1
|
Lees F, Mahbub SB, Gosnell ME, Campbell JM, Weedon H, Habibalahi A, Goldys EM, Wechalekar MD, Hutchinson MR, Crotti TN. Utilising Hyperspectral Autofluorescence Imaging in the Objective Assessment of Disease State and Pain in Patients with Rheumatoid Arthritis. Int J Mol Sci 2024; 25:11996. [PMID: 39596063 PMCID: PMC11593821 DOI: 10.3390/ijms252211996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic inflammatory disease resulting in joint swelling and pain. Treatment options can be reliant on disease activity scores (DAS) incorporating patient global assessments, which are quantified via visual analogue scales (VAS). VAS can be subjective and not necessarily align with clinical symptoms, such as inflammation, resulting in a disconnect between the patient's and practitioners' experience. The development of more objective assessments of pain would enable a more targeted and personalised management of pain within individuals with RA and have the potential to improve the reliability of assessments in research. Using emerging light-based hyperspectral autofluorescence imaging (HAI) technology, we aimed to objectively differentiate disease and pain states based on the analysis of synovial tissue (ST) samples from RA patients. In total, 22 individuals with RA were dichotomised using the DAS in 28-joint counts (DAS-28) into an inactive (IA) or active disease (active-RA) group and then three sub-levels of pain (low, mid, high) based on VAS. HAI was performed on ST sections to identify and quantify the most prominent fluorophores. HAI fluorophore analysis revealed a distinct separation between the IA-RA and active-RA mid-VAS cohort, successfully determining disease state. Additionally, the separation between active-RA Mid-VAS and active RA High-VAS cohort suggests that HAI could be used to objectively separate individuals based on pain severity.
Collapse
Affiliation(s)
- Florence Lees
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia (T.N.C.)
- ARC Centre for Excellence for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA 5005, Australia
| | - Saabah B. Mahbub
- Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (S.B.M.); (J.M.C.); (A.H.)
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Martin E. Gosnell
- Quantitative Pty Ltd., 118 Great Western Highway, Mount Victoria, NSW 2786, Australia;
| | - Jared M. Campbell
- Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (S.B.M.); (J.M.C.); (A.H.)
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Helen Weedon
- Department of Rheumatology, Flinders Medical Centre and Flinders University, Bedford Park, SA 5042, Australia; (H.W.)
| | - Abbas Habibalahi
- Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (S.B.M.); (J.M.C.); (A.H.)
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Ewa M. Goldys
- Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (S.B.M.); (J.M.C.); (A.H.)
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Mihir D. Wechalekar
- Department of Rheumatology, Flinders Medical Centre and Flinders University, Bedford Park, SA 5042, Australia; (H.W.)
| | - Mark R. Hutchinson
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia (T.N.C.)
- ARC Centre for Excellence for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA 5005, Australia
| | - Tania N. Crotti
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia (T.N.C.)
| |
Collapse
|
2
|
Eckert D, Evic M, Schang J, Isbruch M, Er M, Dörrschuck L, Rapp F, Donaubauer AJ, Gaipl US, Frey B, Fournier C. Osteo-immunological impact of radon spa treatment: due to radon or spa alone? Results from the prospective, thermal bath placebo-controlled RAD-ON02 trial. Front Immunol 2024; 14:1284609. [PMID: 38292488 PMCID: PMC10824901 DOI: 10.3389/fimmu.2023.1284609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
Musculoskeletal disorders (MSDs) are associated with pain and lead to reduced mobility and quality of life for patients. Radon therapy is used as alternative or complementary to pharmaceutical treatments. According to previous reports, radon spa leads to analgesic and anti-inflammatory effects, but the cellular and molecular mechanisms are widely unknown. A previous study (RAD-ON01) revealed, that bone erosion markers like collagen fragments (C-terminal telopeptide, CTX) are reduced after radon spa treatment in serum of patients with degenerative MSDs. Within the scope of the prospective, placebo-controlled RAD-ON02 trial presented here, we analyzed the influence of radon and thermal spa treatment on osteoclastogenesis. From patient blood, we isolate monocytes, seeded them on bone slices and differentiated them in the presence of growth factors into mature osteoclasts (mOCs). Subsequent analysis showed a smaller fraction of mOCs after both treatments, which was even smaller after radon spa treatment. A significantly reduced resorbed area on bone slices reflects this result. Only after radon spa treatment, we detected in the serum of patients a significant decrease of receptor activator of NF-κB ligand (RANKL), which indicates reduced differentiation of OCs. However, other markers for bone resorption (CTX) and bone formation (OPG, OCN) were not altered after both treatments. Adipokines, such as visfatin and leptin that play a role in some MSD-types by affecting osteoclastogenesis, were not changed after both treatments. Further, also immune cells have an influence on osteoclastogenesis, by inhibiting and promoting terminal differentiation and activation of OCs, respectively. After radon treatment, the fraction of Treg cells was significantly increased, whereas Th17 cells were not altered. Overall, we observed that both treatments had an influence on osteoclastogenesis and bone resorption. Moreover, radon spa treatment affected the Treg cell population as well as the Th17/Treg ratio were affected, pointing toward a contribution of the immune system after radon spa. These data obtained from patients enrolled in the RAD-ON02 trial indicate that radon is not alone responsible for the effects on bone metabolism, even though they are more pronounced after radon compared to thermal spa treatment.
Collapse
Affiliation(s)
- Denise Eckert
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Megi Evic
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Jasmin Schang
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Maike Isbruch
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Melissa Er
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Lea Dörrschuck
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Felicitas Rapp
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| | - Anna-Jasmina Donaubauer
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Claudia Fournier
- Department of Biophysics, GSI Helmholtzzentrum Für Schwerionenforschung, Darmstadt, Germany
| |
Collapse
|
3
|
Eckert D, Rapp F, Tsedeke AT, Kraft D, Wente I, Molendowska J, Basheer S, Langhans M, Meckel T, Friedrich T, Donaubauer AJ, Becker I, Frey B, Fournier C. Modulation of Differentiation and Bone Resorbing Activity of Human (Pre-) Osteoclasts After X-Ray Exposure. Front Immunol 2022; 13:817281. [PMID: 35603191 PMCID: PMC9116137 DOI: 10.3389/fimmu.2022.817281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Low-dose radiotherapy (LD-RT) is a local treatment option for patients with chronic degenerative and inflammatory diseases, in particular musculoskeletal diseases. Despite reported analgesic and anti-inflammatory effects, cellular and molecular mechanisms related to osteoimmunological effects are still elusive. Here we test the hypothesis that X-irradiation inhibits the differentiation of precursor osteoclasts into mature osteoclasts (mOC) and their bone resorbing activity. Circulating monocytes from healthy donors were isolated and irradiated after attachment with single or fractionated X-ray doses, comparable to an LD-RT treatment scheme. Then monocytes underwent ex vivo differentiation into OC during cultivation up to 21 days, under conditions mimicking the physiological microenvironment of OC on bone. After irradiation, apoptotic frequencies were low, but the total number of OC precursors and mOC decreased up to the end of the cultivation period. On top, we observed an impairment of terminal differentiation, i.e. a smaller fraction of mOC, reduced resorbing activity on bone, and release of collagen fragments. We further analyzed the effect of X-irradiation on multinucleation, resulting from the fusion of precursor OC, which occurs late during OC differentiation. At 21 days after exposure, the observation of smaller cellular areas and a reduced number of nuclei per mOC suggest an impaired fusion of OC precursors to form mOC. Before, at 14 days, the nuclear translocation of Nuclear Factor Of Activated T Cells 1 (NFATc1), a master regulator of osteoclast differentiation and fusion, was decreased. In first results, obtained in the frame of a longitudinal LD-RT study, we previously reported a pain-relieving effect in patients. However, in a subgroup of patients suffering from Calcaneodynia or Achillodynia, we did not observe a consistent decrease of established blood markers for resorption and formation of bone, or modified T cell subtypes involved in regulating these processes. To assess the relevance of changes in bone metabolism for other diseases treated with LD-RT will be subject of further studies. Taken together, we observed that in vitro X-irradiation of monocytes results in an inhibition of the differentiation into bone-resorbing OC and a concomitant reduction of resorbing activity. The detected reduced NFATc1 signaling could be one underlying mechanism.
Collapse
Affiliation(s)
- Denise Eckert
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Felicitas Rapp
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Ayele Taddese Tsedeke
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Daniela Kraft
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Isabell Wente
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Jessica Molendowska
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Sidra Basheer
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Markus Langhans
- Department of Macromolecular and Paper Chemistry and Membrane Dynamics, Technical University Darmstadt, Darmstadt, Germany
| | - Tobias Meckel
- Department of Macromolecular and Paper Chemistry and Membrane Dynamics, Technical University Darmstadt, Darmstadt, Germany
| | - Thomas Friedrich
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Anna-Jasmina Donaubauer
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ina Becker
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Claudia Fournier
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| |
Collapse
|
4
|
Ayón-Pérez MF, Topete-Córdoba JJ, Agraz-Cibrián JM, Ortiz-Martínez L, Durán-Avelar MDJ, Vázquez-Reyes A, Vibanco-Pérez N, Gutiérrez-Franco J, Zambrano-Zaragoza JF. The influence of the -94 Ins/Del ATTG polymorphism of NFkB on the anti-CCP antibody levels in patients with rheumatoid arthritis. Medicine (Baltimore) 2021; 100:e28301. [PMID: 34918708 PMCID: PMC8677897 DOI: 10.1097/md.0000000000028301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/24/2021] [Indexed: 01/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by an inflammatory process that affects mainly synovial tissue in joints, and by the production of cyclic citrullinated peptides (anti-CCP) antibodies. In the inflammatory process the regulation of the nuclear factor kappa B (NFkB) transcription factor activation is a key point in the production of inflammatory cytokines. On the other hand, polymorphisms in several genes could contribute to the promotion of the inflammatory process observed in RA, and the association of the rs28362491 polymorphism in the NFkB gene with RA has been studied in different population. Therefore, it could be one of the interest targets to analyze their association with RA in a Mexican population.This is a case-control study to determine the influence of rs28362491 in the NFkB gene on RA and on clinical features of this disease, such as anti-CCP antibody levels, Disease Activity Score, and Health Assessment Questionnaire-Disability Index.The genotype of rs28362491 in the NFkB gene was determined in 140 RA patients and 135 healthy controls using the polymerase chain reaction-restriction fragment length polymorphism method with the enzyme PflMI. The following clinical variables were also determined: anti-CCP levels, Disease Activity Score, and Spanish version of the Health Assessment Questionnaire Disability-Index.Although no association of the polymorphism as a risk/protection factor with RA was found, the RA patients who carried the Ins/Ins genotype showed higher anti-CCP levels, while those with the Del/Del genotype showed higher Spanish version of the Health Assessment Questionnaire-Disability Index levels, compared to the other genotypes.The NFkB -94 Ins/Del ATTG (rs28362491) polymorphism is, therefore, associated with higher levels of anti-CCP antibodies, though no significant association as a risk or protection factor in RA cases was identified.
Collapse
Affiliation(s)
- Miriam Fabiola Ayón-Pérez
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| | | | - Juan Manuel Agraz-Cibrián
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
- Maestria en Salud Pública, Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| | - Liliana Ortiz-Martínez
- Clinica de Reumatologia, Servicio de Medicina Interna, Instituto Mexicano del Seguro Social HGZ No. 1, Tepic, Nayarit, Mexico
| | - Ma. de Jesús Durán-Avelar
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| | - Alejandro Vázquez-Reyes
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| | - Norberto Vibanco-Pérez
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| | - Jorge Gutiérrez-Franco
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| | - José Francisco Zambrano-Zaragoza
- Unidad Academica de Ciencias Quimico Biológicas y Farmaceuticas-Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
- Maestria en Salud Pública, Universidad Autonoma de Nayarit, Tepic, Nayarit, Mexico
| |
Collapse
|
5
|
Katayama H. Rheumatoid arthritis: Development after the emergence of a chemokine for neutrophils in the synovium. Bioessays 2021; 43:e2100119. [PMID: 34432907 DOI: 10.1002/bies.202100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/09/2022]
Abstract
Rheumatoid arthritis (RA) may not be a multifactorial disease; it can be hypothesized that RA is developed through a series of events following a triggering event, which is the emergence of a chemokine for neutrophils in the synovium. IL-17A, secreted by infiltrated neutrophils, stimulates synoviocytes to produce CCL20, which attracts various CCR6-expressing cells, including Th17 cells. Monocytes (macrophages) appear after neutrophil infiltration according to the natural course of inflammation and secrete IL-1β and TNFα. Then, IL-17A, IL-1β, and TNFα stimulate synoviocytes to produce CCL20, amplifying the inflammation. Varieties of chemokines secreted by infiltrating cells accumulate in the synovium and induce synoviocyte proliferation by binding to the corresponding G protein-coupled receptors, thus expanding the synovial tissue. CCL20 in this tissue attracts circulating monocytes that express both CCR6 and receptor activator of NF-κB (RANK), which differentiate into osteoclasts in the presence of RANKL. In this way, pannus is formed, and bone destruction begins.
Collapse
|
6
|
Williams B, Lees F, Tsangari H, Hutchinson MR, Perilli E, Crotti TN. Effects of Mild and Moderate Monoclonal Antibody Dose on Inflammation, Bone Loss, and Activation of the Central Nervous System in a Female Collagen Antibody-induced Arthritis Mouse Model. J Histochem Cytochem 2021; 69:511-522. [PMID: 34291686 DOI: 10.1369/00221554211033562] [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: 11/22/2022] Open
Abstract
Induction of severe inflammatory arthritis in the collagen antibody-induced arthritis (CAIA) murine model causes extensive joint damage and pain-like behavior compromising analysis. While mild models are less severe, their reduced, variable penetrance makes assessment of treatment efficacy difficult. This study aimed to compare macroscopic and microscopic changes in the paws, along with central nervous system activation between a mild and moderate CAIA model. Balb/c mice (n=18) were allocated to control, mild, and moderate CAIA groups. Paw inflammation, bone volume (BV), and paw volume (PV) were assessed. Histologically, the front paws were assessed for joint inflammation, cartilage damage, and pre/osteoclast-like cells and the lumbar spinal cord and the periaqueductal gray (PAG) region of the brain for glial reactivity. A moderate CAIA dose induced (1) significantly greater local paw inflammation, inflammatory cell infiltration, and PV; (2) significantly more osteoclast-like cells on the bone surface and within the surrounding soft tissue; and (3) significantly greater glial reactivity within the PAG compared with the mild CAIA model. These findings support the use of a moderate CAIA model (higher dose of monoclonal antibodies with low-dose lipopolysaccharide) to induce more consistent histopathological features, without excessive joint destruction.
Collapse
Affiliation(s)
| | - Florence Lees
- Adelaide Medical School.,ARC Centre for Excellence for Nanoscale Biophotonics
| | | | - Mark R Hutchinson
- Adelaide Medical School.,ARC Centre for Excellence for Nanoscale Biophotonics
| | - Egon Perilli
- The University of Adelaide, Adelaide, SA, Australia, and Medical Device Research Institute, College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| | | |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW To highlight the potential uses and applications of imaging in the assessment of the most common and relevant musculoskeletal (MSK) manifestations in systemic lupus erythematosus (SLE). RECENT FINDINGS Ultrasound (US) and magnetic resonance imaging (MRI) are accurate and sensitive in the assessment of inflammation and structural damage at the joint and soft tissue structures in patients with SLE. The US is particularly helpful for the detection of joint and/or tendon inflammation in patients with arthralgia but without clinical synovitis, and for the early identification of bone erosions. MRI plays a key role in the early diagnosis of osteonecrosis and in the assessment of muscle involvement (i.e., myositis and myopathy). Conventional radiography (CR) remains the traditional gold standard for the evaluation of structural damage in patients with joint involvement, and for the study of bone pathology. The diagnostic value of CR is affected by the poor sensitivity in demonstrating early structural changes at joint and soft tissue level. Computed tomography allows a detailed evaluation of bone damage. However, the inability to distinguish different soft tissues and the need for ionizing radiation limit its use to selected clinical circumstances. Nuclear imaging techniques are valuable resources in patients with suspected bone infection (i.e., osteomyelitis), especially when MRI is contraindicated. Finally, dual energy X-ray absorptiometry represents the imaging mainstay for the assessment and monitoring of bone status in patients with or at-risk of osteoporosis. Imaging provides relevant and valuable information in the assessment of MSK involvement in SLE.
Collapse
|
8
|
Di Matteo A, Mankia K, Duquenne L, Cipolletta E, Wakefield RJ, Garcia-Montoya L, Nam JL, Emery P. Ultrasound erosions in the feet best predict progression to inflammatory arthritis in anti-CCP positive at-risk individuals without clinical synovitis. Ann Rheum Dis 2020; 79:901-907. [PMID: 32366522 DOI: 10.1136/annrheumdis-2020-217215] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/23/2020] [Accepted: 04/08/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To investigate, in anti-cyclic citrullinated peptide antibody positive (CCP+) at-risk individuals without clinical synovitis, the prevalence and distribution of ultrasound (US) bone erosions (BE), their correlation with subclinical synovitis and their association with the development of inflammatory arthritis (IA). METHODS Baseline US scans of 419 CCP+ at-risk individuals were analysed. BE were evaluated in the classical sites for rheumatoid arthritis damage: the second and fifth metacarpophalangeal (MCP2 and MCP5) joints, and the fifth metatarsophalangeal (MTP5) joints. US synovitis was defined as synovial hypertrophy (SH) ≥2 or SH ≥1+power Doppler signal ≥1. Subjects with ≥1 follow-up visit were included in the progression analysis (n=400). RESULTS BE were found in ≥1 joint in 41/419 subjects (9.8%), and in 55/2514 joints (2.2%). The prevalence of BE was significantly higher in the MTP5 joints than in the MCP joints (p<0.01). A significant correlation between BE and US synovitis in the MTP5 joints was detected (Cramer's V=0.37, p<0.01). The OR for the development of IA (ever) was highest for the following: BE in >1 joint 10.6 (95% CI 1.9 to 60.4, p<0.01) and BE and synovitis in ≥1 MTP5 joint 5.1 (95% CI 1.4 to 18.9, p=0.02). In high titre CCP+ at-risk individuals, with positive rheumatoid factor and BE in ≥1 joint, the OR increased to 16.9 (95% CI 2.1-132.8, p<0.01). CONCLUSIONS In CCP+ at-risk individuals, BE in the feet appear to precede the onset of clinical synovitis. BE in >1 joint, and BE in combination with US synovitis in the MTP5 joints, are the most predictive for the development of clinical arthritis.
Collapse
Affiliation(s)
- Andrea Di Matteo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Clinica Reumatologica, Polytechnic University of Marche, Ancona, Marche, Italy
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Laurence Duquenne
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Edoardo Cipolletta
- Clinica Reumatologica, Polytechnic University of Marche, Ancona, Marche, Italy
| | - Richard J Wakefield
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Leticia Garcia-Montoya
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Jacqueline Leong Nam
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK .,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| |
Collapse
|
9
|
Assessing the Effects of Parthenolide on Inflammation, Bone Loss, and Glial Cells within a Collagen Antibody-Induced Arthritis Mouse Model. Mediators Inflamm 2020; 2020:6245798. [PMID: 32189995 PMCID: PMC7073477 DOI: 10.1155/2020/6245798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/16/2019] [Accepted: 01/27/2020] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis is characterised by a chronic inflammatory response resulting in destruction of the joint and significant pain. Although a range of treatments are available to control disease activity in RA, bone destruction and joint pain exist despite suppression of inflammation. This study is aimed at assessing the effects of parthenolide (PAR) on paw inflammation, bone destruction, and pain-like behaviour in a mild collagen antibody-induced arthritis (CAIA) mouse model. CAIA was induced in BALB/c mice and treated daily with 1 mg/kg or 4 mg/kg PAR. Clinical paw inflammation was scored daily, and mechanical hypersensitivity was assessed on alternate days. At end point, bone volume and swelling in the paws were assessed using micro-CT. Paw tissue sections were assessed for inflammation and pre-/osteoclast-like cells. The lumbar spinal cord and the periaqueductal grey (PAG) and rostral ventromedulla (RVM) regions of the brain were stained for glial fibrillary acidic protein (GFAP) and ionised calcium-binding adaptor molecule 1 (IBA1) to assess for glial reactivity. Paw scores increased in CAIA mice from days 5-10 and were reduced with 1 mg/kg and 4 mg/kg PAR on days 8-10. Osteoclast-like cells on the bone surface of the radiocarpal joint and within the soft tissue of the hind paw were significantly lower following PAR treatment (p < 0.005). GFAP- and IBA1-positive cells in the PAG and RVM were significantly lower following treatment with 1 mg/kg (p < 0.0001 and p = 0.0004, respectively) and 4 mg/kg PAR (p < 0.0001 and p = 0.001, respectively). In the lumbar spinal cord, IBA1-positive cells were significantly lower in CAIA mice treated with 4 mg/kg PAR (p = 0.001). The findings indicate a suppressive effect of both low- and moderate-dose PAR on paw inflammation, osteoclast presence, and glial cell reactivity in a mild CAIA mouse model.
Collapse
|
10
|
Iwamoto N, Sato S, Sumiyoshi R, Chiba K, Miyamoto N, Arinaga K, Kobayashi M, Yamamoto H, Osaki M, Kawakami A. Comparative study of the inhibitory effect on bone erosion progression with denosumab treatment and conventional treatment in rheumatoid arthritis patients: study protocol for an open-label randomized controlled trial by HR-pQCT. Trials 2019; 20:494. [PMID: 31409388 PMCID: PMC6691552 DOI: 10.1186/s13063-019-3589-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/16/2019] [Indexed: 02/02/2023] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic inflammatory disease of the joints, causes joint destruction, and leads to physical disability. Advances in the treatment of RA, such as biologic disease-modifying anti-rheumatic drugs (DMARDs), have provided better clinical outcomes, including the achievement of remission for patients with RA, but some patients cannot receive these treatments because of their side effects and high cost, and not all patients achieve remission. Although the efficacy of denosumab, which is a human IgG2 monoclonal antibody with a high affinity for the receptor activator of nuclear factor kappa B (RANK) ligand (RANKL), in the treatment of RA has been reported in clinical trials, the efficacy of denosumab in both preventing joint destruction and improving disease activity has not been evaluated in a real-world setting. Methods/design This open-label, randomized, parallel-group study will compare the continued use of conventional synthetic DMARDs (csDMARDs) alone with the combined use of csDMARDs and denosumab in patients whose RA is treated with csDMARDs. In total, 44 patients with RA will be randomly assigned to receive additional treatment with denosumab or to continue RA treatment without additional denosumab. The duration of the intervention will be 12 months. To analyze bone erosion and bone micro-architecture precisely, high-resolution peripheral quantitative computed tomography (HR-pQCT) will be performed every 6 months. The primary endpoint is changes in the depth of bone erosion as measured by HR-pQCT from baseline to 6 months. Important secondary endpoints are the changes from baseline in the width and volume of bone erosion as measured by HR-pQCT and changes from baseline in the depth of bone erosion at 12 months. Changes in bone micro-architecture will also be analyzed as an exploratory endpoint. Discussion The results of this study are expected to provide strong evidence regarding the usefulness of denosumab for the treatment of RA. Moreover, by using HR-pQCT, this study will also reveal the effect of denosumab not only on bone erosion but also on bone micro-architecture. Trial registration This study was registered with the University Hospital Medical Information Network Clinical Trials Registry as UMIN000030575 on December 26, 2017. Electronic supplementary material The online version of this article (10.1186/s13063-019-3589-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Shuntaro Sato
- Nagasaki University Hospital, Clinical Research Center, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Remi Sumiyoshi
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Nanami Miyamoto
- Nagasaki University Hospital, Clinical Research Center, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kumiko Arinaga
- Nagasaki University Hospital, Clinical Research Center, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Makiko Kobayashi
- Medical Science Department, Daiichi Sankyo Co., Ltd, 3-5-1 Nihonbashi honmachi Chuo-ku, Tokyo, 103-8426, Japan
| | - Hiroshi Yamamoto
- Nagasaki University Hospital, Clinical Research Center, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
11
|
Lin ZM, Liu YT, Xu YS, Yang XQ, Zhu FH, Tang W, He SJ, Zuo JP. Cervus and cucumis peptides ameliorates bone erosion in experimental arthritis by inhibiting osteoclastogenesis. Lupus Sci Med 2019; 6:e000331. [PMID: 31168402 PMCID: PMC6519612 DOI: 10.1136/lupus-2019-000331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Rheumatoid arthritis is an autoimmune disease characterised by inflammation and bone loss, leading to joint destruction and deformity. The cervus and cucumis polypeptide (CCP) injection, one of the traditional Chinese medicine injections combined extracts from deer horn and sweet melon seeds, is widely used to treat arthritis and bone fracture in China. The present study investigated the therapeutic efficacy and mechanism of CCP on pathological immune cells and bone homoeostasis in rodent experimental arthritis. METHODS The effects of CCP (4 mg/kg and 2 mg/kg) on clinical arthritis symptoms, bone erosion, proinflammatory cytokines and pathological immune cells induced by complete Freund's adjuvant was evaluated in male Sprague-Dawley rats. The impacts of CCP (2 mg/kg) on joint erythema and swelling, production of pathogenic antibodies and the proportion of inflammatory cells were assessed in collagen-induced arthritis (CIA) in DBA/1J mice. Regulation of osteoclastogenesis by CCP was observed in the murine macrophage-like RAW264.7 cells treated with receptor activator of nuclear factor-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). RESULTS CCP administration significantly prevented disease progression in both adjuvant-induced arthritis (AIA) rats and CIA mice. The therapeutic benefits were accompanied by reduction of paw oedema, reversed bone destruction, decreased pathological changes and osteoclast numbers in joints in AIA rats, as well as attenuated clinical manifestation and autoantibodies production in CIA mice. Meanwhile, in vitro supplemented of CCP concentration dependently inhibited RANKL/M-CSF-induced osteoclast differentiation, without showing cytotoxicity in RAW264.7 cells. Further, the presence of CCP dampened the augmented downstream signalling transduction as well as activation of osteoclast-specific genes and transcription factors induced by RANKL/M-CSF in RAW264.7 cells. CONCLUSION Our study suggested that the therapeutic effects of CCP in experimental arthritis could be attributed to its intervention on RANKL-induced osteoclastogenesis signalling pathway in osteoclast precursor cells.
Collapse
Affiliation(s)
- Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
| | - Yu-Ting Liu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Sheng Xu
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
| | - Wei Tang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shi-Jun He
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
| | - Jian-Ping Zuo
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Xie Z, Liu G, Tang P, Sun X, Chen S, Qin A, Zhu P, Zhang J, Fan S. Bone-targeted methotrexate-alendronate conjugate inhibits osteoclastogenesis in vitro and prevents bone loss and inflammation of collagen-induced arthritis in vivo. Drug Deliv 2018; 25:187-197. [PMID: 29303005 PMCID: PMC6058523 DOI: 10.1080/10717544.2017.1422295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA), a disease that causes joint destruction and bone erosion, is related to osteoclast activity. RA is generally treated with methotrexate (MTX). In this study, a MTX–Alendronate (ALN) conjugate was synthesized and characterized. The conjugate dramatically inhibited osteoclast formation and bone resorption compared with MTX and ALN used alone or in combination. Due to the characteristics of ALN, the MTX–ALN conjugate can adhere to the exposed bone surface and enhance drug accumulation in the pathological region for targeted therapy against osteoclastogenesis. Additionally, MTX was rapidly released in the presence of lysozyme under mildly acidic conditions, similar to inflammatory tissue and osteoclast-surviving conditions, which contributes to inflammatory inhibition; this was confirmed by the presence of pro-inflammatory cytokines. Our study highlights the use of the MTX–ALN conjugate as a potential therapeutic approach for RA by targeting osteoclastogenesis.
Collapse
Affiliation(s)
- Zi'ang Xie
- a Department of Orthopaedics , Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou , China.,b Key Laboratory of Biotherapy of Zhejiang Province , Hangzhou , China
| | - Guanxiong Liu
- c School of Chemistry and Chemical Engineering , Yangzhou University , Jiangsu , China
| | - Pan Tang
- a Department of Orthopaedics , Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou , China.,b Key Laboratory of Biotherapy of Zhejiang Province , Hangzhou , China
| | - Xuewu Sun
- a Department of Orthopaedics , Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou , China.,b Key Laboratory of Biotherapy of Zhejiang Province , Hangzhou , China
| | - Shuai Chen
- a Department of Orthopaedics , Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou , China
| | - An Qin
- d Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant , Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine , Shanghai , PR China
| | - Peizhi Zhu
- c School of Chemistry and Chemical Engineering , Yangzhou University , Jiangsu , China
| | - Jianfeng Zhang
- a Department of Orthopaedics , Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou , China
| | - Shunwu Fan
- a Department of Orthopaedics , Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou , China
| |
Collapse
|
13
|
Skármeta NP, Araneda L, Araya C. Destructive psoriatic arthritis of the temporomandibular joint: a clinical case, an overview of the pathophysiology and its differential diagnoses. Cranio 2018; 38:201-207. [DOI: 10.1080/08869634.2018.1484575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Nicolas Patricio Skármeta
- Faculty of Health Sciences, Occlusion and Orofacial Pain, School of Dental Medicine, Universidad San Sebastian, Santiago, Chile
- Orofacial Pain and TMDs, OPH Dental, Santiago, Chile
- Orofacial Pain and TMDs Hospital del Salvador, SSMO, Santiago, Chile
| | - Luis Araneda
- Maxillofacial Radiology, Hospital del Salvador, SSMO, Santiago, Chile
- Faculty of Health Sciences, Department of Maxillofacial Radiology, School of Dental Medicine, Universidad San Sebastian, Santiago, Chile
- Faculty of Sciences, Maxillofacial Radiology, School of Dental Medicine, Universidad Mayor, Santiago, Chile
- Faculty of Dentistry, Department of Maxillofacial Radiology, Universidad de Chile, Santiago, Chile
| | - Cristobal Araya
- Faculty of Sciences, Oral Medicine and Pathology, School of Dental Medicine, Universidad Mayor, Santiago, Chile
- Oral Medicine and Pathology, Instituto Nacional del Cancer, Santiago, Chile
| |
Collapse
|
14
|
Mechanisms and therapeutic targets for bone damage in rheumatoid arthritis, in particular the RANK-RANKL system. Curr Opin Pharmacol 2018; 40:110-119. [DOI: 10.1016/j.coph.2018.03.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/20/2018] [Indexed: 01/01/2023]
|
15
|
Peters M, de Jong J, Scharmga A, van Tubergen A, Geusens P, Loeffen D, Weijers R, Boyd SK, Barnabe C, Stok KS, van Rietbergen B, van den Bergh J. An automated algorithm for the detection of cortical interruptions and its underlying loss of trabecular bone; a reproducibility study. BMC Med Imaging 2018; 18:13. [PMID: 29764383 PMCID: PMC5952860 DOI: 10.1186/s12880-018-0255-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 04/30/2018] [Indexed: 01/19/2023] Open
Abstract
Background We developed a semi-automated algorithm that detects cortical interruptions in finger joints using high-resolution peripheral quantitative computed tomography (HR-pQCT), and extended it with trabecular void volume measurement. In this study we tested the reproducibility of the algorithm using scan/re-scan data. Methods Second and third metacarpophalangeal joints of 21 subjects (mean age 49 (SD 11) years, 17 early rheumatoid arthritis and 4 undifferentiated arthritis, all diagnosed < 1 year ago) were imaged twice by HR-pQCT on the same day with repositioning between scans. The images were analyzed twice by one operator (OP1) and once by an additional operator (OP2), who independently corrected the bone contours when necessary. The number, surface and volume of interruptions per joint were obtained. Intra- and inter-operator reliability and intra-operator reproducibility were determined by intra-class correlation coefficients (ICC). Intra-operator reproducibility errors were determined as the least significant change (LSCSD). Results Per joint, the mean number of interruptions was 3.1 (SD 3.6), mean interruption surface 4.2 (SD 7.2) mm2, and mean interruption volume 3.5 (SD 10.6) mm3 for OP1. Intra- and inter-operator reliability was excellent for the cortical interruption parameters (ICC ≥0.91), except good for the inter-operator reliability of the interruption surface (ICC = 0.70). The LSCSD per joint was 4.2 for the number of interruptions, 5.8 mm2 for interruption surface, and 3.2 mm3 for interruption volume. Conclusions The algorithm was highly reproducible in the detection of cortical interruptions and their volume. Based on the LSC findings, the potential value of this algorithm for monitoring structural damage in the joints in early arthritis patients needs to be tested in clinical studies. Electronic supplementary material The online version of this article (10.1186/s12880-018-0255-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Peters
- Department of Internal Medicine, Division of Rheumatology, Maastricht University Medical Centre, P.O. Box 5800, NL-6202, Maastricht, AZ, the Netherlands. .,CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands. .,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.
| | - J de Jong
- Department of Internal Medicine, Division of Rheumatology, Maastricht University Medical Centre, P.O. Box 5800, NL-6202, Maastricht, AZ, the Netherlands.,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.,Department of Radiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - A Scharmga
- Department of Internal Medicine, Division of Rheumatology, Maastricht University Medical Centre, P.O. Box 5800, NL-6202, Maastricht, AZ, the Netherlands.,CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands.,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - A van Tubergen
- Department of Internal Medicine, Division of Rheumatology, Maastricht University Medical Centre, P.O. Box 5800, NL-6202, Maastricht, AZ, the Netherlands.,CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - P Geusens
- Department of Internal Medicine, Division of Rheumatology, Maastricht University Medical Centre, P.O. Box 5800, NL-6202, Maastricht, AZ, the Netherlands.,CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands.,Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - D Loeffen
- Department of Radiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - R Weijers
- Department of Radiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - S K Boyd
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - C Barnabe
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - K S Stok
- Department of Biomedical Engineering, the University of Melbourne, Melbourne, Australia
| | - B van Rietbergen
- Faculty of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.,Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - J van den Bergh
- Department of Internal Medicine, Division of Rheumatology, Maastricht University Medical Centre, P.O. Box 5800, NL-6202, Maastricht, AZ, the Netherlands.,NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.,Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.,Department of Internal Medicine, VieCuri Medical Centre, Venlo, the Netherlands
| |
Collapse
|
16
|
Williams B, Dharmapatni A, Crotti T. Intracellular apoptotic pathways: a potential target for reducing joint damage in rheumatoid arthritis. Inflamm Res 2017; 67:219-231. [DOI: 10.1007/s00011-017-1116-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/19/2017] [Accepted: 11/13/2017] [Indexed: 12/24/2022] Open
|
17
|
Boleto G, Dramé M, Lambrecht I, Eschard JP, Salmon JH. Disease-modifying anti-rheumatic drug effect of denosumab on radiographic progression in rheumatoid arthritis: a systematic review of the literature. Clin Rheumatol 2017; 36:1699-1706. [DOI: 10.1007/s10067-017-3722-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/04/2017] [Indexed: 12/31/2022]
|
18
|
Williams B, Tsangari E, Stansborough R, Marino V, Cantley M, Dharmapatni A, Gibson R, Perilli E, Crotti T. Mixed effects of caffeic acid phenethyl ester (CAPE) on joint inflammation, bone loss and gastrointestinal inflammation in a murine model of collagen antibody-induced arthritis. Inflammopharmacology 2017; 25:55-68. [PMID: 28044215 DOI: 10.1007/s10787-016-0306-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 12/16/2016] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To investigate the effect of caffeic acid phenethyl ester (CAPE) on local and systemic inflammation and bone loss in collagen antibody-induced arthritis (CAIA) mice. METHODS Four groups of mice (n = 8 per group) were allocated; control, CAPE (1 mg/kg), CAIA and CAIA + CAPE (1 mg/kg). Local inflammation and bone loss were evaluated using clinical paw scores, in vivo micro-computed tomography (micro-CT), histological assessment and tartrate-resistant acid phosphatase (TRAP) staining. Serum levels of C-reactive protein (CRP) and C-terminal telopeptide (CTX-1) were measured by ELISA. Jejunum and colon sections were evaluated histopathologically for damage and toxicity. RESULTS Greater paw scores and percentage change in paw volume were observed in CAIA + CAPE compared to the control groups (p < 0.05). Bone volume over time remained unchanged (p = 0.94) and the number of multinucleated TRAP-positive cells was greatest in CAIA + CAPE mice (p < 0.05). CRP and CTX-1 levels did not differ between groups. CAIA + CAPE mice exhibited lower colon toxicity scores and a reduced percentage of cavitated goblet cells in the colon crypts compared with CAIA mice (p = 0.026 and p = 0.003, respectively). Histopathology in the jejunum was not altered. CONCLUSION CAPE did not reduce paw inflammation or bone loss in CAIA mice. CAPE reduced histopathological changes in the colon of CAIA mice.
Collapse
Affiliation(s)
- Bonnie Williams
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
| | - Eleni Tsangari
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Romany Stansborough
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Victor Marino
- School of Dentistry, The University of Adelaide, Adelaide, SA, Australia
| | - Melissa Cantley
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Anak Dharmapatni
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Rachel Gibson
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Egon Perilli
- School of Computer Science, Engineering and Mathematics, Flinders University, Adelaide, SA, Australia
| | - Tania Crotti
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
19
|
Strehl C, Maurizi L, Gaber T, Hoff P, Broschard T, Poole AR, Hofmann H, Buttgereit F. Modification of the surface of superparamagnetic iron oxide nanoparticles to enable their safe application in humans. Int J Nanomedicine 2016; 11:5883-5896. [PMID: 27877036 PMCID: PMC5110004 DOI: 10.2147/ijn.s110579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Combined individually tailored methods for diagnosis and therapy (theragnostics) could be beneficial in destructive diseases, such as rheumatoid arthritis. Nanoparticles are promising candidates for theragnostics due to their excellent biocompatibility. Nanoparticle modifications, such as improved surface coating, are in development to meet various requirements, although safety concerns mean that modified nanoparticles require further review before their use in medical applications is permitted. We have previously demonstrated that iron oxide nanoparticles with amino-polyvinyl alcohol (a-PVA) adsorbed on their surfaces have the unwanted effect of increasing human immune cell cytokine secretion. We hypothesized that this immune response was caused by free-floating PVA. The aim of the present study was to prevent unwanted immune reactions by further surface modification of the a-PVA nanoparticles. After cross-linking of PVA to nanoparticles to produce PVA-grafted nanoparticles, and reduction of their zeta potential, the effects on cell viability and cytokine secretion were analyzed. PVA-grafted nanoparticles still stimulated elevated cytokine secretion from human immune cells; however, this was inhibited after reduction of the zeta potential. In conclusion, covalent cross-linking of PVA to nanoparticles and adjustment of the surface charge rendered them nontoxic to immune cells, nonimmunogenic, and potentially suitable for use as theragnostic agents.
Collapse
Affiliation(s)
- Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| | - Lionel Maurizi
- Powder Technology Laboratory, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| | - Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| | - Thomas Broschard
- Non-Clinical Safety, Merck Serono, Merck KGaA, Darmstadt, Germany
| | - A Robin Poole
- Department of Surgery, McGill University, Montreal QC, Canada
| | - Heinrich Hofmann
- Powder Technology Laboratory, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| |
Collapse
|
20
|
Mohammad HR, Bhatti W, Pillai A. An unusual presentation of arteriovenous malformation as an erosive midfoot lesion. J Surg Case Rep 2016; 2016:rjw146. [PMID: 27605662 PMCID: PMC5635607 DOI: 10.1093/jscr/rjw146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Erosive bony lesions are radiographic findings where localized bone resorption and cortical line breakage occurs. One less common cause of bone erosions is arteriovenous malformations (AVMs). This should always be included in the differentials for foot pain. A 33-year-old gentleman presented with a 5-year history of chronic left foot pain. Clinical examination was unremarkable. Magnetic resonance imaging (MRI) and computerized tomography demonstrated erosive bone changes in the navicular, the intermediate and lateral cuneiforms bones and their corresponding metatarsal bases. An ultrasound and magnetic resonance angiogram demonstrated high signal showing the abnormal communication between metatarsal artery and vein at the site of most pain confirming the AVM. This was subsequently successfully treated with sclerotherapy. Clinicians should be aware of the history, symptoms and signs of AVMs and consider the use of MRI with or without digital subtraction angiography in making a definitive diagnosis.
Collapse
Affiliation(s)
- Hasan Raza Mohammad
- Department of Orthopaedics, University Hospital South Manchester, Manchester, UK
| | - Waqar Bhatti
- Department of Musculoskeletal Radiology, University Hospital South Manchester, Manchester, UK
| | - Anand Pillai
- Department of Orthopaedics, University Hospital South Manchester, Manchester, UK
| |
Collapse
|
21
|
Gudmann NS, Munk HL, Christensen AF, Ejstrup L, Sørensen GL, Loft AG, Karsdal MA, Bay-Jensen AC, He Y, Siebuhr AS, Junker P. Chondrocyte activity is increased in psoriatic arthritis and axial spondyloarthritis. Arthritis Res Ther 2016; 18:141. [PMID: 27306080 PMCID: PMC4910260 DOI: 10.1186/s13075-016-1040-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/31/2016] [Indexed: 01/22/2023] Open
Abstract
Background Psoriatic arthritis (PsA) and axial spondyloarthritis (axSpA) are chronic inflammatory rheumatic diseases with complex origins. Both are characterized by altered extracellular matrix remodeling in joints and entheses that results in destructive and osteochondral proliferative lesions. There is a need for biomarkers reflecting core disease pathways for diagnosis and disease mapping. Pro-C2 reflects mature cartilage collagen type IIB formation, while C-Col10 represents turnover of type X collagen, which is exclusively expressed by hypertrophic chondrocytes. The objectives of this study were to study cartilage metabolism in axSpA and PsA by assessing Pro-C2 and C-Col10 and to evaluate their diagnostic utility against a healthy reference population. Methods Patients with PsA (n = 101) or axSpA (n = 110) were recruited consecutively from three rheumatology outpatient clinics. Demographic and clinical disease measures were recorded. Pro-C2 and C-Col10 were quantified in serum by using newly developed and specific competitive enzyme-linked immunosorbent assays based on monoclonal antibodies. One-way analysis of variance and Tukey’s multiple comparison tests were performed on log-transformed data. ROC curve analysis was carried out to evaluate their discriminative power. Results Pro-C2 levels in serum were significantly increased in both axSpA (median concentration 1.11 ng/ml, 0.67–1.64) and PsA (median concentration 1.03 ng/ml, 0.53–1.47) compared with healthy controls (median concentration 0.30 ng/ml, 0.16–0.41) (p < 0.0001). Pro-C2 did not differ according to treatment. C-Col10 was slightly but equally elevated in the PsA and axSpA groups vs. the control group, but it was significantly lower in patients with axSpA undergoing tumor necrosis factor-α inhibitor (TNFi) treatment. ROC curve analysis revealed AUCs of 0.85 (95 % CI 0.79–0.89) for axSpA and 0.81 (95 % CI 0.75–0.86) for PsA. Conclusions These findings indicate that cartilage collagen metabolism was enhanced in the axSpA and PsA groups compared with the healthy control group. The lower C-Col10 level in patients with axSpA undergoing TNFi treatment may reflect that hypertrophic chondrocytes in axSpA are targeted by TNFi. ROC curve analysis showed a diagnostic potential for Pro-C2 in axSpA and PsA.
Collapse
Affiliation(s)
- Natasja Stæhr Gudmann
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, 2730, Denmark.
| | | | | | - Leif Ejstrup
- Department of Rheumatology, Esbjerg Hospital, Esbjerg, Denmark
| | - Grith Lykke Sørensen
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Anne Gitte Loft
- Department of Rheumatology, Vejle Hospital, Vejle, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Asser Karsdal
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, 2730, Denmark
| | | | - Yi He
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, 2730, Denmark
| | - Anne Sofie Siebuhr
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, 2730, Denmark
| | - Peter Junker
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
22
|
Strehl C, Schellmann S, Maurizi L, Hofmann-Amtenbrink M, Häupl T, Hofmann H, Buttgereit F, Gaber T. Effects of PVA-coated nanoparticles on human T helper cell activity. Toxicol Lett 2016; 245:52-8. [DOI: 10.1016/j.toxlet.2016.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/05/2016] [Accepted: 01/08/2016] [Indexed: 10/22/2022]
|
23
|
Park R, Ji JD. Calcium channels: the potential therapeutic targets for inflammatory bone destruction of rheumatoid arthritis. Inflamm Res 2016; 65:347-54. [PMID: 26852086 DOI: 10.1007/s00011-016-0920-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/23/2016] [Accepted: 01/26/2016] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Inflammatory bone resorption causes progressive joint destruction which ultimately leads to functional disability in rheumatoid arthritis (RA). The primary cell responsible for bone resorption is the osteoclast, which means it is a potential therapeutic target against bone destruction. In fact, experimental and clinical findings suggest that blockade of osteoclast differentiation and function is highly effective in inhibiting bone destruction in RA. DISCUSSION AND CONCLUSION In this report, we show several lines of experimental evidence which suggest that a variety of Ca(2+) channels are essential in osteoclast differentiation and function, and present a hypothesis that modulation of Ca(2+) channels is a highly effective therapeutic strategy in preventing osteoclast-induced structural damage in RA.
Collapse
Affiliation(s)
- Robin Park
- Division of Rheumatology, College of Medicine, Korea University, 126-1, Anam-Dong 5-Ga, Sungbuk-Ku, Seoul, 136-705, South Korea
| | - Jong Dae Ji
- Division of Rheumatology, College of Medicine, Korea University, 126-1, Anam-Dong 5-Ga, Sungbuk-Ku, Seoul, 136-705, South Korea.
| |
Collapse
|
24
|
Strehl C, Gaber T, Maurizi L, Hahne M, Rauch R, Hoff P, Häupl T, Hofmann-Amtenbrink M, Poole AR, Hofmann H, Buttgereit F. Effects of PVA coated nanoparticles on human immune cells. Int J Nanomedicine 2015; 10:3429-45. [PMID: 26056442 PMCID: PMC4431506 DOI: 10.2147/ijn.s75936] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Nanotechnology provides new opportunities in human medicine, mainly for diagnostic and therapeutic purposes. The autoimmune disease rheumatoid arthritis (RA) is often diagnosed after irreversible joint structural damage has occurred. There is an urgent need for a very early diagnosis of RA, which can be achieved by more sensitive imaging methods. Superparamagnetic iron oxide nanoparticles (SPION) are already used in medicine and therefore represent a promising tool for early diagnosis of RA. The focus of our work was to investigate any potentially negative effects resulting from the interactions of newly developed amino-functionalized amino-polyvinyl alcohol coated (a-PVA) SPION (a-PVA-SPION), that are used for imaging, with human immune cells. We analyzed the influence of a-PVA-SPION with regard to cell survival and cell activation in human whole blood in general, and in human monocytes and macrophages representative of professional phagocytes, using flow cytometry, multiplex suspension array, and transmission electron microscopy. We found no effect of a-PVA-SPION on the viability of human immune cells, but cytokine secretion was affected. We further demonstrated that the percentage of viable macrophages increased on exposure to a-PVA-SPION. This effect was even stronger when a-PVA-SPION were added very early in the differentiation process. Additionally, transmission electron microscopy analysis revealed that both monocytes and macrophages are able to endocytose a-PVA-SPION. Our findings demonstrate an interaction between human immune cells and a-PVA-SPION which needs to be taken into account when considering the use of a-PVA-SPION in human medicine.
Collapse
Affiliation(s)
- Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany ; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany ; German Rheumatism Research Centre (DRFZ), Berlin, Germany ; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Lionel Maurizi
- Powder Technology Laboratory, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Hahne
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany ; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| | - Roman Rauch
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany ; German Rheumatism Research Centre (DRFZ), Berlin, Germany
| | - Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany ; German Rheumatism Research Centre (DRFZ), Berlin, Germany ; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Thomas Häupl
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - A Robin Poole
- Department of Surgery, McGill University, Montreal, QC, Canada
| | - Heinrich Hofmann
- Powder Technology Laboratory, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany ; German Rheumatism Research Centre (DRFZ), Berlin, Germany ; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| |
Collapse
|
25
|
Cantley MD, Fairlie DP, Bartold PM, Marino V, Gupta PK, Haynes DR. Inhibiting histone deacetylase 1 suppresses both inflammation and bone loss in arthritis. Rheumatology (Oxford) 2015; 54:1713-23. [PMID: 25832610 DOI: 10.1093/rheumatology/kev022] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Histone deacetylase 1 (HDAC1) is highly expressed in the synovium of RA patients. Thus we aimed to investigate a novel HDAC inhibitor (HDACi), NW-21, designed to target HDAC1. The effect of NW-21 on osteoclast formation and activity, cytokine and chemokine expression in vitro and arthritis in mice was assessed. METHODS The effects on human osteoclast formation and activity derived from human blood monocytes stimulated with receptor activator of nuclear factor κB ligand (RANKL) and M-CSF were assessed. The anti-inflammatory activity of NW-21 was assessed using human monocytes stimulated with either TNF-α or lipopolysaccharide for 24 h. mRNA expression of monocyte chemotactic protein 1 (MCP-1), TNF-α, macrophage inflammatory protein 1α (MIP-1α), IL-1 and RANTES (regulated on activation, normal T cell expressed and secreted) was assessed. The effect of NW-21 in the collagen antibody-induced arthritis model was assessed following daily oral administration at 5 mg/kg/day. The HDAC1 inhibitors NW-21 and MS-275 were compared with a broad-acting HDACi, 1179.4b. Effects on inflammation and bone were assessed using paw inflammation scoring, histology and live animal micro-CT. RESULTS NW-21 suppressed osteoclast formation and activity as well as significantly reducing mRNA expression of MCP-1 and MIP-1α in monocytes stimulated by lipopolysaccharide or TNF-α (P < 0.05) in vitro. Only inhibitors that targeted HDAC1 (NW-21 and MS-275) reduced inflammation and bone loss in the arthritis model. CONCLUSION The results indicate that inhibitors targeting HDAC1, such as NW-21 and MS-275, may be useful for treating RA, as such drugs can simultaneously target both inflammation and bone resorption.
Collapse
Affiliation(s)
- Melissa D Cantley
- Discipline of Anatomy and Pathology, School of Medical Sciences, University of Adelaide, Adelaide, SA,
| | - David P Fairlie
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, and
| | - P Mark Bartold
- Colgate Australian Clinical Dental Research Centre, School of Dentistry, University of Adelaide, Adelaide, SA, Australia
| | - Victor Marino
- Colgate Australian Clinical Dental Research Centre, School of Dentistry, University of Adelaide, Adelaide, SA, Australia
| | - Praveer K Gupta
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, and
| | - David R Haynes
- Discipline of Anatomy and Pathology, School of Medical Sciences, University of Adelaide, Adelaide, SA
| |
Collapse
|
26
|
Morado-Urbina CE, Alvarado-Vázquez PA, Montiel-Ruiz RM, Acosta-González RI, Castañeda-Corral G, Jiménez-Andrade JM. Early, Middle, or Late Administration of Zoledronate Alleviates Spontaneous Nociceptive Behavior and Restores Functional Outcomes in a Mouse Model of CFA-Induced Arthritis. Drug Dev Res 2014; 75:438-48. [DOI: 10.1002/ddr.21183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/06/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Carlos Eduardo Morado-Urbina
- Unidad Académica Multidisciplinaria Reynosa Aztlán; Universidad Autónoma de Tamaulipas; Reynosa Tamaulipas México
| | | | - Rosa Mariana Montiel-Ruiz
- Unidad Académica Multidisciplinaria Reynosa Aztlán; Universidad Autónoma de Tamaulipas; Reynosa Tamaulipas México
| | - Rosa Issel Acosta-González
- Unidad Académica Multidisciplinaria Reynosa Aztlán; Universidad Autónoma de Tamaulipas; Reynosa Tamaulipas México
| | | | - Juan Miguel Jiménez-Andrade
- Unidad Académica Multidisciplinaria Reynosa Aztlán; Universidad Autónoma de Tamaulipas; Reynosa Tamaulipas México
| |
Collapse
|
27
|
Kuriyama K, Hashimoto J, Murase T, Fujii M, Nampei A, Hirao M, Tsuboi H, Myoui A, Yoshikawa H. Treatment of juxta-articular intraosseous cystic lesions in rheumatoid arthritis patients with interconnected porous calcium hydroxyapatite ceramic. Mod Rheumatol 2014. [DOI: 10.3109/s10165-008-0147-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Kohji Kuriyama
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital,
Toyonaka, Japan
| | - Jun Hashimoto
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine,
2-2 Yamadaoka, Suita 565-0871, Japan
| | - Tsuyoshi Murase
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine,
2-2 Yamadaoka, Suita 565-0871, Japan
| | - Masakazu Fujii
- Department of Orthopaedic Surgery, Gratia Hospital,
Minoh, Japan
| | - Akihide Nampei
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine,
2-2 Yamadaoka, Suita 565-0871, Japan
| | - Makoto Hirao
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine,
2-2 Yamadaoka, Suita 565-0871, Japan
| | - Hideki Tsuboi
- Department of Orthopaedic Surgery, Osaka Rosai Hospital,
Sakai, Japan
| | - Akira Myoui
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine,
2-2 Yamadaoka, Suita 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine,
2-2 Yamadaoka, Suita 565-0871, Japan
| |
Collapse
|
28
|
Abstract
The transcription factor NF-κB is a family of proteins involved in signaling pathways essential for normal cellular functions and development. Deletion of various components of this pathway resulted with abnormal skeletal development. Research in the last decade has established that NF-κB signaling mediates RANK ligand-induced osteoclastogenesis. Consistently, it was shown that inhibition of NF-κB was an effective approach to inhibit osteoclast formation and bone resorptive activity. Identification of the molecular machinery underlying NF-κB activation permitted osteoclast-specific deletion of the major components of this pathway. As a result, it was clear that deletion of members of the proximal IKK kinase complex and the distal NF-κB subunits and downstream regulators affected skeletal development. These studies provided several targets of therapeutic intervention in osteolytic diseases. NF-κB activity has been also described as the centerpiece of inflammatory responses and is considered a potent mediator of inflammatory osteolysis. Indeed, inflammatory insults exacerbate physiologic RANKL-induced NF-κB signals leading to exaggerated responses and to inflammatory osteolysis. These superimposed NF-κB activities appear to underlie several bone pathologies. This review will describe the individual roles of NF-κB molecules in bone resorption and inflammatory osteolysis.
Collapse
Affiliation(s)
- Y Abu-Amer
- Department of Orthopedic Surgery, Department of Cell Biology & Physiology, Washington University School of Medicine, 660S. Euclid Avenue, Saint Louis, MO 63110, USA.
| |
Collapse
|
29
|
Norisoboldine suppresses osteoclast differentiation through preventing the accumulation of TRAF6-TAK1 complexes and activation of MAPKs/NF-κB/c-Fos/NFATc1 Pathways. PLoS One 2013; 8:e59171. [PMID: 23536866 PMCID: PMC3594163 DOI: 10.1371/journal.pone.0059171] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/12/2013] [Indexed: 11/19/2022] Open
Abstract
Norisoboldine (NOR) is the main alkaloid constituent in the dry root of Lindera aggregata (Sims) Kosterm. (L. strychnifolia Vill.). As reported previously, orally administered NOR displayed a robust inhibition of joint bone destruction present in both mouse collagen-induced arthritis and rat adjuvant-induced arthritis with lower efficacious doses than that required for ameliorating systemic inflammation. This attracted us to assess the effects of NOR on differentiation and function of osteoclasts, primary effector cells for inflammatory bone destruction, to get insight into its anti-rheumatoid arthritis mechanisms. Both RAW264.7 cells and mouse bone marrow-derived macrophages (BMMs) were stimulated with RANKL (100 ng/mL) to establish osteoclast differentiation models. ELISA, RT-PCR, gelatin zymography, western blotting, immunoprecipitation and EMSA were used to reveal related signalling pathways. NOR (10 and 30 µM), without significant cytotoxicity, showed significant reduction of the number of osteoclasts and the resorption pit areas, and it targeted osteoclast differentiation at the early stage. In conjunction with the anti-resorption effect of NOR, mRNA levels of cathepsin K and MMP-9 were decreased, and the activity of MMP-9 was attenuated. Furthermore, our mechanistic studies indicated that NOR obviously suppressed the ubiquitination of TRAF6, the accumulation of TRAF6-TAK1 complexes and the activation of ERK and p38 MAPK, and reduced the nuclear translocation of NF-κB-p65 and DNA-binding activity of NF-κB. However, NOR had little effect on expressions of TRAF6 or the phosphorylation and degradation of IκBα. Moreover, NOR markedly inhibited expressions of transcription factor NFATc1, but not c-Fos. Intriguingly, the subsequent nuclear translocations of c-Fos and NFATc1 were substantially down-regulated. Hence, we demonstrated for the first time that preventing the differentiation and function of osteoclasts at the early stage was an important anti-bone destruction mechanism of NOR, which might be attributed to inhibition of ubiquitination of TRAF6, the accumulation of TRAF6-TAK1 complexes and the activation of MAPKs/NF-κB/c-Fos/NFATc1 pathways.
Collapse
|
30
|
Choi YS, Kang EH, Lee EY, Gong HS, Kang HS, Shin K, Lee EB, Song YW, Lee YJ. Joint-protective effects of compound K, a major ginsenoside metabolite, in rheumatoid arthritis: in vitro evidence. Rheumatol Int 2013; 33:1981-90. [DOI: 10.1007/s00296-013-2664-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 01/02/2013] [Indexed: 12/17/2022]
|
31
|
Forslind K, Kälvesten J, Hafström I, Svensson B. Does digital X-ray radiogrammetry have a role in identifying patients at increased risk for joint destruction in early rheumatoid arthritis? Arthritis Res Ther 2012; 14:R219. [PMID: 23068060 PMCID: PMC4060357 DOI: 10.1186/ar4058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 09/25/2012] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION The aim of this study was to investigate the role of hand bone mineral density (BMD) loss analyzed with digital X-ray radiogrammetry (DXR) in early rheumatoid arthritis (RA) as a predictor for progression of joint damage. METHODS In 379 patients with early RA, baseline and one-year hand BMD was measured with DXR and the hand bone loss (HBL) was analyzed using the smallest detectable change (HBLsdc) and tertiles (HBLtertiles). Joint damage in hands and feet were scored according to the Sharp van der Heijde (SHS) method at baseline and at one, two, five and eight years. At the same time-points Disease Activity Score (DAS28) was calculated and functional disability assessed. Rheumatoid factor (RF) and antibodies against cyclic citrullinated peptides (anti-CCP) were analyzed at baseline. RESULTS Sixty-six percent of the patients had hand BMD loss in the first year of RA determined by HBLsdc and 65% by HBLtertiles. Radiographic progression after two, five and eight years was associated with hand bone loss defined by HBLsdc. By HBLtertiles there were significant associations at all time-points except at eight years. The change in DXR at one year (ChDXR1yr) correlated significantly and inversely with the change in SHS (ChSHS) at two, five and eight years. Multivariate analysis showed that only change in SHS during the first year and the presence of anti-CCP were independent predictors of long-term progressive joint damage. If radiographic scores were not included, DXR-BMD loss was an independent predictor. Patients with great bone loss by HBLtertiles had significantly more often high disease activity after two years. However, neither bone loss by HBLsdc or HBLtertiles nor by ChDXR1yr was an independent predictor of remission after two, five and eight years. CONCLUSIONS This study confirms previous reports of an association of decrease in DXR-BMD during the first disease year with progression of radiographic joint damage over an extended period of time. This association was independent in a regression model only when radiological findings were excluded suggesting a possible predictive role of DXR-BMD in clinical practice when radiographic evaluation is not available. However, further studies are required before this can be established.
Collapse
|
32
|
Karsdal MA, Bay-Jensen AC, Henriksen K, Christiansen C. The pathogenesis of osteoarthritis involves bone, cartilage and synovial inflammation: may estrogen be a magic bullet? ACTA ACUST UNITED AC 2012; 18:139-46. [PMID: 23024184 DOI: 10.1258/mi.2012.012025] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The female predominance of polyarticular osteoarthritis (OA), and in particular the marked increase of OA in women after the menopause points to a likely involvement of female sex hormones in the maintenance of cartilage homeostasis. This perception has inspired many research groups to investigate the role of estrogens in the modulation of cartilage homeostasis with the ultimate aim to clarify whether estrogen replacement therapy (ERT) could provide benefits in preventing the rapid rise in the prevalence of OA in postmenopausal women. The effects of ERT and selective estrogen-receptor modulators on the joint in various experimental models have been investigated. Clinically, the effects of estrogens have been evaluated by post hoc analysis in clinical trials using biochemical markers of cartilage and bone degradation. Lastly, the Women's Health Initiative trial (WHI) investigated the effects of estrogens on the joint and joint replacements. Even though the exact mode of action still needs to be elucidated, the effect involves both direct and indirect mechanisms on the whole joint pathophysiology. Several animal models have demonstrated structural benefits of estrogens, as well as significant effects on joint inflammation. This is in complete alignment with clinical data using biochemical markers of joint degradation which demonstrated approximately 50% inhibition of cartilage destruction. These finding were recently validated in WHI, where women taking estrogens had significantly less joint replacement. In conclusion, the pleiotropic effect of estrogens on several different tissues may match the complicated aetiology of OA in some important aspects.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | | | | | | |
Collapse
|
33
|
Combined therapeutic application of mTOR inhibitor and vitamin D(3) for inflammatory bone destruction of rheumatoid arthritis. Med Hypotheses 2012; 79:757-60. [PMID: 22967804 DOI: 10.1016/j.mehy.2012.08.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 08/21/2012] [Indexed: 11/21/2022]
Abstract
Inflammatory bone destruction is a prominent feature and a cause of substantial morbidity in several inflammatory diseases, including rheumatoid arthritis (RA), periodontitis, and peri-prosthetic loosening. Osteoclasts are unique, multinucleated giant cells that effectively resorb bone and thus are directly responsible for bone destruction in several inflammatory diseases. PI3K/Akt/mTOR pathway has been well known to play important roles in regulating adaptive and innate immune cell function. In addition to play roles in immune responses, several lines of evidence demonstrate that PI3K/Akt/mTOR pathway is critical for osteoclast differentiation and survival. These results suggest that inhibition of PI3K/Akt/mTOR pathway could protect against bone destruction in inflammatory diseases, including RA. However, the clinical use of mTOR inhibitors may be hampered due to limited clinical efficacy and frequent toxic side effects. In the treatment of RA, combination therapy with various disease-modifying antirheumatic drugs (DMARDs) has been suggested to improve the therapeutic efficacy and limit the side effects. In this report, we show several experimental evidences that vitamin D(3) modulates mTOR pathway, and present a hypothesis that the combination of mTOR inhibitor and vitamin D(3) can effectively inhibit osteoclast differentiation and function in chronic inflammatory condition such as RA, therefore this combination will be a powerful therapeutic regimen in preventing the inflammation-induced bone destruction in RA.
Collapse
|
34
|
Karsdal MA, Schett G, Emery P, Harari O, Byrjalsen I, Kenwright A, Bay-Jensen AC, Platt A. IL-6 receptor inhibition positively modulates bone balance in rheumatoid arthritis patients with an inadequate response to anti-tumor necrosis factor therapy: biochemical marker analysis of bone metabolism in the tocilizumab RADIATE study (NCT00106522). Semin Arthritis Rheum 2012; 42:131-9. [PMID: 22397953 DOI: 10.1016/j.semarthrit.2012.01.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/13/2012] [Accepted: 01/16/2012] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To evaluate changes in biochemical markers of bone metabolism in response to tocilizumab in patients with anti-tumor necrosis factor-refractory rheumatoid arthritis (RA). METHODS RADIATE was a randomized, double-blind, placebo-controlled, parallel-group phase 3 trial. C-reactive protein, osteocalcin (OC), C-terminal telopeptides of type-I collagen (C-terminal telopeptides of type-1 collagen (CTX-I) and type-I collagen degradation product), and matrix metalloproteinase-3 (MMP-3) serum levels were analyzed from 299 RA patients. Patients were randomly assigned to either tocilizumab (4 or 8 mg/kg) or placebo intravenously every 4 weeks, along with concomitant stable methotrexate (10 to 25 mg weekly) in all treatment arms. The change in biochemical markers CTX-I and OC in combination was evaluated as a measure of net bone balance, a reflection of the change in equilibrium between resorption and formation. RESULTS Both tocilizumab doses decreased C-reactive protein levels and significantly inhibited cathepsin K-mediated bone resorption in RADIATE subjects, as measured by a decrease in CTX-I. There was a significant overall improvement in net bone balance at week 16 as measured by a decrease in the CTX-I:OC ratio (-25%, P < 0.01). Furthermore, a significant reduction in MMP-3 (43%, P < 0.001) and type-I collagen degradation product levels (18%, P < 0.001) were observed following treatment, both consistent with decreased MMP-mediated type-I collagen catabolism in joint tissue. CONCLUSIONS In anti-tumor necrosis factor-refractory patients, tocilizumab significantly reduced the levels of biochemical markers of cathepsin K-mediated bone resorption and MMP-mediated tissue degradation and remodeling. These observations suggest that tocilizumab has a positive effect on bone balance, which could in part explain the retardation of progressive structural damage observed with tocilizumab. Clinical trial registry number: NCT00106522.
Collapse
|
35
|
Bay-Jensen AC, Leeming DJ, Kleyer A, Veidal SS, Schett G, Karsdal MA. Ankylosing spondylitis is characterized by an increased turnover of several different metalloproteinase-derived collagen species: a cross-sectional study. Rheumatol Int 2011; 32:3565-72. [DOI: 10.1007/s00296-011-2237-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 10/22/2011] [Indexed: 12/17/2022]
|
36
|
Forsblad-d'Elia H, Carlsten H. Bone mineral density by digital X-ray radiogrammetry is strongly decreased and associated with joint destruction in long-standing rheumatoid arthritis: a cross-sectional study. BMC Musculoskelet Disord 2011; 12:242. [PMID: 22024200 PMCID: PMC3226572 DOI: 10.1186/1471-2474-12-242] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 10/24/2011] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The aims were to explore bone mineral density (BMD) by digital X-ray radiogrammetry (DXR) in postmenopausal women with long-lasting rheumatoid arthritis (RA) in relation to dual x-ray absorptiometry (DXA)-BMD, joint destruction by conventional radiographs and disease related variables in a cross-sectional study. METHODS Seventy-five postmenopausal women with RA were examined by DXA measuring DXA-BMD of the forearm, total hip and lumbar spine, by scoring joint destruction on plain radiographs by the method of Larsen and by DXR-BMD in metacarpals two to four. The DXR-BMD results of the RA women were compared with an age and sex-matched reference database. A function of DXR-BMD in relation to age and disease duration was created. Associations were investigated by bivariate and multiple linear regression analyses. RESULTS DXR-BMD was strongly decreased in RA patients compared to the reference database (p < 0.001). Calculations showed that DXR-BMD was not markedly influenced the first years after diagnosis of RA, but between approximately 5-10 years of disease there was a steep decline in DXR-BMD which subsequently levelled off. In multiple regression analyses disease duration, CRP and DXR-BMD were independent variables associated with Larsen score (R2= 0.64). Larsen score and BMD forearm were independent determinants of DXR-BMD (R2 = 0.79). CONCLUSIONS DXR-BMD was strongly reduced and associated with both Larsen score and DXA-BMD forearm in these postmenopausal women with RA implying that DXR-BMD is a technique that reflects both the erosive process and bone loss adjacent to affected joints.
Collapse
Affiliation(s)
- Helena Forsblad-d'Elia
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Centre for Bone and Arthritis Research, Box 480, S-405 30 Gothenburg, Sweden.
| | | |
Collapse
|
37
|
Quirke AM, Fisher BAC, Kinloch AJ, Venables PJ. Citrullination of autoantigens: upstream of TNFα in the pathogenesis of rheumatoid arthritis. FEBS Lett 2011; 585:3681-8. [PMID: 21704035 DOI: 10.1016/j.febslet.2011.06.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 06/06/2011] [Accepted: 06/07/2011] [Indexed: 12/23/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterised by synovial inflammation and destruction of joints. Over 20 years ago, tumour necrosis factor alpha (TNFα) was identified as a key player in a cytokine network, whose multifunctional effects could account for both the inflammation and destruction in RA. The remarkable efficacy of TNF inhibitors in the treatment of RA has resulted in extensive research addressing the regulation of TNFα production responsible for this excessive production. The discovery of autoimmunity to citrullinated protein/peptide antigens (ACPA) has led the concept that ACPA may be the essential link between disease susceptibility factors and the production of TNFα, which ultimately accounts for the disease phenotype. In this review we will consider (1) the mechanisms of citrullination, both physiological and pathological, (2) how known genetic and environmental factors could drive this peculiar form of autoimmunity and (3) how the immune response could lead to excessive production of TNFα by the synovial cells and ultimately to the disease phenotype (Fig. 1).
Collapse
Affiliation(s)
- Anne-Marie Quirke
- Kennedy Institute of Rheumatology, Imperial College London, London, UK
| | | | | | | |
Collapse
|
38
|
Ruperto N, Martini A. Emerging drugs to treat juvenile idiopathic arthritis. Expert Opin Emerg Drugs 2011; 16:493-505. [DOI: 10.1517/14728214.2011.581662] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Karsdal MA, Woodworth T, Henriksen K, Maksymowych WP, Genant H, Vergnaud P, Christiansen C, Schubert T, Qvist P, Schett G, Platt A, Bay-Jensen AC. Biochemical markers of ongoing joint damage in rheumatoid arthritis--current and future applications, limitations and opportunities. Arthritis Res Ther 2011; 13:215. [PMID: 21539724 PMCID: PMC3132026 DOI: 10.1186/ar3280] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease associated with potentially debilitating joint inflammation, as well as altered skeletal bone metabolism and co-morbid conditions. Early diagnosis and aggressive treatment to control disease activity offers the highest likelihood of preserving function and preventing disability. Joint inflammation is characterized by synovitis, osteitis, and/or peri-articular osteopenia, often accompanied by development of subchondral bone erosions, as well as progressive joint space narrowing. Biochemical markers of joint cartilage and bone degradation may enable timely detection and assessment of ongoing joint damage, and their use in facilitating treatment strategies is under investigation. Early detection of joint damage may be assisted by the characterization of biochemical markers that identify patients whose joint damage is progressing rapidly and who are thus most in need of aggressive treatment, and that, alone or in combination, identify those individuals who are likely to respond best to a potential treatment, both in terms of limiting joint damage and relieving symptoms. The aims of this review are to describe currently available biochemical markers of joint metabolism in relation to the pathobiology of joint damage and systemic bone loss in RA; to assess the limitations of, and need for additional, novel biochemical markers in RA and other rheumatic diseases, and the strategies used for assay development; and to examine the feasibility of advancement of personalized health care using biochemical markers to select therapeutic agents to which a patient is most likely to respond.
Collapse
Affiliation(s)
- Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
- Southern University of Denmark (SDU), Campusvej 55DK-5230 Odense M Denmark
| | - Thasia Woodworth
- Leading Edge Clinical Research LLC, 3901 SE St Lucie Blvd unit 20, Stuart, Florida 34997, USA
| | - Kim Henriksen
- Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Walter P Maksymowych
- University of Alberta, 562 Heritage Medical Research Building, Edmonton, AB T6G 2S2, Canada
| | | | | | | | | | - Per Qvist
- Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Georg Schett
- University of Erlangen-Nurnberg Department of Internal Medicine, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Adam Platt
- Roche Products Limited, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, UK
| | | |
Collapse
|
40
|
Kim HR, Kim KW, Jung HG, Yoon KS, Oh HJ, Cho ML, Lee SH. Macrophage migration inhibitory factor enhances osteoclastogenesis through upregulation of RANKL expression from fibroblast-like synoviocytes in patients with rheumatoid arthritis. Arthritis Res Ther 2011; 13:R43. [PMID: 21401926 PMCID: PMC3132025 DOI: 10.1186/ar3279] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 02/09/2011] [Accepted: 03/14/2011] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Macrophage migration inhibitory factor (MIF) is one of key regulators in acute and chronic immune-inflammatory conditions including rheumatoid arthritis (RA). We examined the effect of MIF on osteoclastogenesis, which is known to play a crucial role in bone destruction in RA. METHODS The concentration of MIF and receptor activator of nuclear factor-κB ligand (RANKL) in the synovial fluid was measured by ELISA. MIF-induced RANKL expression of RA synovial fibroblasts was determined by real-time PCR and western blot. Osteoclastogenesis was analyzed in culture of human peripheral blood mononuclear cells (PBMC) with MIF. Osteoclastogenesis was also determined after co-cultures of rhMIF-stimulated RA synovial fibroblasts with human PBMC. RESULTS Synovial fluid MIF concentration in RA patients was significantly higher than in osteoarthritis (OA) patients. The concentration of RANKL correlated with that of MIF in RA synovial fluids (r = 0.6, P < 0.001). MIF stimulated the expression of RANKL mRNA and protein in RA synovial fibroblasts, which was partially reduced by blocking of interleukin (IL)-1β. Osteoclasts were differentiated from PBMC cultures with MIF and M-CSF, even without RANKL. Osteoclastogenesis was increased after co-culture of MIF-stimulated RA synovial fibroblasts with PBMC and this effect was diminished by RANKL neutralization. Blocking of PI3 kinase, p38 MAP kinase, JAK-2, NF-κB, and AP-1 also led to a marked reduction in RANKL expression and osteoclastogenesis. CONCLUSIONS The interactions among MIF, synovial fibroblasts, osteoclasts, RANKL, and IL-1β have a close connection in osteoclastogenesis and they could be a potential gateway leading to new therapeutic approaches in treating bone destruction in RA.
Collapse
Affiliation(s)
- Hae-Rim Kim
- Division of Rheumatology, Medical Immunology Center, Department of Internal Medicine, Konkuk University School of Medicine, 1 Hwayang-dong, Kwangjin-gu, Seoul 143-729, Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Receptor activator of nuclear factor kappa B ligand serum and synovial fluid level. A comparative study between rheumatoid arthritis and osteoarthritis. Rheumatol Int 2011; 32:1589-96. [DOI: 10.1007/s00296-011-1831-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 01/30/2011] [Indexed: 10/18/2022]
|
42
|
Henriksen K, Bollerslev J, Everts V, Karsdal MA. Osteoclast activity and subtypes as a function of physiology and pathology--implications for future treatments of osteoporosis. Endocr Rev 2011; 32:31-63. [PMID: 20851921 DOI: 10.1210/er.2010-0006] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoclasts have traditionally been associated exclusively with catabolic functions that are a prerequisite for bone resorption. However, emerging data suggest that osteoclasts also carry out functions that are important for optimal bone formation and bone quality. Moreover, recent findings indicate that osteoclasts have different subtypes depending on their location, genotype, and possibly in response to drug intervention. The aim of the current review is to describe the subtypes of osteoclasts in four different settings: 1) physiological, in relation to turnover of different bone types; 2) pathological, as exemplified by monogenomic disorders; 3) pathological, as identified by different disorders; and 4) in drug-induced situations. The profiles of these subtypes strongly suggest that these osteoclasts belong to a heterogeneous cell population, namely, a diverse macrophage-associated cell type with bone catabolic and anabolic functions that are dependent on both local and systemic parameters. Further insight into these osteoclast subtypes may be important for understanding cell-cell communication in the bone microenvironment, treatment effects, and ultimately bone quality.
Collapse
Affiliation(s)
- K Henriksen
- Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | | | | | | |
Collapse
|
43
|
Abstract
Osteoarthritis (OA) is a prevalent and disabling condition for which few safe and effective therapeutic options are available. Current approaches are largely palliative and in an effort to mitigate the rising tide of increasing OA prevalence and disease impact, modifying the structural progression of OA has become a focus of drug development. This Review describes disease modification and discusses some of the challenges involved in the discovery and development of disease-modifying OA drugs (DMOADs). A variety of targeted agents are in mature phases of development; specific agents that are beyond preclinical development in phase II and III trials and show promise as potential DMOADs are discussed. A research agenda with respect to disease modification in OA is also provided, and some of the future challenges we face in this field are discussed.
Collapse
Affiliation(s)
- David J Hunter
- Rheumatology Department and Northern Clinical School, Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| |
Collapse
|
44
|
Lee B, Kim TH, Jun JB, Yoo DH, Woo JH, Choi SJ, Lee YH, Song GG, Sohn J, Park-Min KH, Ivashkiv LB, Ji JD. Direct inhibition of human RANK+ osteoclast precursors identifies a homeostatic function of IL-1beta. THE JOURNAL OF IMMUNOLOGY 2010; 185:5926-34. [PMID: 20935210 DOI: 10.4049/jimmunol.1001591] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IL-1β is a key mediator of bone resorption in inflammatory settings, such as rheumatoid arthritis (RA). IL-1β promotes osteoclastogenesis by inducing RANKL expression on stromal cells and synergizing with RANKL to promote later stages of osteoclast differentiation. Because IL-1Rs share a cytosolic Toll-IL-1R domain and common intracellular signaling molecules with TLRs that can directly inhibit early steps of human osteoclast differentiation, we tested whether IL-1β also has suppressive properties on osteoclastogenesis in primary human peripheral blood monocytes and RA synovial macrophages. Early addition of IL-1β, prior to or together with RANKL, strongly inhibited human osteoclastogenesis as assessed by generation of TRAP(+) multinucleated cells. IL-1β acted directly on human osteoclast precursors (OCPs) to strongly suppress expression of RANK, of the costimulatory triggering receptor expressed on myeloid cells 2 receptor, and of the B cell linker adaptor important for transmitting RANK-induced signals. Thus, IL-1β rendered early-stage human OCPs refractory to RANK stimulation. Similar inhibitory effects of IL-1β were observed using RA synovial macrophages. One mechanism of RANK inhibition was IL-1β-induced proteolytic shedding of the M-CSF receptor c-Fms that is required for RANK expression. These results identify a homeostatic function of IL-1β in suppressing early OCPs that contrasts with its well-established role in promoting later stages of osteoclast differentiation. Thus, the rate of IL-1-driven bone destruction in inflammatory diseases, such as RA, can be restrained by its direct inhibitory effects on early OCPs to limit the extent of inflammatory osteolysis.
Collapse
Affiliation(s)
- Bitnara Lee
- Hospital for Rheumatic Diseases, Hanyang University, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cejka D, Hayer S, Niederreiter B, Sieghart W, Fuereder T, Zwerina J, Schett G. Mammalian target of rapamycin signaling is crucial for joint destruction in experimental arthritis and is activated in osteoclasts from patients with rheumatoid arthritis. ACTA ACUST UNITED AC 2010; 62:2294-302. [PMID: 20506288 DOI: 10.1002/art.27504] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Activation of the mammalian target of rapamycin (mTOR) pathway is important for immune cell activation and bone metabolism. To date, the contribution of mTOR signaling to joint inflammation and structural bone and cartilage damage is unknown. The aim of this study was to investigate the potential of inhibiting mTOR as a treatment of inflammatory arthritis. METHODS Human tumor necrosis factor-transgenic mice in which inflammatory arthritis was developing were treated with 2 different mTOR inhibitors, sirolimus or everolimus. The effects of treatment on clinical disease activity, inflammation, and localized joint and cartilage destruction were studied. In addition, the effects of mTOR inhibition on osteoclast survival and expression of key molecules of osteoclast function were analyzed in vitro. Moreover, synovial tissue from patients with rheumatoid arthritis (RA) was assessed for activation of the mTOR pathway. RESULTS Inhibition of mTOR by sirolimus or everolimus reduced synovial osteoclast formation and protected against local bone erosions and cartilage loss. Clinical signs of arthritis improved after mTOR inhibition, and histologic evaluation showed a decrease in synovitis. In vitro, mTOR inhibition down-regulated the expression of digestive enzymes and led to osteoclast apoptosis. Moreover, mTOR signaling was shown to be active in the synovial membrane of patients with RA, particularly in synovial osteoclasts. CONCLUSION Signaling through mTOR is an important link between synovitis and structural damage in inflammatory arthritis. Current pharmacologic inhibitors of mTOR could be effective in protecting joints against structural damage.
Collapse
|
46
|
Le Goff B, Soltner E, Charrier C, Maugars Y, Rédini F, Heymann D, Berthelot JM. A combination of methotrexate and zoledronic acid prevents bone erosions and systemic bone mass loss in collagen induced arthritis. Arthritis Res Ther 2009; 11:R185. [PMID: 20003278 PMCID: PMC3003529 DOI: 10.1186/ar2877] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Revised: 12/03/2009] [Accepted: 12/10/2009] [Indexed: 11/10/2022] Open
Abstract
Introduction Osteoclasts play a key role in the pathogenesis of bone erosion and systemic bone mass loss during rheumatoid arthritis (RA). In this study, we aimed to determine the effect of methotrexate (MTX) and zoledronic acid (ZA), used alone or in combination, on osteoclast-mediated bone erosions and systemic bone mass loss in a rat model of collagen induced arthritis (CIA). We hypothesized that MTX and ZA could have an additive effect to prevent both bone erosion and systemic bone loss. Methods Arthritis was induced in 64 female Sprague-Dawley rats. After the clinical onset of CIA, rats were assigned to treatment with MTX (1 mg/kg/week), ZA (100 μg/kg twice weekly), both treatments at the same regimens, or vehicle. Arthritis score and paw thickness were recorded twice weekly. The rats were sacrificed on D28 and hind paws were removed for radiographic, histological and immunohistochemical analysis. The effects of treatments on osteoclastogenesis were determined by Tartrate resistant acid phosphatase (TRAP) staining. Micro-CT of the tibia was carried out for histomorphometric analysis. Bone mass density was evaluated by densitometry. Results MTX significantly decreased the severity of CIA, whereas ZA slightly exacerbated it. When these two drugs were used in combination, MTX prevented the pro-inflammatory effect of ZA. The combination of ZA with MTX was more effective than MTX alone for reducing structural joint damage with a dramatic decrease of osteoclasts' number in the eroded joints. However, MTX alone also significantly reduced the number of osteoclasts and the number of CD68+ mononuclear cells. ZA alone, or ZA with MTX, significantly increased the systemic bone mass density measured by densitometry and bone volume on histomorphometric analysis. Conclusions A combination of MTX and ZA prevented both bone erosion and systemic bone loss in a rat model of arthritis. Both treatments independently decreased the number of osteoclasts in the eroded joint. However, while MTX probably acts mainly through a decrease of inflammation, ZA has a direct effect on osteoclasts, allowing a dramatic down-regulation of these cells in inflamed joints. These two different mechanisms of action provide support for the use of a combination of these two drugs to improve the prevention of structural joint damage in RA.
Collapse
Affiliation(s)
- Benoit Le Goff
- INSERM UMR-S 957, 1 rue Gaston Veil, 44035, Nantes Cedex 1, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Rheumatoid arthritis (RA) is recognized to be an autoimmune disease that causes preclinical systemic abnormalities and eventually leads to synovial inflammation and destruction of the joint architecture. Recently identified genetic risk factors and novel insights from animal models of spontaneous arthritis have lent support to the concept that thymic selection of an autoreactive T-cell repertoire is an important risk factor for this disease. With advancing age, defects in the homeostatic control of the T-cell pool and in the setting of signaling thresholds lead to the accumulation of pro-inflammatory T-effector cell populations and loss of tolerance to neo-antigens, such as citrullinated peptides. As the breakdown of tolerance to modified self-antigens can precede synovitis by decades, repair of homeostatic defects may open a unique window of opportunity for preventive interventions in RA. The end result of RA, destruction of cartilage and bone, appears to be driven by cytokine- and cell contact-induced activation of synoviocytes and monocytic cells, some of which differentiate into tissue-destructive osteoclasts. Targeting mediators involved in this process has greatly improved the management of this chronic inflammatory syndrome.
Collapse
|
48
|
Cantley MD, Smith MD, Haynes DR. Pathogenic bone loss in rheumatoid arthritis: mechanisms and therapeutic approaches. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/ijr.09.42] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
49
|
McGonagle D, Tan AL, Møller Døhn U, ØStergaard M, Benjamin M. Microanatomic studies to define predictive factors for the topography of periarticular erosion formation in inflammatory arthritis. ACTA ACUST UNITED AC 2009; 60:1042-51. [DOI: 10.1002/art.24417] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
50
|
Abstract
Rheumatoid arthritis is a systemic, inflammatory, autoimmune disorder. Enhanced understanding of molecular pathogenesis has enabled development of innovative biological agents that target specific parts of the immune system. These treatments have changed the course and face of rheumatoid arthritis and outcomes for patients and society. New knowledge has emerged of how environmental factors interact with susceptibility genes and the immune system in the pathogenesis of a major subset of rheumatoid arthritis. Research undertaken on the longitudinal disease process and molecular pathology of joint inflammation has led to new therapeutic strategies that promote early use of disease-modifying drugs with tight disease control and distinct and quantifiable treatment goals. Today, such approaches can halt most cases of joint destruction but not all instances of joint inflammation and comorbidity. Understanding the cause and pathogenesis of different rheumatoid arthritis subsets will lead not only to individualised treatments during early phases of the illness but also, possibly, to disease prevention.
Collapse
Affiliation(s)
- Lars Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | | |
Collapse
|