1
|
Schwager JM, Di Maggio N, Grosso A, Rasadurai A, Minder N, Hubbell JA, Kappos EA, Schaefer DJ, Briquez PS, Banfi A, Burger MG. Semaphorin 3A promotes the long-term persistence of human SVF-derived microvascular networks in engineered grafts. Front Bioeng Biotechnol 2024; 12:1396450. [PMID: 39234267 PMCID: PMC11371724 DOI: 10.3389/fbioe.2024.1396450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction The stromal vascular fraction (SVF) of human adipose tissue is an attractive cell source for engineering grafts with intrinsic vascularization potential, as it is rich in vasculogenic progenitors. However, in order to maintain their functional perfusion it is important to promote the in vivo stabilization of newly assembled microvascular networks. We previously found that Semaphorin 3A (Sema3A) promotes the rapid stabilization of new blood vessels induced by VEGF overexpression in skeletal muscle. Here we investigated whether Sema3A could promote the assembly, connection to circulation and persistence of human SVF-derived microvascular networks in engineered grafts. Methods Recombinant Sema3A was engineered with a transglutaminase substrate sequence (TG-Sema3A) to allow cross-linking into fibrin hydrogels. Grafts were prepared with freshly isolated human SVF cells in fibrin hydrogels decorated with 0, 0.1 or 100 μg/ml TG-Sema3A and implanted subcutaneously in immune-deficient mice. Results After 1 week in vivo, the assembly of human-derived networks was similar in all conditions. The outer part of the grafts was populated by blood vessels of both human and mouse origin, which formed abundant hybrid structures within a common basal lamina. About 90% of human-derived blood vessels were functionally connected to the host circulation in all conditions. However, in the control samples human vessels were unstable. In fact, they significantly regressed by 6 weeks and could no longer be found by 12 weeks. In contrast, a low Sema3A dose (0.1 μg/ml) promoted further human vascular expansion by about 2-fold at 6 weeks and protected them from regression until 12 weeks. From a mechanistic point of view, the stabilization of SVF-derived vessels by 0.1 μg/ml of Sema3A correlated with the recruitment of a specific population of monocytes expressing its receptor Neuropilin-1. Discussion In conclusion, Sema3A is a potent stimulator of in vivo long-term persistence of microvascular networks derived from human SVF. Therefore, decoration of matrices with Sema3a can be envisioned to promote the functional support of tissue engineered grafts.
Collapse
Affiliation(s)
- Juan M Schwager
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Nunzia Di Maggio
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Andrea Grosso
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Abeelan Rasadurai
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Nadja Minder
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Elisabeth A Kappos
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
- Department of Clinical Research, Medical Faculty, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Priscilla S Briquez
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Andrea Banfi
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Maximilian G Burger
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
2
|
Peng W, Chen Q, Zheng F, Xu L, Fang X, Wu Z. The emerging role of the semaphorin family in cartilage and osteoarthritis. Histochem Cell Biol 2024:10.1007/s00418-024-02303-y. [PMID: 38849589 DOI: 10.1007/s00418-024-02303-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2024] [Indexed: 06/09/2024]
Abstract
In the pathogenesis of osteoarthritis, various signaling pathways may influence the bone joint through a common terminal pathway, thereby contributing to the pathological remodeling of the joint. Semaphorins (SEMAs) are cell-surface proteins actively involved in and primarily responsible for regulating chondrocyte function in the pathophysiological process of osteoarthritis (OA). The significance of the SEMA family in OA is increasingly acknowledged as pivotal. This review aims to summarize the mechanisms through which different members of the SEMA family impact various structures within joints. The findings indicate that SEMA3A and SEMA4D are particularly relevant to OA, as they participate in cartilage injury, subchondral bone remodeling, or synovitis. Additionally, other elements such as SEMA4A and SEMA5A may also contribute to the onset and progression of OA by affecting different components of the bone and joint. The mentioned mechanisms demonstrate the indispensable role of SEMA family members in OA, although the detailed mechanisms still require further exploration.
Collapse
Affiliation(s)
- Wenjing Peng
- School of Stomatology, Clinical Research Center for Oral Diseases of Zhejiang Province, Stomatology HospitalZhejiang University School of MedicineKey Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Chen
- School of Stomatology, Clinical Research Center for Oral Diseases of Zhejiang Province, Stomatology HospitalZhejiang University School of MedicineKey Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Fengjuan Zheng
- The Department of Orthodontics, Hangzhou Stomatology Hospital, Hangzhou, China
| | - Li Xu
- School of Stomatology, Clinical Research Center for Oral Diseases of Zhejiang Province, Stomatology HospitalZhejiang University School of MedicineKey Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Xinyi Fang
- School of Stomatology, Clinical Research Center for Oral Diseases of Zhejiang Province, Stomatology HospitalZhejiang University School of MedicineKey Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China.
| | - Zuping Wu
- School of Stomatology, Clinical Research Center for Oral Diseases of Zhejiang Province, Stomatology HospitalZhejiang University School of MedicineKey Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
3
|
Wu Z, Wang Y, Liu W, Lu M, Shi J. The role of neuropilin in bone/cartilage diseases. Life Sci 2024; 346:122630. [PMID: 38614296 DOI: 10.1016/j.lfs.2024.122630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Bone remodeling is the balance between osteoblasts and osteoclasts. Bone diseases such as osteoporosis and osteoarthritis are associated with imbalanced bone remodeling. Skeletal injury leads to limited motor function and pain. Neurophilin was initially identified in axons, and its various ligands and roles in bone remodeling, angiogenesis, neuropathic pain and immune regulation were later discovered. Neurophilin promotes osteoblast mineralization and inhibits osteoclast differentiation and its function. Neuropolin-1 provides channels for immune cell chemotaxis and cytokine diffusion and leads to pain. Neuropolin-1 regulates the proportion of T helper type 17 (Th17) and regulatory T cells (Treg cells), and affects bone immunity. Vascular endothelial growth factors (VEGF) combine with neuropilin and promote angiogenesis. Class 3 semaphorins (Sema3a) compete with VEGF to bind neuropilin, which reduces angiogenesis and rejects sympathetic nerves. This review elaborates on the structure and general physiological functions of neuropilin and summarizes the role of neuropilin and its ligands in bone and cartilage diseases. Finally, treatment strategies and future research directions based on neuropilin are proposed.
Collapse
Affiliation(s)
- Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Wei Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Mingcheng Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Jiejun Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
4
|
Gündoğdu AÇ, Özbayer C, Kar F. Boric Acid Alleviates Gastric Ulcer by Regulating Oxidative Stress and Inflammation-Related Multiple Signaling Pathways. Biol Trace Elem Res 2024; 202:2124-2132. [PMID: 37606879 DOI: 10.1007/s12011-023-03817-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/16/2023] [Indexed: 08/23/2023]
Abstract
Oxidative stress and inflammation have pivotal roles in gastric ulcer development caused by alcohol consumption. Trace element boric acid taken into the human and animal body from dietary sources displays strong antioxidant and anti-inflammatory functions. However, the mechanisms underlying these actions of boric acid remain unclear, and its effectiveness in preventing gastric lesions is unknown. Therefore, the present study was undertaken to evaluate the protective effects of boric acid in alcohol-induced gastric ulcer and elucidate its potential mechanisms. Gastric ulcer was induced by 75% oral ethanol administration in rats, and the effectiveness of prophylactic boric acid treatment at 100 mg/kg concentration was assessed by histopathological examination, ELISA assay and qRT-PCR. Gross macroscopic and histopathological evaluations revealed that boric acid alleviated gastric mucosal lesions. Boric acid decreased reactive oxygen species (ROS) and malondialdehyde (MDA) concentration and the overall oxidation state of the body while improving antioxidant status. It reduced the concentration of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). The mRNA expression of JAK2 and STAT3 was decreased while the expression of AMPK was increased with boric acid pretreatment. Moreover, Sema3A and PlexinA1 levels were elevated upon boric acid pretreatment, and homocysteine levels were reduced. Our results demonstrated that boric acid protects gastric mucosa from ethanol-induced damage by regulating oxidative and inflammatory responses. In addition, our findings suggested that the gastroprotective activity of boric acid could be attributed to its regulatory function in the IL-6/JAK2/STAT3 signaling modulated by AMPK and that Sema3A/PlxnA1 axis and homocysteine are potentially involved in this process.
Collapse
Affiliation(s)
- Ayşe Çakır Gündoğdu
- Department of Histology and Embryology, Faculty of Medicine, Kütahya Health Sciences University, Kütahya, Türkiye
| | - Cansu Özbayer
- Department of Medical Biology, Faculty of Medicine, Kütahya Health Sciences University, Kütahya, Türkiye
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of Medicine, Kütahya Health Sciences University, Evliya Çelebi Campus, 10th km of the Tavşanlı Road, 43100, Kütahya, Türkiye.
| |
Collapse
|
5
|
Rosik J, Kulpa J, Szczepanik M, Pawlik A. The Role of Semaphorins in the Pathogenesis of Rheumatoid Arthritis. Cells 2024; 13:618. [PMID: 38607057 PMCID: PMC11011349 DOI: 10.3390/cells13070618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024] Open
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases. Inflammation of the synovial fluid propagates the pathological process of angiogenesis. Semaphorins play a crucial role in the context of endothelial cell function, and their pleiotropic nature has various effects on the further development of RA. This narrative review summarises the various roles of semaphorins in the pathology of RA and whether they could play a role in developing novel RA treatment options.
Collapse
Affiliation(s)
- Jakub Rosik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.R.); (J.K.); (M.S.)
| | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.R.); (J.K.); (M.S.)
| |
Collapse
|
6
|
Martínez-Ramos S, Rafael-Vidal C, Malvar-Fernández B, Pérez N, Mouriño C, Pérez SG, Maceiras Pan FJ, Conde C, Pego-Reigosa JM, García S. Semaphorin3B promotes an anti-inflammatory and pro-resolving phenotype in macrophages from rheumatoid arthritis patients in a MerTK-dependent manner. Front Immunol 2024; 14:1268144. [PMID: 38283352 PMCID: PMC10811190 DOI: 10.3389/fimmu.2023.1268144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/26/2023] [Indexed: 01/30/2024] Open
Abstract
Previous works from our group show that Semaphorin3B (Sema3B) is reduced in RA and plays a protective role in a mouse arthritis model. In turn, MerTK plays a protective function in murine arthritis models, is expressed by synovial tissue macrophages and is linked to remission in patients with RA. In this study, we examined the role of Sema3B in the phenotypic characteristics of RA macrophages and the implication of MerTK. Peripheral blood monocytes from RA patients were differentiated into IFN-γ (RA MØIFN-γ) or M-CSF (RA MØM-CSF) macrophages and stimulated with LPS, Sema3B or their combination. Alternatively, RA fibroblast like synoviocytes (FLS) were stimulated with RA MØIFN-γ and RA MØM-CSF supernatants. Gene expression was determined by qPCR and protein expression and activation by flow cytometry, ELISA and western blot. Sema3B down-regulated the expression of pro-inflammatory mediators, in both RA MØIFN-γ and RA MØM-CSF. We observed a similar reduction in RA FLS stimulated with the supernatant of Sema3B-treated RA MØIFN-γ and RA MØM-CSF. Sema3B also modulated cell surface markers in macrophages towards an anti-inflammatory phenotype. Besides, MerTK expression and activation was up-regulated by Sema3B, just as GAS6 expression, Resolvin D1 secretion and the phagocytic activity of macrophages. Importantly, the inhibition of MerTK and neuropilins 1 and 2 abrogated the anti-inflammatory effect of Sema3B. Our data demonstrate that Sema3B modulates the macrophage characteristics in RA, inducing a skewing towards an anti-inflammatory/pro-resolving phenotype in a MerTK-dependant manner. Therefore, here we identify a new mechanism supporting the protective role of Sema3B in RA pathogenesis.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Carlos Rafael-Vidal
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Beatriz Malvar-Fernández
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Nair Pérez
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Coral Mouriño
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Sara García Pérez
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Francisco J. Maceiras Pan
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Carmen Conde
- Laboratorio de Reumatologia Experimental y Observacional y Servicio de Reumatologia, Instituto de Investigacion Sanitaria de Santiago (IDIS), Hospital Clinico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Jose María Pego-Reigosa
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Samuel García
- Rheumatology and Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| |
Collapse
|
7
|
Martínez-Ramos S, Rafael-Vidal C, Malvar-Fernández B, Rodriguez-Trillo A, Veale D, Fearon U, Conde C, Conde-Aranda J, Radstake TRDJ, Pego-Reigosa JM, Reedquist KA, García S. HOXA5 is a key regulator of class 3 semaphorins expression in the synovium of rheumatoid arthritis patients. Rheumatology (Oxford) 2023; 62:2621-2630. [PMID: 36398888 PMCID: PMC10321103 DOI: 10.1093/rheumatology/keac654] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/08/2022] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE Class 3 semaphorins are reduced in the synovial tissue of RA patients and these proteins are involved in the pathogenesis of the disease. The aim of this study was to identify the transcription factors involved in the expression of class 3 semaphorins in the synovium of RA patients. METHODS Protein and mRNA expression in synovial tissue from RA and individuals at risk (IAR) patients, human umbilical vein endothelial cells (HUVEC) and RA fibroblast-like synoviocytes (FLS) was determined by ELISA, immunoblotting and quantitative PCR. TCF-3, EBF-1 and HOXA5 expression was knocked down using siRNA. Cell viability, migration and invasion were determined using MTT, calcein, wound closure and invasion assays, respectively. RESULTS mRNA expression of all class 3 semaphorins was significantly lower in the synovium of RA compared with IAR patients. In silico analysis suggested TCF-3, EBF-1 and HOXA5 as transcription factors involved in the expression of these semaphorins. TCF-3, EBF-1 and HOXA5 silencing significantly reduced the expression of several class 3 semaphorin members in FLS and HUVEC. Importantly, HOXA5 expression was significantly reduced in the synovium of RA compared with IAR patients and was negatively correlated with clinical disease parameters. Additionally, TNF-α down-regulated the HOXA5 expression in FLS and HUVEC. Finally, HOXA5 silencing enhanced the migratory and invasive capacities of FLS and the viability of HUVEC. CONCLUSION HOXA5 expression is reduced during the progression of RA and could be a novel therapeutic strategy for modulating the hyperplasia of the synovium, through the regulation of class 3 semaphorins expression.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Carlos Rafael-Vidal
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Beatriz Malvar-Fernández
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Angela Rodriguez-Trillo
- Laboratorio de Reumatología Experimental y Observacional, Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico, Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Douglas Veale
- Rheumatology EULAR Centre of Excellence, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Rheumatology EULAR Centre of Excellence, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
- Department of Molecular Rheumatology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Carmen Conde
- Laboratorio de Reumatología Experimental y Observacional, Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico, Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Timothy R D J Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jose María Pego-Reigosa
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Kris A Reedquist
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Samuel García
- Correspondence to: Samuel García, Rheumatology & Immune-mediated Diseases (IRIDIS) Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Hospital Álvaro Cunqueiro, Estrada Clara Campoamor No. 341, Beade, 36312 Vigo (Pontevedra), Spain. E-mail:
| |
Collapse
|
8
|
Liu Q, Wang X, Chen Y, Ma X, Kang X, He F, Feng D, Zhang Y. Ablation of myeloid discoidin domain receptor 2 exacerbates arthritis and high fat diet induced inflammation. Biochem Biophys Res Commun 2023; 649:47-54. [PMID: 36745969 DOI: 10.1016/j.bbrc.2023.01.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/22/2023] [Indexed: 02/04/2023]
Abstract
Chronic systemic inflammation leads to sever disorders and diseases. It is of great importance to explore novel target for effective treatment. Discoidin domain receptor 2 (Ddr2) is a member of receptor tyrosine kinase (RTK) family and is implicated in skeletal and fat hemostasis. However, the role of Ddr2 in myeloid cells remains obscure. In this study, we conditionally deleted Ddr2 in myeloid lineage cells to generate cKO mice to investigate the role of Ddr2 in myeloid lineage cells. We found that cKO mice exhibited more severe inflammation both in collagen antibody-induced arthritis (CAIA) and high-fat diet (HFD)-induced obesity, indicating the protective role of Ddr2 against inflammation. Mechanistically, Ddr2 promotes macrophage repolarization from the M1 to M2 phenotype, and protect against systemic inflammation. Our study reveals for the first time that Ddr2 modulates macrophage repolarization and plays critical roles in macrophage-mediated inflammation, providing potential target for the intervention of inflammation and related diseases.
Collapse
Affiliation(s)
- Qingyun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaolong Wang
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yazhuo Chen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiao Ma
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaomin Kang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Fang He
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Dongxu Feng
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, People's Republic of China.
| | - Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
9
|
Sema3A Drives Alternative Macrophage Activation in the Resolution of Periodontitis via PI3K/AKT/mTOR Signaling. Inflammation 2023; 46:876-891. [PMID: 36598593 DOI: 10.1007/s10753-022-01777-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023]
Abstract
Macrophages actively participate in immunomodulatory processes throughout periodontal inflammation. Regulation of M1/M2 polarization affects macrophage chemokine and cytokine secretion, resulting in a distinct immunological status that influences prognosis. Semaphorin 3A (Sema3A), a neurite growth factor, exerts anti-inflammatory effects. In this study, we investigated the immunomodulation of Sema3A on macrophage-related immune responses in vivo and in vitro. Topical medications of Sema3A in mice with periodontitis alleviated inflammatory cell infiltration into gingival tissue and reduced areas with positive IL-6 and TNFα expression. We observed that the positive area with the M2 macrophage marker CD206 increased and that of the M1 macrophage marker iNOS decreased in Sema3A-treated mice. It has been postulated that Sema3A alleviates periodontitis by regulating alternative macrophage activation. To understand the mechanism underlying Sema3A modulation of macrophage polarization, an in vitro macrophage research model was established with RAW264.7 cells, and we demonstrated that Sema3A promotes LPS/IFNγ-induced M1 macrophages to polarize into M2 macrophages and activates the PI3K/AKT/mTOR signaling pathways. Inhibition of the PI3K signaling pathway activation might reduce anti-inflammatory activity and boost the expression of the inflammatory cytokines, iNOS, IL-12, TNFα, and IL-6. This study indicated that Sema3A might be a feasible drug to regulate alternative macrophage activation in the inflammatory response and thus alleviate periodontitis.
Collapse
|
10
|
Iwamoto N, Kawakami A. The monocyte-to-osteoclast transition in rheumatoid arthritis: Recent findings. Front Immunol 2022; 13:998554. [PMID: 36172385 PMCID: PMC9510592 DOI: 10.3389/fimmu.2022.998554] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint inflammation leading to joint destruction and deformity. The crucial role of osteoclasts in the bone erosion in RA has been demonstrated. Deregulated osteoclastogenesis which is affected by environmental factors including the inflammatory state, as well as genetic and epigenetic factors, is one of hallmarks of RA pathogenesis. An enhanced-monocyte-to-osteoclast transition plays an important role in osteoclast upregulation in RA because under specific stimuli, circulating monocytes might migrate to a specific location in the bones and fuse with each other to become mature multinucleated osteoclasts. To understand the mechanism of bone damage in RA and to develop novel treatments targeting osteoclast upregulation, it is important to clarify our understanding of the monocyte-to-osteoclast transition in RA. Several potential targets which inhibit both inflammation and osteoclastogenesis, as well as regulators that affect the monocyte-to-osteoclast transition have been revealed by recent studies. Here, we review the factors affecting osteoclastogenesis in RA, summarize the anti-osteoclastogenic effects of current RA treatments, and identify promising therapeutic targets relating to both inflammation and osteoclastogenesis.
Collapse
|
11
|
Bai LK, Su YZ, Wang XX, Bai B, Zhang CQ, Zhang LY, Zhang GL. Synovial Macrophages: Past Life, Current Situation, and Application in Inflammatory Arthritis. Front Immunol 2022; 13:905356. [PMID: 35958604 PMCID: PMC9361854 DOI: 10.3389/fimmu.2022.905356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Inflammatory arthritis is an inflammatory disease that involves the joints and surrounding tissues. Synovial hyperplasia often presents when joints become inflamed due to immune cell infiltration. Synovial membrane is an important as well as a highly specific component of the joint, and its lesions can lead to degeneration of the joint surface, causing pain and joint disability or affecting the patients’ quality of life in severe cases. Synovial macrophages (SMs) are one of the cellular components of the synovial membrane, which not only retain the function of macrophages to engulf foreign bodies in the joint cavity, but also interact with synovial fibroblasts (SFs), T cells, B cells, and other inflammatory cells to promote the production of a variety of pro-inflammatory cytokines and chemokines, such as TNF-α, IL-1β, IL-8, and IL-6, which are involved in the pathogenic process of inflammatory arthritis. SMs from different tissue sources have differently differentiated potentials and functional expressions. This article provides a summary on studies pertaining to SMs in inflammatory arthritis, and explores their role in its treatment, in order to highlight novel treatment modalities for the disease.
Collapse
Affiliation(s)
- Lin-Kun Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Ya-Zhen Su
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xue-Xue Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Bing Bai
- First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Cheng-Qiang Zhang
- Fifth Hospital of Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Li-Yun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Gai-Lian Zhang
- Fifth Hospital of Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
- *Correspondence: Gai-Lian Zhang,
| |
Collapse
|
12
|
Abstract
Semaphorin 3A is a secreted glycoprotein, which was originally identified as axon guidance factor in the neuronal system, but it also possesses immunoregulatory properties. Here, the effect of semaphorin 3A on T-lymphocytes, myeloid dendritic cells and macrophages is systematically analyzed on the bases of all publications available in the literature for 20 years. Expression of semaphorin 3A receptors – neuropilin-1 and plexins A – in these cells is described in details. The data obtained on human and murine cells is described comparatively. A comprehensive overview of the interaction of semaphorin 3A with mononuclear phagocyte system is presented for the first time. Semaphorin 3A signaling mostly results in changes of the cytoskeletal machinery and cellular morphology that regulate pathways involved in migration, adhesion, and cell–cell cooperation of immune cells. Accumulating evidence indicates that this factor is crucially involved in various phases of immune responses, including initiation phase, antigen presentation, effector T cell function, inflammation phase, macrophage activation, and polarization. In recent years, interest in this field has increased significantly because semaphorin 3A is associated with many human diseases and therefore can be used as a target for their treatment. Its involvement in the immune responses is important to study, because semaphorin 3A and its receptors turn to be a promising new therapeutic tools to be applied in many autoimmune, allergic, and oncology diseases.
Collapse
Affiliation(s)
- Ekaterina P Kiseleva
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", St. Petersburg, 197376, Russia.
- Mechnikov North-Western State Medical University, St. Petersburg, 195067, Russia
| | - Kristina V Rutto
- Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", St. Petersburg, 197376, Russia.
| |
Collapse
|
13
|
Komatsu N, Takayanagi H. Mechanisms of joint destruction in rheumatoid arthritis - immune cell-fibroblast-bone interactions. Nat Rev Rheumatol 2022; 18:415-429. [PMID: 35705856 DOI: 10.1038/s41584-022-00793-5] [Citation(s) in RCA: 229] [Impact Index Per Article: 76.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is characterized by inflammation and destruction of bone and cartilage in affected joints. Autoimmune responses lead to increased osteoclastic bone resorption and impaired osteoblastic bone formation, the imbalance of which underlies bone loss in RA, which includes bone erosion, periarticular bone loss and systemic osteoporosis. The crucial role of osteoclasts in bone erosion has been demonstrated in basic studies as well as by the clinical efficacy of antibodies targeting RANKL, an important mediator of osteoclastogenesis. Synovial fibroblasts contribute to joint damage by stimulating both pro-inflammatory and tissue-destructive pathways. New technologies, such as single-cell RNA sequencing, have revealed the heterogeneity of synovial fibroblasts and of immune cells including T cells and macrophages. To understand the mechanisms of bone damage in RA, it is important to clarify how the immune system promotes the tissue-destructive properties of synovial fibroblasts and influences bone cells. The interaction between immune cells and fibroblasts underlies the imbalance between regulatory T cells and T helper 17 cells, which in turn exacerbates not only inflammation but also bone destruction, mainly by promoting RANKL expression on synovial fibroblasts. An improved understanding of the immune mechanisms underlying joint damage and the interplay between the immune system, synovial fibroblasts and bone will contribute to the identification of novel therapeutic targets in RA.
Collapse
Affiliation(s)
- Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
14
|
Gerasimova EV, Popkova TV, Gerasimova DA, Kirichenko TV. Macrophage Dysfunction in Autoimmune Rheumatic Diseases and Atherosclerosis. Int J Mol Sci 2022; 23:4513. [PMID: 35562903 PMCID: PMC9102949 DOI: 10.3390/ijms23094513] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/28/2022] Open
Abstract
One of the problems of modern medical science is cardiovascular pathology caused by atherosclerotic vascular lesions in patients with autoimmune rheumatic diseases (ARDs). The similarity between the mechanisms of the immunopathogenesis of ARD and chronic low-grade inflammation in atherosclerosis draws attention. According to modern concepts, chronic inflammation associated with uncontrolled activation of both innate and acquired immunity plays a fundamental role in all stages of ARDs and atherosclerotic processes. Macrophage monocytes play an important role among the numerous immune cells and mediators involved in the immunopathogenesis of both ARDs and atherosclerosis. An imbalance between M1-like and M2-like macrophages is considered one of the causes of ARDs. The study of a key pathogenetic factor in the development of autoimmune and atherosclerotic inflammation-activated monocyte/macrophages will deepen the knowledge of chronic inflammation pathogenesis.
Collapse
Affiliation(s)
- Elena V. Gerasimova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia; (E.V.G.); (T.V.P.)
| | - Tatiana V. Popkova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Kashirskoe Shosse, 115522 Moscow, Russia; (E.V.G.); (T.V.P.)
| | - Daria A. Gerasimova
- Chair of Organization and Economy of Pharmacy, Institute of Pharmacy, A.P. Nelyubina, I.M. Sechenov First Moscow State Medical University (Sechenov University), 96k1 Ave. Vernadsky, 119526 Moscow, Russia;
| | - Tatiana V. Kirichenko
- Laboratory of Medical Genetics, Chazov National Medical Research Center of Cardiology, 15-a Cherepkovskaya Str., 121552 Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, A.P. Avtsyn Research Institute for Human Morphology, 3 Tsyurupa St., 117418 Moscow, Russia
| |
Collapse
|
15
|
Igea A, Carvalheiro T, Malvar‐Fernández B, Martinez‐Ramos S, Rafael‐Vidal C, Niemantsverdriet E, Varadé J, Fernández‐Carrera A, Jimenez N, McGarry T, Rodriguez‐Trillo A, Veale D, Fearon U, Conde C, Pego‐Reigosa JM, González‐Fernández Á, Reedquist KA, Radstake TRDJ, van der Helm‐Van Mil A, García S. Semaphorin3B plays a central role in serum-induced arthritis model and is reduced in patients with rheumatoid arthritis. Arthritis Rheumatol 2022; 74:972-983. [PMID: 35001548 PMCID: PMC9322571 DOI: 10.1002/art.42065] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/15/2021] [Accepted: 01/04/2022] [Indexed: 11/20/2022]
Abstract
Objective Semaphorin 3B (Sema3B) decreases the migratory and invasive capacities of fibroblast‐like synoviocytes (FLS) in rheumatoid arthritis (RA) and suppresses expression of matrix metalloproteinases. We undertook this study to examine the role of Sema3B in a mouse model of arthritis and its expression in RA patients. Methods Clinical responses, histologic features, and FLS function were examined in wild‐type (WT) and Sema3B−/− mice in a K/BxN serum transfer model of arthritis. Protein and messenger RNA expression of Sema3B in mouse joints and murine FLS, as well as in serum and synovial tissue from patients with arthralgia and patients with RA, was determined using enzyme‐linked immunosorbent assay, immunoblotting, quantitative polymerase chain reaction, and RNA sequencing. FLS migration was determined using a wound closure assay. Results The clinical severity of serum‐induced arthritis was significantly higher in Sema3B−/− mice compared to WT mice. This was associated with increased expression of inflammatory mediators and increased migratory capacity of murine FLS. Administration of recombinant mouse Sema3B reduced the clinical severity of serum‐induced arthritis and the expression of inflammatory mediators. Sema3B expression was significantly lower in the synovial tissue and serum of patients with established RA compared to patients with arthralgia. Serum Sema3B levels were elevated in patients with arthralgia that later progressed to RA, but not in those who did not develop RA; however, these levels drastically decreased 1 and 2 years after RA development. Conclusion Sema3B expression plays a protective role in a mouse model of arthritis. In RA patients, expression levels of Sema3B in the serum depend on the disease stage, suggesting different regulatory roles in disease onset and progression.
Collapse
Affiliation(s)
- Ana Igea
- Universidade de Vigo, Campus Universitario Lagoas Marcosende, and Galicia Sur Health Research Institute, Servicio Galego de Saúde Universidade de VigoVigoSpain
| | | | - Beatriz Malvar‐Fernández
- University of Utrecht, Utrecht, The Netherlands, and Galicia Sur Health Research Institute and University Hospital Complex of VigoVigoSpain
| | - Sara Martinez‐Ramos
- Galicia Sur Health Research InstituteServicio Galego de Saúde Universidade de Vigo, and University Hospital Complex of VigoVigoSpain
| | - Carlos Rafael‐Vidal
- Galicia Sur Health Research InstituteServicio Galego de Saúde Universidade de Vigo, and University Hospital Complex of VigoVigoSpain
| | - Ellis Niemantsverdriet
- Erasmus Medical Center, Rotterdam, The Netherlands, and Leiden University Medical CenterLeidenthe Netherlands
| | - Jezabel Varadé
- Universidade de Vigo, Campus Universitario Lagoas Marcosende, and Galicia Sur Health Research Institute, Servicio Galego de Saúde Universidade de VigoVigoSpain
| | - Andrea Fernández‐Carrera
- Universidade de Vigo, Campus Universitario Lagoas Marcosende, and Galicia Sur Health Research Institute, Servicio Galego de Saúde Universidade de VigoVigoSpain
| | - Norman Jimenez
- Galicia Sur Health Research InstituteServicio Galego de Saúde Universidade de Vigo, and University Hospital Complex of VigoVigoSpain
| | - Trudy McGarry
- St. Vincent's University Hospital and University College Dublin, and Trinity College DublinDublinIreland
| | - Angela Rodriguez‐Trillo
- Hospital Clínico Universitario de Santiago de Compostela, Servicio Galego de SaúdeSantiago de CompostelaSpain
| | - Douglas Veale
- St. Vincent's University Hospital and University College DublinDublinIreland
| | - Ursula Fearon
- St. Vincent's University Hospital and University College Dublin, and Trinity College DublinDublinIreland
| | - Carmen Conde
- Hospital Clínico Universitario de Santiago de Compostela, Servicio Galego de SaúdeSantiago de CompostelaSpain
| | - Jose M. Pego‐Reigosa
- Galicia Sur Health Research InstituteServicio Galego de Saúde Universidade de Vigo, and University Hospital Complex of VigoVigoSpain
| | - África González‐Fernández
- Universidade de Vigo, Campus Universitario Lagoas Marcosende, and Galicia Sur Health Research Institute, Servicio Galego de Saúde Universidade de VigoVigoSpain
| | | | | | | | - Samuel García
- University of Utrecht, Utrecht, The Netherlands, and Galicia Sur Health Research Institute and University Hospital Complex of VigoVigoSpain
| |
Collapse
|
16
|
Zhang H, Lu Y, Wu B, Xia F. Semaphorin 3A mitigates lipopolysaccharide-induced chondrocyte inflammation, apoptosis and extracellular matrix degradation by binding to Neuropilin-1. Bioengineered 2021; 12:9641-9654. [PMID: 34821196 PMCID: PMC8810004 DOI: 10.1080/21655979.2021.1974806] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Semaphorin 3A (SEMA3A) and its receptor neuropilin-1 (NRP-1) are expressed low in chondrocytes under stress, and overexpressing SEMA3A reduces pro-inflammatory cytokine release. This study was aimed at exploring whether SEMA3A participates in lipopolysaccharide (LPS)-induced chondrocyte inflammation, apoptosis and extracellular matrix (ECM) degradation. SEMA3A and NRP-1 expression in LPS-induced ATDC5 cells was determined with RT-qPCR and western blotting. Following stimulation with LPS in the absence or presence of SEMA3A overexpression, the viability of ATDC5 cells was observed through CCK-8 assay. RT-qPCR and western blot were performed to detect the expression of pro-inflammatory cytokines. ATDC5 cell apoptosis was observed through TUNEL, and apoptosis-related proteins were assayed. Expression of ECM-related proteins was measured by RT-qPCR and western blotting. Additionally, the binding of SEMA3A to NRP-1 was verified by co-immunoprecipitation. After interference with NRP-1, cell viability, inflammation and ECM degradation were examined in LPS-induced ATDC5 cells with SEMA3A overexpression. Results revealed that SEMA3A expression in ATDC5 cells decreased following stimulation with LPS. Overexpressing SEMA3A improved cell viability and reduced the inflammatory injury of LPS-stimulated ATDC5 cells. Moreover, SEMA3A overexpression alleviated LPS-induced apoptosis and ECM degradation of ATDC5 chondrocytes. SEMA3A and NRP-1 bound to each other in ATDC5 cells. NRP-1 interference crippled the ameliorative effect of SEMA3A overexpression on LPS-induced chondrocyte inflammation, apoptosis and ECM degradation. To conclude, SEMA3A binds to NRP-1, mitigating LPS-induced chondrocyte inflammation, apoptosis and ECM degradation. This study elucidated the role of SEMA3A in osteoarthritis and illustrated its action mechanism involving NRP-1.
Collapse
Affiliation(s)
- Huiyu Zhang
- Department of Hand Surgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Yue Lu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - BingBing Wu
- Department of Hand Surgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Fei Xia
- Department of Hand Surgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
17
|
Jiao B, Liu S, Tan X, Lu P, Wang D, Xu H. Class-3 semaphorins: Potent multifunctional modulators for angiogenesis-associated diseases. Biomed Pharmacother 2021; 137:111329. [PMID: 33545660 DOI: 10.1016/j.biopha.2021.111329] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 11/29/2022] Open
Abstract
Semaphorins, the neuronal guidance cues, were shown to have broad influences on pathophysiological processes such as bone remodeling, immune responses, and angiogenesis. In particular, Class-3 Semaphorins (SEMA3) is considered a vital regulator involved in angiogenesis. Scientific evidence has pointed to the role of angiogenesis in many diseases, and numerous efforts have been made to explore the possibilities of curing those diseases by targeting angiogenesis. Nevertheless, the efficacies are limited owing to the complex mechanisms of angiogenesis. Hence, investigating the mechanisms of SEMA3 in angiogenesis may contribute to novel therapeutics for diseases. Previous reviews mainly focused on the various functions of semaphorins in one particular disease, and the specific angiogenesis mechanism of SEMA3 in diverse diseases has not been well elucidated. Additionally, the role of SEMA3 in angiogenesis remains elusive, as contradicting results have been found in different disease types. Some evidence from recent studies implies that, while most SEMA3 molecules inhibit pathological angiogenesis in different diseases, occasionally SEMA3 may also promote angiogenesis. This review summarizes the specific role of SEMA3 in a variety of angiogenesis-associated diseases, and documents SEMA3 may be a promising therapeutic target for treating angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Bo Jiao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Tan
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pei Lu
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Danning Wang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hui Xu
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
18
|
Darwish NF, Hablas SA, Baiomy NN, Rageh ESMH. Evaluation of serum14-3-3η protein and Sema3A levels in rheumatoid arthritis: diagnostic and prognostic value. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2020. [DOI: 10.1186/s43166-020-00044-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Serum14-3-3η protein plays an important role in the pathogenesis of rheumatoid arthritis (RA) as it is a joint-derived proinflammatory mediator. Semaphorin3A (Sema3A) plays an immune regulatory and bone remodeling role in many autoimmune diseases. Their role in rheumatoid arthritis needs to be evaluated for diagnostic and prognostic prospective values.
Results
The serum level of protein 14-3-3n was significantly higher in patients with RA than those in healthy controls. Serum 14-3-3η has a significant positive correlation with RF and ACPA, but not with either DAS28, ESR, or CRP. Serum 14-3-3η levels were significantly correlated with radiographically assessed joint damage. Serum Sema3A levels were decreased in rheumatoid arthritis patients compared to controls. There were also negative correlations with disease duration and activity score (DAS28), ESR, CRP, and RF.
Conclusion
The discriminative ability of 14-3-3η was comparable to RF and ACPA enhancing its diagnostic capacity. Sema 3A might serve as a predictive marker for radiographic severity and could have a potential therapeutic role in RA.
Collapse
|
19
|
The role of semaphorins in small vessels of the eye and brain. Pharmacol Res 2020; 160:105044. [PMID: 32590102 DOI: 10.1016/j.phrs.2020.105044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Small vessel diseases, such as ischemic retinopathy and cerebral small vessel disease (CSVD), are increasingly recognized in patients with diabetes, dementia and cerebrovascular disease. The mechanisms of small vessel diseases are poorly understood, but the latest studies suggest a role for semaphorins. Initially identified as axon guidance cues, semaphorins are mainly studied in neuronal morphogenesis, neural circuit assembly, and synapse assembly and refinement. In recent years, semaphorins have been found to play important roles in regulating vascular growth and development and in many pathophysiological processes, including atherosclerosis, angiogenesis after stroke and retinopathy. Growing evidence indicates that semaphorins affect the occurrence, perfusion and regression of both the macrovasculature and microvasculature by regulating the proliferation, apoptosis, migration, barrier function and inflammatory response of endothelial cells, vascular smooth muscle cells (VSMCs) and pericytes. In this review, we concentrate on the regulatory effects of semaphorins on the cell components of the vessel wall and their potential roles in microvascular diseases, especially in the retina and cerebral small vessel. Finally, we discuss potential molecular approaches in targeting semaphorins as therapies for microvascular disorders in the eye and brain.
Collapse
|
20
|
Zhang H, Vreeken D, Junaid A, Wang G, Sol WMPJ, de Bruin RG, van Zonneveld AJ, van Gils JM. Endothelial Semaphorin 3F Maintains Endothelial Barrier Function and Inhibits Monocyte Migration. Int J Mol Sci 2020; 21:ijms21041471. [PMID: 32098168 PMCID: PMC7073048 DOI: 10.3390/ijms21041471] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
In normal physiology, endothelial cells (ECs) form a vital barrier between the blood and underlying tissue controlling leukocyte diapedesis and vascular inflammation. Emerging data suggest that neuronal guidance cues, typically expressed during development, have roles outside the nervous system in vascular biology and immune responses. In particular, Class III semaphorins have been reported to affect EC migration and angiogenesis. While ECs express high levels of semaphorin 3F (SEMA3F), little is known about its function in mature ECs. Here we show that SEMA3F expression is reduced by inflammatory stimuli and increased by laminar flow. Endothelial cells exposed to laminar flow secrete SEMA3F, which subsequently binds to heparan sulfates on the surface of ECs. However, under pro-inflammatory conditions, reduced levels of SEMA3F make ECs more prone to monocyte diapedesis and display impaired barrier function as measured with an electric cell-substrate impedance sensing system and a microfluidic system. In addition, we demonstrate that SEMA3F can directly inhibit the migration of activated monocytes. Taken together, our data suggest an important homeostatic function for EC-expressed SEMA3F, serving as a mediator of endothelial quiescence.
Collapse
|
21
|
Macrophage M1/M2 polarization and rheumatoid arthritis: A systematic review. Autoimmun Rev 2019; 18:102397. [DOI: 10.1016/j.autrev.2019.102397] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023]
|
22
|
Macrophages with regulatory functions, a possible new therapeutic perspective in autoimmune diseases. Autoimmun Rev 2019; 18:102369. [PMID: 31404701 DOI: 10.1016/j.autrev.2019.102369] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022]
Abstract
Macrophages are pivotal cells involved in chronic inflammatory and autoimmune diseases. In fact, during these diseases, activated macrophages may play a critical role, promoting the inflammation as well as mediating the damage resolution. This dichotomy is referred to two end-stage phenotypes of macrophages, conventionally known as M1 and M2, playing a pro-inflammatory and anti-inflammatory role, respectively. The M1 macrophages are the mainly subset involved during inflammatory processes, producing pro-inflammatory mediators. Conversely, the M2 macrophages are proposed to contribute to the resolution phase of inflammation, when cells with pro-resolving property are recruited and activated. In fact, this subset of macrophages may activate regulatory T lymphocytes, which play a critical role in the maintenance of peripheral tolerance and preventing the occurrence of autoimmune diseases. On these bases, the polarization toward the M2 phenotype could play a therapeutic role for autoimmune diseases. In this Review we discussed the characteristic of M1 and M2 macrophages, focusing on the immunoregulatory role of M2 cells and their potential ability to control the inflammation and to promote the immunological tolerance.
Collapse
|
23
|
Mima Y, Suzuki S, Fujii T, Morikawa T, Tamaki S, Takubo K, Shimoda M, Miyamoto T, Watanabe K, Matsumoto M, Nakamura M, Fujita N. Potential involvement of semaphorin 3A in maintaining intervertebral disc tissue homeostasis. J Orthop Res 2019; 37:972-980. [PMID: 30816586 DOI: 10.1002/jor.24258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/20/2019] [Indexed: 02/04/2023]
Abstract
Intervertebral discs (IVDs) are avascular; however, ingrowth of blood vessels into their outer regions has been noted during the progression of degeneration. The mechanisms underlying vascularization in IVD degeneration are not completely understood. Semaphorin 3A (Sema3A), originally characterized as a chemorepulsive factor for growing axons in the developing nervous system, inhibits angiogenesis. This study aimed to elucidate the potential involvement of Sema3A in maintaining tissue homeostasis within the avascular IVD. We demonstrated that the mRNA expression of Sema3A was higher in rat annulus fibrosus (AF) than in nucleus pulposus (NP) and that its expression level decreased with age. Both mRNA and protein expression level of Sema3A was also markedly suppressed in AF tissues of a rat IVD degeneration model. Both real-time RT-PCR and Western blot clearly indicated that Sema3A expression significantly reduced by treating inflammatory cytokines in rat AF cells. In a gain- and loss-of-function study, we observed that Sema3A reduced the catabolic shift in rat AF cells. In addition, our results indicated that Sema3A potentially inhibited the IL-6/JAK/STAT pathway. Finally, BrdU assay and tube formation assay revealed that treatment of recombinant Sema3A significantly blocks both proliferation and tube formation of HUVEC. Our results indicate that Sema3A may help maintain IVD tissue homeostasis. Thus, although further studies are needed, Sema3A may be a potential molecular target for suppressing IVD degeneration. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Yuichiro Mima
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Satoshi Suzuki
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Fujii
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Takayuki Morikawa
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinpei Tamaki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Departments of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Miyamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Kota Watanabe
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Nobuyuki Fujita
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
24
|
Benabdoun HA, Kulbay M, Rondon EP, Vallières F, Shi Q, Fernandes J, Fahmi H, Benderdour M. In vitro and in vivo assessment of the proresolutive and antiresorptive actions of resolvin D1: relevance to arthritis. Arthritis Res Ther 2019; 21:72. [PMID: 30867044 PMCID: PMC6416871 DOI: 10.1186/s13075-019-1852-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Resolvin D1 (RvD1), an important member of resolvins, exerts a wide spectrum of biological effects, including resolution of inflammation, tissue repair, and preservation of cell viability. The aim of the present study is to investigate the anti-arthritic potential and clarify the bone protective actions of RvD1 in vitro and in vivo. METHODS RAW264.7 cells were treated with 50 ng/ml LPS for 72 h in the presence or absence of RvD1 (0-500 nM). Primary human monocytes were treated with M-CSF + RANKL for 14 days ± RvD1 (0-500 nM) with or without siRNA against RvD1 receptor FPR2. Expressions of inflammatory mediators, degrading enzymes, osteoclasts (OC) formation, and bone resorption were analyzed. The therapeutic effect of RvD1 (0-1000 ng) was carried out in murine collagen antibody-induced arthritis. Arthritis scoring, joint histology, and inflammatory and bone turnover markers were measured. RESULTS RvD1 is not toxic and inhibits OC differentiation and activation. It decreases bone resorption, as assessed by the inhibition of TRAP and cathepsin K expression, hydroxyapatite matrix resorption, and bone loss. In addition, RvD1 reduces TNF-α, IL-1β, IFN-γ, PGE2, and RANK and concurrently enhances IL-10 in OC. Moreover, in arthritic mice, RvD1 alleviates clinical score, paw inflammation, and bone and joint destructions. Besides, RvD1 reduces inflammatory mediators and markedly decreases serum markers of bone and cartilage turnover. CONCLUSION Our results provide additional evidence that RvD1 plays a key role in preventing bone resorption and other pathophysiological changes associated with arthritis. The study highlights the clinical relevance of RvD1 as a potential compound for the treatment of inflammatory arthritis and related bone disorders.
Collapse
Affiliation(s)
- Houda Abir Benabdoun
- Department of Pharmacology, Université de Montréal, Montreal,, QC, Canada.,Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Merve Kulbay
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Elsa-Patricia Rondon
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Francis Vallières
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Qin Shi
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Julio Fernandes
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada.,Department of Surgery, Université de Montréal, Montreal, QC, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - Mohamed Benderdour
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada. .,Department of Surgery, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
25
|
Garcia S. Role of Semaphorins in Immunopathologies and Rheumatic Diseases. Int J Mol Sci 2019; 20:ijms20020374. [PMID: 30654587 PMCID: PMC6359241 DOI: 10.3390/ijms20020374] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
Rheumatic diseases are disorders characterized by joint inflammation, in which other organs are also affected. There are more than two hundred rheumatic diseases, the most studied so far are rheumatoid arthritis, osteoarthritis, spondyloarthritis, systemic lupus erythematosus, and systemic sclerosis. The semaphorin family is a large group of proteins initially described as axon guidance molecules involved in nervous system development. Studies have demonstrated that semaphorins play a role in other processes such as the regulation of immunity, angiogenesis, bone remodeling, apoptosis, and cell migration and invasion. Moreover, semaphorins have been related to the pathogenesis of multiple sclerosis, asthma, Alzheimer, myocarditis, atherosclerosis, fibrotic diseases, osteopetrosis, and cancer. The aim of this review is to summarize current knowledge regarding the role of semaphorins in rheumatic diseases, and discuss their potential applications as therapeutic targets to treat these disorders.
Collapse
Affiliation(s)
- Samuel Garcia
- Department of Rheumatology and Clinical Immunology and Laboratory of Translational Immunology, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| |
Collapse
|
26
|
Zhang L, Zheng L, Li C, Wang Z, Li S, Xu L. Sema3a as a Novel Therapeutic Option for High Glucose-Suppressed Osteogenic Differentiation in Diabetic Osteopathy. Front Endocrinol (Lausanne) 2019; 10:562. [PMID: 31481931 PMCID: PMC6710340 DOI: 10.3389/fendo.2019.00562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/02/2019] [Indexed: 12/29/2022] Open
Abstract
Objective: Diabetic osteopathy is a common comorbidity of diabetes mellitus, with skeletal fragility, osteoporosis and bone pain. The aim of our study was to highlight the role of sema3a on osteoblast differentiation of MC3T3-e1 in high-glucose condition and explore its therapeutic effect of diabetic osteopathy in vitro and vivo. Methods: In our study, the expression of osteogenesis-related makers, such as ALP, OCN, OPG, β-catenin and Runx2, were analyzed in MC3T3 osteoblastic cells to explore the effect of sema3a on osteoblast differentiation in high-glucose condition, and as was the staining of ALP and Alizarin Red S. In a diabetic animal model, the expression of serum bone metabolic markers, such as ALP, P1NP, OCN, and β-CTX, were analyzed and micro-CT was used to detect bone architecture, including Tb.N, Tb.Th, Tb.Sp, Tb.Pf, BS/BV, and BV/TV after the treatment of sema3a. Results: High glucose significantly inhibited osteogenic differentiation by decreasing the expression of osteogenesis-related makers, sema3a and its receptor of Nrp-1 in a dose-dependent manner in MC3T3. In high-glucose condition, exogenous sema3a (RPL917Mu01) increased the expression of ALP, OCN, OPG, Runx2, β-catenin, and the positive proportion of ALP and Alizarin Red S staining. In addition, in diabetic animal model, exogenous sema3a could increase bone mass and bone mineral density, and downregulate the expression of ALP, P1NP, OCN, and β-CTX. Conclusion: High glucose suppresses osteogenic differentiation in MC3T3 and sema3a may take part in this process. The application of exogenous sema3a alleviates high glucose-induced inhibition of osteoblast differentiation in diabetic osteopathy.
Collapse
|
27
|
Ha YJ, Han DW, Kim JH, Chung SW, Kang EH, Song YW, Lee YJ. Circulating Semaphorin 4D as a Marker for Predicting Radiographic Progression in Patients with Rheumatoid Arthritis. DISEASE MARKERS 2018; 2018:2318386. [PMID: 30538782 PMCID: PMC6261241 DOI: 10.1155/2018/2318386] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
Abstract
Semaphorin 3A (Sema3A) and semaphorin 4D (Sema4D) are molecules which regulate immune responses as well as bone remodeling process. The aim of this study was to evaluate the serum levels of Sema3A and Sema4D and to investigate their clinical significance in rheumatoid arthritis (RA). The serum levels of Sema3A and Sema4D were measured in 130 patients with RA and 65 sex- and age-matched healthy individuals. Circulating levels of biomarkers of RA-related inflammation and bone turnover such as tumor necrosis factor- (TNF-) α, interleukin- (IL-) 6, IL-22, IL-34, osteopontin, Dkk-1, and sclerostin were also measured. Disease activity was determined by the 28-joint disease activity score (DAS28), and radiographic joint damage was assessed by the modified Sharp van der Heijde score (SHS). The serum levels of Sema3A were significantly higher in patients with RA than those in healthy controls (p < 0.001), whereas serum4D levels did not differ between the two groups. The levels of Sema4D showed a positive correlation with C-reactive protein (p = 0.001) and IL-6 (p < 0.001) levels, whereas the levels of Sema3A showed a negative correlation with Dkk-1 (p = 0.007) and TNF-α (p = 0.001). Even though Sema3A and Sema4D levels were comparable between RA patients with DAS28> 3.2 and with DAS28 ≤ 3.2, RA patients with radiographic progression (ΔSHS change/year ≥ 1) had significantly higher baseline levels of Sema4D than those without progression (p = 0.029). Additionally, when RA patients were divided into 3 groups using tertiles of Sema4D levels, the percentage of progressors was significantly increased (p = 0.045). In multivariate logistic regression analysis, serum Sema4D levels were an independent risk factor for radiographic progression. Our results suggest that the baseline levels of Sema4D might be a useful marker to identify RA patients with subsequent radiographic progression and that Sema4D may be an active mediator involved in RA-induced joint damage.
Collapse
Affiliation(s)
- You-Jung Ha
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dong Woo Han
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Hyoun Kim
- Division of Rheumatology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Sang Wan Chung
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Eun Ha Kang
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yeong Wook Song
- WCU Department of Molecular Medicine and Biopharmaceutical Sciences, Medical Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yun Jong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Translational Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Fang W, Wang Z, Li Q, Wang X, Zhang Y, Sun Y, Tang W, Ma C, Sun J, Li N, Yi F. Gpr97 Exacerbates AKI by Mediating Sema3A Signaling. J Am Soc Nephrol 2018. [PMID: 29531097 DOI: 10.1681/asn.2017080932] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background G protein-coupled receptors (GPCRs) participate in a variety of physiologic functions, and several GPCRs have critical physiologic and pathophysiologic roles in the regulation of renal function. We investigated the role of Gpr97, a newly identified member of the adhesion GPCR family, in AKI.Methods AKI was induced by ischemia-reperfusion or cisplatin treatment in Gpr97-deficient mice. We assessed renal injury in these models and in patients with acute tubular necrosis by histologic examination, and we conducted microarray analysis and in vitro assays to determine the molecular mechanisms of Gpr97 function.Results Gpr97 was upregulated in the kidneys from mice with AKI and patients with biopsy-proven acute tubular necrosis compared with healthy controls. In AKI models, Gpr97-deficient mice had significantly less renal injury and inflammation than wild-type mice. Gpr97 deficiency also attenuated the AKI-induced expression of semaphorin 3A (Sema3A), a potential early diagnostic biomarker of renal injury. In NRK-52E cells subjected to oxygen-glucose deprivation, siRNA-mediated knockdown of Gpr97 further increased the expression of survivin and phosphorylated STAT3 and reduced toll-like receptor 4 expression. Cotreatment with recombinant murine Sema3A protein counteracted these effects. Finally, additional in vivo and in vitro studies, including electrophoretic mobility shift assays and luciferase reporter assays, showed that Gpr97 deficiency attenuates ischemia-reperfusion-induced expression of the RNA-binding protein human antigen R, which post-transcriptionally regulates Sema3A expression.Conclusions Gpr97 is an important mediator of AKI, and pharmacologic targeting of Gpr97-mediated Sema3A signaling at multiple levels may provide a novel approach for the treatment of AKI.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Sun
- Departments of Pharmacology
| | | | | | - Jinpeng Sun
- Biochemistry and Molecular Biology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, and
| | - Ningjun Li
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Fan Yi
- Departments of Pharmacology, .,The State Key Laboratory of Microbial Technology, Shandong University, Jinan, China; and
| |
Collapse
|