Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Eur Respir J ; 63(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38485150

ABSTRACT

There is an unmet need for new therapeutic strategies that target alternative pathways to improve the prognosis of patients with pulmonary arterial hypertension (PAH). As immunity has been involved in the development and progression of vascular lesions in PAH, we review the potential contribution of B-cells in its pathogenesis and evaluate the relevance of B-cell-targeted therapies. Circulating B-cell homeostasis is altered in PAH patients, with total B-cell lymphopenia, abnormal subset distribution (expansion of naïve and antibody-secreting cells, reduction of memory B-cells) and chronic activation. B-cells are recruited to the lungs through local chemokine secretion, and activated by several mechanisms: 1) interaction with lung vascular autoantigens through cognate B-cell receptors; 2) costimulatory signals provided by T follicular helper cells (interleukin (IL)-21), type 2 T helper cells and mast cells (IL-4, IL-6 and IL-13); and 3) increased survival signals provided by B-cell activating factor pathways. This activity results in the formation of germinal centres within perivascular tertiary lymphoid organs and in the local production of pathogenic autoantibodies that target the pulmonary vasculature and vascular stabilisation factors (including angiotensin-II/endothelin-1 receptors and bone morphogenetic protein receptors). B-cells also mediate their effects through enhanced production of pro-inflammatory cytokines, reduced anti-inflammatory properties by regulatory B-cells, immunoglobulin (Ig)G-induced complement activation, and IgE-induced mast cell activation. Precision-medicine approaches targeting B-cell immunity are a promising direction for select PAH conditions, as suggested by the efficacy of anti-CD20 therapy in experimental models and a trial of rituximab in systemic sclerosis-associated PAH.


Subject(s)
B-Lymphocytes , Pulmonary Arterial Hypertension , Humans , B-Lymphocytes/immunology , Pulmonary Arterial Hypertension/immunology , Animals , Lung/immunology , Autoantibodies/immunology , Hypertension, Pulmonary/immunology
2.
Autoimmun Rev ; 23(4): 103514, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38181859

ABSTRACT

Pre-capillary pulmonary arterial hypertension (PAH) is hemodynamically characterized by a mean pulmonary arterial pressure (mPAP) ≥ 20 mmHg, pulmonary capillary wedge pressure (PAWP) ≤15 mmHg and pulmonary vascular resistance (PVR) > 2. PAH is classified in six clinical subgroups, including idiopathic PAH (IPAH) and PAH associated to connective tissue diseases (CTD-PAH), that will be the main object of this review. The aim is to compare these two PAH subgroups in terms of epidemiology, histological and pathogenic findings in an attempt to define disease-specific features, including autoimmunity, that may explain the heterogeneity of response to therapy between IPAH and CTD-PAH.


Subject(s)
Autoimmunity , Connective Tissue Diseases , Humans , Connective Tissue Diseases/immunology , Connective Tissue Diseases/complications , Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/physiopathology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Familial Primary Pulmonary Hypertension/physiopathology , Familial Primary Pulmonary Hypertension/immunology
3.
Front Immunol ; 13: 895501, 2022.
Article in English | MEDLINE | ID: mdl-35757687

ABSTRACT

Introduction: Inflammation is a major pathological feature of pulmonary arterial hypertension (PAH), particularly in the context of inflammatory conditions such as systemic sclerosis (SSc). The endothelin system and anti-endothelin A receptor (ETA) autoantibodies have been implicated in the pathogenesis of PAH, and endothelin receptor antagonists are routinely used treatments for PAH. However, immunological functions of the endothelin B receptor (ETB) remain obscure. Methods: Serum levels of anti-ETB receptor autoantibodies were quantified in healthy donors and SSc patients with or without PAH. Age-dependent effects of overexpression of prepro-endothelin-1 or ETB deficiency on pulmonary inflammation and the cardiovascular system were studied in mice. Rescued ETB-deficient mice (ETB-/-) were used to prevent congenital Hirschsprung disease. The effects of pulmonary T-helper type 2 (Th2) inflammation on PAH-associated pathologies were analyzed in ETB-/- mice. Pulmonary vascular hemodynamics were investigated in isolated perfused mouse lungs. Hearts were assessed for right ventricular hypertrophy. Pulmonary inflammation and collagen deposition were assessed via lung microscopy and bronchoalveolar lavage fluid analyses. Results: Anti-ETB autoantibody levels were elevated in patients with PAH secondary to SSc. Both overexpression of prepro-endothelin-1 and rescued ETB deficiency led to pulmonary hypertension, pulmonary vascular hyperresponsiveness, and right ventricular hypertrophy with accompanying lymphocytic alveolitis. Marked perivascular lymphocytic infiltrates were exclusively found in ETB-/- mice. Following induction of pulmonary Th2 inflammation, PAH-associated pathologies and perivascular collagen deposition were aggravated in ETB-/- mice. Conclusion: This study provides evidence for an anti-inflammatory role of ETB. ETB seems to have protective effects on Th2-evoked pathologies of the cardiovascular system. Anti-ETB autoantibodies may modulate ETB-mediated immune homeostasis.


Subject(s)
Pulmonary Arterial Hypertension , Receptor, Endothelin B , Animals , Autoantibodies/immunology , Endothelin-1/immunology , Familial Primary Pulmonary Hypertension/immunology , Humans , Hypertrophy, Right Ventricular/immunology , Inflammation/immunology , Mice , Pulmonary Arterial Hypertension/immunology , Receptor, Endothelin B/immunology , Scleroderma, Systemic/immunology
4.
Int J Mol Sci ; 23(5)2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35269553

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by (mal)adaptive remodeling of the pulmonary vasculature, which is associated with inflammation, fibrosis, thrombosis, and neovascularization. Vascular remodeling in PAH is associated with cellular metabolic and inflammatory reprogramming that induce profound endothelial and smooth muscle cell phenotypic changes. Multiple signaling pathways and regulatory loops act on metabolic and inflammatory mediators which influence cellular behavior and trigger pulmonary vascular remodeling in vivo. This review discusses the role of bioenergetic and inflammatory impairments in PAH development.


Subject(s)
Muscle, Smooth, Vascular/pathology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/pathology , Animals , Energy Metabolism , Humans , Muscle, Smooth, Vascular/immunology , Pulmonary Arterial Hypertension/immunology , Pulmonary Artery/immunology , Signal Transduction , Vascular Remodeling
5.
Front Immunol ; 12: 684657, 2021.
Article in English | MEDLINE | ID: mdl-34489935

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic, incurable condition characterized by pulmonary vascular remodeling, perivascular inflammation, and right heart failure. Regulatory T cells (Tregs) stave off autoimmunity, and there is increasing evidence for their compromised activity in the inflammatory milieu of PAH. Abnormal Treg function is strongly correlated with a predisposition to PAH in animals and patients. Athymic Treg-depleted rats treated with SU5416, an agent causing pulmonary vascular injury, develop PAH, which is prevented by infusing missing CD4+CD25highFOXP3+ Tregs. Abnormal Treg activity may also explain why PAH disproportionately affects women more than men. This mini review focuses on the role of Tregs in PAH with a special view to sexual dimorphism and the future promise of Treg therapy.


Subject(s)
Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/prevention & control , T-Lymphocytes, Regulatory/immunology , Vascular System Injuries/immunology , Vascular System Injuries/prevention & control , Animals , Autoimmunity , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Indoles/adverse effects , Pulmonary Arterial Hypertension/pathology , Pyrroles/adverse effects , Rats , Sex Characteristics , Vascular System Injuries/pathology
6.
Bioengineered ; 12(1): 2576-2591, 2021 12.
Article in English | MEDLINE | ID: mdl-34233597

ABSTRACT

This study aimed to screen key biomarkers and investigate immune infiltration in pulmonary arterial hypertension (PAH) based on integrated bioinformatics analysis. The Gene Expression Omnibus (GEO) database was used to download three mRNA expression profiles comprising 91 PAH lung specimens and 49 normal lung specimens. Three mRNA expression datasets were combined, and differentially expressed genes (DEGs) were obtained. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses and the protein-protein interaction (PPI) network of DEGs were performed using the STRING and DAVID databases, respectively. The diagnostic value of hub gene expression in PAH was also analyzed. Finally, the infiltration of immune cells in PAH was analyzed using the CIBERSORT algorithm. Total 182 DEGs (117 upregulated and 65 downregulated) were identified, and 15 hub genes were screened. These 15 hub genes were significantly associated with immune system functions such as myeloid leukocyte migration, neutrophil migration, cell chemotaxis, Toll-like receptor signaling pathway, and NF-κB signaling pathway. A 7-gene-based model was constructed and had a better diagnostic value in identifying PAH tissues compared with normal controls. The immune infiltration profiles of the PAH and normal control samples were significantly different. High proportions of resting NK cells, activated mast cells, monocytes, and neutrophils were found in PAH samples, while high proportions of resting T cells CD4 memory and Macrophages M1 cell were found in normal control samples. Functional enrichment of DEGs and immune infiltration analysis between PAH and normal control samples might help to understand the pathogenesis of PAH.


Subject(s)
Biomarkers/metabolism , Computational Biology , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/immunology , Case-Control Studies , Databases, Genetic , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Humans , Models, Biological , Protein Interaction Maps/genetics , ROC Curve , Regression Analysis
7.
Bioengineered ; 12(1): 3137-3147, 2021 12.
Article in English | MEDLINE | ID: mdl-34252346

ABSTRACT

This study aimed to explore the molecular mechanisms related to immune and hub genes related to pulmonary arterial hypertension (PAH). The differentially expressed genes (DEGs) of GSE15197 were identified as filters with adjusted P value <0.05, and |Log2 fold change|> 1. Biofunctional and pathway enrichment annotation of DEGs indicated that immunity and inflammation may play an important role in the molecular mechanism of PAH. The CIBERSORT algorithm further analyzed the immune cell infiltration characteristics of the PAH and control samples. Subsequently, 16 hub genes were identified from DEGs using the least absolute shrinkage and selection operator (LASSO) algorithm. An immune related gene CX3CR1 was further selected from the intersection results of the 16 hub genes and the top 20 genes with the most adjacent nodes in the protein-protein interaction (PPI) network. GSE113439, GSE48149, and GSE33463 datasets were used to validate and proved CX3CR1 with a remarkable score of AUC to distinguish PAH samples caused by various reasons from the control group.


Subject(s)
Computational Biology , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/immunology , Transcriptome/genetics , Transcriptome/immunology , Algorithms , Databases, Genetic , Humans , Protein Interaction Maps , Pulmonary Arterial Hypertension/metabolism
8.
Int J Mol Sci ; 22(9)2021 May 09.
Article in English | MEDLINE | ID: mdl-34065088

ABSTRACT

Loss of function KCNK3 mutation is one of the gene variants driving hereditary pulmonary arterial hypertension (PAH). KCNK3 is expressed in several cell and tissue types on both membrane and endoplasmic reticulum and potentially plays a role in multiple pathological process associated with PAH. However, the role of various stressors driving the susceptibility of KCNK3 mutation to PAH is unknown. Hence, we exposed kcnk3fl/fl animals to hypoxia, metabolic diet and low dose lipopolysaccharide (LPS) and performed molecular characterization of their tissue. We also used tissue samples from KCNK3 patients (skin fibroblast derived inducible pluripotent stem cells, blood, lungs, peripheral blood mononuclear cells) and performed microarray, immunohistochemistry (IHC) and mass cytometry time of flight (CyTOF) experiments. Although a hypoxic insult did not alter vascular tone in kcnk3fl/fl mice, RNASeq study of these lungs implied that inflammatory and metabolic factors were altered, and the follow-up diet study demonstrated a dysregulation of bone marrow cells in kcnk3fl/fl mice. Finally, a low dose LPS study clearly showed that inflammation could be a possible second hit driving PAH in kcnk3fl/fl mice. Multiplex, IHC and CyTOF immunophenotyping studies on human samples confirmed the mouse data and strongly indicated that cell mediated, and innate immune responses may drive PAH susceptibility in these patients. In conclusion, loss of function KCNK3 mutation alters various physiological processes from vascular tone to metabolic diet through inflammation. Our data suggests that altered circulating immune cells may drive PAH susceptibility in patients with KCNK3 mutation.


Subject(s)
Immunomodulation/genetics , Mutation , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/immunology , Animals , Biomarkers , Case-Control Studies , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Hypoxia/genetics , Hypoxia/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Models, Biological , Monocytes/immunology , Monocytes/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Pulmonary Arterial Hypertension/complications , Pulmonary Arterial Hypertension/physiopathology , Transcriptome
9.
Chest ; 160(4): 1442-1458, 2021 10.
Article in English | MEDLINE | ID: mdl-34181952

ABSTRACT

BACKGROUND: Preclinical evidence implicates neutrophil elastase (NE) in pulmonary arterial hypertension (PAH) pathogenesis, and the NE inhibitor elafin is under early therapeutic investigation. RESEARCH QUESTION: Are circulating NE and elafin levels abnormal in PAH and are they associated with clinical severity? STUDY DESIGN AND METHODS: In an observational Stanford University PAH cohort (n = 249), plasma NE and elafin levels were measured in comparison with those of healthy control participants (n = 106). NE and elafin measurements were then related to PAH clinical features and relevant ancillary biomarkers. Cox regression models were fitted with cubic spline functions to associate NE and elafin levels with survival. To validate prognostic relationships, we analyzed two United Kingdom cohorts (n = 75 and n = 357). Mixed-effects models evaluated NE and elafin changes during disease progression. Finally, we studied effects of NE-elafin balance on pulmonary artery endothelial cells (PAECs) from patients with PAH. RESULTS: Relative to control participants, patients with PAH were found to have increased NE levels (205.1 ng/mL [interquartile range (IQR), 123.6-387.3 ng/mL] vs 97.6 ng/mL [IQR, 74.4-126.6 ng/mL]; P < .0001) and decreased elafin levels (32.0 ng/mL [IQR, 15.3-59.1 ng/mL] vs 45.5 ng/mL [IQR, 28.1-92.8 ng/mL]; P < .0001) independent of PAH subtype, illness duration, and therapies. Higher NE levels were associated with worse symptom severity, shorter 6-min walk distance, higher N-terminal pro-type brain natriuretic peptide levels, greater right ventricular dysfunction, worse hemodynamics, increased circulating neutrophil levels, elevated cytokine levels, and lower blood BMPR2 expression. In Stanford patients, NE levels of > 168.5 ng/mL portended increased mortality risk after adjustment for known clinical predictors (hazard ratio [HR], 2.52; CI, 1.36-4.65, P = .003) or prognostic cytokines (HR, 2.63; CI, 1.42-4.87; P = .001), and the NE level added incremental value to established PAH risk scores. Similar prognostic thresholds were identified in validation cohorts. Longitudinal NE changes tracked with clinical trends and outcomes. PAH PAECs exhibited increased apoptosis and attenuated angiogenesis when exposed to NE at the level observed in patients' blood. Elafin rescued PAEC homeostasis, yet the required dose exceeded levels found in patients. INTERPRETATION: Blood levels of NE are increased while elafin levels are deficient across PAH subtypes. Higher NE levels are associated with worse clinical disease severity and outcomes, and this target-specific biomarker could facilitate therapeutic development of elafin.


Subject(s)
Elafin/blood , Leukocyte Elastase/blood , Pulmonary Arterial Hypertension/blood , Adult , Aged , Apoptosis/drug effects , Elafin/pharmacology , Endothelial Cells/drug effects , Female , Humans , Male , Middle Aged , Neovascularization, Physiologic/drug effects , Pancreatic Elastase/pharmacology , Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/cytology , Severity of Illness Index , Vascular Resistance
10.
Respir Res ; 22(1): 137, 2021 May 04.
Article in English | MEDLINE | ID: mdl-33947407

ABSTRACT

BACKGROUND: Recent studies have provided evidence for an important contribution of the immune system in the pathophysiology of pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH). In this report, we investigated whether the inflammatory profile of pulmonary hypertension patients changes over time and correlates with patient WHO subgroups or survival. METHODS: 50 PAH patients (16 idiopathic (I)PAH, 24 Connective Tissue Disease (CTD)-PAH and 10 Congenital Heart Disease (CHD)-PAH), 37 CTEPH patients and 18 healthy controls (HCs) were included in the study. Plasma inflammatory markers at baseline and after 1-year follow-up were measured using ELISAs. Subsequently, correlations with hemodynamic parameters and survival were explored and data sets were subjected to unbiased multivariate analyses. RESULTS: At diagnosis, we found that plasma levels of interleukin-6 (IL-6) and the chemokines (C-X3-C) motif legend CXCL9 and CXCL13 in CTD-PAH patients were significantly increased, compared with HCs. In idiopathic PAH patients the levels of tumor growth factor-ß (TGFß), IL-10 and CXCL9 were elevated, compared with HCs. The increased CXCL9 and IL-8 concentrations in CETPH patients correlated significantly with decreased survival, suggesting that CXCL9 and IL-8 may be prognostic markers. After one year of treatment, IL-10, CXCL13 and TGFß levels changed significantly in the PAH subgroups and CTEPH patients. Unbiased multivariate analysis revealed clustering of PH patients based on inflammatory mediators and clinical parameters, but did not separate the WHO subgroups. Importantly, these multivariate analyses separated patients with < 3 years and > 3 years survival, in particular when inflammatory mediators were combined with clinical parameters. DISCUSSION: Our study revealed elevated plasma levels of inflammatory mediators in different PAH subgroups and CTEPH at baseline and at 1-year follow-up, whereby CXCL9 and IL-8 may prove to be prognostic markers for CTEPH patients. While this study is exploratory and hypothesis generating, our data indicate an important role for IL-8 and CXCL9 in CHD and CTEPH patients considering the increased plasma levels and the observed correlation with survival. CONCLUSION: In conclusion, our studies identified an inflammatory signature that clustered PH patients into WHO classification-independent subgroups that correlated with patient survival.


Subject(s)
Cytokines/blood , Inflammation Mediators/blood , Pulmonary Arterial Hypertension/blood , Adult , Aged , Biomarkers/blood , Case-Control Studies , Female , Humans , Lung Transplantation , Male , Middle Aged , Progression-Free Survival , Prospective Studies , Pulmonary Arterial Hypertension/diagnosis , Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/mortality , Risk Assessment , Risk Factors , Time Factors , Young Adult
11.
Cardiovasc Ther ; 2021: 6651009, 2021.
Article in English | MEDLINE | ID: mdl-33680092

ABSTRACT

BACKGROUND: Pulmonary complications of systemic sclerosis (SSc), including pulmonary arterial hypertension (PAH), are the leading causes of patient death. However, the precise molecular mechanisms of its etiology are unclear. This study's objective was to identify the candidate genes involved in the progression of SSc-PAH and investigate the genes' function. METHODS: The gene expression profiles of GSE33463 were obtained from the Gene Expression Omnibus (GEO) database. A free-scale gene coexpression network was constructed using the weighted gene coexpression network analysis (WGCNA) to explore the association between gene sets and clinical features and identify candidate biomarkers. Then, gene ontology analysis was performed. A second dataset was used, GSE19617, to validate the hub genes. The verified hub genes' potential function was further explored using gene set enrichment analysis (GSEA). RESULTS: Through average link-level clustering, a total of seven modules were classified. A total of 938 hub genes were identified in the key module, and the key module's function mainly enriched was related to chemokine activities. Subsequently, four candidate genes, BTG3, CCR2, RAB10, and TMEM60, were filtered. The expression levels of these four hub genes were consistent in the GSE19617 and GSE33463 datasets. We plotted the ROC curve of the hub genes (all AUC > 0.70). Furthermore, the results of the GSEA for hub genes were correlated with complement and inflammatory responses. CONCLUSIONS: The hub genes (BTG3, CCR2, RAB10, and TMEM60) performed well in distinguishing the SSc-PAH patients from controls, and some biological functions, related to immunity, inflammation, and cytokines, might pave the way for follow-up studies on the diagnosis and treatment of SSc-PAH.


Subject(s)
Cytokines/genetics , Immunity , Inflammation/complications , Pulmonary Arterial Hypertension/etiology , Scleroderma, Systemic/complications , Gene Ontology , Gene Regulatory Networks , Humans , Pulmonary Arterial Hypertension/immunology , Transcriptome
12.
Life Sci ; 271: 119151, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33539912

ABSTRACT

AIMS: Limited cutaneous systemic sclerosis-associated pulmonary arterial hypertension (lcSSc-PAH) is a complex multi-system disease with high morbidity and mortality. The purpose of this study is to identify the hub genes and immune characteristics of limited cutaneous systemic sclerosis (lcSSc) and lcSSc-PAH through bioinformatics. MAIN METHODS: LcSSc-PAH raw data were obtained from the GEO database (GSE19617). Weighted gene Co-expression Network analysis (WGCNA) was used to evaluate key modules. Then, we performed Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis with R software and verified the diagnostic value of the hub genes. Finally, Immune Cell Abundance Identifier (ImmuCellAI) was used to analyze the immune characteristics of the normal subjects, lcSSc and lcSSc-PAH patients, the results were displayed graphically. KEY FINDINGS: Enrichment of two important modules by GO and KEGG identified key biological processes and pathways related to pathogen infection and immune function. Three hub genes (BID, IFNGR1, ZAP70) related to immune function were identified. The analysis of immune characteristics showed that the correlation and abundance of immune cells such as inducible regulatory T (iTreg) cells, B cells, macrophages, natural killer (NK) cells, CD8T cells, mucosal-associated invariant T(MAIT) cells and dendritic cells(DCs) were significantly different in the normal subjects, lcSSc and lcSSc-PAH patients. SIGNIFICANCE: Pathogen infection, changes in the number and function of immune cells, and interactions among immune cells may preliminarily reveal the pathological mechanism of lcSSc-PAH. The hub genes, pathways and immune characteristics identified in this research remains to be further studied.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/immunology , Scleroderma, Systemic/genetics , Scleroderma, Systemic/immunology , Databases, Genetic , Humans , T-Lymphocytes/immunology
13.
Mod Rheumatol Case Rep ; 5(1): 152-155, 2021 01.
Article in English | MEDLINE | ID: mdl-32697139

ABSTRACT

Antisynthetase Syndrome (ASS) is a subset of idiopathic inflammatory myopathies characterised by specific clinical features such as interstitial lung disease (ILD), fever, myositis, Raynaud's phenomenon, cutaneous involvement and arthritis related to the presence of anti-aminoacyl-tRNA-synthetase (anti-ARS) autoantibodies. Moreover, Pulmonary arterial hypertension (PAH) is a life-threatening complication associated with connective tissue diseases mainly systemic sclerosis (SSc-PAH). It has been suggested that PAH can complicate ASS patients but little is known about the prevalence and risk factors to develop this complication. Here we report on two patients with ASS and PH. The first one represents a complete picture of ASS anti-Jo-1 positive, the second an amyophatic ASS anti-PL-12 positive. In one of our ASS-PAH patients, specific treatment lead to improvement of PAH. There are no specific recommendations on current guidelines regarding either PAH screening or treatment in ASS, but performing echocardiogram, ECG, pulmonary function test and prompt initiation of specific therapies seems to improve right heart catheterisation (RHC) parameters and survival.


Subject(s)
Myositis/complications , Pulmonary Arterial Hypertension/complications , Amino Acyl-tRNA Synthetases/immunology , Autoantibodies/blood , Female , Humans , Middle Aged , Myositis/immunology , Pulmonary Arterial Hypertension/immunology , Tomography, X-Ray Computed
14.
Front Immunol ; 11: 608883, 2020.
Article in English | MEDLINE | ID: mdl-33362796

ABSTRACT

Pulmonary arterial hypertension (PAH) is a disease of the lung blood vessels that results in right heart failure. PAH is thought to occur in about 5% to 10% of patients with hepatosplenic schistosomiasis, particularly due to S. mansoni. The lung blood vessel injury may result from a combination of embolization of eggs through portocaval shunts into the lungs causing localized Type 2 inflammatory response and vessel remodeling, triggering of autonomous pathology that becomes independent of the antigen, and high cardiac output as seen in portopulmonary hypertension. The condition is likely underdiagnosed as there is little systematic screening, and risk factors for developing PAH are not known. Screening is done by echocardiography, and formal diagnosis requires invasive right heart catheterization. Patients with Schistosoma-associated PAH show reduced functional capacity and can be treated with pulmonary vasodilators, which improves symptoms and may improve survival. There are animal models of this disease that might help in understanding disease pathogenesis and identify novel targets to screen and treatment. Pathogenic mechanisms include Type 2 immunity and activation and signaling in the TGF-ß pathway. There are still major uncertainties regarding Schistosoma-associated PAH development, course and treatment.


Subject(s)
Pulmonary Arterial Hypertension/pathology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/pathology , Animals , Humans , Lung/immunology , Lung/pathology , Pulmonary Arterial Hypertension/immunology , Schistosomiasis mansoni/immunology , Transforming Growth Factor beta/immunology , Vascular Remodeling/immunology , Vascular Remodeling/physiology
15.
Front Immunol ; 11: 588212, 2020.
Article in English | MEDLINE | ID: mdl-33281818

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.


Subject(s)
B-Lymphocytes/immunology , Membrane Glycoproteins/immunology , Pulmonary Arterial Hypertension/immunology , Aged , Animals , Female , Humans , Male , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Peritoneal Cavity/cytology , Spleen/cytology , Spleen/immunology
16.
J Cell Mol Med ; 24(19): 11409-11421, 2020 10.
Article in English | MEDLINE | ID: mdl-32860486

ABSTRACT

Inflammation and immunity play a causal role in the pathogenesis of pulmonary vascular remodelling and pulmonary arterial hypertension (PAH). However, the pathways and mechanisms by which inflammation and immunity contribute to pulmonary vascular remodelling remain unknown. RNA sequencing was used to analyse the transcriptome in control and rats injected with monocrotaline (MCT) for various weeks. Using the transcriptional profiling of MCT-induced PAH coupled with bioinformatics analysis, we clustered the differentially expressed genes (DEGs) and chose the increased expression patterns associated with inflammatory and immune response. We found the enrichment of Toll-like receptor (TLR) and Nod-like receptor (NLR) pathways and identified NF-κB-mediated inflammatory and immune profiling in MCT-induced PAH. Pathway-based data integration and visualization showed the dysregulated TLR and NLR pathways, including increased expression of TLR2 and NLRP3, and their downstream molecules. Further analysis revealed that the activation of TLR and NLR pathways was associated with up-regulation of damage-associated molecular patterns (DAMPs) and RIPK3-mediated necroptosis was involved in the generation of DAMPs in MCT-induced PAH. Collectively, we identify RIPK3-mediated necroptosis and its triggered TLR and NLR pathways in the progression of pulmonary vascular remodelling, thus providing novel insights into the mechanisms underlying inflammation and immunity in the pathogenesis of PAH.


Subject(s)
NLR Proteins/metabolism , Necroptosis/genetics , Pulmonary Arterial Hypertension/genetics , Signal Transduction , Toll-Like Receptors/metabolism , Transcriptome/genetics , Alarmins/metabolism , Animals , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation , Immunity/genetics , Inflammation/genetics , Models, Biological , Monocrotaline , Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/pathology , Rats, Sprague-Dawley , Signal Transduction/genetics
17.
Autoimmun Rev ; 19(9): 102602, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32659476

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) in systemic sclerosis (SSc) is a lethal complication affecting 8-15% of patients. Screening tests such as echocardiography and pulmonary function tests allow for triaging patients for diagnosis by right heart catheterization. Understanding risk factors of SSc-PAH could help differentiate high-risk patients. METHODS: A systematic review was conducted to determine associations with SSc-PAH, including clinical/disease characteristics, antibodies, labs and biomarkers. The frequencies of publications featuring each risk/association were reported. RESULTS: Among 2654 articles, 984 duplicates and 1578 irrelevant articles were removed, leaving 92 articles for manual screening. After excluding 55 papers with small sample sizes, publications from identical cohorts, not English language, or PAH not ascertained by RHC, 37 articles were eligible. A total of 43 factors for SSc-PAH were identified within seven categories. Several associations were due to PAH and risk factors such as dynpnea, right heart failure, and short 6-minute walk distance. Patient characteristics (14), pulmonary physiology (6), antibody profiles (6) and genetics/epigenetics (6) had the most numerous and diverse factors, while biomarkers (4) and other labs (2) features were infrequent. Low carbon monoxide (DLCO) (6), older age (4), longer disease duration (4), positive anticentromere antibodies (ACA) (4), telangiectasias (4), high brain natriuretic peptide (4) were frequent associations. CONCLUSIONS: Risk factors for SSc-PAH such as ACA, older age, longer disease duration limited cutaneous SSc subset and presence of ILD may enrich screening programs. Genes and other antibody profiles are inconsistent and requires further validation.


Subject(s)
Pulmonary Arterial Hypertension , Scleroderma, Systemic , Aging , Humans , Natriuretic Peptide, Brain , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/immunology , Risk Factors , Scleroderma, Systemic/genetics , Scleroderma, Systemic/immunology
18.
Biomed Pharmacother ; 129: 110355, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32559622

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease with limited effective therapies that can be lethal in the terminal stage. Since the universal use of traditional PAH-specific drugs affecting the nitric oxide pathway, endothelin pathway and prostacyclin pathway, major advances to treat this devastating condition have been made. For example, it has been recently demonstrated that PAH is associated with various dysfunctional genes and molecular signaling pathways, abnormal metabolism and endocrine systems, inflammation and immune dysfunction. Numerous descriptions of novel mechanisms in PAH have been presented, and some new drugs targeted to ease, reverse or cure PAH have been developed at the laboratory-level and/or tested in clinical trials. There are also some investigated drugs that have shown promising efficacy and safety in animal models but have failed in human beings. Inspiringly, immunotherapies also seem to be valuable therapies for PAH patients. In this review, we update the advances in investigational pharmacotherapy and immunotherapy of PAH. Hopefully this work can help promote more non-invasive treatment for PAH patients.


Subject(s)
Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Immunotherapy , Pulmonary Arterial Hypertension/therapy , Pulmonary Artery/drug effects , Animals , Antihypertensive Agents/adverse effects , Humans , Immunosuppressive Agents/therapeutic use , Immunotherapy/adverse effects , Molecular Targeted Therapy , Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/immunology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Signal Transduction , Vaccines/therapeutic use
19.
Aging (Albany NY) ; 12(6): 4953-4969, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32176619

ABSTRACT

Pulmonary arterial hypertension (PAH) is a serious disease characterized by elevated pulmonary artery pressure, inflammatory cell infiltration and pulmonary vascular remodeling. However, little is known about the pathogenic mechanisms underlying the disease onset and progression. RNA sequencing (RNA-seq) was used to identify the transcriptional profiling in control and rats injected with monocrotaline (MCT) for 1, 2, 3 and 4 weeks. A total of 23200 transcripts and 280, 1342, 908 and 3155 differentially expressed genes (DEGs) were identified at the end of week 1, 2, 3 and 4, of which Svop was the common top 10 DEGs over the course of PAH progression. Functional enrichment analysis of DEGs showed inflammatory/immune response occurred in the early stage of PAH development. KEGG pathway enrichment analysis of DEGs showed that cytokine-cytokine receptor interaction and neuroactive ligand-receptor interaction were in the initiation and progression of PAH. Further analysis revealed impaired expression of cholinergic receptors, adrenergic receptors including alpha1, beta1 and beta2 receptor, and dysregulated expression of γ-aminobutyric acid receptors. In summary, the dysregulated inflammation/immunity and neuroactive ligand receptor signaling pathways may be involved in the onset and progression of PAH.


Subject(s)
Chemokines/metabolism , Monocrotaline/administration & dosage , Pulmonary Arterial Hypertension/metabolism , Animals , Gene Expression , Inflammation/chemically induced , Inflammation/metabolism , Ligands , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/immunology , Rats, Sprague-Dawley , Sequence Analysis, RNA , Signal Transduction
20.
Am J Respir Crit Care Med ; 201(2): 224-239, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31545648

ABSTRACT

Rationale: Pulmonary hypertension (PH) is a life-threatening cardiopulmonary disorder in which inflammation and immunity have emerged as critical early pathogenic elements. Although proinflammatory processes in PH and pulmonary arterial hypertension (PAH) are the focus of extensive investigation, the initiating mechanisms remain elusive.Objectives: We tested whether activation of the complement cascade is critical in regulating proinflammatory and pro-proliferative processes in the initiation of experimental hypoxic PH and can serve as a prognostic biomarker of outcome in human PAH.Methods: We used immunostaining of lung tissues from experimental PH models and patients with PAH, analyses of genetic murine models lacking specific complement components or circulating immunoglobulins, cultured human pulmonary adventitial fibroblasts, and network medicine analysis of a biomarker risk panel from plasma of patients with PAH.Measurements and Main Results: Pulmonary perivascular-specific activation of the complement cascade was identified as a consistent critical determinant of PH and PAH in experimental animal models and humans. In experimental hypoxic PH, proinflammatory and pro-proliferative responses were dependent on complement (alternative pathway and component 5), and immunoglobulins, particularly IgG, were critical for activation of the complement cascade. We identified Csf2/GM-CSF as a primary complement-dependent inflammatory mediator. Furthermore, using network medicine analysis of a biomarker risk panel from plasma of patients with PAH, we demonstrated that complement signaling can serve as a prognostic factor for clinical outcome in PAH.Conclusions: This study establishes immunoglobulin-driven dysregulated complement activation as a critical pathobiological mechanism regulating proinflammatory and pro-proliferative processes in the initiation of experimental hypoxic PH and demonstrates complement signaling as a critical determinant of clinical outcome in PAH.


Subject(s)
Complement Activation/immunology , Fibroblasts/immunology , Hypertension, Pulmonary/immunology , Immunoglobulin G/immunology , Vascular Remodeling/immunology , Animals , Complement C3/immunology , Complement C5/immunology , Complement Factor B/immunology , Complement Pathway, Alternative/immunology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Hypertension, Pulmonary/etiology , Hypoxia/complications , Immunoglobulins/immunology , Inflammation , Mice , Mice, Knockout , Prognosis , Pulmonary Arterial Hypertension/immunology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...