1
|
Valdés N, Espinoza D, Pareja-Barrueto C, Olate N, Barraza-Rojas F, Benavides-Larenas A, Cortés M, Imarai M. Expression and regulation of the CXCL9-11 chemokines and CXCR3 receptor in Atlantic salmon (Salmo salar). Front Immunol 2024; 15:1455457. [PMID: 39301034 PMCID: PMC11410577 DOI: 10.3389/fimmu.2024.1455457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024] Open
Abstract
Chemokines are cytokines that mediate leukocyte traffic between the lymphoid organs, the bloodstream, and the site of tissue damage, which is essential for an efficient immune response. In particular, the gamma interferon (IFN- γ) inducible chemokines CXCL9, CXCL10, and CXCL11, and their receptor CXCR3, are involved in T cell and macrophage recruitment to the site of infection. The nature and function of these chemokines and their receptor are well-known in mammals, but further research is needed to achieve a similar level of understanding in fish immunity. Thus, in this study, we seek to identify the genes encoding the components of the Atlantic salmon (Salmo salar) CXCL9, CXCL10, CXCL11/CXCR3 axis (CXCL9-11/CXCR3), predict the protein structure from the amino acid sequence, and explore the regulation of gene expression as well as the response of these chemokines and their receptor to viral infections. The cxcl9, cxcl10, cxcl11, and cxcr3 gene sequences were retrieved from the databases, and the phylogenetic analysis was conducted to determine the evolutionary relationships. The study revealed an interesting pattern of clustering and conservation among fish and mammalian species. The salmon chemokine sequences clustered with orthologs from other fish species, while the mammalian sequences formed separate clades. This indicates a divergent evolution of chemokines between mammals and fish, possibly due to different evolutionary pressures. While the structural analysis of the chemokines and the CXCR3 receptor showed the conservation of critical motifs and domains, suggesting preserved functions and stability throughout evolution. Regarding the regulation of gene expression, some components of the CXCL9-11/CXCR3 axis are induced by recombinant gamma interferon (rIFN-γ) and by Infectious pancreatic necrosis virus (IPNV) infection in Atlantic salmon cells. Further studies are needed to explore the role of Atlantic salmon CXCL9-11 chemokines in regulating immune cell migration and endothelial activation, as seen in mammals. To the best of our knowledge, there have been no functional studies of chemokines to understand these effects in Atlantic salmon.
Collapse
Affiliation(s)
- Natalia Valdés
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Daniela Espinoza
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Claudia Pareja-Barrueto
- Departamento de Hematología y Oncología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicole Olate
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe Barraza-Rojas
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Almendra Benavides-Larenas
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Marcos Cortés
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Mónica Imarai
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
2
|
Peng D, Zhuge F, Wang M, Zhang B, Zhuang Z, Zhou R, Zhang Y, Li J, Yu Z, Shi J. Morus alba L. (Sangzhi) alkaloids mitigate atherosclerosis by regulating M1/M2 macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155526. [PMID: 38564921 DOI: 10.1016/j.phymed.2024.155526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/01/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Atherosclerosis (AS) is an important cause of cardiovascular disease, posing a substantial health risk. Recognized as a chronic inflammatory disorder, AS hinges on the pivotal involvement of macrophages in arterial inflammation, participating in its formation and progression. Sangzhi alkaloid (SZ-A) is a novel natural alkaloid extracted from the mulberry branches, has extensive pharmacological effects and stable pharmacokinetic characteristics. However, the effects and mechanisms of SZ-A on AS remain unclear. PURPOSE To explore the effect and underlying mechanisms of SZ-A on inflammation mediated by macrophages and its role in AS development. METHODS Atherosclerosis was induced in vivo in apolipoprotein E-deficient mice through a high-fat and high-choline diet. We utilized macrophages and vascular endothelial cells to investigate the effects of SZ-A on macrophage polarization and its anti-inflammatory properties on endothelial cells in vitro. The transcriptomic analyses were used to investigate the major molecule that mediates cell-cell interactions and the antiatherogenic mechanisms of SZ-A based on AS, subsequently validated in vivo and in vitro. RESULTS SZ-A demonstrated a significant inhibition in vascular inflammation and alleviation of AS severity by mitigating macrophage infiltration and modulating M1/M2 macrophage polarization in vitro and in vivo. Moreover, SZ-A effectively reduced the release of the proinflammatory mediator C-X-C motif chemokine ligand (CXCL)-10, predominantly secreted by M1 macrophages. This reduction in CXCL-10 contributed to improved endothelial cell function, reduced recruitment of additional macrophages, and inhibited the inflammatory amplification effect. This ultimately led to the suppression of atherogenesis. CONCLUSION SZ-A exhibited potent anti-inflammatory effects by inhibiting macrophage-mediated inflammation, providing a new therapeutic avenue against AS. This is the first study demonstrating the efficacy of SZ-A in alleviating AS severity and offers novel insights into its anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Dandan Peng
- Department of Endocrinology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Guizhou Medical University, Guiyang, Guizhou, China
| | - Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Mingwei Wang
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Binbin Zhang
- Department of Infectious Diseases and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhenjie Zhuang
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Run Zhou
- College of Nursing, Hangzhou Normal University, Zhejiang, China
| | - Yuanyuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Jie Li
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Zhenqiu Yu
- Guizhou Medical University, Guiyang, Guizhou, China; The Department of Hypertension, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.
| | - Junping Shi
- Department of Infectious Diseases and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Medical Epigenetics, Hangzhou, Zhejiang, China; Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Lozano-Vicario L, Muñoz-Vázquez ÁJ, Ramírez-Vélez R, Galbete-Jiménez A, Fernández-Irigoyen J, Santamaría E, Cedeno-Veloz BA, Zambom-Ferraresi F, Van Munster BC, Ortiz-Gómez JR, Hidalgo-Ovejero ÁM, Romero-Ortuno R, Izquierdo M, Martínez-Velilla N. Association of postoperative delirium with serum and cerebrospinal fluid proteomic profiles: a prospective cohort study in older hip fracture patients. GeroScience 2024; 46:3235-3247. [PMID: 38236313 PMCID: PMC11009174 DOI: 10.1007/s11357-024-01071-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024] Open
Abstract
Postoperative delirium (POD) is a common neuropsychiatric complication in geriatric inpatients after hip fracture surgery and its occurrence is associated with poor outcomes. The purpose of this study was to investigate the relationship between preoperative biomarkers in serum and cerebrospinal fluid (CSF) and the development of POD in older hip fracture patients, exploring the possibility of integrating objective methods into future predictive models of delirium. Sixty hip fracture patients were recruited. Blood and CSF samples were collected at the time of spinal anesthesia when none of the subjects had delirium. Patients were assessed daily using the 4AT scale, and based on these results, they were divided into POD and non-POD groups. The Olink® platform was used to analyze 45 cytokines. Twenty-one patients (35%) developed POD. In the subsample of 30 patients on whom proteomic analyses were performed, a proteomic profile was associated with the incidence of POD. Chemokine (C-X-C motif) ligand 9 (CXCL9) had the strongest correlation between serum and CSF samples in patients with POD (rho = 0.663; p < 0.05). Although several cytokines in serum and CSF were associated with POD after hip fracture surgery in older adults, there was a significant association with lower preoperative levels of CXCL9 in CSF and serum. Despite the small sample size, this study provides preliminary evidence of the potential role of molecular biomarkers in POD, which may provide a basis for the development of new delirium predictive models.
Collapse
Affiliation(s)
- Lucía Lozano-Vicario
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Pamplona, Spain.
| | | | - Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Arkaitz Galbete-Jiménez
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Enrique Santamaría
- Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | | | - Fabricio Zambom-Ferraresi
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara C Van Munster
- Department of Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - José Ramón Ortiz-Gómez
- Department of Anesthesiology and Reanimation, Hospital Universitario de Navarra (HUN), Pamplona, Spain
| | | | - Román Romero-Ortuno
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Nicolás Martínez-Velilla
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Pamplona, Spain
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Alserawan L, Mulet M, Anguera G, Riudavets M, Zamora C, Osuna-Gómez R, Serra-López J, Barba Joaquín A, Sullivan I, Majem M, Vidal S. Kinetics of IFNγ-Induced Cytokines and Development of Immune-Related Adverse Events in Patients Receiving PD-(L)1 Inhibitors. Cancers (Basel) 2024; 16:1759. [PMID: 38730712 PMCID: PMC11083441 DOI: 10.3390/cancers16091759] [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: 02/29/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have the potential to trigger unpredictable immune-related adverse events (irAEs), which can be severe. The underlying mechanisms of these events are not fully understood. As PD-L1 is upregulated by IFN, the heightened immune activation resulting from PD-1/PD-L1 inhibition may enhance the IFN response, triggering the expression of IFN-inducible genes and contributing to irAE development and its severity. In this study, we investigated the interplay between irAEs and the expression of IFN-inducible chemokines and cytokines in 134 consecutive patients with solid tumours treated with PD-(L)1 inhibitors as monotherapy or in combination with chemotherapy or other immunotherapy agents. We compared the plasma levels of IFN-associated cytokines (CXCL9/10/11, IL-18, IL-10, IL-6 and TGFβ) at various time points (at baseline, at the onset of irAE and previous to irAE onset) in three patient groups categorized by irAE development and severity: patients with serious irAEs, mild irAEs and without irAEs after PD-(L)1 inhibitors. No differences were observed between groups at baseline. However, patients with serious irAEs exhibited significant increases in CXCL9/10/11, IL-18 and IL-10 levels at the onset of the irAE compared to baseline. A network analysis and correlation patterns highlighted a robust relationship among these chemokines and cytokines at serious-irAE onset. Combining all of the analysed proteins in a cluster analysis, we identified a subgroup of patients with a higher incidence of serious irAEs affecting different organs or systems. Finally, an ROC analysis and a decision tree model proposed IL-18 levels ≥ 807 pg/mL and TGFβ levels ≤ 114 pg/mL as predictors for serious irAEs in 90% of cases. In conclusion, our study elucidates the dynamic changes in cytokine profiles associated with serious irAE development during treatment with PD-(L)1 inhibitors. The study's findings offer valuable insights into the intricate IFN-induced immune responses associated with irAEs and propose potential predictive markers for their severity.
Collapse
Affiliation(s)
- Leticia Alserawan
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
- Department of Immunology, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Maria Mulet
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| | - Geòrgia Anguera
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Mariona Riudavets
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
- Department of Pneumologie, Hôpital Cochin—APHP Centre, 75014 Paris, France
| | - Carlos Zamora
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| | - Rubén Osuna-Gómez
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| | - Jorgina Serra-López
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Andrés Barba Joaquín
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Ivana Sullivan
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Silvia Vidal
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| |
Collapse
|
5
|
Yang J, Xu S, Cheng J, Yin X, Yan D, Li X. CXCL10 and its receptor in patients with chronic hepatitis B and their ability to predict HBeAg seroconversion during antiviral treatment with TDF. J Med Virol 2024; 96:e29516. [PMID: 38469895 DOI: 10.1002/jmv.29516] [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: 08/19/2023] [Revised: 12/19/2023] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
The serum chemokine C-X-C motif ligand-10 (CXCL10) and its unique receptor (CXCR3) may predict the prognosis of patients with chronic hepatitis B (CHB) treated with tenofovir disoproxil fumarate (TDF). Nevertheless, there are few reports on the profile of CXCL10 and CXCR3 and their clinical application in HBeAg (+) CHB patients during TDF antiviral therapy. CXCL10 and CXCR3 were determined in 118 CHB patients naively treated with TDF for at least 96 weeks at baseline and at treatment weeks 12 and 24. In addition, gene set enrichment analysis was used to examine the associated dataset from Gene Expression Omnibus and explore the gene sets associated with HBeAg seroconversion (SC). The change of CXCL10 (ΔCXCL10, baseline to 48-week TDF treatment) and CXCR3 (ΔCXCR3) is closely related to the possibility of HBeAg SC of CHB patients under TDF treatment. Immunohistochemical analysis of CXCL10/CXCR3 protein in liver tissue shows that there is a significant difference between paired liver biopsy samples taken before and after 96 weeks of successful TDF treatment of CHB patients (11 pairs) but no significance for unsuccessful TDF treatment (14 pairs). Multivariate Cox analysis suggests that the ΔCXCL10 is an independent predictive indicator of HBeAg SC, and the area under the receiver operating characteristic curve of the ΔCXCL10 in CHB patients is 0.8867 (p < 0.0001). Our results suggest that a lower descending CXCL10 level is associated with an increased probability of HBeAg SC of CHB patients during TDF therapy. Moreover, liver tissue CXCL10 might be involved in the immunological process of HBeAg SC.
Collapse
Affiliation(s)
- Jiezuan Yang
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Shaoyan Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinlin Cheng
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Xuying Yin
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Dong Yan
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Xuefen Li
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Drews SJ. Commentary on "Inhibition of cellular activation induced by platelet factor 4 via the CXCR3 pathway ameliorates Japanese encephalitis and dengue viral infections". J Thromb Haemost 2024; 22:609-612. [PMID: 38417982 DOI: 10.1016/j.jtha.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 03/01/2024]
Affiliation(s)
- Steven J Drews
- Canadian Blood Services, Microbiology Department, Donor Policy and Studies, Edmonton, Alberta, Canada; Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
van Puijfelik F, Blok KM, Klein Kranenbarg RAM, Rip J, de Beukelaar J, Wierenga-Wolf AF, Wokke B, van Luijn MM, Smolders J. Ocrelizumab associates with reduced cerebrospinal fluid B and CD20 dim CD4 + T cells in primary progressive multiple sclerosis. Brain Commun 2024; 6:fcae021. [PMID: 38385000 PMCID: PMC10881107 DOI: 10.1093/braincomms/fcae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/01/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
The anti-CD20 monoclonal antibody ocrelizumab reduces disability progression in primary progressive multiple sclerosis. CD20 is a prototypical B-cell marker; however, subpopulations of CD4+ and CD8+ T cells in peripheral blood and cerebrospinal fluid also express low levels of CD20 (CD20dim). Therefore, direct targeting and depletion of these CD20dim T-cell subpopulations may contribute to the therapeutic effect of ocrelizumab. The aim of this observational cohort study was to compare CD20+ B-cell and CD20dim T-cell distributions between peripheral blood and cerebrospinal fluid of ocrelizumab-treated or ocrelizumab-untreated people with primary progressive multiple sclerosis. Ocrelizumab treatment was associated with depletion of circulating B cells and CD20dim CD4+ and CD20dim CD8+ T cells (P < 0.0001, P = 0.0016 and P = 0.0008, respectively) but, in cerebrospinal fluid, only with lower proportions of B cells and CD20dim memory CD4+ T cells (P < 0.0001 and P = 0.0043, respectively). The proportional prevalence of cerebrospinal fluid CD20dim memory CD8+ T cells was not significantly reduced (P = 0.1333). Only in cerebrospinal fluid, the proportions of CD20dim cells within CD4+ and not CD8+ T cells positive for CCR5, CCR6 and CXCR3 were reduced in ocrelizumab-treated participants. The proportion of CD20dim CD4+ T cells and abundance of CD4+ relative to CD8+ T cells in cerebrospinal fluid correlated positively with age (R = 0.6799, P = 0.0150) and Age-Related Multiple Sclerosis Severity score (R = 0.8087, P = 0.0014), respectively. We conclude that, in contrast to cerebrospinal fluid CD20dim CD8+ T cells, B cells and CD20dim CD4+ T cells are reduced in cerebrospinal fluid of people with primary progressive multiple sclerosis with an ocrelizumab-associated depletion of circulating B cells and CD20dim T cells. Therefore, these cells are likely to contribute to the therapeutic effects of ocrelizumab in people with primary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Fabiënne van Puijfelik
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Katelijn M Blok
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
- Department of Neurology, Albert Schweitzer Hospital, 3318 AT, Dordrecht, The Netherlands
| | - Romy A M Klein Kranenbarg
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
- Department of Neurology, Albert Schweitzer Hospital, 3318 AT, Dordrecht, The Netherlands
| | - Jasper Rip
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Janet de Beukelaar
- Department of Neurology, Albert Schweitzer Hospital, 3318 AT, Dordrecht, The Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Beatrijs Wokke
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Jo G, Chae JB, Jung SA, Lyu J, Chung H, Lee JH. Sulfated CXCR3 Peptide Trap Use as a Promising Therapeutic Approach for Age-Related Macular Degeneration. Biomedicines 2024; 12:241. [PMID: 38275412 PMCID: PMC10813770 DOI: 10.3390/biomedicines12010241] [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: 12/07/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Chemokines have various biological functions and potential roles in the development or progression of neuroinflammatory diseases. However, the specific pathogenic roles of chemokines in the major cause for vision loss among the elderly, the leading cause of blindness in older individuals, remain elusive. Chemokines interact with their receptors expressed in the endothelium and on leukocytes. The sulfation of tyrosine residues in chemokine receptors increases the strength of ligand-receptor interaction and modulates signaling. Therefore, in the present study, we aimed to construct a human recombinant sulfated CXCR3 peptide trap (hCXCR3-S2) and mouse recombinant sulfated CXCR3 peptide trap (mCXCR3-S2) to demonstrate in vivo effects in preventing choroidal neovascularization (CNV) and chemotaxis. MATERIALS AND METHODS We generated expression vectors for mCXCR3-S2 and hCXCR3-S2 with GST domains and their respective cDNA sequences. Following overexpression in E. coli BL21 (DE3), we purified the fusion proteins from cell lysates using affinity chromatography. First, the impact of hCXCR3-S2 was validated in vitro. Subsequently, the in vivo efficacy of mCXCR3-S2 was investigated using a laser-induced CNV mouse model, a mouse model of neovascular age-related macular degeneration (AMD). RESULTS hCXCR3-S2 inhibited the migration and invasion of two human cancer cell lines. Intravitreal injection of mCXCR3-S2 attenuated CNV and macrophage recruitment in neovascular lesions of mouse models. These in vitro and in vivo effects were significantly stronger with CXCR3-S2 than with wild-type CXCR3 peptides. CONCLUSION These findings demonstrate that the sulfated form of the CXCR3 peptide trap is a valuable tool that could be supplemented with antivascular endothelial growth factors in AMD treatment.
Collapse
Affiliation(s)
- Gukheui Jo
- Myung-Gok Eye Research Institute, Kim’s Eye Hospital, Konyang University College of Medicine, Seoul 07301, Republic of Korea; (G.J.); (S.-A.J.)
| | - Jae-Byoung Chae
- Department of Ophthalmology, Konkuk University College of Medicine, Seoul 05029, Republic of Korea;
| | - Sun-Ah Jung
- Myung-Gok Eye Research Institute, Kim’s Eye Hospital, Konyang University College of Medicine, Seoul 07301, Republic of Korea; (G.J.); (S.-A.J.)
| | - Jungmook Lyu
- Department of Medical Science, Konyang University, Seo-gu, Daejeon 35365, Republic of Korea;
| | - Hyewon Chung
- Department of Ophthalmology, Konkuk University College of Medicine, Seoul 05029, Republic of Korea;
| | - Joon H. Lee
- Myung-Gok Eye Research Institute, Kim’s Eye Hospital, Konyang University College of Medicine, Seoul 07301, Republic of Korea; (G.J.); (S.-A.J.)
| |
Collapse
|
9
|
Livne-Bar I, Maurya S, Gronert K, Sivak JM. Lipoxins A 4 and B 4 inhibit glial cell activation via CXCR3 signaling in acute retinal neuroinflammation. J Neuroinflammation 2024; 21:18. [PMID: 38212822 PMCID: PMC10782675 DOI: 10.1186/s12974-024-03010-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/01/2024] [Indexed: 01/13/2024] Open
Abstract
Lipoxins are small lipids that are potent endogenous mediators of systemic inflammation resolution in a variety of diseases. We previously reported that Lipoxins A4 and B4 (LXA4 and LXB4) have protective activities against neurodegenerative injury. Yet, lipoxin activities and downstream signaling in neuroinflammatory processes are not well understood. Here, we utilized a model of posterior uveitis induced by lipopolysaccharide endotoxin (LPS), which results in rapid retinal neuroinflammation primarily characterized by activation of resident macroglia (astrocytes and Müller glia), and microglia. Using this model, we observed that each lipoxin reduces acute inner retinal inflammation by affecting endogenous glial responses in a cascading sequence beginning with astrocytes and then microglia, depending on the timing of exposure; prophylactic or therapeutic. Subsequent analyses of retinal cytokines and chemokines revealed inhibition of both CXCL9 (MIG) and CXCL10 (IP10) by each lipoxin, compared to controls, following LPS injection. CXCL9 and CXCL10 are common ligands for the CXCR3 chemokine receptor, which is prominently expressed in inner retinal astrocytes and ganglion cells. We found that CXCR3 inhibition reduces LPS-induced neuroinflammation, while CXCR3 agonism alone induces astrocyte reactivity. Together, these data uncover a novel lipoxin-CXCR3 pathway to promote distinct anti-inflammatory and proresolution cascades in endogenous retinal glia.
Collapse
Affiliation(s)
- Izhar Livne-Bar
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada
| | - Shubham Maurya
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Karsten Gronert
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
- Vision Science Program, University of California Berkeley, Berkeley, CA, USA
- Infectious Disease and Immunity Program, University of California Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada.
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto School of Medicine, Toronto, Canada.
| |
Collapse
|
10
|
Tang WHW. Targeting anti-CXCR3 autoantibodies as potential cardioprotective therapy: promises and challenges. Eur Heart J 2023; 44:4950-4952. [PMID: 37944029 DOI: 10.1093/eurheartj/ehad665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2023] Open
Affiliation(s)
- Wai Hong Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
| |
Collapse
|
11
|
Hochuli N, Kadyan S, Park G, Patoine C, Nagpal R. Pathways linking microbiota-gut-brain axis with neuroinflammatory mechanisms in Alzheimer's pathophysiology. MICROBIOME RESEARCH REPORTS 2023; 3:9. [PMID: 38455083 PMCID: PMC10917618 DOI: 10.20517/mrr.2023.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/03/2023] [Accepted: 11/30/2023] [Indexed: 03/09/2024]
Abstract
Disturbances in the local and peripheral immune systems are closely linked to a wide range of diseases. In the context of neurodegenerative disorders such as Alzheimer's disease (AD), inflammation plays a crucial role, often appearing as a common manifestation despite the variability in the occurrence of other pathophysiological hallmarks. Thus, combating neuroinflammation holds promise in treating complex pathophysiological diseases like AD. Growing evidence suggests the gut microbiome's crucial role in shaping the pathogenesis of AD by influencing inflammatory mediators. Gut dysbiosis can potentially activate neuroinflammatory pathways through bidirectional signaling of the gut-brain axis; however, the precise mechanisms of this complex interweaved network remain largely unclear. In these milieus, this review attempts to summarize the contributing role of gut microbiome-mediated neuroinflammatory signals in AD pathophysiology, while also pondering potential mechanisms through which commensal and pathogenic gut microbes affect neuroinflammation. While certain taxa such as Roseburia and Escherichia have been strongly correlated with AD, other clades such as Bacteroides and Faecalibacterium exhibit variations at the species and strain levels. In order to disentangle the inflammatory aspects of neurodegeneration attributed to the gut microbiome, it is imperative that future mechanistic studies investigate the species/strain-level dependency of commensals, opportunistic, and pathogenic gut microbes that consistently show correlations with AD patients across multiple associative studies.
Collapse
Affiliation(s)
| | | | | | | | - Ravinder Nagpal
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
12
|
Dragan P, Joshi K, Atzei A, Latek D. Keras/TensorFlow in Drug Design for Immunity Disorders. Int J Mol Sci 2023; 24:15009. [PMID: 37834457 PMCID: PMC10573944 DOI: 10.3390/ijms241915009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Homeostasis of the host immune system is regulated by white blood cells with a variety of cell surface receptors for cytokines. Chemotactic cytokines (chemokines) activate their receptors to evoke the chemotaxis of immune cells in homeostatic migrations or inflammatory conditions towards inflamed tissue or pathogens. Dysregulation of the immune system leading to disorders such as allergies, autoimmune diseases, or cancer requires efficient, fast-acting drugs to minimize the long-term effects of chronic inflammation. Here, we performed structure-based virtual screening (SBVS) assisted by the Keras/TensorFlow neural network (NN) to find novel compound scaffolds acting on three chemokine receptors: CCR2, CCR3, and one CXC receptor, CXCR3. Keras/TensorFlow NN was used here not as a typically used binary classifier but as an efficient multi-class classifier that can discard not only inactive compounds but also low- or medium-activity compounds. Several compounds proposed by SBVS and NN were tested in 100 ns all-atom molecular dynamics simulations to confirm their binding affinity. To improve the basic binding affinity of the compounds, new chemical modifications were proposed. The modified compounds were compared with known antagonists of these three chemokine receptors. Known CXCR3 compounds were among the top predicted compounds; thus, the benefits of using Keras/TensorFlow in drug discovery have been shown in addition to structure-based approaches. Furthermore, we showed that Keras/TensorFlow NN can accurately predict the receptor subtype selectivity of compounds, for which SBVS often fails. We cross-tested chemokine receptor datasets retrieved from ChEMBL and curated datasets for cannabinoid receptors. The NN model trained on the cannabinoid receptor datasets retrieved from ChEMBL was the most accurate in the receptor subtype selectivity prediction. Among NN models trained on the chemokine receptor datasets, the CXCR3 model showed the highest accuracy in differentiating the receptor subtype for a given compound dataset.
Collapse
Affiliation(s)
- Paulina Dragan
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
| | - Kavita Joshi
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
| | - Alessandro Atzei
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
- Department of Life and Environmental Science, Food Toxicology Unit, University of Cagliari, University Campus of Monserrato, SS 554, 09042 Cagliari, Italy
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
| |
Collapse
|
13
|
Ding W, Xu D, Li F, Huang C, Song T, Zhong N, Lai K, Deng Z. Intrapulmonary IFN-γ instillation causes chronic lymphocytic inflammation in the spleen and lung through the CXCR3 pathway. Int Immunopharmacol 2023; 122:110675. [PMID: 37481849 DOI: 10.1016/j.intimp.2023.110675] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
Some patients with chronic refractory cough have high levels of pulmonary IFN-γ and IFN-γ-producing T lymphocytes. Pulmonary IFN-γ administration causes acute airway lymphocytic inflammation and cough hypersensitivity by increasing the number of pulmonary IFN-γ-producing T lymphocytes, but these lymphocytes may be recruited from other organs. Intraperitoneal IFN-γ injection can increase the spleen weight of mice. It remains elusive whether pulmonary IFN-γ can induce chronic airway lymphocytic inflammation and cough hypersensitivity by stimulating the proliferation of IFN-γ -producing T lymphocytes in the spleen. Here, we found that pulmonary IFN-γ administration induced chronic airway inflammation and chronic cough hypersensitivity with an increased number of IFN-γ-producing T lymphocytes in the spleen, blood and lung. Pulmonary IFN-γ administration also increased 1) the proliferation of spleen lymphocytes in vivo and 2) the IP-10 level and CXCR3+ T lymphocyte numbers in the spleen and lung of mice. IP-10 could promote the proliferation of spleen lymphocytes in vitro but not blood lymphocytes or lung-resident lymphocytes. AMG487, a potent inhibitor of binding between IP-10 and CXCR3, could block pulmonary IFN-γ instillation-induced chronic airway lymphocytic inflammation and the proliferation of IFN-γ-producing T lymphocytes in mouse spleens. In conclusion, intrapulmonary IFN-γ instillation may induce the proliferation of splenic IFN-γ-producing T lymphocytes through IP-10 and the CXCR3 pathway. The IFN-γ-producing T lymphocytes in blood, partly released from the mouse spleen, may be partly attracted to the lung by pulmonary IP-10 through the CXCR3 pathway. IFN-γ-producing T lymphocytes and IFN-γ in the lung may cause chronic airway lymphocytic inflammation and chronic cough hypersensitivity.
Collapse
Affiliation(s)
- Wenbin Ding
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongting Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fengying Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chuqin Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tongtong Song
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kefang Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zheng Deng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
14
|
Hanin A, Cespedes J, Pulluru Y, Gopaul M, Aronica E, Decampo D, Helbig I, Howe CL, Huttner A, Koh S, Navarro V, Taraschenko O, Vezzani A, Wilson MR, Xian J, Gaspard N, Hirsch LJ. Review and standard operating procedures for collection of biospecimens and analysis of biomarkers in new onset refractory status epilepticus. Epilepsia 2023; 64:1444-1457. [PMID: 37039049 PMCID: PMC10756682 DOI: 10.1111/epi.17600] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/12/2023]
Abstract
New onset refractory status epilepticus (NORSE), including its subtype with a preceding febrile illness known as febrile infection-related epilepsy syndrome (FIRES), is one of the most severe forms of status epilepticus. The exact causes of NORSE are currently unknown, and there is so far no disease-specific therapy. Identifying the underlying pathophysiology and discovering specific biomarkers, whether immunologic, infectious, genetic, or other, may help physicians in the management of patients with NORSE. A broad spectrum of biomarkers has been proposed for status epilepticus patients, some of which were evaluated for patients with NORSE. Nonetheless, none has been validated, due to significant variabilities in study cohorts, collected biospecimens, applied analytical methods, and defined outcome endpoints, and to small sample sizes. The NORSE Institute established an open NORSE/FIRES biorepository for health-related data and biological samples allowing the collection of biospecimens worldwide, promoting multicenter research and sharing of data and specimens. Here, we suggest standard operating procedures for biospecimen collection and biobanking in this rare condition. We also propose criteria for the appropriate use of previously collected biospecimens. We predict that the widespread use of standardized procedures will reduce heterogeneity, facilitate the future identification of validated biomarkers for NORSE, and provide a better understanding of the pathophysiology and best clinical management for these patients.
Collapse
Affiliation(s)
- Aurélie Hanin
- Department of Neurology and Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Sorbonne Université, Institut du Cerveau ICM, Paris Brain Institute, Inserm, CNRS, Assistance Publique -Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, DMU Neurosciences 6, Paris, France
- Assistance Publique -Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, DMU Neurosciences 6, Epilepsy Unit and Department of Clinical Neurophysiology, Paris, France
| | - Jorge Cespedes
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Universidad Autonoma de Centro America, School of Medicine, San Jose, Costa Rica
| | - Yashwanth Pulluru
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Nebraska Medical Center, Omaha, Nebraska, USA
| | - Margaret Gopaul
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Danielle Decampo
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Epilepsy NeuroGenetics Initiative, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ingo Helbig
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Epilepsy NeuroGenetics Initiative, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles L. Howe
- Division of Experimental Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sookyong Koh
- Department of Pediatrics, Children’s Hospital Medical Center, University of Nebraska, Omaha, Nebraska, USA
| | - Vincent Navarro
- Sorbonne Université, Institut du Cerveau ICM, Paris Brain Institute, Inserm, CNRS, Assistance Publique -Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, DMU Neurosciences 6, Paris, France
- Assistance Publique -Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, DMU Neurosciences 6, Epilepsy Unit and Department of Clinical Neurophysiology, Paris, France
| | - Olga Taraschenko
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Annamaria Vezzani
- Department of Acute Brain Injury, Istituto di Recerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michael R. Wilson
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, California, San Francisco, USA
| | - Julie Xian
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Epilepsy NeuroGenetics Initiative, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicolas Gaspard
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Université Libre de Bruxelles, Hôpital Erasme, Brussels, Belgium
| | - Lawrence J. Hirsch
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Zhang J, Zhang X, Shi X, Liu Y, Cheng D, Tian Q, Lin N, Wei W, Wu H. CXCL9, 10, 11/CXCR3 Axis Contributes to the Progress of Primary Sjogren's Syndrome by Activating GRK2 to Promote T Lymphocyte Migration. Inflammation 2023; 46:1047-1060. [PMID: 36801996 DOI: 10.1007/s10753-023-01791-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/21/2023]
Abstract
Primary Sjogren's syndrome (pSS) is a systemic autoimmune disease that causes dysfunction of secretory glands and the specific pathogenesis is still unknown. The CXCL9, 10, 11/CXCR3 axis and G protein-coupled receptor kinase 2 (GRK2) involved in many inflammation and immunity processes. We used NOD/Ltj mice, a spontaneous SS animal model, to elucidate the pathological mechanism of CXCL9, 10, 11/CXCR3 axis promoting T lymphocyte migration by activating GRK2 in pSS. We found that CD4 + GRK2, Th17 + CXCR3 was apparently increased and Treg + CXCR3 was significantly decreased in the spleen of 4W NOD mice without sicca symptom compared to ICR mice (control group). The protein levels of IFN-γ, CXCL9, 10, 11 increased in submandibular gland (SG) tissue accompanied by obvious lymphocytic infiltration and Th17 cells overwhelmingly infiltrated relative to Treg cells at the sicca symptom occurs, and we found that the proportion of Th17 cells was increased, whereas that of Treg cells was decreased in spleen. In vitro, we used IFN-γ to stimulate human salivary gland epithelial cells (HSGECs) co-cultured with Jurkat cells, and the results showed that CXCL9, 10, 11 was increased by IFN-γ activating JAK2/STAT1 signal pathway and Jurkat cell migration increased with the raised of cell membrane GRK2 expression. HSGECs with tofacitinib or Jurkat cells with GRK2 siRNA can reduce the migration of Jurkat cells. The results indicate that CXCL9, 10, 11 significantly increased in SG tissue through IFN-γ stimulating HSGECs, and the CXCL9, 10, 11/CXCR3 axis contributes to the progress of pSS by activating GRK2 to promote T lymphocyte migration.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China
| | - Xiao Zhang
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China
| | - Xingjie Shi
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, 12 Zhongyou Road, Chuzhou, 239001, China
| | - Yuqi Liu
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China
| | - Danqian Cheng
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China
| | - Qianwen Tian
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China
| | - Ning Lin
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, 12 Zhongyou Road, Chuzhou, 239001, China.
| | - Wei Wei
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China.
| | - Huaxun Wu
- Institute of Clinical PharmacologyKey Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune MedicineShushan District, Anhui Medical University, 81# Meishan Road, 230032Anhui Province, Hefei City, China. .,Anhui Provincial Institute of Translation Medicine, Hefei, 230032, China.
| |
Collapse
|