1
|
Muñoz-Callejas A, Sánchez-Abad I, Ramos-Manzano A, San Antonio E, González-Sánchez E, Silván J, González-Tajuelo R, González-Álvaro I, García-Pérez J, Tomero EG, García-Vicuña R, Vicente-Rabaneda EF, Castañeda S, Urzainqui A. Regulation of monocyte apoptosis and DNA extrusion in monocyte extracellular traps by PSGL-1: Relevance in systemic lupus erythematosus. Transl Res 2024; 274:10-20. [PMID: 39182668 DOI: 10.1016/j.trsl.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease characterized by severe organ damage and lacking curative treatment. While various immune cell types, especially dysfunctional B and T cells and neutrophils, have been related with disease pathogenesis, limited research has focused on the role of monocytes in SLE. Increased DNA extracellular traps, apoptosis and necrosis have been related to lupus pathogenesis. Our goal is to analyze the contribution of P-selectin glycoprotein ligand 1 (PSGL-1) in SLE monocytes to disease pathogenesis by investigating the control exerted by PSGL-1 on monocyte apoptosis and DNA extrusion in extracellular traps (METs). Monocytes from active disease patients (aSLE) exhibited reduced levels of PSGL-1. Importantly, lower PSGL-1 levels in SLE monocytes associated with several clinical characteristics, including anti-dsDNA autoantibodies, lupus anticoagulant, clinical lung involvement, and anemia. Monocytes from SLE patients showed higher susceptibility to apoptosis than healthy donors (HD) monocytes and PSGL-1/P-selectin interaction decreased secondary necrosis in HD but not in aSLE monocytes. Regarding METs, aSLE monocytes exhibited higher susceptibility to generate METs than HD monocytes. The interaction of HD monocytes with P-selectin induced Syk activation and reduced the levels of DNA extruded in METs. However, in aSLE monocytes, PSGL-1/P-selectin interaction did not activate Syk or reduce the amount of extruded DNA. Our data suggest a dysfunctional PSGL-1/P-selectin axis in aSLE monocytes, unable to reduce secondary necrosis or the amount of DNA released into the extracellular medium in METs, potentially contributing to lupus pathogenesis.
Collapse
Affiliation(s)
- Antonio Muñoz-Callejas
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain; Facultad de Medicina y Biomedicina, Universidad Alfonso X El Sabio, 28691, Madrid, Spain
| | - Inés Sánchez-Abad
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Alejandra Ramos-Manzano
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain; Facultad de Medicina, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Esther San Antonio
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Elena González-Sánchez
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Javier Silván
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Rafael González-Tajuelo
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Isidoro González-Álvaro
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Javier García-Pérez
- Pulmonology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Eva G Tomero
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Rosario García-Vicuña
- Facultad de Medicina, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Esther F Vicente-Rabaneda
- Facultad de Medicina, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain
| | - Santos Castañeda
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain; Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain.
| | - Ana Urzainqui
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006, Madrid, Spain.
| |
Collapse
|
2
|
San Antonio E, Silván J, Sevilla-Montero J, González-Sánchez E, Muñoz-Callejas A, Sánchez-Abad I, Ramos-Manzano A, Muñoz-Calleja C, González-Álvaro I, Tomero EG, García-Pérez J, García-Vicuña R, Vicente-Rabaneda EF, Castañeda S, Urzainqui A. PSGL-1, ADAM8, and selectins as potential biomarkers in the diagnostic process of systemic lupus erythematosus and systemic sclerosis: an observational study. Front Immunol 2024; 15:1403104. [PMID: 39100683 PMCID: PMC11297358 DOI: 10.3389/fimmu.2024.1403104] [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: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 08/06/2024] Open
Abstract
Background Early diagnosis and treatment of Systemic lupus erythematosus (SLE) and Systemic sclerosis (SSc) present significant challenges for clinicians. Although various studies have observed changes in serum levels of selectins between healthy donors and patients with autoimmune diseases, including SLE and SSc, their potential as biomarkers has not been thoroughly explored. We aimed to investigate serum profiles of PSGL-1 (sPSGL-1), ADAM8 (sADAM8) and P-, E- and L-selectins (sP-, sE- and sL-selectins) in defined SLE and SSc patient cohorts to identify disease-associated molecular patterns. Methods We collected blood samples from 64 SLE patients, 58 SSc patients, and 81 healthy donors (HD). Levels of sPSGL-1, sADAM8 and selectins were analyzed by ELISA and leukocyte membrane expression of L-selectin and ADAM8 by flow cytometry. Results Compared to HD, SLE and SSc patients exhibited elevated sE-selectin and reduced sL-selectin levels. Additionally, SLE patients exhibited elevated sPSGL-1 and sADAM8 levels. Compared to SSc, SLE patients had decreased sL-selectin and increased sADAM8 levels. Furthermore, L-selectin membrane expression was lower in SLE and SSc leukocytes than in HD leukocytes, and ADAM8 membrane expression was lower in SLE neutrophils compared to SSc neutrophils. These alterations associated with some clinical characteristics of each disease. Using logistic regression analysis, the sL-selectin/sADAM8 ratio in SLE, and a combination of sL-selectin/sE-selectin and sE-selectin/sPSGL-1 ratios in SSc were identified and cross-validated as potential serum markers to discriminate these patients from HD. Compared to available diagnostic biomarkers for each disease, both sL-selectin/sADAM8 ratio for SLE and combined ratios for SSc provided higher sensitivity (98% SLE and and 67% SSc correctly classified patients). Importantly, the sADAM8/% ADAM8(+) neutrophils ratio discriminated between SSc and SLE patients with the same sensitivity and specificity than current disease-specific biomarkers. Conclusion SLE and SSc present specific profiles of sPSGL-1, sE-, sL-selectins, sADAM8 and neutrophil membrane expression which are potentially relevant to their pathogenesis and might aid in their early diagnosis.
Collapse
Affiliation(s)
- Esther San Antonio
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Javier Silván
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Javier Sevilla-Montero
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Elena González-Sánchez
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Antonio Muñoz-Callejas
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Inés Sánchez-Abad
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Alejandra Ramos-Manzano
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
| | - Isidoro González-Álvaro
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Eva G. Tomero
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Javier García-Pérez
- Pulmonology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Rosario García-Vicuña
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Esther F. Vicente-Rabaneda
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Santos Castañeda
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| |
Collapse
|
3
|
Pereira JL, Ferreira F, Dos Santos NR. Antibody targeting of surface P-selectin glycoprotein ligand 1 leads to lymphoma apoptosis and tumorigenesis inhibition. Hematol Oncol 2024; 42:e3257. [PMID: 38415859 DOI: 10.1002/hon.3257] [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: 10/09/2023] [Revised: 01/11/2024] [Accepted: 02/03/2024] [Indexed: 02/29/2024]
Abstract
Lymphomas are a heterogeneous group of diseases that originate from T, B or natural killer cells. Lymphoma treatment is based on chemotherapy, radiotherapy, and monoclonal antibody (mAb) or other immunotherapies. The P-selectin glycoprotein ligand 1 (PSGL-1) is expressed at the surface of hematological malignant cells and has been shown to have a pro-oncogenic role in multiple myeloma and lymphoma. Here, we investigated the expression and therapeutic potential of PSGL-1 in T and B cell lymphomas. By flow cytometry analysis, we found that PSGL-1 was expressed in both T and B cell-derived lymphoma cell lines but generally at higher levels in T cell lymphoma cell lines. For most T and B cell-derived lymphoma cell lines, in vitro targeting with the PL1 mAb, which recognizes the PSGL-1 N-terminal extracellular region and blocks functional interactions with selectins, resulted in reduced cell viability. The PL1 mAb pro-apoptotic activity was shown to be dose-dependent, to be linked to increased ERK kinase phosphorylation, and to be dependent on the MAP kinase signaling pathway. Importantly, anti-PSGL-1 treatment of mice xenografted with the HUT-78 cutaneous T-cell lymphoma cell line resulted in decreased tumor growth, had no effect on in vivo proliferation, but increased the levels of apoptosis in tumors. Anti-PSGL-1 treatment of mice xenografted with a Burkitt lymphoma cell line that was resistant to anti-PSGL-1 treatment in vitro, had no impact on tumorigenesis. These findings show that PSGL-1 antibody targeting triggers lymphoma cell apoptosis and substantiates PSGL-1 as a potential target for lymphoma therapy.
Collapse
Affiliation(s)
- João L Pereira
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- FMUP-Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Francisca Ferreira
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Master´s degree in Bioengineering, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar and Faculty of Engineering, University of Porto, Porto, Portugal
| | - Nuno R Dos Santos
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| |
Collapse
|
4
|
Kauffman K, Manfra D, Nowakowska D, Zafari M, Nguyen PA, Phennicie R, Vollmann EH, O'Nuallain B, Basinski S, Komoroski V, Rooney K, Culyba EK, Wahle J, Ries C, Brehm M, Sazinsky S, Feldman I, Novobrantseva TI. PSGL-1 Blockade Induces Classical Activation of Human Tumor-associated Macrophages. CANCER RESEARCH COMMUNICATIONS 2023; 3:2182-2194. [PMID: 37819238 PMCID: PMC10601817 DOI: 10.1158/2767-9764.crc-22-0513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/22/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
The immune suppressive microenvironment is a major culprit for difficult-to-treat solid cancers. Particularly, inhibitory tumor-associated macrophages (TAM) define the resistant nature of the tumor milieu. To define tumor-enabling mechanisms of TAMs, we analyzed molecular clinical datasets correlating cell surface receptors with the TAM infiltrate. Though P-selectin glycoprotein ligand-1 (PSGL-1) is found on other immune cells and functions as an adhesion molecule, PSGL-1 is highly expressed on TAMs across multiple tumor types. siRNA-mediated knockdown and antibody-mediated inhibition revealed a role for PSGL-1 in maintaining an immune suppressed macrophage state. PSGL-1 knockdown or inhibition enhanced proinflammatory mediator release across assays and donors in vitro. In several syngeneic mouse models, PSGL-1 blockade alone and in combination with PD-1 blockade reduced tumor growth. Using a humanized tumor model, we observed the proinflammatory TAM switch following treatment with an anti-PSGL-1 antibody. In ex vivo patient-derived tumor cultures, a PSGL-1 blocking antibody increased expression of macrophage-derived proinflammatory cytokines, as well as IFNγ, indicative of T-cell activation. Our data demonstrate that PSGL-1 blockade reprograms TAMs, offering a new therapeutic avenue to patients not responding to T-cell immunotherapies, as well as patients with tumors devoid of T cells. SIGNIFICANCE This work is a significant and actionable advance, as it offers a novel approach to treating patients with cancer who do not respond to T-cell checkpoint inhibitors, as well as to patients with tumors lacking T-cell infiltration. We expect that this mechanism will be applicable in multiple indications characterized by infiltration of TAMs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kate Rooney
- Verseau Therapeutics, Auburndale, Massachusetts
| | | | | | - Carola Ries
- Dr. Carola Ries Consulting, Penzberg, Germany
| | - Michael Brehm
- University of Massachusetts Medical School, Worcester, Massachusetts
| | | | - Igor Feldman
- Verseau Therapeutics, Auburndale, Massachusetts
- Currently employed by Moderna Therapeutics, Cambridge, Massachusetts
| | - Tatiana I. Novobrantseva
- Verseau Therapeutics, Auburndale, Massachusetts
- Currently employed by Moderna Therapeutics, Cambridge, Massachusetts
| |
Collapse
|
5
|
Díaz-García E, García-Sánchez A, Alfaro E, López-Fernández C, Mañas E, Cano-Pumarega I, López-Collazo E, García-Río F, Cubillos-Zapata C. PSGL-1: a novel immune checkpoint driving T-cell dysfunction in obstructive sleep apnea. Front Immunol 2023; 14:1277551. [PMID: 37854605 PMCID: PMC10579800 DOI: 10.3389/fimmu.2023.1277551] [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: 08/14/2023] [Accepted: 09/12/2023] [Indexed: 10/20/2023] Open
Abstract
Introduction Although higher incidence of cancer represents a major burden for obstructive sleep apnea (OSA) patients, the molecular pathways driving this association are not completely understood. Recently, the adhesion receptor P-selectin glycoprotein-1 (PSGL 1) has been identified as a novel immune checkpoint, which are recognized major hallmarks in several types of cancer and have revolutionized cancer therapy. Methods The expression of PSGL-1 and its ligands VISTA and SIGLEC-5 was assessed in the leucocytes of OSA patients and control subjects exploring the role of intermittent hypoxia (IH) using in vitro models. In addition, PSGL-1 impact on T-cells function was evaluated by ex vivo models. Results Data showed PSGL-1 expression is upregulated in the T-lymphocytes from patients with severe OSA, indicating a relevant role of hypoxemia mediated by intermittent hypoxia. Besides, results suggest an inhibitory role of PSGL-1 on T-cell proliferation capacity. Finally, the expression of SIGLEC-5 but not VISTA was increased in monocytes from OSA patients, suggesting a regulatory role of intermittent hypoxia. Discussion In conclusion, PSGL-1 might constitute an additional immune checkpoint leading to T-cell dysfunction in OSA patients, contributing to the disruption of immune surveillance, which might provide biological plausibility to the higher incidence and aggressiveness of several tumors in these patients.
Collapse
Affiliation(s)
- Elena Díaz-García
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, Hospital La Paz Institute for Health Research – IdiPAZ, Madrid, Spain
| | - Aldara García-Sánchez
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Enrique Alfaro
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, Hospital La Paz Institute for Health Research – IdiPAZ, Madrid, Spain
| | - Cristina López-Fernández
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, Hospital La Paz Institute for Health Research – IdiPAZ, Madrid, Spain
| | - Eva Mañas
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Eduardo López-Collazo
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- The Innate Immune Response Group, Hospital La Paz Institute for Health Research – IdiPAZ, Madrid, Spain
| | - Francisco García-Río
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, Hospital La Paz Institute for Health Research – IdiPAZ, Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | - Carolina Cubillos-Zapata
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, Hospital La Paz Institute for Health Research – IdiPAZ, Madrid, Spain
| |
Collapse
|
6
|
Muñoz-Callejas A, González-Sánchez E, Silván J, San Antonio E, González-Tajuelo R, Ramos-Manzano A, Sánchez-Abad I, González-Alvaro I, García-Pérez J, Tomero EG, de Vicuña RG, Vicente-Rabaneda EF, Castañeda S, Urzainqui A. Low P-Selectin Glycoprotein Ligand-1 Expression in Neutrophils Associates with Disease Activity and Deregulated NET Formation in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:ijms24076144. [PMID: 37047117 PMCID: PMC10093849 DOI: 10.3390/ijms24076144] [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: 01/19/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease characterized by the generation of anti-DNA autoantibodies due to exposure of immune cells to excessive amounts of extracellular DNA. Lack of P-selectin in mice induces the development of a lupus-like syndrome and patients with cutaneous lupus have reduced P-selectin expression in skin vessels. Using flow cytometry we analyzed in healthy donors and patients the expression of P-selectin Glycoprotein Ligand-1 (PSGL-1) in circulating neutrophils and the implication of PSGL-1/P-selectin interaction in neutrophil extracellular traps (NETs) generation. We found a statistical significance that neutrophils from active SLE patients have a reduced expression of PSGL-1 and low levels of PSGL-1 in neutrophils from SLE patients associated with the presence of anti-dsDNA antibodies, clinical lung involvement, Raynaud's phenomenon, and positive lupus anticoagulant. PSGL-1 is present along the DNA in the NET. In healthy donors, neutrophil interaction with immobilized P-selectin triggers Syk activation, increases the NETs percentage and reduces the amount of DNA extruded in the NETs. In active SLE patients, neutrophil interaction with P-selectin does not activate Syk or reduce the amount of DNA extruded in the NETs, that might contribute to increase the extracellular level of DNA and hence, to disease pathogenesis.
Collapse
Affiliation(s)
- Antonio Muñoz-Callejas
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Elena González-Sánchez
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Javier Silván
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Esther San Antonio
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Rafael González-Tajuelo
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Alejandra Ramos-Manzano
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Inés Sánchez-Abad
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Isidoro González-Alvaro
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Javier García-Pérez
- Pulmonology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Eva G Tomero
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Rosario García de Vicuña
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Esther F Vicente-Rabaneda
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Santos Castañeda
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
- Catedra UAM-Roche, EPID-Future, Department of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| |
Collapse
|
7
|
Klaus T, Wilson AS, Vicari E, Hadaschik E, Klein M, Helbich SSC, Kamenjarin N, Hodapp K, Schunke J, Haist M, Butsch F, Probst HC, Enk AH, Mahnke K, Waisman A, Bednarczyk M, Bros M, Bopp T, Grabbe S. Impaired Treg-DC interactions contribute to autoimmunity in leukocyte adhesion deficiency type 1. JCI Insight 2022; 7:162580. [PMID: 36346673 PMCID: PMC9869970 DOI: 10.1172/jci.insight.162580] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Leukocyte adhesion deficiency type 1 (LAD-1) is a rare disease resulting from mutations in the gene encoding for the common β-chain of the β2-integrin family (CD18). The most prominent clinical symptoms are profound leukocytosis and high susceptibility to infections. Patients with LAD-1 are prone to develop autoimmune diseases, but the molecular and cellular mechanisms that result in coexisting immunodeficiency and autoimmunity are still unresolved. CD4+FOXP3+ Treg are known for their essential role in preventing autoimmunity. To understand the role of Treg in LAD-1 development and manifestation of autoimmunity, we generated mice specifically lacking CD18 on Treg (CD18Foxp3), resulting in defective LFA-1 expression. Here, we demonstrate a crucial role of LFA-1 on Treg to maintain immune homeostasis by modifying T cell-DC interactions and CD4+ T cell activation. Treg-specific CD18 deletion did not impair Treg migration into extralymphatic organs, but it resulted in shorter interactions of Treg with DC. In vivo, CD18Foxp3 mice developed spontaneous hyperplasia in lymphatic organs and diffuse inflammation of the skin and in multiple internal organs. Thus, LFA-1 on Treg is required for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Tanja Klaus
- Department of Dermatology,,Research Center for Immunotherapy, and
| | - Alicia S. Wilson
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Elisabeth Vicari
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Eva Hadaschik
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany.,Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Matthias Klein
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | | | - Nadine Kamenjarin
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Katrin Hodapp
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Jenny Schunke
- Department of Dermatology,,Research Center for Immunotherapy, and
| | - Maximilian Haist
- Department of Dermatology,,Research Center for Immunotherapy, and
| | | | - Hans Christian Probst
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Alexander H. Enk
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Karsten Mahnke
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Ari Waisman
- Research Center for Immunotherapy, and,Institute for Molecular Medicine, University of Mainz Medical Center, Mainz, Germany
| | | | - Matthias Bros
- Department of Dermatology,,Research Center for Immunotherapy, and
| | - Tobias Bopp
- Research Center for Immunotherapy, and,Institute of Immunology, University of Mainz Medical Center, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology,,Research Center for Immunotherapy, and
| |
Collapse
|
8
|
The Role of T Cells in Systemic Sclerosis: An Update. IMMUNO 2022. [DOI: 10.3390/immuno2030034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic disease characterized by microvasculopathy, autoantibodies (autoAbs), and fibrosis. The pathogenesis of the disease is incompletely understood. Microvasculopathy and autoAbs appear very early in the disease process. AutoAbs, such as those directed against DNA topoisomerase I (Topo I), are disease specific and associated with disease manifestations, and indicate activation of the adaptive immune system. B cells are involved in fibrosis in SSc. T cells are also involved in disease pathogenesis. T cells show signs of antigen-induced activation; T cells of TH2 type are increased and produce profibrotic cytokines interleukin (IL)-4, IL-13, and IL-31; CD4+ cytotoxic T lymphocytes are increased in skin lesions, and cause fibrosis and endothelial cell apoptosis; circulating T follicular helper (TFH) cells are increased in SSc produce IL-21 and promote plasmablast antibody production. On the other hand, regulatory T cells are impaired in SSc. These findings provide strong circumstantial evidence for T cell implication in SSc pathogenesis and encourage new T cell-directed therapeutic strategies for the disease.
Collapse
|
9
|
Abid F, Rubab Z, Fatima S, Qureshi A, Azhar A, Jafri A. In-silico analysis of interacting pathways through KIM-1 protein interaction in diabetic nephropathy. BMC Nephrol 2022; 23:254. [PMID: 35843953 PMCID: PMC9290293 DOI: 10.1186/s12882-022-02876-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Background Human Kidney Injury Molecule-1, also known as HAVCR-1 (Hepatitis A virus cellular receptor 1), belongs to the cell-surface protein of immunoglobulin superfamily involved in the phagocytosis by acting as scavenger receptor epithelial cells. The study focused on pinpointing the mechanisms and genes that interact with KIM-1. Methods This in-silico study was done from March 2019 to December 2019. The Enrichment and protein-protein interaction (PPI) network carefully choose proteins. In addition, the diagramed gene data sets were accomplished using FunRich version 3.1.3. It was done to unveil the proteins that may affect the regulation of HAVCR1 or may be regulated by this protein. These genes were then further considered in pathway analysis to discover the dysregulated pathways in diabetic nephropathy. The long list of differentially expressed genes is meaningless without pathway analysis. Results Critical pathways that are dysregulated in diabetic nephropathy patients have been identified. These include Immune System (Total = 237, P < 0.05), Innate Immune System (Total = 140, P < 0.05), Cytokine Signaling Immune system (Total = 116, P < 0.05), Adaptive Immune System (Total = 85) and Neutrophil degranulation (Total = 78). Conclusion The top 5 genes that are interacting directly with HIVCR1 include CASP3, CCL2, SPP1, B2M, and TIMP1 with degrees 161, 144, 108, 107, and 105 respectively for Immune system pathways (Innate Immune System, Cytokine Signaling Immune system, Adaptive Immune System and Neutrophil degranulation).
Collapse
Affiliation(s)
- F Abid
- Department Physiology, Jinnah Sindh Medical University, Karachi, Pakistan.
| | - Z Rubab
- Ziauddin Medical College-Ziauddin University, Karachi, Pakistan
| | - S Fatima
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - A Qureshi
- Department Physiology, Jinnah Sindh Medical University, Karachi, Pakistan
| | - A Azhar
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - A Jafri
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
10
|
González-Sánchez E, Muñoz-Callejas A, Gómez-Román J, San Antonio E, Marengo A, Tsapis N, Bohne-Japiassu K, González-Tajuelo R, Pereda S, García-Pérez J, Cavagna L, González-Gay MÁ, Vicente-Rabaneda E, Meloni F, Fattal E, Castañeda S, Urzainqui A. Everolimus targeted nanotherapy reduces inflammation and fibrosis in scleroderma-related interstitial lung disease (SSc-ILD) developed by PSGL-1 deficient mice. Br J Pharmacol 2022; 179:4534-4548. [PMID: 35726496 DOI: 10.1111/bph.15898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/21/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Interstitial lung disease (ILD) is the main cause of mortality in systemic sclerosis (SSc) and current therapies available are of low efficacy or high toxicity. Thus, the identification of innovative less toxic and high efficacy therapeutic approaches to ILD treatment is a crucial point. P-selectin Glycoprotein Ligand-1 (PSGL-1) interaction with P-selectin initiates leukocyte extravasation and the lack of its expression brings to SSc-like syndrome with high incidence of ILD in aged mice. EXPERIMENTAL APPROACH Aged PSGL-1-/- mice were used to assay the therapeutic efficacy of an innovative nanotherapy with everolimus (Ev), included in liposomes decorated with high MW hyaluronic acid (LipHA+Ev) and administrated intratracheally to specifically target CD44-expressing lung cells. KEY RESULTS PSGL-1-/- mice had increased number of CD45+ and CD45- cells, including alveolar and interstitial macrophages, eosinophils, granulocytes and NK cells, and elevated number of myofibroblasts in broncoalveolar lavage (BAL). CD45+ and CD45- cells expressing proinflammatory and profibrotic cytokines were also increased. PSGL-1-/- mice lung histopathology showed increased immune cell infiltration and apoptosis and exacerbated interstitial and peribronchial fibrosis. Targeted nanotherapy with LipHA+Ev reduced BAL number of myofibroblast, cells producing proinflammatory and profibrotic cytokines, and the degree of lung inflammation at histology. LipHA+Ev treatment also provided an important decrease in severity of peribronchial and interstitial lung fibrosis from moderate to mild injury score. CONCLUSIONS AND IMPLICATIONS Our preclinical study in PSGL-1-/- mice indicates that targeted nanotherapy with LipHA+Ev represents an effective treatment for SSc-ILD, reducing the number of inflammatory and fibrotic cells in BAL and reducing inflammation and fibrosis in lungs.
Collapse
Affiliation(s)
- Elena González-Sánchez
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Antonio Muñoz-Callejas
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Javier Gómez-Román
- Pathology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Esther San Antonio
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Alessandro Marengo
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Nicolas Tsapis
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Kamila Bohne-Japiassu
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Rafael González-Tajuelo
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Saray Pereda
- Pathology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Javier García-Pérez
- Pneumology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital Universitario de la Princesa, Madrid, Spain
| | - Lorenzo Cavagna
- Rheumatology Department, University and IRCCS Policlinico S. Matteo Foundation, Università degli Studi di Pavia, Pavia, Italy
| | - Miguel Ángel González-Gay
- Rheumatology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Esther Vicente-Rabaneda
- Rheumatology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| | - Federica Meloni
- Internal Medicine Department, Pneumology Division, IRCCS San Matteo Foundation and Università degli Studi di Pavia, Pavia, Italy
| | - Elias Fattal
- Institut Galien Paris Sud, UMR CNRS 8612. School of Pharmacy at University Paris-Saclay, Châtenay-Malabry, France
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain.,Cathedra UAM-Roche, EPID-Future, Department of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Hospital Universitario de la Princesa, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Madrid, Spain
| |
Collapse
|
11
|
DeRogatis JM, Viramontes KM, Neubert EN, Henriquez ML, Guerrero-Juarez CF, Tinoco R. Targeting the PSGL-1 Immune Checkpoint Promotes Immunity to PD-1 Resistant Melanoma. Cancer Immunol Res 2022; 10:612-625. [PMID: 35303066 DOI: 10.1158/2326-6066.cir-21-0690] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/10/2021] [Accepted: 03/15/2022] [Indexed: 11/16/2022]
Abstract
Immune checkpoint inhibitors have had impressive efficacy in some cancer patients, reinvigorating long-term durable immune responses against tumors. Despite the clinical success of these therapies, most cancer patients continue to be unresponsive to these treatments, highlighting the need for novel therapeutic options. Although P-selectin glycoprotein ligand-1 (PSGL-1) has been shown to inhibit immune responses in a variety of disease models, previous work has yet to address whether PSGL-1 can be targeted therapeutically to promote antitumor immunity. Using an aggressive melanoma tumor model, we targeted PSGL-1 in tumor-bearing mice and found increased effector CD4+ and CD8+ T-cell responses and decreased regulatory T cells (Tregs) in tumors. T cells exhibited increased effector function, activation, and proliferation, which delayed tumor growth in mice after anti-PSGL-1 treatment. Targeting PD-1 in PSGL-1-deficient, tumor-bearing mice led to an increased frequency of mice with complete tumor eradication. Targeting both PSGL-1 and PD-1 in wild-type tumor-bearing mice also showed enhanced anti-tumor immunity and slowed melanoma tumor growth. Our findings showed that therapeutically targeting the PSGL-1 immune checkpoint can reinvigorate anti-tumor immunity and suggest that targeting PSGL-1 may represent a new therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberto Tinoco
- University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
12
|
Asano Y. Insights Into the Preclinical Models of SSc. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2021. [DOI: 10.1007/s40674-021-00187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
13
|
Manfredi AA, Ramirez GA, Godino C, Capobianco A, Monno A, Franchini S, Tombetti E, Corradetti S, Distler JHW, Bianchi ME, Rovere-Querini P, Maugeri N. Platelet Phagocytosis via P-selectin Glycoprotein Ligand 1 and Accumulation of Microparticles in Systemic Sclerosis. Arthritis Rheumatol 2021; 74:318-328. [PMID: 34279048 DOI: 10.1002/art.41926] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/29/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE It is unclear why activated platelets and platelet-derived microparticles (MPs) accumulate in the blood of patients with systemic sclerosis (SSc). This study was undertaken to investigate whether defective phagocytosis might contribute to MP accumulation in the blood of patients with SSc. METHODS Blood samples were obtained from a total of 81 subjects, including 25 patients with SSc and 26 patients with stable coronary artery disease (CAD). Thirty sex- and age-matched healthy volunteers served as controls. Studies were also conducted in NSG mice, in which the tail vein of the mice was injected with MPs, and samples of the lung parenchyma were obtained for analysis of the pulmonary microvasculature. Tissue samples from human subjects and from mice were assessed by flow cytometry and immunochemical analyses for determination of platelet-neutrophil interactions, phagocytosis, levels and distribution of P-selectin, P-selectin glycoprotein ligand 1 (PSGL-1), and HMGB1 on platelets and MPs, and concentration of byproducts of neutrophil extracellular trap (NET) generation/catabolism. RESULTS Activated P-selectin+ platelets and platelet-derived HMGB1+ MPs accumulated in the blood of SSc patients but not in the blood of healthy controls. Patients with CAD, a vasculopathy independent of systemic inflammation, had fewer P-selectin+ platelets and a negligible number of MPs. The expression of the receptor for P-selectin, PSGL-1, in neutrophils from SSc patients was significantly decreased, raising the possibility that phagocytes in SSc do not recognize activated platelets, leading to a failure of phagocytosis and continued neutrophil release of MPs. As evidence of this process, activated platelets were not detected in the neutrophils from SSc patients, whereas they were consistently present in the neutrophils from patients with CAD. HMGB1+ MPs elicited generation of NETs, which were only detected in the plasma of SSc patients. In mice, P-selectin-PSGL-1 interaction resulted in platelet phagocytosis in vitro and influenced the ability of MPs to elicit NETs, endothelial activation, and migration of leukocytes through the pulmonary microvasculature. CONCLUSION The clearance of activated platelets via PSGL-1 limits the undesirable effects of MP-elicited neutrophil activation. This balance is disrupted in patients with SSc. Its reconstitution might curb vascular inflammation and prevent fibrosis.
Collapse
Affiliation(s)
- Angelo A Manfredi
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe A Ramirez
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cosmo Godino
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Annalisa Capobianco
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Monno
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Franchini
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrico Tombetti
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Corradetti
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jörg H W Distler
- Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Marco E Bianchi
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Norma Maugeri
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
14
|
DeRogatis JM, Viramontes KM, Neubert EN, Tinoco R. PSGL-1 Immune Checkpoint Inhibition for CD4 + T Cell Cancer Immunotherapy. Front Immunol 2021; 12:636238. [PMID: 33708224 PMCID: PMC7940186 DOI: 10.3389/fimmu.2021.636238] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023] Open
Abstract
Immune checkpoint inhibition targeting T cells has shown tremendous promise in the treatment of many cancer types and are now standard therapies for patients. While standard therapies have focused on PD-1 and CTLA-4 blockade, additional immune checkpoints have shown promise in promoting anti-tumor immunity. PSGL-1, primarily known for its role in cellular migration, has also been shown to function as a negative regulator of CD4+ T cells in numerous disease settings including cancer. PSGL-1 is highly expressed on T cells and can engage numerous ligands that impact signaling pathways, which may modulate CD4+ T cell differentiation and function. PSGL-1 engagement in the tumor microenvironment may promote CD4+ T cell exhaustion pathways that favor tumor growth. Here we highlight that blocking the PSGL-1 pathway on CD4+ T cells may represent a new cancer therapy approach to eradicate tumors.
Collapse
Affiliation(s)
| | | | | | - Roberto Tinoco
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
15
|
González-Tajuelo R, González-Sánchez E, Silván J, Muñoz-Callejas A, Vicente-Rabaneda E, García-Pérez J, Castañeda S, Urzainqui A. Relevance of PSGL-1 Expression in B Cell Development and Activation. Front Immunol 2020; 11:588212. [PMID: 33281818 PMCID: PMC7689347 DOI: 10.3389/fimmu.2020.588212] [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: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 12/03/2022] Open
Abstract
PSGL-1 is expressed in all plasma cells, but only in a small percentage of circulating B cells. Patients with systemic sclerosis (SSc) show reduced expression of PSGL-1 in B cells and increased prevalence of pulmonary arterial hypertension. PSGL-1 deficiency leads to a SSc-like syndrome and SSc-associated pulmonary hypertension in female mice. In this work, the expression of PSGL-1 was assessed during murine B cell development in the bone marrow and in several peripheral and spleen B cell subsets. The impact of PSGL-1 absence on B cell biology was also evaluated. Interestingly, the percentage of PSGL-1 expressing cells and PSGL-1 expression levels decreased in the transition from common lymphoid progenitors to immature B cells. PSGL-1−/− mice showed reduced frequencies of peripheral B cells and reduced B cell lineage-committed precursors in the bone marrow. In the spleen of WT mice, the highest percentages of PSGL-1+ populations were shown by Breg (90%), B1a (34.7%), and B1b (19.1%), while only 2.5–8% of B2 cells expressed PSGL-1; however, within B2 cells, the class-switched subsets showed the highest percentages of PSGL-1+ cells. Interestingly, PSGL-1−/− mice had increased IgG+ and IgD+ subsets and decreased IgA+ population. Of note, the percentage of PSGL-1+ cells was increased in all the B cell subclasses studied in peritoneal fluid. Furthermore, PSGL-1 engagement during in vitro activation with anti-IgM and anti-CD40 antibodies of human peripheral B cells, blocked IL-10 expression by activated human B cells. Remarkably, PSGL-1 expression in circulating plasma cells was reduced in pulmonary arterial hypertension patients. In summary, although the expression of PSGL-1 in mature B cells is low, the lack of PSGL-1 compromises normal B cell development and it may also play a role in the maturation and activation of peripheral naïve B cells.
Collapse
Affiliation(s)
- Rafael González-Tajuelo
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| | - Elena González-Sánchez
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| | - Javier Silván
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| | - Antonio Muñoz-Callejas
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| | - Esther Vicente-Rabaneda
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| | - Javier García-Pérez
- Pulmunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| | - Santos Castañeda
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain.,Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain.,Cátedra UAM-Roche, EPID-Future, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, Madrid, Spain
| |
Collapse
|
16
|
González‐Tajuelo R, de la Fuente‐Fernández M, Morales‐Cano D, Muñoz‐Callejas A, González‐Sánchez E, Silván J, Serrador JM, Cadenas S, Barreira B, Espartero‐Santos M, Gamallo C, Vicente‐Rabaneda EF, Castañeda S, Pérez‐Vizcaíno F, Cogolludo Á, Jiménez‐Borreguero LJ, Urzainqui A. Spontaneous Pulmonary Hypertension Associated With Systemic Sclerosis in P-Selectin Glycoprotein Ligand 1-Deficient Mice. Arthritis Rheumatol 2020; 72:477-487. [PMID: 31509349 PMCID: PMC7065124 DOI: 10.1002/art.41100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Pulmonary arterial hypertension (PAH), one of the major complications of systemic sclerosis (SSc), is a rare disease with unknown etiopathogenesis and noncurative treatments. As mice deficient in P-selectin glycoprotein ligand 1 (PSGL-1) develop a spontaneous SSc-like syndrome, we undertook this study to analyze whether they develop PAH and to examine the molecular mechanisms involved. METHODS Doppler echocardiography was used to estimate pulmonary pressure, immunohistochemistry was used to assess vascular remodeling, and myography of dissected pulmonary artery rings was used to analyze vascular reactivity. Angiotensin II (Ang II) levels were quantified by enzyme-linked immunosorbent assay, and Western blotting was used to measure Ang II type 1 receptor (AT1 R), AT2 R, endothelial cell nitric oxide synthase (eNOS), and phosphorylated eNOS expression in lung lysates. Flow cytometry allowed us to determine cytokine production by immune cells and NO production by endothelial cells. In all cases, there were 4-8 mice per experimental group. RESULTS PSGL-1-/- mice showed lung vessel wall remodeling and a reduced mean ± SD expression of pulmonary AT2 R (expression ratio [relative to β-actin] in female mice age >18 months: wild-type mice 0.799 ± 0.508 versus knockout mice 0.346 ± 0.229). With aging, female PSGL-1-/- mice had impaired up-regulation of estrogen receptor α (ERα) and developed lung vascular endothelial dysfunction coinciding with an increase in mean ± SEM pulmonary Ang II levels (wild-type 48.70 ± 5.13 pg/gm lung tissue versus knockout 78.02 ± 28.09 pg/gm lung tissue) and a decrease in eNOS phosphorylation, leading to reduced endothelial NO production. These events led to a reduction in the pulmonary artery acceleration time:ejection time ratio in 33% of aged female PSGL-1-/- mice, indicating pulmonary hypertension. Importantly, we found expanded populations of interferon-γ-producing PSGL-1-/- T cells and B cells and a reduced presence of regulatory T cells. CONCLUSION The absence of PSGL-1 induces a reduction in Treg cells, NO production, and ERα expression and causes an increase in Ang II in the lungs of female mice, favoring the development of PAH.
Collapse
Affiliation(s)
- Rafael González‐Tajuelo
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | | | - Daniel Morales‐Cano
- University Complutense of Madrid School of Medicine and Ciber Enfermedades RespiratoriasMadridSpain
| | - Antonio Muñoz‐Callejas
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | - Elena González‐Sánchez
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | - Javier Silván
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | - Juan Manuel Serrador
- Centro de Biología Molecular Severo Ochoa (CBMSO) and Instituto de Física Teórica CSIC/Universidad Autónoma de Madrid (UAM)MadridSpain
| | - Susana Cadenas
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, and CBMSO, CSIC‐UAMMadridSpain
| | - Bianca Barreira
- University Complutense of Madrid School of Medicine and Ciber Enfermedades RespiratoriasMadridSpain
| | - Marina Espartero‐Santos
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | - Carlos Gamallo
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | - Esther F. Vicente‐Rabaneda
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| | - Santos Castañeda
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, and Catedra UAM‐ROCHEMadridSpain
| | - Francisco Pérez‐Vizcaíno
- University Complutense of Madrid School of Medicine and Ciber Enfermedades RespiratoriasMadridSpain
| | - Ángel Cogolludo
- University Complutense of Madrid School of Medicine and Ciber Enfermedades RespiratoriasMadridSpain
| | | | - Ana Urzainqui
- Fundación de Investigación Biomédica‐Hospital de la PrincesaIIS‐Princesa, Servicio de InmunlogíaMadridSpain
| |
Collapse
|
17
|
VISTA is an acidic pH-selective ligand for PSGL-1. Nature 2019; 574:565-570. [PMID: 31645726 DOI: 10.1038/s41586-019-1674-5] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 09/20/2019] [Indexed: 12/20/2022]
Abstract
Co-inhibitory immune receptors can contribute to T cell dysfunction in patients with cancer1,2. Blocking antibodies against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) partially reverse this effect and are becoming standard of care in an increasing number of malignancies3. However, many of the other axes by which tumours become inhospitable to T cells are not fully understood. Here we report that V-domain immunoglobulin suppressor of T cell activation (VISTA) engages and suppresses T cells selectively at acidic pH such as that found in tumour microenvironments. Multiple histidine residues along the rim of the VISTA extracellular domain mediate binding to the adhesion and co-inhibitory receptor P-selectin glycoprotein ligand-1 (PSGL-1). Antibodies engineered to selectively bind and block this interaction in acidic environments were sufficient to reverse VISTA-mediated immune suppression in vivo. These findings identify a mechanism by which VISTA may engender resistance to anti-tumour immune responses, as well as an unexpectedly determinative role for pH in immune co-receptor engagement.
Collapse
|
18
|
Deregulated PSGL-1 Expression in B Cells and Dendritic Cells May Be Implicated in Human Systemic Sclerosis Development. J Invest Dermatol 2018; 138:2123-2132. [PMID: 29689251 DOI: 10.1016/j.jid.2018.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 01/13/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disorder with high morbidity and mortality, is difficult to diagnose early, and has no curative treatment. PSGL-1 is a leukocyte receptor whose deficiency in mice promotes an SSc-like disease. ADAM8, a metalloprotease that cleaves PSGL-1, is implicated in inflammatory processes. Our goal was to evaluate whether PSGL-1 and ADAM8 contribute to the pathogenesis of human SSc. We found that patients with SSc presented increased PSGL-1 expression on monocytes, dendritic cells, and T cells and decreased expression of PSGL-1 on B cells. PSGL-1 on monocytes from SSc patients failed to induce Syk phosphorylation or IL-10 production after interaction with P-selectin. Up to 60% of the IL-10-producing B cells expressed PSGL-1, pointing to a regulatory role for PSGL-1 in B cells, and PSGL-1+ B cells from SSc patients had decreased IL-10 production. ADAM8 expression was increased on antigen-presenting cells and T lymphocytes of SSc patients. Patients treated with calcium antagonists had lower levels of ADAM8 on APCs and T lymphocytes. Multivariate analysis indicated that the high percentage of ADAM8-expressing plasmacytoid dendritic cells discriminated patients from healthy donors. High PSGL-1 expression on dendritic cells was associated with the presence of interstitial lung disease.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW We review P-selectin glycoprotein ligand-1 (PSGL-1) as a selectin and chemokine-binding adhesion molecule. PSGL-1 is widely studied in neutrophils. Here, we focus on T cells, because PSGL-1 was recently described as a major immunomodulatory molecule during viral infection. PSGL-1 also plays a crucial role in T-cell homeostasis by binding to lymphoid chemokines, and can induce tolerance by enhancing the functions of regulatory T cells. RECENT FINDINGS PSGL-1 was originally described as a leukocyte ligand for P-selectin, but it is actually a ligand for all selectins (P-, L- and E-selectin), binds chemokines, activates integrins and profoundly affects T-cell biology. It has been shown recently that PSGL-1 can modulate T cells during viral infection by acting as a negative regulator for T-cell functions. Absence of PSGL-1 promotes effector CD4 and CD8 T-cell differentiation and prevents T-cell exhaustion. Consistent with this, tumor growth was significantly reduced in PSGL-1-deficient mice because of an enhanced number of effector T cells together with reduced levels of inhibitory receptors that induce T-cell exhaustion. SUMMARY PSGL-1 is the best-studied selectin ligand and has become a posterchild of versatility in leukocyte adhesion, inflammation and immunology. The direct involvement of PSGL-1 in T-cell biology suggests that it might be a drug target. Indeed, PSGL-1 has been tested in some clinical trials and recently, PSGL-1 blockers were proposed as a potential cotherapy in cancer immunotherapy.
Collapse
|
20
|
PSGL-1: A New Player in the Immune Checkpoint Landscape. Trends Immunol 2017; 38:323-335. [PMID: 28262471 DOI: 10.1016/j.it.2017.02.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/02/2017] [Accepted: 02/08/2017] [Indexed: 02/07/2023]
Abstract
P-selectin glycoprotein ligand-1 (PSGL-1) has long been studied as an adhesion molecule involved in immune cell trafficking and is recognized as a regulator of many facets of immune responses by myeloid cells. PSGL-1 also regulates T cell migration during homeostasis and inflammatory settings. However, recent findings indicate that PSGL-1 can also negatively regulate T cell function. Because T cell differentiation is finely tuned by multiple positive and negative regulatory signals that appropriately scale the magnitude of the immune response, PSGL-1 has emerged as an important checkpoint during this process. We summarize what is known regarding PSGL-1 structure and function and highlight how it may act as an immune checkpoint inhibitor in T cells.
Collapse
|
21
|
P-Selectin preserves immune tolerance in mice and is reduced in human cutaneous lupus. Sci Rep 2017; 7:41841. [PMID: 28150814 PMCID: PMC5288776 DOI: 10.1038/srep41841] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022] Open
Abstract
Mice deficient in P-Selectin presented altered immunity/tolerance balance. We have observed that the absence of P-Selectin promotes splenomegaly with reduced naïve T cell population, elevated activated/effector T cell subset, increased germinal center B and Tfh populations and high production of autoreactive antibodies. Moreover, 1.5-3-month-old P-selectin KO mice showed reduced IL-10-producing leukocytes in blood and a slightly reduced Treg population in the skin. With aging and, coinciding with disease severity, there is an increase in the IL17+ circulating and dermal T cell subpopulations and reduction of dermal Treg. As a consequence, P-Selectin deficient mice developed a progressive autoimmune syndrome showing skin alterations characteristic of lupus prone mice and elevated circulating autoantibodies, including anti-dsDNA. Similar to human SLE, disease pathogenesis was characterized by deposition of immune complexes in the dermoepidermal junction and renal glomeruli, and a complex pattern of autoantibodies. More important, skin biopsies of cutaneous lupus erythematosus patients did not show increased expression of P-Selectin, as described for other inflammatory diseases, and the number of vessels expressing P-Selectin was reduced.
Collapse
|
22
|
Abstract
An important underlying mechanism that contributes to autoimmunity is the loss of inhibitory signaling in the immune system. Sialic acid-recognizing Ig superfamily lectins or Siglecs are a family of cell surface proteins largely expressed in hematopoietic cells. The majority of Siglecs are inhibitory receptors expressed in immune cells that bind to sialic acid-containing ligands and recruit SH2-domain-containing tyrosine phosphatases to their cytoplasmic tails. They deliver inhibitory signals that can contribute to the constraining of immune cells, and thus protect the host from autoimmunity. The inhibitory functions of CD22/Siglec-2 and Siglec-G and their contributions to tolerance and autoimmunity, primarily in the B lymphocyte context, are considered in some detail in this review. The relevance to autoimmunity and unregulated inflammation of modified sialic acids, enzymes that modify sialic acid, and other sialic acid-binding proteins are also reviewed.
Collapse
Affiliation(s)
- Vinay S Mahajan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Departments of Medicine and Pathology, Harvard Medical School, Boston, MA, USA.,Deaprtment of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Departments of Medicine and Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Selectin-mediated leukocyte trafficking during the development of autoimmune disease. Autoimmun Rev 2015; 14:984-95. [DOI: 10.1016/j.autrev.2015.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/18/2015] [Indexed: 12/18/2022]
|
24
|
De Langhe E, Lories R. Fibrogenesis, novel lessons from animal models. Semin Immunopathol 2015; 37:565-74. [PMID: 26141608 DOI: 10.1007/s00281-015-0510-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/16/2015] [Indexed: 12/15/2022]
Abstract
Systemic sclerosis (SSc) is a devastating chronic autoimmune connective tissue disease characterized by vasculopathy, autoimmunity with inflammation, and progressive fibrogenesis. The current paradigm of the pathogenesis of SSc is that of an unknown initial trigger, leading to a complex interaction of immune cells, endothelial cells, and fibroblasts, producing cytokines, growth and angiogenic factors, and resulting in uncontrolled and persistent tissue fibrogenesis by an altered mesenchymal cell compartment. Animal models are of utmost importance to investigate the different steps in the pathogenesis. This review will elaborate on recent findings in established and more recently developed animal models, presenting data on compounds that are in or ready to be translated into clinical trials, or provide interesting new findings in the understanding of the pathophysiology of SSc. We focus on recent findings concerning the vessel-extracellular matrix interaction, the initial triggering aggressor, the concept of autoimmunity and inflammatory changes, the effector cells and their origins, and the complex interaction of the different signaling pathways in fibrogenesis.
Collapse
Affiliation(s)
- Ellen De Langhe
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | | |
Collapse
|