piestany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
18
|
Abdallah YEH, Chahal S, Jamali F, Mahmoud SH. Drug-disease interaction: Clinical consequences of inflammation on drugs action and disposition. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2023; 26:11137. [PMID: 36942294 PMCID: PMC9990632 DOI: 10.3389/jpps.2023.11137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023]
Abstract
Inflammation is a culprit in many conditions affecting millions of people worldwide. A plethora of studies has revealed that inflammation and inflammatory mediators such as cytokines and chemokines are associated with altered expression and activity of various proteins such as those involved in drug metabolism, specifically cytochrome P450 enzymes (CYPs). Emphasis of most available reports is on the inflammation-induced downregulation of CYPs, subsequently an increase in their substrate concentrations, and the link between the condition and the inflammatory mediators such as interleukin-6 and tumor necrosis factor alpha. However, reports also suggest that inflammation influences expression and/or activity of other proteins such as those involved in the drug-receptor interaction. These multifaced involvements render the clinical consequence of the inflammation unexpected. Such changes are shown in many inflammatory conditions including rheumatoid arthritis, Crohn's disease, acute respiratory illnesses as well as natural processes such as aging, among others. For example, some commonly used cardiovascular drugs lose their efficacy when patients get afflicted with inflammatory conditions such as rheumatoid arthritis and Crohn's disease. Interestingly, this is despite increased concentration subsequent to reduced clearance. The observation is attributed to a simultaneous reduction in the expression of target receptor proteins such as the calcium and potassium channel and β-adrenergic receptor as well as the metabolic enzymes. This narrative review summarizes the current understanding and clinical implications of the inflammatory effects on both CYPs and drug-receptor target proteins.
Collapse
|
19
|
Engineering Closed-Loop, Autoregulatory Gene Circuits for Osteoarthritis Cell-Based Therapies. Curr Rheumatol Rep 2022; 24:96-110. [PMID: 35404006 DOI: 10.1007/s11926-022-01061-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Genetic engineering offers the possibility to simultaneously target multiple cellular pathways in the joints affected by osteoarthritis (OA). The purpose of this review is to summarize the ongoing efforts to develop disease-modifying osteoarthritis drugs (DMOADs) using genetic engineering, including targeting approaches, genome editing techniques, and delivery methods. RECENT FINDINGS Several gene circuits have been developed that reprogram cells to autonomously target inflammation, and their efficacy has been demonstrated in chondrocytes and stem cells. Gene circuits developed for metabolic disorders, such as those targeting insulin resistance and obesity, also have the potential to mitigate the impact of these conditions on OA onset and/or progression. Despite the strides made in characterizing the inflammatory environment of the OA joint, our incomplete understanding of how the multiple regulators interact to control signal transduction, gene transcription, and translation to protein limits the development of targeted disease-modifying therapeutics. Continuous advances in targeted genome editing, combined with online toolkits that simplify the design and production of gene circuits, have the potential to accelerate the discovery and clinical application of multi-target gene circuits with disease-modifying properties for the treatment of OA.
Collapse
|
20
|
Yoo SJ, Lee HR, Kim J, Yoo IS, Park CK, Kang SW. Hypoxia-Inducible Factor-2 Alpha Regulates the Migration of Fibroblast-like Synoviocytes via Oxidative Stress-Induced CD70 Expression in Patients with Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms23042342. [PMID: 35216458 PMCID: PMC8877612 DOI: 10.3390/ijms23042342] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
This study aimed to examine the role of CD70, which is highly expressed on fibroblast-like synoviocytes (FLS), in rheumatoid arthritis (RA) patients. FLS isolated from RA (n = 14) and osteoarthritis (OA, n = 4) patients were stimulated with recombinant interleukin-17 (IL-17; 5 ng/mL) and tumor necrosis factor alpha (TNF-α; 5 ng/mL) for 24 h. Expression of CD70, CD27/soluble CD27 (sCD27), and hypoxia-inducible factor-2 alpha (HIF-2α) was analyzed by RT-qPCR, flow cytometry, and ELISA assays, respectively. Reactive oxygen species (ROS) expression and cell migration were also examined. The HIF-2α inhibitor PT-2385 and CD70 inhibitor BU69 were used to specifically suppress these pathways. Stimulation with IL-17 and TNF-α significantly induced CD70 expression in RA FLS. Although the synovial fluids from patients with RA contained high levels of sCD27, surface expression of CD27, a ligand of CD70, was rarely detected in RA FLS. Cytokine-induced CD70 expression was significantly decreased following antioxidant treatment. Following HIF-2α inhibition, RA FLS had decreased expression of CD70 and ROS levels. Migration of RA FLS was also inhibited by inhibition of CD70 or HIF-2α. The surface expression of CD70 is regulated by HIF-2α and ROS levels and is a key contributor to cytokine-enhanced migration in RA FLS.
Collapse
Affiliation(s)
- Su-Jin Yoo
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (S.-J.Y.); (H.-R.L.); (J.K.)
- Research Institute for Medical Sciences, Chungnam National University School of Medicine, 266 Munhwaro, Daejeon 35015, Korea
| | - Ha-Reum Lee
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (S.-J.Y.); (H.-R.L.); (J.K.)
- Research Institute for Medical Sciences, Chungnam National University School of Medicine, 266 Munhwaro, Daejeon 35015, Korea
| | - Jinhyun Kim
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (S.-J.Y.); (H.-R.L.); (J.K.)
| | - In Seol Yoo
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Sejong Hospital, 20 Bodeum-7-ro, Sejong 30099, Korea; (I.S.Y.); (C.K.P.)
| | - Chan Keol Park
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Sejong Hospital, 20 Bodeum-7-ro, Sejong 30099, Korea; (I.S.Y.); (C.K.P.)
| | - Seong Wook Kang
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (S.-J.Y.); (H.-R.L.); (J.K.)
- Research Institute for Medical Sciences, Chungnam National University School of Medicine, 266 Munhwaro, Daejeon 35015, Korea
- Correspondence: ; Tel.: +82-42-338-2428
| |
Collapse
|
21
|
Liu S, Deng Z, Chen K, Jian S, Zhou F, Yang Y, Fu Z, Xie H, Xiong J, Zhu W. Cartilage tissue engineering: From proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments (Review). Mol Med Rep 2022; 25:99. [PMID: 35088882 PMCID: PMC8809050 DOI: 10.3892/mmr.2022.12615] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA), one of the most common joint diseases, is characterized by fibrosis, rhagadia, ulcers and attrition of articular cartilage due to a number of factors. The etiology of OA remains unclear, but its occurrence has been associated with age, obesity, inflammation, trauma and genetic factors. Inflammatory cytokines are crucial for the occurrence and progression of OA. The intra-articular proinflammatory and anti-inflammatory cytokines jointly maintain a dynamic balance, in accordance with the physiological metabolism of articular cartilage. However, dynamic imbalance between proinflammatory and anti-inflammatory cytokines can cause abnormal metabolism in knee articular cartilage, which leads to deformation, loss and abnormal regeneration, and ultimately destroys the normal structure of the knee joint. The ability of articular cartilage to self-repair once damaged is limited, due to its inability to obtain nutrients from blood vessels, nerves and lymphatic vessels, as well as limitations in the extracellular matrix. There are several disadvantages inherent to conventional repair methods, while cartilage tissue engineering (CTE), which combines proinflammatory and anti-inflammatory cytokines, offers a new therapeutic approach for OA. The aim of the present review was to examine the proinflammatory factors implicated in OA, including IL-1β, TNF-α, IL-6, IL-15, IL-17 and IL-18, as well as the key anti-inflammatory factors reducing OA-related articular damage, including IL-4, insulin-like growth factor and TGF-β. The predominance of proinflammatory over anti-inflammatory cytokine effects ultimately leads to the development of OA. CTE, which employs mesenchymal stem cells and scaffolding technology, may prevent OA by maintaining the homeostasis of pro- and anti-inflammatory factors.
Collapse
Affiliation(s)
- Shuyu Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Kang Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Shengsheng Jian
- Department of Orthopedics, Luo Hu Hospital, Shenzhen, Guangdong 518001, P.R. China
| | - Feifei Zhou
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Yuan Yang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zicai Fu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Huanyu Xie
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Jianyi Xiong
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
22
|
Önnheim K, Huang S, Holmertz AS, Andersson S, Lönnblom E, Jonsson C, Holmdahl R, Gjertsson I. Rheumatoid arthritis chondrocytes produce increased levels of pro-inflammatory proteins. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100235. [DOI: 10.1016/j.ocarto.2022.100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/14/2022] [Indexed: 10/19/2022] Open
|
23
|
Debreova M, Culenova M, Smolinska V, Nicodemou A, Csobonyeiova M, Danisovic L. Rheumatoid arthritis: From synovium biology to cell-based therapy. Cytotherapy 2022; 24:365-375. [DOI: 10.1016/j.jcyt.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/23/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022]
|
24
|
Tang J, Cheng X, Yi S, Zhang Y, Tang Z, Zhong Y, Zhang Q, Pan B, Luo Y. Euphorbia Factor L2 ameliorates the Progression of K/BxN Serum-Induced Arthritis by Blocking TLR7 Mediated IRAK4/IKKβ/IRF5 and NF-kB Signaling Pathways. Front Pharmacol 2021; 12:773592. [PMID: 34950033 PMCID: PMC8691750 DOI: 10.3389/fphar.2021.773592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/03/2021] [Indexed: 02/05/2023] Open
Abstract
Toll like receptor (TLR)s have a central role in regulating innate immunity and their activation have been highlighted in the pathogenesis of rheumatoid arthritis (RA). EFL2, one of diterpenoids derived from Euphorbia seeds, is nearly unknown expect for its improving effect on acute lung injury. Our present study aimed to investigate EFL2's pharmacokinetic features, its therapeutic effect on rheumatoid arthritis, and explored the potential anti-arthritic mechanisms. K/BxN serum transfer arthritis (STA) murine model was used to assess EFL2's anti-arthritic effects. We also applied UPLC-MS method to measure the concentrations of EFL2 in plasma. The inhibitory effects of this compound on inflammatory cells infiltration and activation were determined by flow cytometry analysis and quantitative real-time polymerase chain reaction (qRT-PCR) in vivo, and immunochemistry staining and ELISA in murine macrophages and human PBMCs in vitro, respectively. The mechanism of EFL2 on TLRs mediated signaling pathway was evaluated by PCR array, Western blot, plasmid transfection and confocal observation. Intraperitoneal (i.p.) injection of EFL2, instead of oral administration, could effectively ameliorate arthritis severity of STA mice. The inflammatory cells migration and infiltration into ankles were also significantly blocked by EFL2, accompanied with dramatically reduction of chemokines mRNA expression and pro-inflammatory cytokines production. In vivo PCR microarray indicated that EFL2 exerted anti-arthritis bioactivity by suppressing TLR7 mediated signaling pathway. In vitro study confirmed the inhibitory effects of EFL2 on TLR7 or TLR3/7 synergistically induced inflammatory cytokines secretion in murine macrophages and human PBMCs. In terms of molecular mechanism, we further verified that EFL2 robustly downregulated TLR7 mediated IRAK4-IKKβ-IRF5 and NF-κB signaling pathways activation, and blocked IRF5 and p65 phosphorylation and translocation activity. Taken together, our data indicate EFL2's therapeutic potential as a candidate for rheumatoid arthritis and other TLR7-dependent diseases.
Collapse
Affiliation(s)
- Jing Tang
- Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Sichuan, China.,Department of Rheumatology and Immunology, Luzhou's People's Hospital, Luzhou, China
| | - Xiaolan Cheng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyu Yi
- Department of Rheumatology and Immunology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Yuanyuan Zhang
- Sichuan Food and Drug Inspection and Testing Institute, Chengdu, China
| | - Zhigang Tang
- Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Sichuan, China
| | - Yutong Zhong
- Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Sichuan, China
| | - Qiuping Zhang
- Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Sichuan, China
| | - Bin Pan
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang, China
| | - Yubin Luo
- Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
25
|
Molnar V, Matišić V, Kodvanj I, Bjelica R, Jeleč Ž, Hudetz D, Rod E, Čukelj F, Vrdoljak T, Vidović D, Starešinić M, Sabalić S, Dobričić B, Petrović T, Antičević D, Borić I, Košir R, Zmrzljak UP, Primorac D. Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis. Int J Mol Sci 2021; 22:9208. [PMID: 34502117 PMCID: PMC8431625 DOI: 10.3390/ijms22179208] [Citation(s) in RCA: 308] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/15/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis is a common cause of disability worldwide. Although commonly referred to as a disease of the joint cartilage, osteoarthritis affects all joint tissues equally. The pathogenesis of this degenerative process is not completely understood; however, a low-grade inflammation leading to an imbalance between anabolic and katabolic processes is a well-established factor. The complex network of cytokines regulating these processes and cell communication has a central role in the development and progression of osteoarthritis. Concentrations of both proinflammatory and anti-inflammatory cytokines were found to be altered depending on the osteoarthritis stage and activity. In this review, we analyzed individual cytokines involved in the immune processes with an emphasis on their function in osteoarthritis.
Collapse
Affiliation(s)
- Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Ivan Kodvanj
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Roko Bjelica
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Nursing, University North, 48000 Varaždin, Croatia
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Orthopaedic Surgery, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
- Department of Health Studies, University of Split, 21000 Split, Croatia
- Department of Traumatology, Medical University Merkur Hospital, 10000 Zagreb, Croatia
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Orthopaedic Surgery, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Dinko Vidović
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
| | | | - Srećko Sabalić
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
| | - Borut Dobričić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Orthopaedics and Traumatology, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Tadija Petrović
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Darko Antičević
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Traumatology, Medical University Merkur Hospital, 10000 Zagreb, Croatia
- Medical School, University of Split, 21000 Split, Croatia;
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
| | - Rok Košir
- Molecular Biology Laboratory, BIA Separations CRO, Labena Ltd., 1000 Ljubljana, Slovenia; (R.K.); (U.P.Z.)
| | - Uršula Prosenc Zmrzljak
- Molecular Biology Laboratory, BIA Separations CRO, Labena Ltd., 1000 Ljubljana, Slovenia; (R.K.); (U.P.Z.)
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Split, 21000 Split, Croatia;
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96450 Coburg, Germany
- Eberly College of Science, State College, The Pennsylvania State University, University Park, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
| |
Collapse
|
26
|
Huang J, Fu X, Chen X, Li Z, Huang Y, Liang C. Promising Therapeutic Targets for Treatment of Rheumatoid Arthritis. Front Immunol 2021; 12:686155. [PMID: 34305919 PMCID: PMC8299711 DOI: 10.3389/fimmu.2021.686155] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic poly-articular chronic autoimmune joint disease that mainly damages the hands and feet, which affects 0.5% to 1.0% of the population worldwide. With the sustained development of disease-modifying antirheumatic drugs (DMARDs), significant success has been achieved for preventing and relieving disease activity in RA patients. Unfortunately, some patients still show limited response to DMARDs, which puts forward new requirements for special targets and novel therapies. Understanding the pathogenetic roles of the various molecules in RA could facilitate discovery of potential therapeutic targets and approaches. In this review, both existing and emerging targets, including the proteins, small molecular metabolites, and epigenetic regulators related to RA, are discussed, with a focus on the mechanisms that result in inflammation and the development of new drugs for blocking the various modulators in RA.
Collapse
Affiliation(s)
- Jie Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xuekun Fu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xinxin Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Zheng Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Yuhong Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China.,Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
27
|
Deng C, Zhang Q, He P, Zhou B, He K, Sun X, Lei G, Gong T, Zhang Z. Targeted apoptosis of macrophages and osteoclasts in arthritic joints is effective against advanced inflammatory arthritis. Nat Commun 2021; 12:2174. [PMID: 33846342 PMCID: PMC8042091 DOI: 10.1038/s41467-021-22454-z] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Insufficient apoptosis of inflammatory macrophages and osteoclasts (OCs) in rheumatoid arthritis (RA) joints contributes toward the persistent progression of joint inflammation and destruction. Here, we deliver celastrol (CEL) to selectively induce apoptosis of OCs and macrophages in arthritic joints, with enzyme-responsive nanoparticles (termed PRNPs) composed of RGD modified nanoparticles (termed RNPs) covered with cleavable PEG chains. CEL-loaded PRNPs (CEL-PRNPs) dually target OCs and inflammatory macrophages derived from patients with RA via an RGD-αvβ3 integrin interaction after PEG cleavage by matrix metalloprotease 9, leading to increased apoptosis of these cells. In an adjuvant-induced arthritis rat model, PRNPs have an arthritic joint-specific distribution and CEL-PRNPs efficiently reduce the number of OCs and inflammatory macrophages within these joints. Additionally, rats with advanced arthritis go into inflammatory remission with bone erosion repair and negligible side effects after CEL-PRNPs treatment. These findings indicate potential for targeting chemotherapy-induced apoptosis in the treatment of advanced inflammatory arthritis.
Collapse
Affiliation(s)
- Caifeng Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Quan Zhang
- Institute of Materia Medica, School of Pharmacy, Chengdu Medical College, Chengdu, 610500, China
- Development and Regeneration Key Lab of Sichuan Province, Department of Pathology, Department of Anatomy and Histology and Embryology, Chengdu Medical College, Chengdu, 610500, China
| | - Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Bin Zhou
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, China
| | - Ke He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
28
|
Interleukin-6 in Rheumatoid Arthritis. Int J Mol Sci 2020; 21:ijms21155238. [PMID: 32718086 PMCID: PMC7432115 DOI: 10.3390/ijms21155238] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 01/08/2023] Open
Abstract
The role of interleukin (IL)-6 in health and disease has been under a lot of scrutiny in recent years, particularly during the recent COVID-19 pandemic. The inflammatory pathways in which IL-6 is involved are also partly responsible of the development and progression of rheumatoid arthritis (RA), opening interesting perspectives in terms of therapy. Anti-IL-6 drugs are being used with variable degrees of success in other diseases and are being tested in RA. Results have been encouraging, particularly when anti-IL-6 has been used with other drugs, such as metothrexate (MTX). In this review we discuss the main immunologic aspects that make anti-IL-6 a good candidate in RA, but despite the main therapeutic options available to target IL-6, no gold standard treatment has been established so far.
Collapse
|
29
|
Bone Control of Muscle Function. Int J Mol Sci 2020; 21:ijms21041178. [PMID: 32053970 PMCID: PMC7072735 DOI: 10.3390/ijms21041178] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/21/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023] Open
Abstract
Bone and muscle represent a single functional system and are tightly connected to each other. Indeed, diseases characterized by alterations of muscle physiology have effects on bone remodeling and structure and vice versa. Muscle influence on bone has been deeply studied, and recent studies identified irisin as new molecule involved in this crosstalk. Muscle regulation by bone needs to be extensively investigated since in the last few years osteocalcin was recognized as a key molecule in the bone–muscle interaction. Osteocalcin can exist in two forms with different degrees of carboxylation. The undercarboxylated form of osteocalcin is a hormone released by the bone matrix during the osteoclast bone resorption and can bind its G-protein coupled receptor GPRC6A expressed in the muscle, thus regulating its function. Recently, this hormone was described as an antiaging molecule for its ability to regulate bone, muscle and cognitive functions. Indeed, the features of this bone-related hormone were used to test a new therapeutic approach for sarcopenia, since injection of osteocalcin in older mice induces the acquirement of physical abilities of younger animals. Even if this approach should be tested in humans, osteocalcin represents the most surprising molecule in endocrine regulation by the skeleton.
Collapse
|
30
|
Mahendra A, Yang X, Abnouf S, Adolacion JRT, Park D, Soomro S, Roszik J, Coarfa C, Romain G, Wanzeck K, Bridges SL, Aggarwal A, Qiu P, Agarwal SK, Mohan C, Varadarajan N. Beyond Autoantibodies: Biologic Roles of Human Autoreactive B Cells in Rheumatoid Arthritis Revealed by RNA-Sequencing. Arthritis Rheumatol 2019; 71:529-541. [PMID: 30407753 PMCID: PMC6741783 DOI: 10.1002/art.40772] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 11/01/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To obtain the comprehensive transcriptome profile of human citrulline-specific B cells from patients with rheumatoid arthritis (RA). METHODS Citrulline- and hemagglutinin-specific B cells were sorted by flow cytometry using peptide-streptavidin conjugates from the peripheral blood of RA patients and healthy individuals. The transcriptome profile of the sorted cells was obtained by RNA-sequencing, and expression of key protein molecules was evaluated by aptamer-based SOMAscan assay and flow cytometry. The ability of these proteins to effect differentiation of osteoclasts and proliferation and migration of synoviocytes was examined by in vitro functional assays. RESULTS Citrulline-specific B cells, in comparison to citrulline-negative B cells, from patients with RA differentially expressed the interleukin-15 receptor α (IL-15Rα) gene as well as genes related to protein citrullination and cyclic AMP signaling. In analyses of an independent cohort of cyclic citrullinated peptide-seropositive RA patients, the expression of IL-15Rα protein was enriched in citrulline-specific B cells from the patients' peripheral blood, and surprisingly, all B cells from RA patients were capable of producing the epidermal growth factor ligand amphiregulin (AREG). Production of AREG directly led to increased migration and proliferation of fibroblast-like synoviocytes, and, in combination with anti-citrullinated protein antibodies, led to the increased differentiation of osteoclasts. CONCLUSION To the best of our knowledge, this is the first study to document the whole transcriptome profile of autoreactive B cells in any autoimmune disease. These data identify several genes and pathways that may be targeted by repurposing several US Food and Drug Administration-approved drugs, and could serve as the foundation for the comparative assessment of B cell profiles in other autoimmune diseases.
Collapse
Affiliation(s)
- Ankit Mahendra
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Xingyu Yang
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Shaza Abnouf
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Jay R T Adolacion
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Daechan Park
- Department of Biological Sciences, College of Natural Sciences, Ajou University, Republic of Korea
| | - Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, TX
| | - Jason Roszik
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cristian Coarfa
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX
| | - Gabrielle Romain
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Keith Wanzeck
- Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - S. Louis Bridges
- Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - Amita Aggarwal
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Immunology, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX
| | - Navin Varadarajan
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| |
Collapse
|
31
|
Zhang S, Ren Q, Qi H, Liu S, Liu Y. Adverse Effects of Fine-Particle Exposure on Joints and Their Surrounding Cells and Microenvironment. ACS NANO 2019; 13:2729-2748. [PMID: 30773006 DOI: 10.1021/acsnano.8b08517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Current understanding of the health risks and adverse effects upon exposure to fine particles is premised on the direct association of particles with target organs, particularly the lung; however, fine-particle exposure has also been found to have detrimental effects on sealed cavities distant to the portal-of-entry, such as joints. Moreover, the fundamental toxicological issues have been ascribed to the direct toxic mechanisms, in particular, oxidative stress and proinflammatory responses, without exploring the indirect mechanisms, such as compensated, adaptive, and secondary effects. In this Review, we recapitulate the current findings regarding the detrimental effects of fine-particle exposure on joints, the surrounding cells, and microenvironment, as well as their deteriorating impact on the progression of arthritis. We also elaborate the likely molecular mechanisms underlying the particle-induced detrimental influence on joints, not limited to direct toxicity, but also considering the other indirect mechanisms. Because of the similarities between fine air particles and engineered nanomaterials, we compare the toxicities of engineered nanomaterials to those of fine air particles. Arthritis and joint injuries are prevalent, particularly in the elderly population. Considering the severity of global exposure to fine particles and limited studies assessing the detrimental effects of fine-particle exposure on joints and arthritis, this Review aims to appeal to a broad interest and to promote more research efforts in this field.
Collapse
Affiliation(s)
- Shuping Zhang
- Institute for Medical Engineering and Science , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Quanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
| | - Hui Qi
- Beijing Jishuitan Hospital , Peking University Health Science Center , Beijing 100035 , P. R. China
- Beijing Research Institute of Traumatology and Orthopaedics , Beijing 100035 , P. R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
| | - Yajun Liu
- Beijing Jishuitan Hospital , Peking University Health Science Center , Beijing 100035 , P. R. China
| |
Collapse
|
32
|
Matsumura T, Saito Y, Suzuki T, Teramoto A, Ozasa Y, Yamashita T, Fujimiya M, Saito-Chikenji T. Phosphorylated Platelet-Derived Growth Factor Receptor-Positive Cells With Anti-apoptotic Properties Accumulate in the Synovium of Patients With Rheumatoid Arthritis. Front Immunol 2019; 10:241. [PMID: 30828336 PMCID: PMC6384265 DOI: 10.3389/fimmu.2019.00241] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/28/2019] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease caused by inflammation of the synovium and characterized by chronic polyarthritis that destroys bone and cartilage. Fibroblast-like synoviocytes (FLSs) in the synovium of patients with RA can promote cartilage and bone destruction by producing proteins such as matrix metalloproteinases and receptor activator of NF-κB ligand, thereby representing an important therapeutic target for RA. FLSs have several phenotypes depending on which cell surface proteins and adhesion factors are expressed. Identifying the cellular functions associated with different phenotypes and methods of controlling them are considered essential for developing therapeutic strategies for RA. In this study, synovial tissue was collected from patients with RA and control subjects who required surgery due to ligament injury or fracture. Immunohistological analysis was used to investigate the rates of positivity for phosphorylated platelet-derived growth factor receptor-αβ (pPDGFRαβ) and cadherin-11 (CDH11) expression, and apoptosis-related markers were assessed for each cell phenotype. Next, FLSs were isolated in vitro and stimulated with tumor necrosis factor-α (TNF-α) in addition to a combination of PDGF and transforming growth factor (2GF) to investigate pPDGFRαβ and CDH11 expression and the effects of the inhibition of TNF and cyclin-dependent kinase (CDK) 4/6 on FLSs. Immunohistological analysis showed a large percentage of pPDGFRαβ+CDH11– cells in the sub-lining layer (SL) of patients with RA. These cells exhibited increased B-cell lymphoma-2 expression, reduced TNF receptor-1 expression, resistance to cell death, and abnormal proliferation, suggesting a tendency to accumulate in the synovium. Further, in vitro 2GF stimulation of FLSs lowered, whereas 2GF + TNF stimulation increased the pPDGFRαβ/CDH11 ratio. Hypothesizing that FLSs stimulated with 2GF + TNF would accumulate in vivo in RA, we determined the therapeutic effects of TNF and CDK4/6 inhibitors. The TNF inhibitor lowered the pPDGFRαβ/CDH11 ratio, whereas the CDK4/6 inhibitor suppressed cell proliferation. However, a synergistic effect was not observed by combining both the drugs. We observed an increase in pPDGFRαβ+CDH11– cells in the SL of the RA synovium and accumulation of these cells in the synovium. We found that the TNF inhibitor suppressed FLS activity and the CDK4/6 inhibitor reduced cell proliferation.
Collapse
Affiliation(s)
- Takashi Matsumura
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomoyuki Suzuki
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Teramoto
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuhiro Ozasa
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako Saito-Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
33
|
Lima AC, Cunha C, Carvalho A, Ferreira H, Neves NM. Interleukin-6 Neutralization by Antibodies Immobilized at the Surface of Polymeric Nanoparticles as a Therapeutic Strategy for Arthritic Diseases. ACS APPLIED MATERIALS & INTERFACES 2018; 10:13839-13850. [PMID: 29614225 DOI: 10.1021/acsami.8b01432] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Arthritic diseases are disabling conditions affecting millions of patients worldwide. Pro-inflammatory cytokines, particularly interleukin-6 (IL-6), plays a crucial role in inflammation and cartilage destruction. Although the beneficial effects of antibody therapy, its efficacy is limited. Therefore, this work proposes the immobilization of antibodies at the surface of biodegradable polymeric nanoparticles (NPs) to capture and neutralize IL-6. Our system is intended to protect, extend and enhance the therapeutic efficacy after delivery. Chitosan-hyaluronic acid NPs are synthesized as a stable monodisperse population. After determining the maximum immobilization capacity (10 μg/mL), the capture ability was confirmed. Biological assays demonstrate the NPs cytocompatibility with human articular chondrocytes (hACs) and human macrophages. hACs stimulated with macrophage conditioned medium shows the beneficial role of IL-6 capture and neutralization. Biofunctionalized NPs exhibit a prolonged action and stronger efficacy than the free antibody. In conclusion, this system can be an effective and long lasting treatment for arthritic diseases.
Collapse
Affiliation(s)
- Ana Cláudia Lima
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics , University of Minho , Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark , 4805-017 Barco, Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Braga/Guimarães , Portugal
| | - Cristina Cunha
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Braga/Guimarães , Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences , University of Minho , Campus de Gualtar , 4710-057 Braga , Portugal
| | - Agostinho Carvalho
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Braga/Guimarães , Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences , University of Minho , Campus de Gualtar , 4710-057 Braga , Portugal
| | - Helena Ferreira
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics , University of Minho , Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark , 4805-017 Barco, Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Braga/Guimarães , Portugal
| | - Nuno M Neves
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics , University of Minho , Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark , 4805-017 Barco, Guimarães , Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Braga/Guimarães , Portugal
- The Discoveries Centre for Regenerative and Precision Medicine , Headquarters at University of Minho , Avepark 4805-017 Barco, Guimarães , Portugal
| |
Collapse
|
34
|
Livshits G, Kalinkovich A. Hierarchical, imbalanced pro-inflammatory cytokine networks govern the pathogenesis of chronic arthropathies. Osteoarthritis Cartilage 2018; 26:7-17. [PMID: 29074297 DOI: 10.1016/j.joca.2017.10.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/15/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic inflammatory arthropathies, such as rheumatoid arthritis (RA), spondyloarthritis, including psoriatic arthritis (PsA), ankylosing spondyloarthritis (AS), osteoarthritis (OA), and intervertebral disc degenerative disease (DDD) constitute major public health problems that are anticipated to grow significantly as the human population ages. However, many aspects concerning the molecular mechanisms underlying their onset and progression remain unclear. DESIGN This narrative review critically analyzes the molecular mechanisms underlying the inflammation-associated pathogenesis of the aforementioned joint diseases. This includes, in particular, the major role played by several key soluble factors (such as cytokines and the associated signaling pathways, designated as "fragile nodes") produced by local cells and recruited to the joints' immune cells, whose elimination by specific drugs has dramatically improved the diseases' symptomatology and outcome in human clinical trials or in rodent arthritis models. HYPOTHESIS AND THE AIM OF THIS REVIEW We hypothesize that the pathogenesis of chronic inflammatory arthropathies is governed by hierarchical, imbalanced pro-inflammatory cytokine networks (HIPICNs) (comprising a combination of fragile nodes) that are created during the development of both autoimmune (RA, PsA, and AS) and non-autoimmune (OA and DDD) disorders. The main aim of this review is to provide evidence that despite substantial pathobiological differences between these arthropathies, the HIPICNs created are quite common, thus justifying the merging of these disorders mechanistically and suggesting that these common mechanisms exist in the onset and progression of different joint diseases.
Collapse
Affiliation(s)
- G Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel.
| | - A Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
35
|
Patel B, Wi CI, Hasassri ME, Divekar R, Absah I, Almallouhi E, Ryu E, King K, Juhn YJ. Heterogeneity of asthma and the risk of celiac disease in children. Allergy Asthma Proc 2018; 39:51-58. [PMID: 29279060 DOI: 10.2500/aap.2018.39.4100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Although human leukocyte antigen (HLA)-DR and HLA-DQ genes and gluten play crucial roles in developing celiac disease (CD), most patients with these risk factors still do not develop CD, which indicates additional unrecognized risk factors. OBJECTIVE To determine the association between asthma and the risk of CD in children. METHODS We conducted a population-based retrospective case-control study in children who resided in Olmsted County, Minnesota. We identified children with CD (cases) between January 1, 1997, and December 31, 2014, and compared these with children without CD (controls) (1:2 matching). Asthma status was ascertained by using the predetermined asthma criteria (PAC) and the asthma predictive index (API). Data analysis included conditional logistic regression models and an unsupervised network analysis by using an independent phenome-wide association scan (PheWAS) data set. RESULTS Although asthma status as determined by using PAC was not associated with the risk of CD (odds ratio [OR] 1.4 [95% confidence interval {CI}, 0.8-2.5]; p = 0.2), asthma status by using the API was significantly associated (OR 2.8 [95% CI, 1.3-6.0]; p = 0.008). A subgroup analysis indicated that children with both asthma as determined by using PAC and a family history of asthma had an increased risk of CD compared with those without asthma (OR 2.28 [95% CI, 1.11-4.67]; p = 0.024). PheWAS data showed a cluster of asthma single nucleotide polymorphisms and patients with CD. CONCLUSION A subgroup of children with asthma who also had a family history of asthma seemed to be at an increased risk of CD, and, thus, the third factor that underlies the risk of CD might be related to genetic factors for asthma. Heterogeneity of asthma plays a role in determining the risk of asthma-related comorbidity.
Collapse
|
36
|
Loro E, Ramaswamy G, Chandra A, Tseng WJ, Mishra MK, Shore EM, Khurana TS. IL15RA is required for osteoblast function and bone mineralization. Bone 2017; 103:20-30. [PMID: 28602725 PMCID: PMC5598756 DOI: 10.1016/j.bone.2017.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022]
Abstract
Interleukin-15 receptor alpha (IL15RA) is an important component of interleukin-15 (IL15) pro-inflammatory signaling. In addition, IL15 and IL15RA are present in the circulation and are detected in a variety of tissues where they influence physiological functions such as muscle contractility and overall metabolism. In the skeletal system, IL15RA was previously shown to be important for osteoclastogenesis. Little is known, however, about its role in osteoblast function and bone mineralization. In this study, we evaluated bone structural and mechanical properties of an Il15ra whole-body knockout mouse (Il15ra-/-) and used in vitro and bioinformatic analyses to understand the role IL15/IL15RA signaling on osteoblast function. We show that lack of IL15RA decreased bone mineralization in vivo and in isolated primary osteogenic cultures, suggesting a cell-autonomous effect. Il15ra-/- osteogenic cultures also had reduced Rankl/Opg mRNA ratio, indicating defective osteoblast/osteoclast coupling. We analyzed the transcriptome of primary pre-osteoblasts from normal and Il15ra-/- mice and identified 1150 genes that were differentially expressed at a FDR of 5%. Of these, 844 transcripts were upregulated and 306 were downregulated in Il15ra-/- cells. The largest functional clusters, highlighted using DAVID analysis, were related to metabolism, immune response, bone mineralization and morphogenesis. The transcriptome analysis was validated by qPCR of some of the most significant hits. Using bioinformatic approaches, we identified candidate genes, including Cd200 and Enpp1, that could contribute to the reduced mineralization. Silencing Il15ra using shRNA in the calvarial osteoblast MC3T3-E1 cell line decreased ENPP1 activity. Taken together, these data support that IL15RA plays a cell-autonomous role in osteoblast function and bone mineralization.
Collapse
Affiliation(s)
- Emanuele Loro
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Girish Ramaswamy
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abhishek Chandra
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Division of Geriatric Medicine and Gerontology, Mayo Clinic, Rochester, MN, USA
| | - Wei-Ju Tseng
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Manoj K Mishra
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Tejvir S Khurana
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Shen ZT, Sigalov AB. Rationally designed ligand-independent peptide inhibitors of TREM-1 ameliorate collagen-induced arthritis. J Cell Mol Med 2017; 21:2524-2534. [PMID: 28382703 PMCID: PMC5618672 DOI: 10.1111/jcmm.13173] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 02/24/2017] [Indexed: 12/24/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 1 (TREM‐1) is critically involved in the pathogenesis of rheumatoid arthritis (RA). In contrast to cytokine blockers, therapeutic blockade of TREM‐1 can blunt excessive inflammation while preserving the capacity for microbial control. However, the nature of the TREM‐1 ligand(s) and mechanisms of TREM‐1 signalling are still not yet well understood, impeding the development of clinically relevant inhibitors of TREM‐1. The aim of this study was to evaluate the anti‐arthritic activity of a novel, ligand‐independent TREM‐1 inhibitory nonapeptide GF9 that was rationally designed using the signalling chain homo oligomerization (SCHOOL) model of cell signalling. Free GF9 and GF9 bound to macrophage‐targeted nanoparticles that mimic human high‐density lipoproteins (GF9‐HDL) were used to treat collagen‐induced arthritis (CIA). We also tested if 31‐mer peptides with sequences from GF9 and helices 4 (GE31) and 6 (GA31) of the major HDL protein, apolipoprotein A‐I, are able to perform three functions: assist in the self‐assembly of GA/E31‐HDL, target these particles to macrophages and block TREM‐1 signalling. We showed that GF9, but not control peptide, ameliorated CIA and protected against bone and cartilage damage. The therapeutic effect of GF9 was accompanied by a reduction in the plasma levels of macrophage colony‐stimulating factor and pro‐inflammatory cytokines such as tumour necrosis factor‐α, interleukin (IL)‐1 and IL‐6. Incorporation of GF9 alone or as a part of GE31 and GA31 peptides into HDL significantly increased its therapeutic efficacy. Collectively, our findings suggest that TREM‐1 inhibitory SCHOOL sequences may be promising alternatives for the treatment of RA.
Collapse
|
38
|
Casado JG, Blázquez R, Vela FJ, Álvarez V, Tarazona R, Sánchez-Margallo FM. Mesenchymal Stem Cell-Derived Exosomes: Immunomodulatory Evaluation in an Antigen-Induced Synovitis Porcine Model. Front Vet Sci 2017; 4:39. [PMID: 28377922 PMCID: PMC5359696 DOI: 10.3389/fvets.2017.00039] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/06/2017] [Indexed: 12/28/2022] Open
Abstract
Synovitis is an inflammatory process associated with pain, disability, and discomfort, which is usually treated with anti-inflammatory drugs or biological agents. Mesenchymal stem cells (MSCs) have been also successfully used in the treatment of inflammatory-related diseases such as synovitis or arthritis. In the last years, the exosomes derived from MSCs have become a promising tool for the treatment of inflammatory-related diseases and their therapeutic effect is thought to be mediated (at least in part) by their immunomodulatory potential. In this work, we aimed to evaluate the anti-inflammatory effect of these exosomes in an antigen-induced synovitis animal model. To our knowledge, this is the first report where exosomes derived from MSCs have been evaluated in an animal model of synovitis. Our results demonstrated a decrease of synovial lymphocytes together with a downregulation of TNF-α transcripts in those exosome-treated joints. These results support the immunomodulatory effect of these exosomes and point out that they may represent a promising therapeutic option for the treatment of synovitis.
Collapse
Affiliation(s)
- Javier G Casado
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain; Immunology Unit, Department of Physiology, University of Extremadura, Cáceres, Spain; CIBER de Enfermedades Cardiovasculares, Cáceres, Spain
| | - Rebeca Blázquez
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain; CIBER de Enfermedades Cardiovasculares, Cáceres, Spain
| | - Francisco Javier Vela
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre , Cáceres , Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre , Cáceres , Spain
| | - Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura , Cáceres , Spain
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain; CIBER de Enfermedades Cardiovasculares, Cáceres, Spain
| |
Collapse
|
39
|
Rodríguez-Álvarez Y, Morera-Díaz Y, Gerónimo-Pérez H, Castro-Velazco J, Martínez-Castillo R, Puente-Pérez P, Besada-Pérez V, Hardy-Rando E, Chico-Capote A, Martínez-Cordovez K, Santos-Savio A. Active immunization with human interleukin-15 induces neutralizing antibodies in non-human primates. BMC Immunol 2016; 17:30. [PMID: 27671547 PMCID: PMC5036325 DOI: 10.1186/s12865-016-0168-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Interleukin-15 is an immunostimulatory cytokine overexpressed in several autoimmune and inflammatory diseases such as Rheumatoid Arthritis, psoriasis and ulcerative colitis; thus, inhibition of IL-15-induced signaling could be clinically beneficial in these disorders. Our approach to neutralize IL-15 consisted in active immunization with structurally modified human IL-15 (mhIL-15) with the aim to induce neutralizing antibodies against native IL-15. In the present study, we characterized the antibody response in Macaca fascicularis, non-human primates that were immunized with a vaccine candidate containing mhIL-15 in Aluminum hydroxide (Alum), Montanide and Incomplete Freund’s Adjuvant. Results Immunization with mhIL-15 elicited a specific antibodies response that neutralized native IL-15-dependent biologic activity in a CTLL-2 cell proliferation assay. The highest neutralizing response was obtained in macaques immunized with mhIL-15 adjuvanted in Alum. This response, which was shown to be transient, also inhibited the activity of simian IL-15 and did not affect the human IL-2-induced proliferation of CTLL-2 cells. Also, in a pool of synovial fluid cells from two Rheumatoid Arthritis patients, the immune sera slightly inhibited TNF-α secretion. Finally, it was observed that this vaccine candidate neither affect animal behavior, clinical status, blood biochemistry nor the percentage of IL-15-dependent cell populations, specifically CD56+ NK and CD8+ T cells. Conclusion Our results indicate that vaccination with mhIL-15 induced neutralizing antibodies to native IL-15 in non-human primates. Based on this fact, we propose that this vaccine candidate could be potentially beneficial for treatment of diseases where IL-15 overexpression is associated with their pathogenesis.
Collapse
Affiliation(s)
- Yunier Rodríguez-Álvarez
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba.
| | - Yanelys Morera-Díaz
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| | - Haydee Gerónimo-Pérez
- Quality Control Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Jorge Castro-Velazco
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Rafael Martínez-Castillo
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Pedro Puente-Pérez
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Vladimir Besada-Pérez
- Chemistry and Physics Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Eugenio Hardy-Rando
- Biotechnology Laboratory, Study Center for Research and Biological Evaluations, Institute of Pharmacy and Foods, Havana University, Avenue 222, PO Box 13600, Havana, 10600, Cuba
| | - Araceli Chico-Capote
- Rheumatology Department, Hermanos Ameijeiras Hospital, San Lazaro 701, PO Box 6122, Havana, 10600, Cuba
| | - Klaudia Martínez-Cordovez
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| | - Alicia Santos-Savio
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| |
Collapse
|
40
|
Wang XD, Wang YL, Gao WF. Honokiol possesses potential anti-inflammatory effects on rheumatoid arthritis and GM-CSF can be a target for its treatment. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:7929-7936. [PMID: 26339358 PMCID: PMC4555686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/26/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To observe the anti-inflammatory effects of honokiol in primary cultures of peripheral blood mononuclear cells of rheumatoid arthritis patients, the pro-inflammatory cytokines and potential targets were investigated. METHODS The levels of GM-CSF, IL-1β, TNF-α and IL-8 were determined by ELISA assay. The genes and proteins expression were analyzed by real-time PCR and Western blotting respectively. RESULTS The serum IL-1β, TNF-α and GM-CSF levels were 1.76-, 2.16- and 3.57-fold increased in patients with RA as compared to those of control group. Honokiol inhibited the expression levels of IL-1β, TNF-α, GM-CSF and IL-8 in PBMCs with a dose-dependent manner. Measurements obtained from supernatants were positively correlated between TNF-α and IL-1β, moreover, similar results found TNF-α levels positively correlated with GM-CSF and IL-8 activity in the supernatants of PBMCs isolated from RA patients. Furthermore, the mRNA and protein expression of IL-1β, GM-CSF and IL-8 were up-regulated when the PBMCs exposure to TNF-α, however, honokiol treatment significantly reversed the expression of IL-1β, TNF-α and GM-CSF in response to TNF-α with a dose-dependent manner. CONCLUSIONS This study demonstrates that honokiol could possess potential anti-inflammatory effects and inhibits TNF-α-induced IL-1β, GM-CSF and IL-8 production in PBMCs from rheumatoid arthritis patients.
Collapse
Affiliation(s)
- Xiao-Dong Wang
- Department of Immunology and Rheumatology, Affiliated Hospital of Weifang Medical UniversityWeifang 261031, Shandong, China
| | - Ying-Liang Wang
- Department of Immunology and Rheumatology, Affiliated Hospital of Weifang Medical UniversityWeifang 261031, Shandong, China
| | - Wen-Feng Gao
- Department of Immunology and Rheumatology, Clinical Faculty (Affiliated Hospital), Weifang Medical UniversityWeifang 261053, Shandong, China
| |
Collapse
|