1
|
Shao Y, Mei Y, Tan Y, Yang M, Wu H. The regulatory functions of G protein-coupled receptors signaling pathways in B cell differentiation and development contributing to autoimmune diseases. Cell Biosci 2025; 15:57. [PMID: 40307944 PMCID: PMC12042570 DOI: 10.1186/s13578-025-01398-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
Autoimmune diseases are characterized by a dysfunction of the immune system. Disruptions in the balance of B-cell dynamics and the increase in auto-antibody levels are pivotal in the triggering of several autoimmune disorders. All of this is inextricably linked to the differentiation, development, migration, and functional regulation of B cells in the human immune response. G protein-coupled receptors (GPCR) are recognized as crucial targets in drug development and play pivotal roles in both B cell differentiation and the underlying mechanisms of autoimmune diseases. However, there has been an inadequate comprehension of how GPCR intricately modulate B cell development and impact the pathogenesis of autoimmune diseases. Ligands and functions of GPCR-chemokine receptors including CXCR3, CXCR4, CXCR5 and CCR7, lipid receptors including S1PR1-5, cannabinoid receptor CB2 as well as orphan GPCR including GPR132, GPR183, GPR174, and P2RY8 in B cell differentiation and development, will be elaborated in this review. The roles these GPCR play in mediating B cells in several autoimmune diseases will also be discussed. The elucidation of the multifaceted mechanisms controlled by GPCR not only enriches our comprehension of immune responses but also provides a promising avenue for therapeutic interventions in the domain of autoimmune disorders.
Collapse
Affiliation(s)
- Yongqi Shao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Yang Mei
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Yixin Tan
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.
| | - Ming Yang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.
| |
Collapse
|
2
|
Lee ES, Kim HJ, Lee D, Kang JY, Shin DM, Hong JH. Rheumatoid arthritis severity is mediated by crosstalk between synoviocytes and mature osteoclasts through a calcium and cytokine feedback loop. Exp Mol Med 2025; 57:402-419. [PMID: 39894824 PMCID: PMC11873226 DOI: 10.1038/s12276-025-01401-8] [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: 03/04/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 02/04/2025] Open
Abstract
Fibroblast-like synoviocytes (FLSs) and osteoclasts are central cells in the maintenance of joint homeostasis. Rheumatoid arthritis (RA) is a chronic inflammatory disease of joints that induces cytokine-activated FLSs and progressive bone erosion. Interactions between FLSs and other cells, such as T cells and B cells, have been recognized in the development of RA. Here we hypothesized that calcium released from bone by mature osteoclasts might activate FLSs, which are also affected by inflammatory cytokines in the inflamed synovium. Osteoclastogenesis occurs in the presence of cytokine-stimulated FLS medium, and calcium released from the bone disc activates FLS migration. We first investigated the calcium and cytokine feedback loop between FLSs and osteoclast maturation. Moreover, by addressing the role of the sodium-bicarbonate cotransporter NBCn1 in osteoclastogenesis, we found that the inhibition of NBCn1 attenuated the infinite calcium and cytokine feedback loop between FLSs and osteoclasts. In a collagen-induced arthritis mouse model, the inhibition of NBC reduced the RA pathological phenotype and bone resorption area in the femur. These results suggest that modulation of the crosstalk between FLSs and osteoclasts by inhibiting the calcium and cytokine feedback loop could be considered to develop pioneering strategies to combat RA severity and dysregulated bone homeostasis.
Collapse
Affiliation(s)
- Eun Sun Lee
- Department of Physiology, College of Medicine, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea
| | - Hyeong Jae Kim
- Department of Physiology, College of Medicine, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea
| | - Dongun Lee
- Department of Health Sciences and Technology, GAIHST, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea
| | - Jung Yun Kang
- Department of Dental Hygiene, College of Software Digital Healthcare Convergence, Yonsei University, Wonju, South Korea
| | - Dong Min Shin
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.
| | - Jeong Hee Hong
- Department of Physiology, College of Medicine, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea.
- Department of Health Sciences and Technology, GAIHST, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea.
| |
Collapse
|
3
|
Chen S, Zhao W, Du J, Chen S, Li J, Shen B, Zhou Y, Chen S. The expression of RBPJ and its potential role in rheumatoid arthritis. BMC Genomics 2024; 25:899. [PMID: 39350019 PMCID: PMC11441141 DOI: 10.1186/s12864-024-10804-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Recombination signal-binding protein for immunoglobulin kappa J region (RBPJ) is a transcriptional regulator that plays an important role in maintaining immune homeostasis. This study aimed to estimate the expression of RBPJ in rheumatoid arthritis (RA) patients and investigate its relationship with RA. METHODS A total of 83 newly diagnosed RA patients and 70 healthy controls were included. mRNA was extracted from peripheral blood mononuclear cells (PBMCs), and the expression of RBPJ was detected using quantitative real-time PCR (qRT‒PCR). An RA dataset (GSE89408) was obtained from the Gene Expression Omnibus (GEO) database, and RA synovial tissues were divided into two groups. The differentially expressed genes (DEGs) were selected with the "DESeq2" R package. RESULTS RBPJ expression was lower in RA patients than in health controls and was negatively correlated with the DAS28 score, C-reactive protein (CRP) level and erythrocyte sedimentation rate (ESR). RA synovial tissues from GSE89408 were classified into RBPJ-low (≤ 25%) and RBPJ-high (≥ 75%) groups according to RBPJ expression, and 562 DEGs were identified. Gene Ontology (GO) enrichment analyses revealed that the DEGs significantly affected the regulation of T cell activation and lymphocyte/mononuclear cell differentiation. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that the most enriched pathways of DEGs were the T cell receptor signaling pathway, Th1/2 and Th17 cell differentiation, the PI3K - Akt signaling pathway and cytokine‒cytokine receptor interaction. CytoHubba Plugin revealed that most of the top 10 genes were involved in osteoclast differentiation, the T cell receptor signaling pathway and cytokine‒cytokine receptor interaction. CONCLUSIONS RBPJ expression was significantly lower in RA patients and negatively correlated with disease activity. GEO dataset analysis demonstrated that RBPJ may be involved in osteoclast differentiation, T cell activation and differentiation, and the T cell receptor signaling pathway. Our research may contribute to understanding the potential mechanisms by which RBPJ regulates T cell differentiation and cytokine‒cytokine receptor interaction in RA patients.
Collapse
Affiliation(s)
- Shuaishuai Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Weibo Zhao
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
| | - Juping Du
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Suyun Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Jun Li
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Bo Shen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Yuanlin Zhou
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China.
| | - Shiyong Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China.
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China.
| |
Collapse
|
4
|
Chen YJ, Chen Y, Chen P, Jia YQ, Wang H, Hong XP. Characteristics of PD-1 +CD4 + T cells in peripheral blood and synovium of rheumatoid arthritis patients. Clin Transl Immunology 2024; 13:e70006. [PMID: 39345753 PMCID: PMC11427813 DOI: 10.1002/cti2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 07/29/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Objectives PD-1 plays a crucial role in the immune dysregulation of rheumatoid arthritis (RA), but the specific characteristics of PD-1+CD4+ T cells remain unclear and require further investigation. Methods Circulating PD-1+CD4+ T cells from RA patients were analysed using flow cytometry. Plasma levels of soluble PD-1 (sPD-1) were measured using enzyme-linked immunosorbent assay (ELISA). Single-cell RNA sequence data from peripheral blood mononuclear cells (PBMCs) and synovial tissue of patients were obtained from the GEO and the ImmPort databases. Bioinformatics analyses were performed in the R studio to characterise PD-1+CD4+ T cells. Expression of CCR7, KLF2 and IL32 in PD-1+CD4+ T cells was validated by flow cytometry. Results RA patients showed an elevated proportion of PD-1+CD4+ T cells in peripheral blood, along with increased plasma sPD-1 levels, which positively correlated with TNF-α and erythrocyte sedimentation rate. Bioinformatic analysis revealed PD-1 expression on CCR7+CD4+ T cells in PBMCs, and on both CCR7+CD4+ T cells and CXCL13+CD4+ T cells in RA synovium. PD-1 was co-expressed with CCR7, KLF2, and IL32 in peripheral CD4+ T cells. In synovium, PD-1+CCR7+CD4+ T cells had higher expression of TNF and LCP2, while PD-1+CXCL13+CD4+ T cells showed elevated levels of ARID5A and DUSP2. PD-1+CD4+ T cells in synovium also appeared to interact with B cells and fibroblasts through BTLA and TNFSF signalling pathways. Conclusion This study highlights the increased proportion of PD-1+CD4+ T cells and elevated sPD-1 levels in RA. The transcriptomic profiles and signalling networks of PD-1+CD4+ T cells offer new insights into their role in RA pathogenesis.
Collapse
Affiliation(s)
- Yan-Juan Chen
- Department of Rheumatology and Immunology The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital Shenzhen China
- Integrated Chinese and Western Medicine Postdoctoral Research Station Jinan University Guangzhou China
| | - Yong Chen
- Department of Rheumatology and Immunology Affiliated Hospital of Zunyi Medical University Zunyi China
| | - Ping Chen
- Department of Rheumatology and Immunology Shenzhen People's Hospital Shenzhen China
| | - Yi-Qun Jia
- Stomatology Center, The Second Clinical Medical College of Jinan University Shenzhen People's Hospital Shenzhen China
| | - Hua Wang
- Department of Orthopaedics, The Second Clinical Medical College of Jinan University, The First Afiliated Hospital of Southern University of Science and Technology Shenzhen People's Hospital Shenzhen China
| | - Xiao-Ping Hong
- Department of Rheumatology and Immunology The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital Shenzhen China
| |
Collapse
|
5
|
Zhang Z, Shao Z, Xu Z, Wang J. Similarities and differences between osteoarthritis and rheumatoid arthritis: insights from Mendelian randomization and transcriptome analysis. J Transl Med 2024; 22:851. [PMID: 39304950 DOI: 10.1186/s12967-024-05643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) and rheumatoid arthritis (RA) are often difficult to distinguish in the early stage of the disease. The purpose of this study was to explore the similarities and differences between the two diseases through Mendelian randomization (MR) and transcriptome analysis. METHODS We first performed a correlation analysis of phenotypic data from genome-wide association studies (GWAS) of OA and RA. Then, we performed functional and pathway enrichment of differentially expressed genes in OA, RA, and normal patients. The infiltration of immune cells in arthritis was analyzed according to gene expression. Finally, MR analysis was performed with inflammatory cytokines and immune cells as exposures and arthritis as the outcome. The same and different key cytokines and immune cells were obtained by the two analysis methods. RESULTS GWAS indicated that there was a genetic correlation between OA and RA. The common function of OA and RA is enriched in their response to cytokines, while the difference is enriched in lymphocyte activation. T cells are the main immune cells that differentiate between OA and RA. MR analysis further revealed that OA is associated with more protective cytokines, and most of the cytokines in RA are pathogenic. In addition, CCR7 on naive CD4 + T cell was positively correlated with OA. SSC-A on CD4 + T cell was negatively correlated with RA, while HLA DR on CD33- HLA DR + was positively correlated with RA. CONCLUSION Our study demonstrated the similarities and differences of immune inflammation between OA and RA, allowing us to better understand these two diseases.
Collapse
Affiliation(s)
- Zhixiang Zhang
- Department of Orthopedic, Yancheng No.1 People's Hospital, Affiliated Hospital of Medical School, Nanjing University, The First People's Hospital of Yancheng, Yancheng, 224000, China
| | - Zhiqiang Shao
- Department of Orthopedic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, 215008, China
| | - Zonghan Xu
- Department of Orthopedic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, 215008, China.
| | - Jiaqian Wang
- Department of Orthopedic, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Patente TA, Gasan TA, Scheenstra M, Ozir-Fazalalikhan A, Obieglo K, Schetters S, Verwaerde S, Vergote K, Otto F, Wilbers RHP, van Bloois E, Wijck YV, Taube C, Hammad H, Schots A, Everts B, Yazdanbakhsh M, Guigas B, Hokke CH, Smits HH. S. mansoni -derived omega-1 prevents OVA-specific allergic airway inflammation via hampering of cDC2 migration. PLoS Pathog 2024; 20:e1012457. [PMID: 39186814 PMCID: PMC11379383 DOI: 10.1371/journal.ppat.1012457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/06/2024] [Accepted: 07/27/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic infection with Schistosoma mansoni parasites is associated with reduced allergic sensitization in humans, while schistosome eggs protects against allergic airway inflammation (AAI) in mice. One of the main secretory/excretory molecules from schistosome eggs is the glycosylated T2-RNAse Omega-1 (ω1). We hypothesized that ω1 induces protection against AAI during infection. Peritoneal administration of ω1 prior to sensitization with Ovalbumin (OVA) reduced airway eosinophilia and pathology, and OVA-specific Th2 responses upon challenge, independent from changes in regulatory T cells. ω1 was taken up by monocyte-derived dendritic cells, mannose receptor (CD206)-positive conventional type 2 dendritic cells (CD206+ cDC2), and by recruited peritoneal macrophages. Additionally, ω1 impaired CCR7, F-actin, and costimulatory molecule expression on myeloid cells and cDC2 migration in and ex vivo, as evidenced by reduced OVA+ CD206+ cDC2 in the draining mediastinal lymph nodes (medLn) and retainment in the peritoneal cavity, while antigen processing and presentation in cDC2 were not affected by ω1 treatment. Importantly, RNAse mutant ω1 was unable to reduce AAI or affect DC migration, indicating that ω1 effects are dependent on its RNAse activity. Altogether, ω1 hampers migration of OVA+ cDC2 to the draining medLn in mice, elucidating how ω1 prevents allergic airway inflammation in the OVA/alum mouse model.
Collapse
Affiliation(s)
- Thiago A Patente
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Thomas A Gasan
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Maaike Scheenstra
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Arifa Ozir-Fazalalikhan
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Katja Obieglo
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Sjoerd Schetters
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Stijn Verwaerde
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Karl Vergote
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Frank Otto
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Ruud H P Wilbers
- Laboratory of Nematology, Plant Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Eline van Bloois
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - Christian Taube
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Arjen Schots
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart Everts
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center (LUMC), Leiden, Netherlands
| |
Collapse
|
7
|
Khokhar M, Dey S, Tomo S, Jaremko M, Emwas AH, Pandey RK. Unveiling Novel Drug Targets and Emerging Therapies for Rheumatoid Arthritis: A Comprehensive Review. ACS Pharmacol Transl Sci 2024; 7:1664-1693. [PMID: 38898941 PMCID: PMC11184612 DOI: 10.1021/acsptsci.4c00067] [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: 02/07/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic debilitating autoimmune disease, that causes joint damage, deformities, and decreased functionality. In addition, RA can also impact organs like the skin, lungs, eyes, and blood vessels. This autoimmune condition arises when the immune system erroneously targets the joint synovial membrane, resulting in synovitis, pannus formation, and cartilage damage. RA treatment is often holistic, integrating medication, physical therapy, and lifestyle modifications. Its main objective is to achieve remission or low disease activity by utilizing a "treat-to-target" approach that optimizes drug usage and dose adjustments based on clinical response and disease activity markers. The primary RA treatment uses disease-modifying antirheumatic drugs (DMARDs) that help to interrupt the inflammatory process. When there is an inadequate response, a combination of biologicals and DMARDs is recommended. Biological therapies target inflammatory pathways and have shown promising results in managing RA symptoms. Close monitoring for adverse effects and disease progression is critical to ensure optimal treatment outcomes. A deeper understanding of the pathways and mechanisms will allow new treatment strategies that minimize adverse effects and maintain quality of life. This review discusses the potential targets that can be used for designing and implementing precision medicine in RA treatment, spotlighting the latest breakthroughs in biologics, JAK inhibitors, IL-6 receptor antagonists, TNF blockers, and disease-modifying noncoding RNAs.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department
of Biochemistry, All India Institute of
Medical Sciences, Jodhpur, 342005 Rajasthan, India
| | - Sangita Dey
- CSO
Department, Cellworks Research India Pvt
Ltd, Bengaluru, 560066 Karnataka, India
| | - Sojit Tomo
- Department
of Biochemistry, All India Institute of
Medical Sciences, Jodhpur, 342005 Rajasthan, India
| | - Mariusz Jaremko
- Smart-Health
Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological
and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955 Jeddah, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core
Laboratories, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Rajan Kumar Pandey
- Department
of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
8
|
Schälter F, Azizov V, Frech M, Dürholz K, Schmid E, Hendel A, Sarfati I, Maeda Y, Sokolova M, Miyagawa I, Focke K, Sarter K, van Baarsen LGM, Krautwald S, Schett G, Zaiss MM. CCL19-Positive Lymph Node Stromal Cells Govern the Onset of Inflammatory Arthritis via Tropomyosin Receptor Kinase. Arthritis Rheumatol 2024; 76:857-868. [PMID: 38268500 DOI: 10.1002/art.42807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 10/30/2023] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVE The study objective was to assess the role of CCL19+ lymph node stromal cells of the joint-draining popliteal lymph node (pLN) for the development of arthritis. METHODS CCL19+ lymph node stromal cells were spatiotemporally depleted for five days in the pLN before the onset of collagen-induced arthritis (CIA) using Ccl19-Cre × iDTR mice. In addition, therapeutic treatment with recombinant CCL19-immunoglobulin G (IgG), locally injected in the footpad, was used to confirm the results. RNA sequencing of lymph node stromal cells combined with T cell coculture assays using tropomyosin receptor kinase (Trk) family inhibitors together with in vivo local pLN small interfering RNA (siRNA) treatments were used to elucidate the pathway by which CCL19+ lymph node stromal cells initiate the onset of arthritis. RESULTS Spatiotemporal depletion of CCL19+ lymph node stromal cells prevented disease onset in CIA mice. These inhibitory effects could be mimicked by local CCL19-IgG treatment. The messenger RNA sequencing analyses showed that CCL19+ lymph node stromal cells down-regulated the expression of the tropomyosin receptor kinase A (TrkA) just before disease onset. Blocking TrkA in lymph node stromal cells led to increased T cell proliferation in in vitro coculture assays. Similar effects were observed with the pan-Trk inhibitor larotrectinib in cocultures of lymph node stromal cells of patients with rheumatoid arthritis and T cells. Finally, local pLN treatment with TrkA inhibitor and TrkA siRNA led to exacerbated arthritis scores. CONCLUSION CCL19+ lymph node stromal cells are crucially involved in the development of inflammatory arthritis. Therefore, targeting of CCL19+ lymph node stromal cells via TRK could provide a tool to prevent arthritis.
Collapse
Affiliation(s)
- Fabian Schälter
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Vugar Azizov
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Frech
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kerstin Dürholz
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Eva Schmid
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anna Hendel
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ilann Sarfati
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yuichi Maeda
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany, and Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Maria Sokolova
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ippei Miyagawa
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany, and The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Kristin Focke
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kerstin Sarter
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lisa G M van Baarsen
- Department of Rheumatology and Clinical Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC and University of Amsterdam, Amsterdam, Netherlands
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, and Deutsches Zentrumlmmuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
9
|
Cheng XJ, Ji R, Huan RH, Huang SQ, Fan W, Zhao YC, Yuan RD, Wang XQ, Zhang X. [Clinical study of the cytokine panel in the diagnosis of ocular chronic graft-versus-host disease]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:242-248. [PMID: 38716595 PMCID: PMC11078663 DOI: 10.3760/cma.j.cn121090-20231031-00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Indexed: 08/21/2024]
Abstract
Objective: To investigate the association between cytokines and ocular chronic graft-versus-host disease (cGVHD) and identify specific biomarkers for ocular cGVHD to enhance clinical diagnosis, treatment, and evaluation. Methods: A mouse model of cGVHD was established to explore the correlation between cGVHD and serum cytokines. Based on the findings from the animal experiments and literature review, a panel of 16 cytokine combinations was identified. Enzyme-linked immunosorbent assay (ELISA) was used to compare the cytokine concentrations in the serum and tear samples from patients who underwent allogeneic hematopoietic stem cell transplantation from June 2017 to March 2022 at the Medical Center of Hematology, Xinqiao Hospital, Army Medical University. Results: ① Compared with the control group, mice with cGVHD exhibited elevated serum IL-1β, IL-6, IL-8, IL-17, IFN-γ, CX3CL1, CXCL11, CXCL13, CCL11, and CCL19 concentrations (all P<0.05). ② Analysis of the cytokine profiles of the serum and tear samples revealed that compared with patients without ocular cGVHD, those with ocular cGVHD exhibited increased serum IL-8 [P=0.032, area under the curve (AUC) =0.678]; decreased serum IL-10 (P=0.030, AUC=0.701) ; elevated IL-8, IFN-γ, CXCL9, and CCL17 in tear samples; and lower IL-10 and CCL19 in tear samples (all P<0.05, all AUC>0.7). Moreover, cytokines in tear samples showed correlations with ocular surface parameters related to ocular cGVHD. Conclusions: Tear fluid demonstrates greater specificity and sensitivity as a biomarker for diagnosing ocular cGVHD than serum biomarkers. Among the identified cytokines in tear samples, IL-8, IL-10, IFN-γ, CXCL9, CCL17, and CCL19 serve as diagnostic biomarkers for ocular cGVHD post-transplantation, offering practical reference value for diagnosis.
Collapse
Affiliation(s)
- X J Cheng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Micoenvironment, State Key Laboatory of Trauma and Chemical Poisoning, Chongqing 400037, China
| | - R Ji
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Micoenvironment, State Key Laboatory of Trauma and Chemical Poisoning, Chongqing 400037, China
| | - R H Huan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Micoenvironment, State Key Laboatory of Trauma and Chemical Poisoning, Chongqing 400037, China
| | - S Q Huang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Micoenvironment, State Key Laboatory of Trauma and Chemical Poisoning, Chongqing 400037, China
| | - W Fan
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Y C Zhao
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - R D Yuan
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - X Q Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Micoenvironment, State Key Laboatory of Trauma and Chemical Poisoning, Chongqing 400037, China
| | - X Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Micoenvironment, State Key Laboatory of Trauma and Chemical Poisoning, Chongqing 400037, China Jinfeng Laboratory, Chongqing 400037, China
| |
Collapse
|
10
|
Qin Y, Jin J, Zhang J, Wang H, Liu L, Zhang Y, Ling S, Hu J, Li N, Wang J, Lv C, Yang X. A fully human monoclonal antibody targeting Semaphorin 5A alleviates the progression of rheumatoid arthritis. Biomed Pharmacother 2023; 168:115666. [PMID: 37832409 DOI: 10.1016/j.biopha.2023.115666] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is the most common chronic autoimmune disease worldwide. Although progress has been made in RA treatment in recent decades, remission cannot be effectively achieved for a considerable proportion of RA patients. Thus, novel potential targets for therapeutic strategies are needed. Semaphorin 5A (SEMA5A) plays a pivotal role in RA progression by facilitating pannus formation, and it is a promising therapeutic target. In this study, we sought to develop an antibody treatment strategy targeting SEMA5A and evaluate its therapeutic effect using a collagen-induced arthritis (CIA) model. We generated SYD12-12, a fully human SEMA5A blocking antibody, through phage display technology. SYD12-12 intervention effectively inhibited angiogenesis and aggressive phenotypes of RA synoviocytes in vitro and dose-dependently inhibited synovial hyperplasia, pannus formation, bone destruction in CIA mice. Notably, SYD12-12 also improved the Treg/Th17 imbalance in CIA mice. We confirmed through immunofluorescence and molecular docking that SYD12-12 integrated with the unique TSP-1 domain of SEMA5A. In conclusion, we developed and characterized a fully human SEMA5A-blocking antibody for the first time. SYD12-12 effectively alleviated disease progression in CIA mice by inhibiting pannus formation and improving the Treg/Th17 imbalance, demonstrating its potential for the RA treatment.
Collapse
Affiliation(s)
- Yang Qin
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Jin
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiani Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanwen Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunwang Ling
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinzhu Hu
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Nuan Li
- Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianguang Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Chen Lv
- Department of Orthopedics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xinyu Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
11
|
Sachdeva M, Taneja S, Sachdeva N. Stem cell-like memory T cells: Role in viral infections and autoimmunity. World J Immunol 2023; 13:11-22. [DOI: 10.5411/wji.v13.i2.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/06/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023] Open
Abstract
Stem cell-like memory T (TSCM) cells possess stem cell properties including multipotency and self-renewal and are being recognized as emerging players in various human diseases. Advanced technologies such as multiparametric flowcytometry and single cell sequencing have enabled their identification and molecular characterization. In case of chronic viral diseases such as human immunodeficiency virus-1, CD4+ TSCM cells, serve as major reservoirs of the latent virus. However, during immune activation and functional exhaustion of effector T cells, these cells also possess the potential to replenish the pool of functional effector cells to curtail the infection. More recently, these cells are speculated to play important role in protective immunity following acute viral infections such as coronavirus disease 2019 and might be amenable for therapeutics by ex vivo expansion. Similarly, studies are also investigating their pathological role in driving autoimmune responses. However, there are several gaps in the understanding of the role of TSCM cells in viral and autoimmune diseases to make them potential therapeutic targets. In this minireview, we have attempted an updated compilation of the dyadic role of these complex TSCM cells during such human diseases along with their biology and transcriptional programs.
Collapse
Affiliation(s)
- Meenakshi Sachdeva
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Shivangi Taneja
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| |
Collapse
|
12
|
Li W, Crouse KK, Alley J, Frisbie RK, Fish SC, Andreyeva TA, Reed LA, Thorn M, DiMaggio G, Donovan CB, Bennett D, Garren J, Oziolor E, Qian J, Newman L, Vargas AP, Kumpf SW, Steyn SJ, Schnute ME, Thorarensen A, Hegen M, Stevens E, Collinge M, Lanz TA, Vincent F, Vincent MS, Berstein G. A Novel C-C Chemoattractant Cytokine (Chemokine) Receptor 6 (CCR6) Antagonist (PF-07054894) Distinguishes between Homologous Chemokine Receptors, Increases Basal Circulating CCR6 + T Cells, and Ameliorates Interleukin-23-Induced Skin Inflammation. J Pharmacol Exp Ther 2023; 386:80-92. [PMID: 37142443 DOI: 10.1124/jpet.122.001452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Abstract
Blocking chemokine receptor C-C chemoattractant cytokine (chemokine) receptor (CCR) 6-dependent T cell migration has therapeutic promise in inflammatory diseases. PF-07054894 is a novel CCR6 antagonist that blocked only CCR6, CCR7, and C-X-C chemoattractant cytokine (chemokine) receptor (CXCR) 2 in a β-arrestin assay panel of 168 G protein-coupled receptors. Inhibition of CCR6-mediated human T cell chemotaxis by (R)-4-((2-(((1,4-Dimethyl-1H-pyrazol-3-yl)(1-methylcyclopentyl)methyl)amino)-3,4-dioxocyclobut-1-en-1-yl)amino)-3-hydroxy-N,N-dimethylpicolinamide (PF-07054894) was insurmountable by CCR6 ligand, C-C motif ligand (CCL) 20. In contrast, blockade of CCR7-dependent chemotaxis in human T cells and CXCR2-dependent chemotaxis in human neutrophils by PF-07054894 were surmountable by CCL19 and C-X-C motif ligand 1, respectively. [3H]-PF-07054894 showed a slower dissociation rate for CCR6 than for CCR7 and CXCR2 suggesting that differences in chemotaxis patterns of inhibition could be attributable to offset kinetics. Consistent with this notion, an analog of PF-07054894 with fast dissociation rate showed surmountable inhibition of CCL20/CCR6 chemotaxis. Furthermore, pre-equilibration of T cells with PF-07054894 increased its inhibitory potency in CCL20/CCR6 chemotaxis by 10-fold. The functional selectivity of PF-07054894 for inhibition of CCR6 relative to CCR7 and CXCR2 is estimated to be at least 50- and 150-fold, respectively. When administered orally to naïve cynomolgus monkeys, PF-07054894 increased the frequency of CCR6+ peripheral blood T cells, suggesting that blockade of CCR6 inhibited homeostatic migration of T cells from blood to tissues. PF-07054894 inhibited interleukin-23-induced mouse skin ear swelling to a similar extent as genetic ablation of CCR6. PF-07054894 caused an increase in cell surface CCR6 in mouse and monkey B cells, which was recapitulated in mouse splenocytes in vitro. In conclusion, PF-07054894 is a potent and functionally selective CCR6 antagonist that blocks CCR6-mediated chemotaxis in vitro and in vivo. SIGNIFICANCE STATEMENT: The chemokine receptor, C-C chemoattractant cytokine (chemokine) receptor 6 (CCR6) plays a key role in the migration of pathogenic lymphocytes and dendritic cells into sites of inflammation. (R)-4-((2-(((1,4-Dimethyl-1H-pyrazol-3-yl)(1-methylcyclopentyl)methyl)amino)-3,4-dioxocyclobut-1-en-1-yl)amino)-3-hydroxy-N,N-dimethylpicolinamide (PF-07054894) is a novel CCR6 small molecule antagonist that illustrates the importance of binding kinetics in achieving pharmacological potency and selectivity. Orally administered PF-07054894 blocks homeostatic and pathogenic functions of CCR6, suggesting that it is a promising therapeutic agent for the treatment of a variety of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Wei Li
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Kimberly K Crouse
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Jennifer Alley
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Richard K Frisbie
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Susan C Fish
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Tatyana A Andreyeva
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Lori A Reed
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Mitchell Thorn
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Giovanni DiMaggio
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Carol B Donovan
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Donald Bennett
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Jeonifer Garren
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Elias Oziolor
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Jesse Qian
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Leah Newman
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Amanda P Vargas
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Steven W Kumpf
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Stefan J Steyn
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Mark E Schnute
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Atli Thorarensen
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Martin Hegen
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Erin Stevens
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Mark Collinge
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Thomas A Lanz
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Fabien Vincent
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Michael S Vincent
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| | - Gabriel Berstein
- Inflammation and Immunology Research Unit (W.L., K.K.C., J.A., S.C.F., T.A.A., M.H., M.S.V., G.B.), Biostatistics (D.B., J.G.), and Medicine Design (S.J.S., M.E.S., A.T.), Pfizer, Inc., Cambridge, Massachusetts, and Primary Pharmacology Group (R.K.F., F.V.), Clinical Biomarkers (M.T., E.S.), and Drug Safety Research and Development (L.A.R., G.D., C.B.D., E.O., J.Q., L.N., A.P.V., S.W.K., M.C., T.A.L.), Pfizer, Inc., Groton, Connecticut
| |
Collapse
|
13
|
Cheng H, Chen W, Lin Y, Zhang J, Song X, Zhang D. Signaling pathways involved in the biological functions of dendritic cells and their implications for disease treatment. MOLECULAR BIOMEDICINE 2023; 4:15. [PMID: 37183207 PMCID: PMC10183318 DOI: 10.1186/s43556-023-00125-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/02/2023] [Indexed: 05/16/2023] Open
Abstract
The ability of dendritic cells (DCs) to initiate and regulate adaptive immune responses is fundamental for maintaining immune homeostasis upon exposure to self or foreign antigens. The immune regulatory function of DCs is strictly controlled by their distribution as well as by cytokines, chemokines, and transcriptional programming. These factors work in conjunction to determine whether DCs exert an immunosuppressive or immune-activating function. Therefore, understanding the molecular signals involved in DC-dependent immunoregulation is crucial in providing insight into the generation of organismal immunity and revealing potential clinical applications of DCs. Considering the many breakthroughs in DC research in recent years, in this review we focused on three basic lines of research directly related to the biological functions of DCs and summarized new immunotherapeutic strategies involving DCs. First, we reviewed recent findings on DC subsets and identified lineage-restricted transcription factors that guide the development of different DC subsets. Second, we discussed the recognition and processing of antigens by DCs through pattern recognition receptors, endogenous/exogenous pathways, and the presentation of antigens through peptide/major histocompatibility complexes. Third, we reviewed how interactions between DCs and T cells coordinate immune homeostasis in vivo via multiple pathways. Finally, we summarized the application of DC-based immunotherapy for autoimmune diseases and tumors and highlighted potential research prospects for immunotherapy that targets DCs. This review provides a useful resource to better understand the immunomodulatory signals involved in different subsets of DCs and the manipulation of these immune signals can facilitate DC-based immunotherapy.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenjing Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yubin Lin
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jianan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoshuang Song
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
14
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
15
|
Murayama MA, Shimizu J, Miyabe C, Yudo K, Miyabe Y. Chemokines and chemokine receptors as promising targets in rheumatoid arthritis. Front Immunol 2023; 14:1100869. [PMID: 36860872 PMCID: PMC9968812 DOI: 10.3389/fimmu.2023.1100869] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that commonly causes inflammation and bone destruction in multiple joints. Inflammatory cytokines, such as IL-6 and TNF-α, play important roles in RA development and pathogenesis. Biological therapies targeting these cytokines have revolutionized RA therapy. However, approximately 50% of the patients are non-responders to these therapies. Therefore, there is an ongoing need to identify new therapeutic targets and therapies for patients with RA. In this review, we focus on the pathogenic roles of chemokines and their G-protein-coupled receptors (GPCRs) in RA. Inflamed tissues in RA, such as the synovium, highly express various chemokines to promote leukocyte migration, tightly controlled by chemokine ligand-receptor interactions. Because the inhibition of these signaling pathways results in inflammatory response regulation, chemokines and their receptors could be promising targets for RA therapy. The blockade of various chemokines and/or their receptors has yielded prospective results in preclinical trials using animal models of inflammatory arthritis. However, some of these strategies have failed in clinical trials. Nonetheless, some blockades showed promising results in early-phase clinical trials, suggesting that chemokine ligand-receptor interactions remain a promising therapeutic target for RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Jun Shimizu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Chie Miyabe
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazuo Yudo
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yoshishige Miyabe
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| |
Collapse
|
16
|
Jiang Y, Dai S, Jia L, Qin L, Zhang M, Liu H, Wang X, Pang R, Zhang J, Peng G, Li W. Single-cell transcriptomics reveals cell type-specific immune regulation associated with anti-NMDA receptor encephalitis in humans. Front Immunol 2022; 13:1075675. [PMID: 36544777 PMCID: PMC9762154 DOI: 10.3389/fimmu.2022.1075675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Anti-N-methyl-D-aspartate receptor encephalitis (anti-NMDARE) is a rare autoimmune disease, and the peripheral immune characteristics associated with anti-NMDARE antibodies remain unclear. Methods Herein, we characterized peripheral blood mononuclear cells from patients with anti-NMDARE and healthy individuals by single-cell RNA sequencing (scRNA-seq). Results The transcriptional profiles of 129,217 cells were assessed, and 21 major cell clusters were identified. B-cell activation and differentiation, plasma cell expansion, and excessive inflammatory responses in innate immunity were all identified. Patients with anti-NMDARE showed higher expression levels of CXCL8, IL1B, IL6, TNF, TNFSF13, TNFSF13B, and NLRP3. We observed that anti-NMDARE patients in the acute phase expressed high levels of DC_CCR7 in human myeloid cells. Moreover, we observed that anti-NMDARE effects include oligoclonal expansions in response to immunizing agents. Strong humoral immunity and positive regulation of lymphocyte activation were observed in acute stage anti-NMDARE patients. Discussion This high-dimensional single-cell profiling of the peripheral immune microenvironment suggests that potential mechanisms are involved in the pathogenesis and recovery of anti-NMDAREs.
Collapse
Affiliation(s)
- Yushu Jiang
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China,*Correspondence: Wei Li, ; Yushu Jiang,
| | - Shuhua Dai
- Department of Neurology, Henan Provincial People’s Hospital, Xinxiang Medical University, Zhengzhou, Henan, China
| | - Linlin Jia
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lingzhi Qin
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Milan Zhang
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huiqin Liu
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaojuan Wang
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Pang
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiewen Zhang
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Gongxin Peng
- China Center for Bioinformatics, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and School of Basic Medicine, Beijing, China
| | - Wei Li
- Department of Neurology, Henan Joint International Research Laboratory of Accurate Diagnosis, Treatment, Research and Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China,*Correspondence: Wei Li, ; Yushu Jiang,
| |
Collapse
|
17
|
Anthony-Gonda K, Ray A, Su H, Wang Y, Xiong Y, Lee D, Block A, Chilunda V, Weiselberg J, Zemelko L, Wang YY, Kleinsorge-Block S, Reese JS, de Lima M, Ochsenbauer C, Kappes JC, Dimitrov DS, Orentas R, Deeks SG, Rutishauser RL, Berman JW, Goldstein H, Dropulić B. In vivo killing of primary HIV-infected cells by peripheral-injected early memory-enriched anti-HIV duoCAR T cells. JCI Insight 2022; 7:e161698. [PMID: 36345941 PMCID: PMC9675454 DOI: 10.1172/jci.insight.161698] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/14/2022] [Indexed: 11/09/2022] Open
Abstract
HIV-specific chimeric antigen receptor-T cell (CAR T cell) therapies are candidates to functionally cure HIV infection in people with HIV (PWH) by eliminating reactivated HIV-infected cells derived from latently infected cells within the HIV reservoir. Paramount to translating such therapeutic candidates successfully into the clinic will require anti-HIV CAR T cells to localize to lymphoid tissues in the body and eliminate reactivated HIV-infected cells such as CD4+ T cells and monocytes/macrophages. Here we show that i.v. injected anti-HIV duoCAR T cells, generated using a clinical-grade anti-HIV duoCAR lentiviral vector, localized to the site of active HIV infection in the spleen of humanized mice and eliminated HIV-infected PBMCs. CyTOF analysis of preinfusion duoCAR T cells revealed an early memory phenotype composed predominantly of CCR7+ stem cell-like/central memory T cells (TSCM/TCM) with expression of some effector-like molecules. In addition, we show that anti-HIV duoCAR T cells effectively sense and kill HIV-infected CD4+ T cells and monocytes/macrophages. Furthermore, we demonstrate efficient genetic modification of T cells from PWH on suppressive ART into anti-HIV duoCAR T cells that subsequently kill autologous PBMCs superinfected with HIV. These studies support the safety and efficacy of anti-HIV duoCAR T cell therapy in our presently open phase I/IIa clinical trial (NCT04648046).
Collapse
Affiliation(s)
- Kim Anthony-Gonda
- Caring Cross, Gaithersburg, Maryland, USA
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Alex Ray
- Department of Microbiology & Immunology and
| | - Hang Su
- Department of Microbiology & Immunology and
| | - Yuge Wang
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Ying Xiong
- Caring Cross, Gaithersburg, Maryland, USA
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Danica Lee
- Department of Microbiology & Immunology and
| | | | - Vanessa Chilunda
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jessica Weiselberg
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lily Zemelko
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Yen Y. Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Sarah Kleinsorge-Block
- Stem Cell Transplant Program and Center for Regenerative Medicine, University Hospitals Seidman Cancer Center and Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jane S. Reese
- Stem Cell Transplant Program and Center for Regenerative Medicine, University Hospitals Seidman Cancer Center and Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Marcos de Lima
- Stem Cell Transplant Program and Center for Regenerative Medicine, University Hospitals Seidman Cancer Center and Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Christina Ochsenbauer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John C. Kappes
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Medical Center, Research Service, Birmingham, Alabama, USA
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rimas Orentas
- Caring Cross, Gaithersburg, Maryland, USA
- Department of Pediatrics, University of Washington School of Medicine, and Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research lnstitute, Seattle, Washington, USA
| | - Steven G. Deeks
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Joan W. Berman
- Department of Microbiology & Immunology and
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Harris Goldstein
- Department of Microbiology & Immunology and
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Boro Dropulić
- Caring Cross, Gaithersburg, Maryland, USA
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| |
Collapse
|
18
|
Lu X, Crowley SD. Actions of Dendritic Cells in the Kidney during Hypertension. Compr Physiol 2022; 12:4087-4101. [PMID: 35950656 DOI: 10.1002/cphy.c210050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The immune response plays a critical role in the pathogenesis of hypertension, and immune cell populations can promote blood pressure elevation via actions in the kidney. Among these cell lineages, dendritic cells (DCs), the most potent antigen-presenting cells, play a central role in regulating immune response during hypertension and kidney disease. DCs have different subtypes, and renal DCs are comprised of the CD103+ CD11b- and CD103- CD11b+ subsets. DCs become mature and express costimulatory molecules on their surface once they encounter antigen. Isolevuglandin-modified proteins function as antigens to activate DCs and trigger them to stimulate T cells. Activated T cells accumulate in the hypertensive kidney, release effector cytokines, promote renal oxidative stress, and promote renal salt and water retention. Individual subsets of activated T cells can secrete tumor necrosis factor-alpha, interleukin-17A, and interferon-gamma, each of which has augmented the elevation of blood pressure in hypertensive models by enhancing renal sodium transport. Fms-like tyrosine kinase 3 ligand-dependent classical DCs are required to sustain the full hypertensive response, but C-X3 -C chemokine receptor 1 positive DCs do not regulate blood pressure. Excess sodium enters the DC through transporters to activate DCs, whereas the ubiquitin editor A20 in dendritic cells constrains blood pressure elevation by limiting T cell activation. By contrast, activation of the salt sensing kinase, serum/glucocorticoid kinase 1 in DCs exacerbates salt-sensitive hypertension. This article discusses recent studies illustrating mechanisms through which DC-T cell interactions modulate levels of pro-hypertensive mediators to regulate blood pressure via actions in the kidney. © 2022 American Physiological Society. Compr Physiol 12:1-15, 2022.
Collapse
Affiliation(s)
- Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| |
Collapse
|
19
|
Zhao J, Wei K, Jiang P, Chang C, Xu L, Xu L, Shi Y, Guo S, He D. G-Protein-Coupled Receptors in Rheumatoid Arthritis: Recent Insights into Mechanisms and Functional Roles. Front Immunol 2022; 13:907733. [PMID: 35874704 PMCID: PMC9304905 DOI: 10.3389/fimmu.2022.907733] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to joint damage and even disability. Although there are various clinical therapies for RA, some patients still have poor or no response. Thus, the development of new drug targets remains a high priority. In this review, we discuss the role of G-protein-coupled receptors (GPCRs), including chemokine receptors, melanocortin receptors, lipid metabolism-related receptors, adenosine receptors, and other inflammation-related receptors, on mechanisms of RA, such as inflammation, lipid metabolism, angiogenesis, and bone destruction. Additionally, we summarize the latest clinical trials on GPCR targeting to provide a theoretical basis and guidance for the development of innovative GPCR-based clinical drugs for RA.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
20
|
Dendritic cells in systemic lupus erythematosus: From pathogenesis to therapeutic applications. J Autoimmun 2022; 132:102856. [DOI: 10.1016/j.jaut.2022.102856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
|
21
|
Hu H, Tang N, Zhang F, Li L, Li L. Bioinformatics and System Biology Approach to Identify the Influences of COVID-19 on Rheumatoid Arthritis. Front Immunol 2022; 13:860676. [PMID: 35464423 PMCID: PMC9021444 DOI: 10.3389/fimmu.2022.860676] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Background Severe coronavirus disease 2019 (COVID -19) has led to a rapid increase in mortality worldwide. Rheumatoid arthritis (RA) was a high-risk factor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas the molecular mechanisms underlying RA and CVOID-19 are not well understood. The objectives of this study were to analyze potential molecular mechanisms and identify potential drugs for the treatment of COVID-19 and RA using bioinformatics and a systems biology approach. Methods Two Differentially expressed genes (DEGs) sets extracted from GSE171110 and GSE1775544 datasets were intersected to generate common DEGs, which were used for functional enrichment, pathway analysis, and candidate drugs analysis. Results A total of 103 common DEGs were identified in the two datasets between RA and COVID-19. A protein-protein interaction (PPI) was constructed using various combinatorial statistical methods and bioinformatics tools. Subsequently, hub genes and essential modules were identified from the PPI network. In addition, we performed functional analysis and pathway analysis under ontological conditions and found that there was common association between RA and progression of COVID-19 infection. Finally, transcription factor-gene interactions, protein-drug interactions, and DEGs-miRNAs coregulatory networks with common DEGs were also identified in the datasets. Conclusion We successfully identified the top 10 hub genes that could serve as novel targeted therapy for COVID-19 and screened out some potential drugs useful for COVID-19 patients with RA.
Collapse
Affiliation(s)
- Huan Hu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Nana Tang
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Facai Zhang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Long Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
22
|
Li K, Lai C, Liu C, Li Z, Guo K, Xu K. WGCNA and molecular docking reveal key hub genes and potential natural inhibitor in interstitial cystitis/bladder pain syndrome. Int Urogynecol J 2022; 33:2241-2249. [PMID: 35333927 DOI: 10.1007/s00192-022-05113-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION AND HYPOTHESIS The etiology and treatment of interstitial cystitis/bladder pain syndrome are still controversial. The purpose of this study is to determine the key genes and specific regulatory pathways related to it and to find potential drug-active components through integrated bioinformatics. METHODS The data set GSE11783 was downloaded from GEO database. The modules significantly related to interstitial cystitis/bladder pain syndrome were identified by weighted correlation network analysis. The genes in the key modules were analyzed by functional enrichment and protein interaction by Cytoscape software, and finally the core hub genes were screened. Furthermore, the molecular docking verification of active components and key proteins was carried out by using AutoDock Vin software. RESULTS Among the 14 modules derived from WGCNA, turquoise module had the highest correlation with IC/BPS (r = 0.85, P < 0.001). The genes in the module were mainly enriched in the biological processes such as the interaction between cytokines and cytokine receptors and chemokine signaling pathway. The genes in the related modules of differentially expressed genes and WGCNA traits were intersected to obtain the core hub genes. Protein-protein interaction network analysis showed that the key genes were upregulated genes CCR7 and CCL19. In terms of molecular docking, triptolide, the active component in the traditional anti-inflammatory drug Tripterygium wilfordii, can form effective molecular binding with both core hub genes. CONCLUSIONS Our study identified the core hub genes CCR7 and CCL19, which acted as essential components in interstitial cystitis/bladder pain syndrome. Furthermore, CCR7 and CCL19 can form effective binding with triptolide, which will provide new insights into the development of new therapies for interstitial cystitis/bladder pain syndrome.
Collapse
Affiliation(s)
- Kuiqing Li
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Cong Lai
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Cheng Liu
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Zhuohang Li
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Kaixuan Guo
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Kewei Xu
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China.
| |
Collapse
|
23
|
Hong W, Yang B, He Q, Wang J, Weng Q. New Insights of CCR7 Signaling in Dendritic Cell Migration and Inflammatory Diseases. Front Pharmacol 2022; 13:841687. [PMID: 35281921 PMCID: PMC8914285 DOI: 10.3389/fphar.2022.841687] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
CCR7, collaborated with its ligands CCL19 and CCL21, controls extensive migratory events in the immune system. CCR7-bearing dendritic cells can swarm into T-cell zones in lymph nodes, initiating the antigen presentation and T-cell response. Abnormal expression of CCR7 in dendritic cells will cause a series of inflammatory diseases due to the chaotic dendritic cell trafficking. In this review, we take an in-depth look at the structural–functional domains of CCR7 and CCR7-bearing dendritic cell trajectory to lymph nodes. Then, we summarize the regulatory network of CCR7, including transcriptional regulation, translational and posttranslational regulation, internalization, desensitization, and recycling. Furthermore, the potential strategies of targeting the CCR7 network to regulate dendritic cell migration and to deal with inflammatory diseases are integrated, which not only emphasizes the possibility of CCR7 to be a potential target of immunotherapy but also has an implication on the homing of dendritic cells to benefit inflammatory diseases.
Collapse
Affiliation(s)
- Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- *Correspondence: Qinjie Weng, ; Jiajia Wang,
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Qinjie Weng, ; Jiajia Wang,
| |
Collapse
|
24
|
Gowhari Shabgah A, Al-Obaidi ZMJ, Sulaiman Rahman H, Kamal Abdelbasset W, Suksatan W, Bokov DO, Thangavelu L, Turki Jalil A, Jadidi-Niaragh F, Mohammadi H, Mashayekhi K, Gholizadeh Navashenaq J. Does CCL19 act as a double-edged sword in cancer development? Clin Exp Immunol 2021; 207:164-175. [PMID: 35020885 PMCID: PMC8982982 DOI: 10.1093/cei/uxab039] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is considered a life-threatening disease, and several factors are involved in its development. Chemokines are small proteins that physiologically exert pivotal roles in lymphoid and non-lymphoid tissues. The imbalance or dysregulation of chemokines has contributed to the development of several diseases, especially cancer. CCL19 is one of the homeostatic chemokines that is abundantly expressed in the thymus and lymph nodes. This chemokine, which primarily regulates immune cell trafficking, is involved in cancer development. Through the induction of anti-tumor immune responses and inhibition of angiogenesis, CCL19 exerts tumor-suppressive functions. In contrast, CCL19 also acts as a tumor-supportive factor by inducing inflammation, cell growth, and metastasis. Moreover, CCL19 dysregulation in several cancers, including colorectal, breast, pancreatic, and lung cancers, has been considered a tumor biomarker for diagnosis and prognosis. Using CCL19-based therapeutic approaches has also been proposed to overcome cancer development. This review will shed more light on the multifarious function of CCL19 in cancer and elucidate its application in diagnosis, prognosis, and even therapy. It is expected that the study of CCL19 in cancer might be promising to broaden our knowledge of cancer development and might introduce novel approaches in cancer management.
Collapse
Affiliation(s)
| | - Zaid Mahdi Jaber Al-Obaidi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Alkafeel, Najaf, Iraq,Department of Chemistry and Biochemistry, College of Medicine, University of Kerbala, Karbala, Iraq
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Dmitry O Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation,Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha institute of medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abduladheem Turki Jalil
- Faculty of Biology and Ecology, Yanka Kupala State University of Grodno, Grodno, Belarus,College of Technical Engineering, The Islamic University, Najaf, Iraq
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran,Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Kazem Mashayekhi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Jamshid Gholizadeh Navashenaq
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran,Correspondence: Jamshid Gholizadeh Navashenaq, Bam University of Medical Sciences, Bam, Kerman, Iran. E-mail: ;
| |
Collapse
|
25
|
Cuesta-Mateos C, Terrón F, Herling M. CCR7 in Blood Cancers - Review of Its Pathophysiological Roles and the Potential as a Therapeutic Target. Front Oncol 2021; 11:736758. [PMID: 34778050 PMCID: PMC8589249 DOI: 10.3389/fonc.2021.736758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
According to the classical paradigm, CCR7 is a homing chemokine receptor that grants normal lymphocytes access to secondary lymphoid tissues such as lymph nodes or spleen. As such, in most lymphoproliferative disorders, CCR7 expression correlates with nodal or spleen involvement. Nonetheless, recent evidence suggests that CCR7 is more than a facilitator of lymphatic spread of tumor cells. Here, we review published data to catalogue CCR7 expression across blood cancers and appraise which classical and novel roles are attributed to this receptor in the pathogenesis of specific hematologic neoplasms. We outline why novel therapeutic strategies targeting CCR7 might provide clinical benefits to patients with CCR7-positive hematopoietic tumors.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto la Princesa (IIS-IP), Madrid, Spain.,Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Fernando Terrón
- Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Marco Herling
- Clinic of Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
26
|
Dendritic cell migration in inflammation and immunity. Cell Mol Immunol 2021; 18:2461-2471. [PMID: 34302064 PMCID: PMC8298985 DOI: 10.1038/s41423-021-00726-4] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023] Open
Abstract
Dendritic cells (DCs) are the key link between innate immunity and adaptive immunity and play crucial roles in both the promotion of immune defense and the maintenance of immune tolerance. The trafficking of distinct DC subsets across lymphoid and nonlymphoid tissues is essential for DC-dependent activation and regulation of inflammation and immunity. DC chemotaxis and migration are triggered by interactions between chemokines and their receptors and regulated by multiple intracellular mechanisms, such as protein modification, epigenetic reprogramming, metabolic remodeling, and cytoskeletal rearrangement, in a tissue-specific manner. Dysregulation of DC migration may lead to abnormal positioning or activation of DCs, resulting in an imbalance of immune responses and even immune pathologies, including autoimmune responses, infectious diseases, allergic diseases and tumors. New strategies targeting the migration of distinct DC subsets are being explored for the treatment of inflammatory and infectious diseases and the development of novel DC-based vaccines. In this review, we will discuss the migratory routes and immunological consequences of distinct DC subsets, the molecular basis and regulatory mechanisms of migratory signaling in DCs, and the association of DC migration with the pathogenesis of autoimmune and infectious diseases.
Collapse
|
27
|
Brandum EP, Jørgensen AS, Rosenkilde MM, Hjortø GM. Dendritic Cells and CCR7 Expression: An Important Factor for Autoimmune Diseases, Chronic Inflammation, and Cancer. Int J Mol Sci 2021; 22:ijms22158340. [PMID: 34361107 PMCID: PMC8348795 DOI: 10.3390/ijms22158340] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
Chemotactic cytokines-chemokines-control immune cell migration in the process of initiation and resolution of inflammatory conditions as part of the body's defense system. Many chemokines also participate in pathological processes leading up to and exacerbating the inflammatory state characterizing chronic inflammatory diseases. In this review, we discuss the role of dendritic cells (DCs) and the central chemokine receptor CCR7 in the initiation and sustainment of selected chronic inflammatory diseases: multiple sclerosis (MS), rheumatoid arthritis (RA), and psoriasis. We revisit the binary role that CCR7 plays in combatting and progressing cancer, and we discuss how CCR7 and DCs can be harnessed for the treatment of cancer. To provide the necessary background, we review the differential roles of the natural ligands of CCR7, CCL19, and CCL21 and how they direct the mobilization of activated DCs to lymphoid organs and control the formation of associated lymphoid tissues (ALTs). We provide an overview of DC subsets and, briefly, elaborate on the different T-cell effector types generated upon DC-T cell priming. In the conclusion, we promote CCR7 as a possible target of future drugs with an antagonistic effect to reduce inflammation in chronic inflammatory diseases and an agonistic effect for boosting the reactivation of the immune system against cancer in cell-based and/or immune checkpoint inhibitor (ICI)-based anti-cancer therapy.
Collapse
|
28
|
Carlsson E, Beresford MW, Ramanan AV, Dick AD, Hedrich CM. Juvenile Idiopathic Arthritis Associated Uveitis. CHILDREN-BASEL 2021; 8:children8080646. [PMID: 34438537 PMCID: PMC8393258 DOI: 10.3390/children8080646] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 01/31/2023]
Abstract
Juvenile idiopathic arthritis (JIA) is the most common childhood rheumatic disease. The development of associated uveitis represents a significant risk for serious complications, including permanent loss of vision. Initiation of early treatment is important for controlling JIA-uveitis, but the disease can appear asymptomatically, making frequent screening procedures necessary for patients at risk. As our understanding of pathogenic drivers is currently incomplete, it is difficult to assess which JIA patients are at risk of developing uveitis. Identification of specific risk factors for JIA-associated uveitis is an important field of research, and in this review, we highlight the genomic, transcriptomic, and proteomic factors identified as potential uveitis risk factors in JIA, and discuss therapeutic strategies.
Collapse
Affiliation(s)
- Emil Carlsson
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L14 5AB, UK;
- Correspondence: (E.C.); (C.M.H.); Tel.: +44-151-228-4811 (ext. 2690) (E.C.); +44-151-252-5849 (C.M.H.)
| | - Michael W. Beresford
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L14 5AB, UK;
- Department of Rheumatology, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool L14 5AB, UK
- National Institute for Health Research Alder Hey Clinical Research Facility, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool L14 5AB, UK
| | - Athimalaipet V. Ramanan
- Bristol Royal Hospital for Children & Translational Health Sciences, University of Bristol, Bristol BS2 8DZ, UK;
| | - Andrew D. Dick
- Translational Health Sciences, University of Bristol, Bristol BS2 8DZ, UK;
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital, London EC1V 2PD, UK
| | - Christian M. Hedrich
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L14 5AB, UK;
- Department of Rheumatology, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool L14 5AB, UK
- National Institute for Health Research Alder Hey Clinical Research Facility, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool L14 5AB, UK
- Correspondence: (E.C.); (C.M.H.); Tel.: +44-151-228-4811 (ext. 2690) (E.C.); +44-151-252-5849 (C.M.H.)
| |
Collapse
|
29
|
Huang J, Fu X, Chen X, Li Z, Huang Y, Liang C. Promising Therapeutic Targets for Treatment of Rheumatoid Arthritis. Front Immunol 2021; 12:686155. [PMID: 34305919 PMCID: PMC8299711 DOI: 10.3389/fimmu.2021.686155] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic poly-articular chronic autoimmune joint disease that mainly damages the hands and feet, which affects 0.5% to 1.0% of the population worldwide. With the sustained development of disease-modifying antirheumatic drugs (DMARDs), significant success has been achieved for preventing and relieving disease activity in RA patients. Unfortunately, some patients still show limited response to DMARDs, which puts forward new requirements for special targets and novel therapies. Understanding the pathogenetic roles of the various molecules in RA could facilitate discovery of potential therapeutic targets and approaches. In this review, both existing and emerging targets, including the proteins, small molecular metabolites, and epigenetic regulators related to RA, are discussed, with a focus on the mechanisms that result in inflammation and the development of new drugs for blocking the various modulators in RA.
Collapse
Affiliation(s)
- Jie Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xuekun Fu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xinxin Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Zheng Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Yuhong Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China.,Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
30
|
Mohd Shukri ND, Farah Izati A, Wan Ghazali WS, Che Hussin CM, Wong KK. CD3 +CD4 +gp130 + T Cells Are Associated With Worse Disease Activity in Systemic Lupus Erythematosus Patients. Front Immunol 2021; 12:675250. [PMID: 34149710 PMCID: PMC8213373 DOI: 10.3389/fimmu.2021.675250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023] Open
Abstract
The receptors for IL-35, IL-12Rβ2 and gp130, have been implicated in the inflammatory pathophysiology of autoimmune diseases. In this study, we set out to investigate the serum IL-35 levels and the surface levels of IL-12Rβ2 and gp130 in CD3+CD4+, CD3+CD4─ and CD3─CD4─ lymphocyte subpopulations in systemic lupus erythematosus (SLE) patients (n=50) versus healthy controls (n=50). The potential T cell subsets associated with gp130 transcript (i.e. IL6ST) expression in CD4+ T cells of SLE patients was also examined in publicly-available gene expression profiling (GEP) datasets. Here, we report that serum IL-35 levels were significantly higher in SLE patients than healthy controls (p=0.038) but it was not associated with SLEDAI-2K scores. The proportions of IL-12Rβ2+ and gp130+ cells in SLE patients did not differ significantly with those of healthy controls in all lymphocyte subpopulations investigated. Essentially, higher SLEDAI-2K scores were positively correlated with increased proportion of gp130+ cells, but not IL-12Rβ2+ cells, on CD3+CD4+ T cells (r=0.425, p=0.002, q=0.016). Gene Set Enrichment Analysis (GSEA) of a GEP dataset of CD4+ T cells isolated from SLE patients (n=8; GSE4588) showed that IL6ST expression was positively associated with genes upregulated in CD4+ T cells vs myeloid or B cells (q<0.001). In an independent GEP dataset of CD4+ T cells isolated from SLE patients (n=9; GSE1057), IL6ST expression was induced upon anti-CD3 stimulation, and that Treg, TCM and CCR7+ T cells gene sets were significantly enriched (q<0.05) by genes highly correlated with IL6ST expression (n=92 genes; r>0.75 with IL6ST expression) upon anti-CD3 stimulation in these SLE patients. In conclusion, gp130 signaling in CD3+CD4+ T cell subsets may contribute to increased disease activity in SLE patients, and it represents a promising therapeutic target for inhibition in the disease.
Collapse
Affiliation(s)
- Nur Diyana Mohd Shukri
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Aziz Farah Izati
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Wan Syamimee Wan Ghazali
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Che Maraina Che Hussin
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
31
|
Jeon M, Jagodnik KM, Kropiwnicki E, Stein DJ, Ma'ayan A. Prioritizing Pain-Associated Targets with Machine Learning. Biochemistry 2021; 60:1430-1446. [PMID: 33606503 DOI: 10.1021/acs.biochem.0c00930] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
While hundreds of genes have been associated with pain, much of the molecular mechanisms of pain remain unknown. As a result, current analgesics are limited to few clinically validated targets. Here, we trained a machine learning (ML) ensemble model to predict new targets for 17 categories of pain. The model utilizes features from transcriptomics, proteomics, and gene ontology to prioritize targets for modulating pain. We focused on identifying novel G-protein-coupled receptors (GPCRs), ion channels, and protein kinases because these proteins represent the most successful drug target families. The performance of the model to predict novel pain targets is 0.839 on average based on AUROC, while the predictions for arthritis had the highest accuracy (AUROC = 0.929). The model predicts hundreds of novel targets for pain; for example, GPR132 and GPR109B are highly ranked GPCRs for rheumatoid arthritis. Overall, gene-pain association predictions cluster into three groups that are enriched for cytokine, calcium, and GABA-related cell signaling pathways. These predictions can serve as a foundation for future experimental exploration to advance the development of safer and more effective analgesics.
Collapse
Affiliation(s)
- Minji Jeon
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Kathleen M Jagodnik
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Eryk Kropiwnicki
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Daniel J Stein
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| |
Collapse
|
32
|
Cuesta-Mateos C, Brown JR, Terrón F, Muñoz-Calleja C. Of Lymph Nodes and CLL Cells: Deciphering the Role of CCR7 in the Pathogenesis of CLL and Understanding Its Potential as Therapeutic Target. Front Immunol 2021; 12:662866. [PMID: 33841445 PMCID: PMC8024566 DOI: 10.3389/fimmu.2021.662866] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
The lymph node (LN) is an essential tissue for achieving effective immune responses but it is also critical in the pathogenesis of chronic lymphocytic leukemia (CLL). Within the multitude of signaling pathways aberrantly regulated in CLL the homeostatic axis composed by the chemokine receptor CCR7 and its ligands is the main driver for directing immune cells to home into the LN. In this literature review, we address the roles of CCR7 in the pathophysiology of CLL, and how this chemokine receptor is of critical importance to develop more rational and effective therapies for this malignancy.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- Biomarkers, Tumor
- Chemotaxis/genetics
- Chemotaxis/immunology
- Disease Susceptibility
- Gene Expression
- Humans
- Immune Tolerance
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Ligands
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Molecular Targeted Therapy
- Protein Binding
- Receptors, CCR7/antagonists & inhibitors
- Receptors, CCR7/genetics
- Receptors, CCR7/metabolism
- Tumor Microenvironment
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto de La Princesa (IIS-IP), Madrid, Spain
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
- Catapult Therapeutics BV, Lelystad, Netherlands
| | - Jennifer R. Brown
- Chronic Lymphocytic Leukemia (CLL) Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Fernando Terrón
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
- Catapult Therapeutics BV, Lelystad, Netherlands
| | - Cecilia Muñoz-Calleja
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto de La Princesa (IIS-IP), Madrid, Spain
- School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
33
|
ACPA Status Correlates with Differential Immune Profile in Patients with Rheumatoid Arthritis. Cells 2021; 10:cells10030647. [PMID: 33799480 PMCID: PMC8000255 DOI: 10.3390/cells10030647] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a progressive erosive autoimmune disease that affects 1% of the world population. Anti-citrullinated protein autoantibodies (ACPA) are routinely used for the diagnosis of RA, however 20–30% of patients are ACPA negative. ACPA status is a delineator of RA disease endotypes with similar clinical manifestation but potentially different pathophysiology. Profiling of key peripheral blood and synovial tissue immune populations including B cells, T follicular helper (Tfh) cells and CD4 T cell proinflammatory cytokine responses could elucidate the underlying immunological mechanisms involved and inform a treat to target approach for both ACPA-positive and ACPA-negative RA. Detailed high dimensionality flow cytometric analysis with supervised and unsupervised algorithm analysis revealed unique RA patient peripheral blood B cell and Tfh cell profiles. Synovial tissue single cell analysis of B cell subpopulation distribution was similar between ACPA− and ACPA+ RA patients, highlighting a key role for specific B cell subsets in both disease endotypes. Interestingly, synovial tissue single cell analysis of CD4 T cell proinflammatory cytokine production was markedly different between ACPA− and APCA+ RA patients. RNAseq analysis of RA patient synovial tissue highlighted disease endotype specific gene signatures. ACPA status associates with unique immune profile signatures that reinforce the need for a treat to target approach for both endotypes of RA.
Collapse
|
34
|
Cuesta-Mateos C, Juárez-Sánchez R, Mateu-Albero T, Loscertales J, Mol W, Terrón F, Muñoz-Calleja C. Targeting cancer homing into the lymph node with a novel anti-CCR7 therapeutic antibody: the paradigm of CLL. MAbs 2021; 13:1917484. [PMID: 33944659 PMCID: PMC8098074 DOI: 10.1080/19420862.2021.1917484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 11/25/2022] Open
Abstract
Lymph node (LN) is a key tissue in the pathophysiology of mature blood cancers, especially for chronic lymphocytic leukemia (CLL). Within the multiple de-regulated pathways affecting CLL homeostasis, the CC-chemokine receptor 7 (CCR7) grants homing of CLL cells into the LN where protective environments foster tumor progression. To cover the lack of specific therapies targeting the CCR7-dependence of CLL to enter into the LN, and aiming to displace the disease from LN, we generated CAP-100, an antibody that specifically binds to hCCR7 and neutralizes its ligand-binding site and signaling. In various in vitro and in vivo preclinical models CAP-100 strongly inhibited CCR7-induced migration, extravasation, homing, and survival in CLL samples. Moreover, it triggered potent tumor cell killing, mediated by host immune mechanisms, and was effective in xenograft models of high-risk disease. Additionally, CAP-100 showed a favorable toxicity profile on relevant hematopoietic subsets. Our results validated CAP-100 as a novel therapeutic tool to prevent the access of CLL cells, and other neoplasia with nodal-dependence, into the LN niches, thus hitting a central hub in the pathogenesis of cancer. The first-in-human clinical trial (NCT04704323), which will evaluate this novel therapeutic approach in CLL patients, is pending.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immed S.L., Immunological and Medicinal Products, Madrid, Spain
- Catapult Therapeutics, Lelystad, The Netherlands
- Immunology Department, Hospital Universitario De La Princesa, IIS-IP, Madrid, Spain
| | - Raquel Juárez-Sánchez
- Immed S.L., Immunological and Medicinal Products, Madrid, Spain
- Immunology Department, Hospital Universitario De La Princesa, IIS-IP, Madrid, Spain
| | - Tamara Mateu-Albero
- Immunology Department, Hospital Universitario De La Princesa, IIS-IP, Madrid, Spain
| | - Javier Loscertales
- Hematology Department, Hospital Universitario De La Princesa, IIS-IP, Madrid, Spain
| | - Wim Mol
- Catapult Therapeutics, Lelystad, The Netherlands
- Pepscan, Lelystad, The Netherlands
| | - Fernando Terrón
- Immed S.L., Immunological and Medicinal Products, Madrid, Spain
- Catapult Therapeutics, Lelystad, The Netherlands
| | - Cecilia Muñoz-Calleja
- Immunology Department, Hospital Universitario De La Princesa, IIS-IP, Madrid, Spain
- Medicine Faculty, Universidad Autónoma De Madrid, Madrid, Spain
| |
Collapse
|
35
|
Napier RJ, Lee EJ, Davey MP, Vance EE, Furtado JM, Snow PE, Samson KA, Lashley SJ, Brown BR, Horai R, Mattapallil MJ, Xu B, Callegan MC, Uebelhoer LS, Lancioni CL, Vehe RK, Binstadt BA, Smith JR, Caspi RR, Rosenzweig HL. T cell-intrinsic role for Nod2 in protection against Th17-mediated uveitis. Nat Commun 2020; 11:5406. [PMID: 33106495 PMCID: PMC7589501 DOI: 10.1038/s41467-020-18961-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/20/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations in nucleotide-binding oligomerization domain-containing protein 2 (NOD2) cause Blau syndrome, an inflammatory disorder characterized by uveitis. The antimicrobial functions of Nod2 are well-established, yet the cellular mechanisms by which dysregulated Nod2 causes uveitis remain unknown. Here, we report a non-conventional, T cell-intrinsic function for Nod2 in suppression of Th17 immunity and experimental uveitis. Reconstitution of lymphopenic hosts with Nod2-/- CD4+ T cells or retina-specific autoreactive CD4+ T cells lacking Nod2 reveals a T cell-autonomous, Rip2-independent mechanism for Nod2 in uveitis. In naive animals, Nod2 operates downstream of TCR ligation to suppress activation of memory CD4+ T cells that associate with an autoreactive-like profile involving IL-17 and Ccr7. Interestingly, CD4+ T cells from two Blau syndrome patients show elevated IL-17 and increased CCR7. Our data define Nod2 as a T cell-intrinsic rheostat of Th17 immunity, and open new avenues for T cell-based therapies for Nod2-associated disorders such as Blau syndrome.
Collapse
Affiliation(s)
- Ruth J Napier
- VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Ellen J Lee
- VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Michael P Davey
- VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Emily E Vance
- VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - João M Furtado
- Division of Ophthalmology, Ribeirão Preto Medical School, University of São Paulo, Butanta, Ribeirão Preto, Brazil
| | - Paige E Snow
- Department of Public Health, Oregon Health and Science University, Portland, OR, 97239, USA
| | | | - Sydney J Lashley
- VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, 97239, USA
| | | | - Reiko Horai
- Laboratory of Immunology, NEI, NIH, Bethesda, MD, 20814, USA
| | | | - Biying Xu
- Laboratory of Immunology, NEI, NIH, Bethesda, MD, 20814, USA
| | - Michelle C Callegan
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma, OK, 73104, USA.,Dean A. McGee Institute, Oklahoma City, OK, 73104, USA
| | - Luke S Uebelhoer
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Christina L Lancioni
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Richard K Vehe
- Department of Pediatrics, University of Minnesota and the University of Minnesota Masonic Children's Hospital, Minneapolis, MN, 55455, USA
| | - Bryce A Binstadt
- Department of Pediatrics, University of Minnesota and the University of Minnesota Masonic Children's Hospital, Minneapolis, MN, 55455, USA.,Center for Immunology and Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Justine R Smith
- College of Medicine and Public Health, Flinders University, Adelaide, SA, 5042, Australia
| | - Rachel R Caspi
- Laboratory of Immunology, NEI, NIH, Bethesda, MD, 20814, USA
| | - Holly L Rosenzweig
- VA Portland Health Care System, Portland, OR, 97239, USA. .,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
36
|
Van Raemdonck K, Umar S, Shahrara S. The pathogenic importance of CCL21 and CCR7 in rheumatoid arthritis. Cytokine Growth Factor Rev 2020; 55:86-93. [PMID: 32499193 PMCID: PMC10018533 DOI: 10.1016/j.cytogfr.2020.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/12/2020] [Indexed: 02/08/2023]
Abstract
Innate and adaptive immunity regulate the inflammatory and erosive phenotypes observed in rheumatoid arthritis (RA) patients. Hence, identifying novel pathways that participate in different stages of RA pathology will provide valuable insights concerning the mechanistic behavior of different joint leukocytes and the strategy to restrain their activity. Recent findings have revealed that CCL21 poses as a risk factor for RA and expression of its receptor, CCR7, on circulating monocytes is representative of the patient's disease activity score. Expression of CCR7 was found to be the hallmark of RA synovial fluid (SF) M1 macrophages (MФs) and its levels were potentiated in response to M1 mediating factors and curtailed by M2 mediators in naïve MФs. Intriguingly, although both CCR7 ligands, CCL19 and CCL21, are elevated in RA specimens, only CCL21 was predominately responsible for CCR7's pathological manifestation of RA. Unique subset of MФs differentiated in response to CCL21 stimulation, exhibited upregulation in Th17-polarizing monokines. Moreover, CCL21-activated monokines were capable of differentiating naïve T cells into joint Th17 cells, which also partook in RA osteoclastogenesis. Finally, to conserve chronic inflammation, SF CCL21 amplified RA neovascularization directly and indirectly by promoting RA FLS and MΦs to secrete proangiogenic factors, VEGF and IL-17. This review aims to shed light on the broad pathogenic impact of CCL21, linking immunostimulatory MФs with Th17 cells, while concurrently advancing RA bone destruction and neovascularization.
Collapse
Affiliation(s)
- Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, United States
| | - Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, United States
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, United States.
| |
Collapse
|
37
|
Guo X, Xu T, Zheng J, Cui X, Li M, Wang K, Su M, Zhang H, Zheng K, Sun C, Song S, Liu H. Accumulation of synovial fluid CD19 +CD24 hiCD27 + B cells was associated with bone destruction in rheumatoid arthritis. Sci Rep 2020; 10:14386. [PMID: 32873834 PMCID: PMC7462986 DOI: 10.1038/s41598-020-71362-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/10/2020] [Indexed: 12/29/2022] Open
Abstract
Regulatory CD19+CD24hiCD27+ B cells were proved to be numerically decreased and functionally impaired in the peripheral blood (PB) from rheumatoid arthritis (RA), with the potential of converting into osteoclast-priming cells. However, the distribution and function of CD19+CD24hiCD27+ B cells in RA synovial fluid (SF) were unclear. In this study, we investigated whether RA SF CD19+CD24hiCD27+ B cells were increased and associated with bone destruction. We found that the proportion of RA SF CD19+CD24hiCD27+ B cells was increased significantly, and was positively correlated with swollen joint counts, tender joint counts and disease activity. CXCL12, CXCL13, CCL19 contributed to the recruitment of CD19+CD24hiCD27+ B cells in RA SF. Notably, CD19+CD24hiCD27+ B cells in the SF from RA expressed significantly more RANKL compared to OA and that in the PB from RA. Critically, RA CD19+CD24hiCD27+ B cells promoted osteoclast (OC) differentiation in vitro, and the number of OCs was higher in cultures with RA SF CD19+CD24hiCD27+ B cells than in those derived from RA PB. Collectively, these findings revealed the accumulation of CD19+CD24hiCD27+ B cells in SF and their likely contribution to joint destruction in RA. Modulating the status of CD19+CD24hiCD27+ B cells might provide novel therapeutic strategies for RA.
Collapse
Affiliation(s)
- Xiaofeng Guo
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Tingting Xu
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Jing Zheng
- Department of Hematology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Yichang, 443000, Hubei Province, China
| | - Xiangjun Cui
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Ming Li
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Kai Wang
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Min Su
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Huifang Zhang
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Ke Zheng
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Chongling Sun
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Shulin Song
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China.
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China.
| |
Collapse
|
38
|
Miyabe Y, Miyabe C, Iwai Y, Luster AD. Targeting the Chemokine System in Rheumatoid Arthritis and Vasculitis. JMA J 2020; 3:182-192. [PMID: 33150252 PMCID: PMC7590389 DOI: 10.31662/jmaj.2020-0019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/27/2020] [Indexed: 01/01/2023] Open
Abstract
Arrest of circulating leukocytes and subsequent diapedesis is a fundamental component of inflammation. In general, the leukocyte migration cascade is tightly regulated by chemoattractants, such as chemokines. Chemokines, small secreted chemotactic cytokines, as well as their G-protein-coupled seven transmembrane spanning receptors, control the migratory patterns, positioning and cellular interactions of immune cells. Increased levels of chemokines and their receptors are found in the blood and within inflamed tissue in patients with rheumatoid arthritis (RA) and vasculitis. Chemokine ligand-receptor interactions regulate the recruitment of leukocytes into tissue, thus contributing in important ways to the pathogenesis of RA and vasculitis. Despite the fact that blockade of chemokines and chemokine receptors in animal models have yielded promising results, human clinical trials in RA using inhibitors of chemokines and their receptors have generally failed to show clinical benefits. However, recent early phase clinical trials suggest that strategies blocking specific chemokines may have clinical benefits in RA, demonstrating that the chemokine system remains a promising therapeutic target for rheumatic diseases, such as RA and vasuculitis and requires further study.
Collapse
Affiliation(s)
- Yoshishige Miyabe
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Chie Miyabe
- Department of Dermatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoshiko Iwai
- Department of Cell Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
39
|
Jaeger K, Bruenle S, Weinert T, Guba W, Muehle J, Miyazaki T, Weber M, Furrer A, Haenggi N, Tetaz T, Huang CY, Mattle D, Vonach JM, Gast A, Kuglstatter A, Rudolph MG, Nogly P, Benz J, Dawson RJP, Standfuss J. Structural Basis for Allosteric Ligand Recognition in the Human CC Chemokine Receptor 7. Cell 2020; 178:1222-1230.e10. [PMID: 31442409 PMCID: PMC6709783 DOI: 10.1016/j.cell.2019.07.028] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 11/15/2022]
Abstract
The CC chemokine receptor 7 (CCR7) balances immunity and tolerance by homeostatic trafficking of immune cells. In cancer, CCR7-mediated trafficking leads to lymph node metastasis, suggesting the receptor as a promising therapeutic target. Here, we present the crystal structure of human CCR7 fused to the protein Sialidase NanA by using data up to 2.1 Å resolution. The structure shows the ligand Cmp2105 bound to an intracellular allosteric binding pocket. A sulfonamide group, characteristic for various chemokine receptor ligands, binds to a patch of conserved residues in the Gi protein binding region between transmembrane helix 7 and helix 8. We demonstrate how structural data can be used in combination with a compound repository and automated thermal stability screening to identify and modulate allosteric chemokine receptor antagonists. We detect both novel (CS-1 and CS-2) and clinically relevant (CXCR1-CXCR2 phase-II antagonist Navarixin) CCR7 modulators with implications for multi-target strategies against cancer.
Collapse
Affiliation(s)
- Kathrin Jaeger
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI
| | - Steffen Bruenle
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI
| | - Tobias Weinert
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI
| | - Wolfgang Guba
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Jonas Muehle
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI; Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Takuya Miyazaki
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland; Chugai Pharmaceutical Co., Ltd., Research Division, Kamakura Research Labs, Kamakura, Kanagawa, Japan
| | - Martin Weber
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Antonia Furrer
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI
| | - Noemi Haenggi
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Tim Tetaz
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Chia-Ying Huang
- Macromolecular Crystallography, Swiss Light Source, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland
| | - Daniel Mattle
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI; Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Jean-Marie Vonach
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Alain Gast
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Andreas Kuglstatter
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Markus G Rudolph
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Przemyslaw Nogly
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI
| | - Joerg Benz
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Roger J P Dawson
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland.
| | - Joerg Standfuss
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen PSI.
| |
Collapse
|
40
|
Van Raemdonck K, Umar S, Palasiewicz K, Volkov S, Volin MV, Arami S, Chang HJ, Zanotti B, Sweiss N, Shahrara S. CCL21/CCR7 signaling in macrophages promotes joint inflammation and Th17-mediated osteoclast formation in rheumatoid arthritis. Cell Mol Life Sci 2020; 77:1387-1399. [PMID: 31342120 PMCID: PMC10040247 DOI: 10.1007/s00018-019-03235-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/08/2019] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
Abstract
In rheumatoid arthritis (RA), synovial tissue abundantly expresses CCL21, a chemokine strongly associated with RA susceptibility. In this study, we aimed to characterize the functional significance of CCL21/CCR7 signaling in different phases of RA pathogenesis. We determined that CCR7 is a hallmark of RA M1 synovial fluid (SF) macrophages, and its expression in RA monocytes and in vitro differentiated macrophages is closely associated with disease activity score (DAS28). In early stages of RA, monocytes infiltrate the synovial tissue. However, blockade of SF CCL21 or CCR7 prevents RA SF-mediated monocyte migration. CCR7 expression in the newly migrated macrophages can be accentuated by LPS and IFNγ and suppressed by IL-4 treatment. We also uncovered that CCL21 stimulation increases the number of M1-polarized macrophages (CD14+CD86+), resulting in elevated transcription of IL-6 and IL-23. These CCL21-induced M1 cytokines differentiate naïve T cells to Th17 cells, without affecting Th1 cell polarization. In the erosive stages of disease, CCL21 potentiates RA osteoclastogenesis through M1-driven Th17 polarization. Disruption of this intricate crosstalk, by blocking IL-6, IL-23, or IL-17 function, impairs the osteoclastogenic capacity of CCL21. Consistent with our in vitro findings, we establish that arthritis mediated by CCL21 expands the joint inflammation to bone erosion by connecting the differentiation of M1 macrophages with Th17 cells. Disease progression is further exacerbated by CCL21-induced neovascularization. We conclude that CCL21 is an attractive novel target for RA therapy, as blockade of its function may abrogate erosive arthritis modulated by M1 macrophages and Th17 cell crosstalk.
Collapse
Affiliation(s)
- Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Suncica Volkov
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Shiva Arami
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Huan J Chang
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA. .,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA.
| |
Collapse
|
41
|
Kasper M, Walscheid K, Laffer B, Bauer D, Busch M, Loser K, Vogl T, Langmann T, Ganser G, Rath T, Heiligenhaus A. Phenotype of Innate Immune Cells in Uveitis Associated with Axial Spondyloarthritis- and Juvenile Idiopathic Arthritis-associated Uveitis. Ocul Immunol Inflamm 2020; 29:1080-1089. [PMID: 32160102 DOI: 10.1080/09273948.2020.1715449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose: To analyze circulating immune cells in patients with anterior uveitis (AU) associated to axial spondyloarthritis (SpA), or juvenile idiopathic arthritis (JIA).Methods: Venous blood samples were collected from healthy controls (n = 16), and either SpA (n = 19) or JIA (n = 23) patients with associated anterior uveitis (AU) during active flare, or after ≥3 months of inactivity. Frequencies of CD56+, MHC-I+, and S100A9+ monocytes, CCR7+ dendritic cells, CD56+dim natural killer (NK) cells and CD3+CD56bright T-cells were analyzed via flow cytometry. Serum S100A8/A9 levels were determined via ELISA.Results: SpA patients showed a reduced frequency of CD56+dim NK cells during uveitis activity, a constitutively activated monocyte phenotype, and elevated S100A8/A9 serum levels. In contrast, JIAU patients showed elevated frequencies of CD56+ monocytes and CCR7+ DC.Conclusion: Phenotype of peripheral immune cells differ between patients, probably contributing to different courses of acute onset AU in SpA and insidious onset AU in JIAU patients.Abbreviations: AU: anterior uveitis, AR: arthritis, JIA: juvenile idiopathic arthritis, SpA: axial spondyloarthritis.
Collapse
Affiliation(s)
- Maren Kasper
- Department of Ophthalmology, Ophtha-Lab at St. Franziskus Hospital, Münster, Germany
| | - Karoline Walscheid
- Department of Ophthalmology, Ophtha-Lab at St. Franziskus Hospital, Münster, Germany.,University of Duisburg-Essen, Essen, Germany
| | - Björn Laffer
- Department of Ophthalmology, Ophtha-Lab at St. Franziskus Hospital, Münster, Germany.,University of Duisburg-Essen, Essen, Germany
| | - Dirk Bauer
- Department of Ophthalmology, Ophtha-Lab at St. Franziskus Hospital, Münster, Germany
| | - Martin Busch
- Department of Ophthalmology, Ophtha-Lab at St. Franziskus Hospital, Münster, Germany
| | - Karin Loser
- Department of Dermatology, Experimental Dermatology and Immunobiology of the Skin University of Münster, Münster, Germany
| | - Thomas Vogl
- Department of Immunology, University of Münster, Münster, Germany
| | - Thomas Langmann
- Experimental Immunology of the Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Gerd Ganser
- Department of Pediatric Rheumatology, St. Josef-Stift Sendenhorst, Sendenhorst, Germany
| | - Thomas Rath
- Department of Nephrology, Immunology and Osteology of St. Franziskus Hospital, Münster, Germany
| | - Arnd Heiligenhaus
- Department of Ophthalmology, Ophtha-Lab at St. Franziskus Hospital, Münster, Germany.,University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
42
|
Cuesta-Mateos C, Portero-Sainz I, García-Peydró M, Alcain J, Fuentes P, Juárez-Sánchez R, Pérez-García Y, Mateu-Albero T, Díaz-Fernández P, Vega-Piris L, Sánchez-López BA, Marcos-Jiménez A, Cardeñoso L, Gómez-García de Soria V, Toribio ML, Muñoz-Calleja C. Evaluation of therapeutic targeting of CCR7 in acute graft-versus-host disease. Bone Marrow Transplant 2020; 55:1935-1945. [PMID: 32086495 DOI: 10.1038/s41409-020-0830-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 01/22/2020] [Accepted: 02/04/2020] [Indexed: 12/19/2022]
Abstract
Graft-versus-host disease (GVHD) is the main complication after allogeneic hematopoietic stem cell transplantation. We previously unveiled a correlation between proportions of C-C motif chemokine receptor 7 (CCR7)+ T cells in the apheresis and the risk of developing GVHD. We wanted to evaluate in vivo whether apheresis with low proportion of CCR7+ cells or treatment with an anti-human CCR7 monoclonal antibody (mAb) were suitable strategies to prevent or treat acute GVHD in preclinical xenogeneic models. Therapeutic anti-CCR7 mAb was the most effective strategy in both prophylactic and therapeutic settings where antibody drastically reduced in vivo lymphoid organ infiltration of donor CCR7+ T cells, extended lifespan and solved clinical signs. The antibody neutralized in vitro migration of naïve and central memory T cells toward CCR7 ligands and depleted target CCR7+ subsets through complement activation. Both mechanisms of action spared CCR7- subsets, including effector memory and effector memory CD45RA+ T cells which may mediate graft versus leukemia effect and immunity against infections. Accordingly, the numbers of donor CCR7+ T cells in the apheresis were not associated to cytomegalovirus reactivation or the recurrence of the underlying disease. These findings provide a promising new strategy to prevent and treat acute GVHD, a condition where new specific, safety and effective treatment is needed.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain.,Immunological and Medicinal Products S.L. (IMMED), Madrid, Spain
| | - Itxaso Portero-Sainz
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Juan Alcain
- Centro de Biología Molecular Severo Ochoa (CBM-SO), Madrid, Spain
| | - Patricia Fuentes
- Centro de Biología Molecular Severo Ochoa (CBM-SO), Madrid, Spain
| | - Raquel Juárez-Sánchez
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain.,Immunological and Medicinal Products S.L. (IMMED), Madrid, Spain
| | - Yaiza Pérez-García
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Tamara Mateu-Albero
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Paula Díaz-Fernández
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Lorena Vega-Piris
- Methodology Unit, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Blanca A Sánchez-López
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Ana Marcos-Jiménez
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Laura Cardeñoso
- Department of Microbiology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Valle Gómez-García de Soria
- Department of Hematology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Cecilia Muñoz-Calleja
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain.
| |
Collapse
|
43
|
Chemokines in rheumatic diseases: pathogenic role and therapeutic implications. Nat Rev Rheumatol 2019; 15:731-746. [PMID: 31705045 DOI: 10.1038/s41584-019-0323-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2019] [Indexed: 12/20/2022]
Abstract
Chemokines, a family of small secreted chemotactic cytokines, and their G protein-coupled seven transmembrane spanning receptors control the migratory patterns, positioning and cellular interactions of immune cells. The levels of chemokines and their receptors are increased in the blood and within inflamed tissue of patients with rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, vasculitis or idiopathic inflammatory myopathies. Chemokine ligand-receptor interactions control the recruitment of leukocytes into tissue, which are central to the pathogenesis of these rheumatic diseases. Although the blockade of various chemokines and chemokine receptors has yielded promising results in preclinical animal models of rheumatic diseases, human clinical trials have, in general, been disappointing. However, there have been glimmers of hope from several early-phase clinical trials that suggest that sufficiently blocking the relevant chemokine pathway might in fact have clinical benefits in rheumatic diseases. Hence, the chemokine system remains a promising therapeutic target for rheumatic diseases and requires further study.
Collapse
|
44
|
Martínez-Burgo B, Cobb SL, Pohl E, Kashanin D, Paul T, Kirby JA, Sheerin NS, Ali S. A C-terminal CXCL8 peptide based on chemokine-glycosaminoglycan interactions reduces neutrophil adhesion and migration during inflammation. Immunology 2019; 157:173-184. [PMID: 31013364 DOI: 10.1111/imm.13063] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/27/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Leucocyte recruitment is critical during many acute and chronic inflammatory diseases. Chemokines are key mediators of leucocyte recruitment during the inflammatory response, by signalling through specific chemokine G-protein-coupled receptors (GPCRs). In addition, chemokines interact with cell-surface glycosaminoglycans (GAGs) to generate a chemotactic gradient. The chemokine interleukin-8/CXCL8, a prototypical neutrophil chemoattractant, is characterized by a long, highly positively charged GAG-binding C-terminal region, absent in most other chemokines. To examine whether the CXCL8 C-terminal peptide has a modulatory role in GAG binding during neutrophil recruitment, we synthesized the wild-type CXCL8 C-terminal [CXCL8 (54-72)] (Peptide 1), a peptide with a substitution of glutamic acid (E) 70 with lysine (K) (Peptide 2) to increase positive charge; and also, a scrambled sequence peptide (Peptide 3). Surface plasmon resonance showed that Peptide 1, corresponding to the core CXCL8 GAG-binding region, binds to GAG but Peptide 2 binding was detected at lower concentrations. In the absence of cellular GAG, the peptides did not affect CXCL8-induced calcium signalling or neutrophil chemotaxis along a diffusion gradient, suggesting no effect on GPCR binding. All peptides equally inhibited neutrophil adhesion to endothelial cells under physiological flow conditions. Peptide 2, with its greater positive charge and binding to polyanionic GAG, inhibited CXCL8-induced neutrophil transendothelial migration. Our studies suggest that the E70K CXCL8 peptide, may serve as a lead molecule for further development of therapeutic inhibitors of neutrophil-mediated inflammation based on modulation of chemokine-GAG binding.
Collapse
Affiliation(s)
- Beatriz Martínez-Burgo
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| | | | - Ehmke Pohl
- Chemistry Department, Durham University, Durham, UK
| | | | | | - John A Kirby
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| | - Neil S Sheerin
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| | - Simi Ali
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| |
Collapse
|
45
|
Khan MA, Khurana N, Ahmed RS, Umar S, Md G Sarwar AH, Alam Q, Kamal MA, Ashraf GM. Chemokines: A Potential Therapeutic Target to Suppress Autoimmune Arthritis. Curr Pharm Des 2019; 25:2937-2946. [PMID: 31580792 DOI: 10.2174/1381612825666190709205028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 06/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chemokines are a family of low molecular weight proteins that induce chemotaxis of inflammatory cells, which mainly depends on the recognition of a chemo-attractant gradient and interaction with the substratum. In Rheumatoid Arthritis (RA), abundant chemokines are expressed in synovial tissue, cause inflammatory cells migration into the inflamed joint that necessitates the formation of new blood vessels i.e. angiogenesis. Over the decades, studies showed that continuous inflammation may lead to the loss of tissue architecture and function, causing severe disability and cartilage destruction. In spite of the advancement of modern drug therapy, thousands of arthritic patients suffer mortality and morbidity globally. Thus, there is an urgent need for the development of novel therapeutic agents for the treatment of RA. METHODS This review is carried out throughout a non-systematic search of the accessible literature, will provide an overview of the current information of chemokine in RA and also exploring the future perspective of the vital role of targeting chemokine in RA treatment. RESULTS Since, chemokines are associated with inflammatory cells/leucocyte migration at the site of inflammation in chronic inflammatory diseases and hence, blockade or interference with chemokines activity showing a potential approach for the development of new anti-inflammatory agents. Currently, results obtained from both preclinical and clinical studies showed significant improvement in arthritis. CONCLUSION This review summarizes the role of chemokines and their receptors in the pathogenesis of RA and also indicates possible interactions of chemokines/receptors with various synthetic and natural compounds that may be used as a potential therapeutic target in the future for the treatment of RA.
Collapse
Affiliation(s)
- Mahmood A Khan
- Department of Biochemistry, University College of Medical Sciences & GTB Hospital, Dilshad Garden, Delhi 110095, India
| | - Nikhil Khurana
- Department of Biochemistry, University College of Medical Sciences & GTB Hospital, Dilshad Garden, Delhi 110095, India
| | - Rafat S Ahmed
- Department of Biochemistry, University College of Medical Sciences & GTB Hospital, Dilshad Garden, Delhi 110095, India
| | - Sadiq Umar
- Division of Rheumatology, University of Illinois, Clinical Science Building (CSB), Chicago, IL-60612, United States
| | - Abu H Md G Sarwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi-110025, India
| | - Qamre Alam
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Novel Global Community Educational Foundation, NSW, Australia
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
46
|
Xi J, Wang L, Yan C, Song J, Song Y, Chen J, Zhu Y, Chen Z, Jin C, Ding J, Zhao C. The Cancer Genome Atlas dataset-based analysis of aberrantly expressed genes by GeneAnalytics in thymoma associated myasthenia gravis: focusing on T cells. J Thorac Dis 2019; 11:2315-2323. [PMID: 31372268 DOI: 10.21037/jtd.2019.06.01] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Myasthenia gravis (MG) is a group of autoimmune disease which could be accompanied by thymoma. Many differences have been observed between thymoma-associated MG (TAMG) and non-MG thymoma (NMG). However, the molecular difference between them remained unknown. This study aimed to explore the differentially expressed genes (DEGs) between the two categories and to elucidate the possible pathogenesis of TAMG further. Methods DEGs were calculated using the RNA-Sequencing data from 11 TAMG and 10 NMG in The Cancer Genome Atlas (TCGA) database. GeneAnalytics was performed to characterize the associations between DEGs and tissues and cells, diseases, gene ontology (GO) terms, pathways, phenotypes, and drug/compounds, respectively. Genes related to T cells were sorted out using LifeMapDiscovery Cells and Tissues Database. Genes directly related to the phenotype of autoimmune diseases that were identified by VarElect were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Results The expression level of 169 genes showed a significant difference between the two groups, with 94 up-regulated and 75 down-regulated. Overexpression of six genes (ATM, SFTPB, ANKRD55, BTLA, CCR7, TNFRSF25), which are expressed in T cells and directly related to autoimmune disease through VarElect, was identified. The overexpression of soluble BTLA (sBTLA) (P=0.027), CCR7 (P=0.0018), TNFRSF25 (P=0.0013) and ANKRD55 (P=0.0026) was validated by RT-qPCR in thymoma tissues from our center. Conclusions Overexpression of sBTLA, CCR7, TNFRSF25 and ANKRD55 was identified and validated by RT-qPCR, which could partly explain the underlying pathogenesis in TAMG.
Collapse
Affiliation(s)
- Jianying Xi
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chong Yan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Song
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yang Song
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongjun Zhu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhiming Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chun Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Jianyong Ding
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Department of Neurology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai 200040, China
| |
Collapse
|
47
|
Miranda JP, Camões SP, Gaspar MM, Rodrigues JS, Carvalheiro M, Bárcia RN, Cruz P, Cruz H, Simões S, Santos JM. The Secretome Derived From 3D-Cultured Umbilical Cord Tissue MSCs Counteracts Manifestations Typifying Rheumatoid Arthritis. Front Immunol 2019; 10:18. [PMID: 30804924 PMCID: PMC6370626 DOI: 10.3389/fimmu.2019.00018] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder whose treatment is mostly restricted to pain and symptom management and to the delay of joint destruction. Mesenchymal stem/stromal cells from the umbilical cord tissue (UC-MSCs) have previously been proven to be immunomodulatory and more efficient than bone marrow-derived MSCs in causing remission of local and systemic arthritic manifestations in vivo. Given the paracrine nature of UC-MSC activity, their application as active substances can be replaced by their secretome, thus avoiding allogeneic rejection and safety issues related to unwanted grafting. In this work, we aimed at demonstrating the viability of applying the 3D-primed UC-MSC secretome for the amelioration of arthritic signs. A proteomic analysis was performed to both, media conditioned by UC-MSC monolayer (CM2D) and 3D cultures (CM3D). The analysis of relevant trophic factors confirmed secretome profiles with very significant differences in terms of therapeutic potential. Whereas, CM3D was characterised by a prevailing expression of anti-inflammatory cytokines such as IL-10 and LIF, along with trophic factors involved in different mechanisms leading to tissue regeneration, such as PDGF-BB, FGF-2, I-309, SCF, and GM-CSF; CM2D presented relatively higher levels of IL-6, MCP-1, and IL-21, with recognised pro-inflammatory roles in joint disease and pleiotropic effects in the progression of rheumatoid arthritis (RA). Accordingly, different motogenic effects over mouse chondrocytes and distinct capacities of inducing glycosaminoglycan synthesis in vitro were observed between CM3D and CM2D. Finally, the evaluation of arthritic manifestations in vivo, using an adjuvant-induced model for arthritis (AIA), suggested a significantly higher therapeutic potential of CM3D over CM2D and even UC-MSCs. Histological analysis confirmed a faster remission of local and systemic arthritic manifestations of CM3D-treated animals. Overall, the results show that the use of UC-MSC CM3D is a viable and better strategy than direct UC-MSC administration for counteracting AIA-related signs. This strategy represents a novel MSC-based but nonetheless cell-free treatment for arthritic conditions such as those characterising RA.
Collapse
Affiliation(s)
- Joana P Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Sérgio P Camões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Maria M Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Joana S Rodrigues
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Manuela Carvalheiro
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | | | | | | | - Sandra Simões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Jorge M Santos
- ECBio S.A., Amadora, Portugal.,Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente, Universidade do Porto, Porto, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|