Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.762
1.
Curr Rheumatol Rev ; 20(3): 304-316, 2024.
Article En | MEDLINE | ID: mdl-38807470

INTRODUCTION: The "scleroderma" type capillaroscopic pattern is a reference pattern in rheumatology that is a diagnostic sign for systemic sclerosis (SSc) in an appropriate clinical context and is observed in more than 90% of scleroderma patients. Similar microvascular changes, the so-called "scleroderma-like", have been described albeit in a lower proportion of patients with other rheumatic diseases, such as dermatomyositis (DM), undifferentiated connective tissue diseases (UCTD), systemic lupus erythematosus (SLE), etc. Three distinct stages of "scleroderma" pattern have been suggested by Cutolo et al., i.e., "early", "active", and "late". However, disease duration is just one of the factors that contributes to the progression of microvascular changes, and in this regard, "active" or even "late" pattern could be observed in patients with shorter disease duration. In addition, stable microvascular changes could be found for long periods in other cases. OBJECTIVE: The aim of the study was to assess the presence of differentiating features between "scleroderma" pattern in SSc and "scleroderma-like" pattern in other rheumatic diseases. METHODS: 684 capillaroscopic images demonstrating a "scleroderma" and "scleroderma-like" pattern have been analysed in the current retrospective cross-sectional study. 479 capillaroscopic pictures were obtained from 50 SSc patients, 105 from 7 DM patients, 38 from 10 rheumatoid arthritis (RA) patients, 36 images from 5 patients with SLE, and 26 images from 9 patients with UCTD. All capillaroscopic images used in the current analysis have fulfilled the criteria for "sclerderma/scleroderma-like" pattern, as the pathological changes in the capillaroscopic parameters have also been confirmed by quantitative measurement of capillary diameters, capillary density, and intercapillary distance. All the images have been categorized into one of the following groups, i.e., "early", "active" and "late" phases (according to the definition of Cutolo et al.), or "other" findings, the latter being specifically described as they could not be attributed to one of the other three categories. RESULTS: 479 capillaroscopic pictures were obtained from 50 scleroderma patients. 31 of them showed an "early", 391 an "active" phase, and 57 a "late" phase "scleroderma" type microangiopathy. In 69 images assessed as an "active" pattern, neoangiogenesis was found. In 43 out of 105 capillaroscopic pictures from DM patients, an "active" phase was detected; in 2 of the images, a "late" pattern was found, and in 60 capillaroscopic pictures, neoangiogenesis in combination with giant capillary loops was observed. Early microangiopathy was not found in this group. Among capillaroscopic images from SLE patients, "late" phase microangiopathy was not found. "Early" phase was present in 3 images, "active" phase in 29, neoangiogenesis in "active" phase in 4 pictures. Early microangiopathy was detected in 11 capillaroscopic pictures from RA patients (8 out of 9 patients), an "active" phase in 4 images (3 patients), and in 23 capillaroscopic images, neoangiogenesis with mild capillary derangement and capillary loss and single giant capillaries ("rheumatoid neoangiogenic pattern") were observed. Classic "late" type microangiopathy was not found in RA patients as well as among patients with UCTD. The predominant capillaroscopic pattern in UCTD patients was early microangiopathy (n = 23). The rest images from UCTD exhibited features of the "active" phase. CONCLUSION: In conclusion, early microangiopathy was observed in RA, SLE, and UCTD patients, but not in patients with DM. An "active" phase "scleroderma" type capillaroscopic pattern was detected in all patient groups other than SSc, i.e., DM, SLE, RA, and UCTD. "Late" phase "scleroderma" type microangiopathy was present in patients with scleroderma and DM and was not observed in SLE, RA, and UCTD. Despite the fact that in some cases, microangiopathy in scleroderma and other rheumatic diseases may be indistinguishable, the results of the current research have shown the presence of some differentiating features between "scleroderma" and "scleroderma-like" microangiopathy that might be a morphological phenomenon associated with differences in the pathogenesis and the degree of microvascular pathology in various rheumatic diseases.


Microscopic Angioscopy , Scleroderma, Systemic , Humans , Microscopic Angioscopy/methods , Scleroderma, Systemic/diagnostic imaging , Scleroderma, Systemic/pathology , Male , Female , Middle Aged , Adult , Aged , Retrospective Studies
2.
J Clin Invest ; 134(10)2024 Mar 26.
Article En | MEDLINE | ID: mdl-38747285

Transforming growth factor ß (TGF-ß) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-ß remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-ß in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-ß. The activation of latent TGF-ß requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-ß, rebalanced TGF-ß signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-ß in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.


Fibroblasts , Fibrosis , Transforming Growth Factor beta , Wnt-5a Protein , rho-Associated Kinases , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Animals , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Mice , Humans , Fibroblasts/metabolism , Fibroblasts/pathology , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Scleroderma, Systemic/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/genetics , Mice, Knockout , Wnt Proteins/metabolism , Wnt Proteins/genetics , MAP Kinase Signaling System , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/genetics
3.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38731946

Systemic sclerosis (SSc) is a heterogeneous disease characterized by autoimmunity, vasculopathy, and fibrosis which affects the skin and internal organs. One key aspect of SSc vasculopathy is pulmonary arterial hypertension (SSc-PAH) which represents a leading cause of morbidity and mortality in patients with SSc. The pathogenesis of pulmonary hypertension is complex, with multiple vascular cell types, inflammation, and intracellular signaling pathways contributing to vascular pathology and remodeling. In this review, we focus on shared molecular features of pulmonary hypertension and those which make SSc-PAH a unique entity. We highlight advances in the understanding of the clinical and translational science pertinent to this disease. We first review clinical presentations and phenotypes, pathology, and novel biomarkers, and then highlight relevant animal models, key cellular and molecular pathways in pathogenesis, and explore emerging treatment strategies in SSc-PAH.


Pulmonary Arterial Hypertension , Scleroderma, Systemic , Humans , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology , Animals , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/metabolism , Biomarkers , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Disease Models, Animal , Translational Research, Biomedical , Signal Transduction
4.
Arthritis Res Ther ; 26(1): 94, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702742

BACKGROUND: Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular injury and inflammation, followed by excessive fibrosis of the skin and other internal organs, including the lungs. CX3CL1 (fractalkine), a chemokine expressed on endothelial cells, supports the migration of macrophages and T cells that express its specific receptor CX3CR1 into targeted tissues. We previously reported that anti-CX3CL1 monoclonal antibody (mAb) treatment significantly inhibited transforming growth factor (TGF)-ß1-induced expression of type I collagen and fibronectin 1 in human dermal fibroblasts. Additionally, anti-mouse CX3CL1 mAb efficiently suppressed skin inflammation and fibrosis in bleomycin- and growth factor-induced SSc mouse models. However, further studies using different mouse models of the complex immunopathology of SSc are required before the initiation of a clinical trial of CX3CL1 inhibitors for human SSc. METHODS: To assess the preclinical utility and functional mechanism of anti-CX3CL1 mAb therapy in skin and lung fibrosis, a sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse model was analyzed with immunohistochemical staining for characteristic infiltrating cells and RNA sequencing assays. RESULTS: On day 42 after bone marrow transplantation, Scl-cGVHD mice showed increased serum CX3CL1 level. Intraperitoneal administration of anti-CX3CL1 mAb inhibited the development of fibrosis in the skin and lungs of Scl-cGVHD model, and did not result in any apparent adverse events. The therapeutic effects were correlated with the number of tissue-infiltrating inflammatory cells and α-smooth muscle actin (α-SMA)-positive myofibroblasts. RNA sequencing analysis of the fibrotic skin demonstrated that cGVHD-dependent induction of gene sets associated with macrophage-related inflammation and fibrosis was significantly downregulated by mAb treatment. In the process of fibrosis, mAb treatment reduced cGVHD-induced infiltration of macrophages and T cells in the skin and lungs, especially those expressing CX3CR1. CONCLUSIONS: Together with our previous findings in other SSc mouse models, the current results indicated that anti-CX3CL1 mAb therapy could be a rational therapeutic approach for fibrotic disorders, such as human SSc and Scl-cGVHD.


Antibodies, Monoclonal , Chemokine CX3CL1 , Disease Models, Animal , Graft vs Host Disease , Pulmonary Fibrosis , Scleroderma, Systemic , Skin , Animals , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/pathology , Scleroderma, Systemic/immunology , Mice , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Skin/pathology , Skin/drug effects , Skin/metabolism , Skin/immunology , Fibrosis , Female , Mice, Inbred C57BL , Humans , Lung/pathology , Lung/drug effects , Lung/metabolism , Lung/immunology
5.
JCI Insight ; 9(10)2024 Apr 23.
Article En | MEDLINE | ID: mdl-38652537

NKX2-5 is a member of the homeobox-containing transcription factors critical in regulating tissue differentiation in development. Here, we report a role for NKX2-5 in vascular smooth muscle cell phenotypic modulation in vitro and in vascular remodeling in vivo. NKX2-5 is upregulated in scleroderma patients with pulmonary arterial hypertension. Suppression of NKX2-5 expression in smooth muscle cells halted vascular smooth muscle proliferation and migration, enhanced contractility, and blocked the expression of extracellular matrix genes. Conversely, overexpression of NKX2-5 suppressed the expression of contractile genes (ACTA2, TAGLN, CNN1) and enhanced the expression of matrix genes (COL1) in vascular smooth muscle cells. In vivo, conditional deletion of NKX2-5 attenuated blood vessel remodeling and halted the progression to hypertension in a mouse chronic hypoxia model. This study revealed that signals related to injury such as serum and low confluence, which induce NKX2-5 expression in cultured cells, is potentiated by TGF-ß and further enhanced by hypoxia. The effect of TGF-ß was sensitive to ERK5 and PI3K inhibition. Our data suggest a pivotal role for NKX2-5 in the phenotypic modulation of smooth muscle cells during pathological vascular remodeling and provide proof of concept for therapeutic targeting of NKX2-5 in vasculopathies.


Homeobox Protein Nkx-2.5 , Muscle, Smooth, Vascular , Vascular Remodeling , Animals , Mice , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Humans , Vascular Remodeling/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Male , Scleroderma, Systemic/pathology , Scleroderma, Systemic/complications , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/etiology , Female , Transforming Growth Factor beta/metabolism , Disease Models, Animal , Cell Proliferation/genetics , Middle Aged , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology
6.
In Vivo ; 38(3): 1451-1453, 2024.
Article En | MEDLINE | ID: mdl-38688588

BACKGROUND: Immunotherapy using immune checkpoint inhibitors is not devoid of immune-related adverse events (irAEs) including rheumatological conditions. CASE REPORT: We report a rare case of a 47-year-old woman with metastatic melanoma who developed systemic scleroderma after initiating nivolumab. The patient displayed inflammatory arthralgias, morning stiffness, and classical cutaneous manifestations of the disease. Clinical evaluations also revealed carpal tunnel syndrome, cardiac involvement, and dyspnea. RNA-Polymerase III antibodies were positive. Nivolumab, an anti-PD-1 antibody, was considered as a potential trigger for this condition. CONCLUSION: To our knowledge, this is the first case of nivolumab-induced systemic scleroderma in the context of melanoma described in the literature that fulfills the classification criteria of the disease. This case underscores the need for increased awareness of immune-related adverse events in patients receiving immune checkpoint inhibitors, emphasizing timely intervention and further research.


Melanoma , Nivolumab , Scleroderma, Systemic , Humans , Nivolumab/adverse effects , Melanoma/drug therapy , Melanoma/pathology , Female , Middle Aged , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/pathology , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Antineoplastic Agents, Immunological/adverse effects
7.
Nat Commun ; 15(1): 3302, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658535

Uncontrolled secretion of ECM proteins, such as collagen, can lead to excessive scarring and fibrosis and compromise tissue function. Despite the widespread occurrence of fibrotic diseases and scarring, effective therapies are lacking. A promising approach would be to limit the amount of collagen released from hyperactive fibroblasts. We have designed membrane permeant peptide inhibitors that specifically target the primary interface between TANGO1 and cTAGE5, an interaction that is required for collagen export from endoplasmic reticulum exit sites (ERES). Application of the peptide inhibitors leads to reduced TANGO1 and cTAGE5 protein levels and a corresponding inhibition in the secretion of several ECM components, including collagens. Peptide inhibitor treatment in zebrafish results in altered tissue architecture and reduced granulation tissue formation during cutaneous wound healing. The inhibitors reduce secretion of several ECM proteins, including collagens, fibrillin and fibronectin in human dermal fibroblasts and in cells obtained from patients with a generalized fibrotic disease (scleroderma). Taken together, targeted interference of the TANGO1-cTAGE5 binding interface could enable therapeutic modulation of ERES function in ECM hypersecretion, during wound healing and fibrotic processes.


Cicatrix , Collagen , Fibroblasts , Wound Healing , Zebrafish , Humans , Animals , Fibroblasts/metabolism , Fibroblasts/drug effects , Collagen/metabolism , Wound Healing/drug effects , Cicatrix/metabolism , Cicatrix/pathology , Cicatrix/drug therapy , Skin/metabolism , Skin/pathology , Skin/drug effects , Fibrosis , Peptides/pharmacology , Peptides/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects
8.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L736-L753, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38651940

Systemic sclerosis (SSc) with interstitial lung disease (SSc-ILD) lacks curative pharmacological treatments, thus necessitating effective animal models for candidate drug discovery. Existing bleomycin (BLM)-induced SSc-ILD mouse models feature spatially limited pulmonary fibrosis, spontaneously resolving after 28 days. Here, we present an alternative BLM administration approach in female C57BL/6 mice, combining oropharyngeal aspiration (OA) and subcutaneous mini-pump delivery (pump) of BLM to induce a sustained and more persistent fibrosis, while retaining stable skin fibrosis. A dose-finding study was performed with BLM administered as 10 µg (OA) +80 mg/kg (pump) (10 + 80), 10 + 100, and 15 + 100. Forty-two days after OA, micro-computed tomography (micro-CT) imaging and histomorphometric analyses showed that the 10 + 100 and 15 + 100 treatments induced significant alterations in lung micro-CT-derived readouts, Ashcroft score, and more severe fibrosis grades compared with saline controls. In addition, a marked reduction in hypodermal thickness was observed in the 15 + 100 group. A time-course characterization of the BLM 15 + 100 treatment at days 28, 35, and 42, including longitudinal micro-CT imaging, revealed progressing alterations in lung parameters. Lung histology highlighted a sustained fibrosis accompanied by a reduction in hypodermis thickness throughout the explored time-window, with a time-dependent increase in fibrotic biomarkers detected by immunofluorescence analysis. BLM-induced alterations were partly mitigated by Nintedanib treatment. Our optimized BLM delivery approach leads to extensive and persistent lung fibrotic lesions coupled with cutaneous fibrotic alterations: it thus represents a significant advance compared with current preclinical models of BLM-induced SSc-ILD.NEW & NOTEWORTHY This study introduces an innovative approach to enhance the overall performance of the mouse bleomycin (BLM)-induced model for systemic sclerosis with interstitial lung disease (SSc-ILD). By combining oropharyngeal aspiration and subcutaneous mini-pump delivery of BLM, our improved model leads to sustained lung fibrosis and stable skin fibrosis in female C57BL/6 mice. The optimized 15 + 100 treatment results in extensive and persistent lung fibrotic lesions and thus represents a significant improvement over existing preclinical models of BLM-induced SSc-ILD.


Bleomycin , Disease Models, Animal , Mice, Inbred C57BL , Pulmonary Fibrosis , Animals , Bleomycin/administration & dosage , Bleomycin/toxicity , Female , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Mice , Scleroderma, Systemic/pathology , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/complications , X-Ray Microtomography , Skin/pathology , Skin/drug effects , Lung/pathology , Lung/drug effects , Lung/diagnostic imaging , Oropharynx/pathology , Oropharynx/drug effects , Oropharynx/diagnostic imaging , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/chemically induced , Lung Diseases, Interstitial/diagnostic imaging
9.
Article Zh | MEDLINE | ID: mdl-38664034

Skin fibrosis diseases mainly include hypertrophic scar, keloid, and systemic sclerosis, etc. The main pathological features are excessive activation of fibroblasts and abnormal deposition of extracellular matrix. In recent years, studies have shown that aerobic glycolysis is closely related to the occurrence and development of skin fibrosis diseases. Drugs targeting aerobic glycolysis has provided new ideas for skin anti-fibrosis treatment. This article reviews the role of enzymes and products related to aerobic glycolysis in the occurrence and development of skin fibrosis diseases and the drugs targeting aerobic glycolysis for the treatment of skin fibrosis diseases.


Fibrosis , Glycolysis , Humans , Fibrosis/metabolism , Fibrosis/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Skin Diseases/drug therapy , Skin/pathology , Skin/metabolism , Keloid/metabolism , Keloid/pathology , Keloid/drug therapy , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/drug therapy
10.
Int J Mol Sci ; 25(5)2024 Feb 28.
Article En | MEDLINE | ID: mdl-38474040

Two-dimensional in vitro cultures have represented a milestone in biomedical and pharmacological research. However, they cannot replicate the architecture and interactions of in vivo tissues. Moreover, ethical issues regarding the use of animals have triggered strategies alternative to animal models. The development of three-dimensional (3D) models offers a relevant tool to investigate disease pathogenesis and treatment, modeling in vitro the in vivo environment. We aimed to develop a dynamic 3D in vitro model for culturing human endothelial cells (ECs) and skin fibroblasts, simulating the structure of the tissues mainly affected in systemic sclerosis (SSc), a prototypical autoimmune fibrotic vasculopathy. Dermal fibroblasts and umbilical vein ECs grown in scaffold or hydrogel, respectively, were housed in bioreactors under flow. Fibroblasts formed a tissue-like texture with the deposition of a new extracellular matrix (ECM) and ECs assembled tube-shaped structures with cell polarization. The fine-tuned dynamic modular system allowing 3D fibroblast/EC culture connection represents a valuable model of the in vivo interplay between the main players in fibrotic vasculopathy as SSc. This model can lead to a more accurate study of the disease's pathogenesis, avoiding the use of animals, and to the development of novel therapies, possibly resulting in improved patient management.


Scleroderma, Systemic , Vascular Diseases , Animals , Humans , Endothelial Cells/pathology , Skin/pathology , Scleroderma, Systemic/pathology , Fibrosis , Vascular Diseases/pathology , Fibroblasts/pathology , Cells, Cultured
11.
Int J Mol Sci ; 25(6)2024 Mar 09.
Article En | MEDLINE | ID: mdl-38542130

Systemic Sclerosis (SSc) is a heterogeneous autoimmune disease characterized by widespread vasculopathy, the presence of autoantibodies and the progressive fibrosis of skin and visceral organs. There are still many questions about its pathogenesis, particularly related to the complex regulation of the fibrotic process, and to the factors that trigger its onset. Our recent studies supported a key role of N-formyl peptide receptors (FPRs) and their crosstalk with uPAR in the fibrotic phase of the disease. Here, we found that dermal fibroblasts acquire a proliferative phenotype after the activation of FPRs and their interaction with uPAR, leading to both Rac1 and ERK activation, c-Myc phosphorylation and Cyclin D1 upregulation which drive cell cycle progression. The comparison between normal and SSc fibroblasts reveals that SSc fibroblasts exhibit a higher proliferative rate than healthy control, suggesting that an altered fibroblast proliferation could contribute to the initiation and progression of the fibrotic process. Finally, a synthetic compound targeting the FPRs/uPAR interaction significantly inhibits SSc fibroblast proliferation, paving the way for the development of new targeted therapies in fibrotic diseases.


Receptors, Formyl Peptide , Scleroderma, Systemic , Humans , Receptors, Formyl Peptide/metabolism , Scleroderma, Systemic/pathology , Fibrosis , Fibroblasts/metabolism , Autoantibodies/metabolism , Skin/metabolism , Cells, Cultured
12.
Circ Res ; 134(7): 875-891, 2024 Mar 29.
Article En | MEDLINE | ID: mdl-38440901

BACKGROUND: Systemic sclerosis (SSc) is a connective tissue disease that can serve as a model to study vascular changes in response to inflammation, autoimmunity, and fibrotic remodeling. Although microvascular changes are the earliest histopathologic manifestation of SSc, the vascular pathophysiology remains poorly understood. METHODS: We applied spatial proteomic approaches to deconvolute the heterogeneity of vascular cells at the single-cell level in situ and characterize cellular alterations of the vascular niches of patients with SSc. Skin biopsies of patients with SSc and control individuals were analyzed by imaging mass cytometry, yielding a total of 90 755 cells including 2987 endothelial cells and 4096 immune cells. RESULTS: We identified 7 different subpopulations of blood vascular endothelial cells (VECs), 2 subpopulations of lymphatic endothelial cells, and 3 subpopulations of pericytes. A novel population of CD34+;αSMA+ (α-smooth muscle actin);CD31+ VECs was more common in SSc, whereas endothelial precursor cells were decreased. Co-detection by indexing and tyramide signal amplification confirmed these findings. The microenvironment of CD34+;αSMA+;CD31+ VECs was enriched for immune cells and myofibroblasts, and CD34+;αSMA+;CD31+ VECs expressed markers of endothelial-to-mesenchymal transition. The density of CD34+;αSMA+;CD31+ VECs was associated with clinical progression of fibrosis in SSc. CONCLUSIONS: Using spatial proteomics, we unraveled the heterogeneity of vascular cells in control individuals and patients with SSc. We identified CD34+;αSMA+;CD31+ VECs as a novel endothelial cell population that is increased in patients with SSc, expresses markers for endothelial-to-mesenchymal transition, and is located in close proximity to immune cells and myofibroblasts. CD34+;αSMA+;CD31+ VEC counts were associated with clinical outcomes of progressive fibrotic remodeling, thus providing a novel cellular correlate for the crosstalk of vasculopathy and fibrosis.


Endothelial Progenitor Cells , Scleroderma, Systemic , Humans , Proteomics , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology , Fibrosis , Myofibroblasts/pathology
13.
Clin Rheumatol ; 43(5): 1703-1709, 2024 May.
Article En | MEDLINE | ID: mdl-38509242

INTRODUCTION: Mixed connective tissue disease (MCTD) is a rare systemic disease characterized by overlapping features of systemic lupus erythematosus (SLE), systemic sclerosis (SSc), dermato-/polymyositis (DM/PM), and rheumatoid arthritis (RA). Naifold capillaroscopy (NFC) is a non-invasive test for evaluating the capillaries of the nail shaft used in the diagnosis of rheumatic diseases. OBJECTIVES: To determine whether there are characteristic abnormalities in NFC in MCTD patients, and whether the type of NFC lesions correlates with organ involvement in these patients. METHODS: Clinical picture and NFC patterns were analyzed in 43 patients with MCTD. Capillaroscopic images were divided into scleroderma-like pattern (SD-like pattern) according to the Cutolo classification, non-specific lesions, and normal images. Relationships between the clinical aspects considered in the MCTD classification criteria and the changes in the capillaroscopic images were evaluated. RESULTS: SD-like pattern was present in 20 MCTD patients (46.51%) with a predominance of the "early" pattern. Giant, branched, dilated capillaries and reduced capillary density were found more frequently in MCTD patients compared to the control group (p-values 0.0005, 0.005, 0.02, < 0.0001 respectively). There were associations found between the presence of a reduced number of vessels, avascular areas, and SD-like pattern with the presence of sclerodactyly in MCTD patients (p = 0.002, p = 0.006, p = 0.02, respectively), alongside an association between the presence of branched vessels and the subpapillary plexus with pulmonary arterial hypertension (PAH) (p = 0.04 and p = 0.005, respectively). CONCLUSIONS: MCTD patients are significantly more likely to have abnormalities upon NFC. It is worthwhile to perform capillaroscopic examination in MCTD patients. Key Points • Scleroderma-like pattern was found in more than half of the MCTD patients. • Reduced capillary density was found to be a significant predictor of the diagnosis of MCTD. • There were relationships between the presence of reduced capillary density, avascular areas, and SD-like with the presence of sclerodactyly in the MCTD patients. • There was an association between the presence of branched vessels and the visibility of the subpapillary plexus and pulmonary arterial hypertension (PAH).


Lupus Erythematosus, Systemic , Mixed Connective Tissue Disease , Pulmonary Arterial Hypertension , Scleroderma, Localized , Scleroderma, Systemic , Humans , Microscopic Angioscopy/methods , Mixed Connective Tissue Disease/diagnostic imaging , Mixed Connective Tissue Disease/pathology , Capillaries/diagnostic imaging , Capillaries/pathology , Lupus Erythematosus, Systemic/pathology , Scleroderma, Systemic/diagnostic imaging , Scleroderma, Systemic/pathology , Scleroderma, Localized/pathology
14.
Medicina (Kaunas) ; 60(3)2024 Mar 18.
Article En | MEDLINE | ID: mdl-38541223

Both scleroderma and immunoglobulin G4-related disease (IgG4-RD) are systemic fibro-inflammatory diseases characterised by lymphoplasmacytic infiltrates. IgG4-RD and systemic sclerosis (SSc) may share common pathophysiological mechanisms, but no examples of co-occurrence of the diseases have been found. Autologous haematopoietic stem cell transplantation (AHSCT) is implemented in selected rapidly progressive SSc with a high risk of organ failure. However, existing guidelines are based on clinical trials that do not represent the entire patient population and exclude critically ill patients with no therapeutic alternatives. Examples of AHSCT in IgG4-RD are absent. We report the case of a 44-year-old female patient with overlapping progressive diffuse SSc and sinonasal IgG4-RD. After 11 years of ineffective SSc treatment, AHSCT was performed. The 63-month follow-up showed a regression of SSc symptoms. AHSCT was not intended as treatment in the case of IgG4RD, although the first symptoms of the disease developed before transplantation. The sinus lesions progressed after AHSCT and remained indolent only after surgical treatment (bilateral ethmoidectomy, sphenoidotomy, intranasal buccal antrostomy), which allowed histopathological confirmation of IgG4-RD.


Hematopoietic Stem Cell Transplantation , Immunoglobulin G4-Related Disease , Scleroderma, Systemic , Female , Humans , Adult , Scleroderma, Systemic/complications , Scleroderma, Systemic/therapy , Scleroderma, Systemic/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Transplantation, Autologous
15.
Reumatismo ; 76(1)2024 Mar 22.
Article En | MEDLINE | ID: mdl-38523580

OBJECTIVE: Scleroderma, or systemic sclerosis (SSc), is a chronic autoimmune connective disease with an unknown etiology and poorly understood pathogenesis. The striking array of autoimmune, vascular, and fibrotic changes that develop in almost all patients makes SSc unique among connective tissue diseases. Although no animal model developed for SSc to date fully represents all features of human disease, some animal models that demonstrate features of SSc may help to better understand the pathogenesis of the disease and to develop new therapeutic options. In this review, we aimed to evaluate skin fibrosis and lung involvement in a bleomycin (BLM)-induced mouse model and to evaluate the differences between studies. METHODS: A systematic literature review (PRISMA guideline) on PubMed and EMBASE (until May 2023, without limits) was performed. A primary literature search was conducted using the PubMed and EMBASE databases for all articles published from 1990 to May 2023. Review articles, human studies, and non-dermatological studies were excluded. Of the 38 non-duplicated studies, 20 articles were included. RESULTS: Among inducible animal models, the BLM-induced SSc is still the most widely used. In recent years, the measurement of tissue thickness between the epidermal-dermal junction and the dermal-adipose tissue junction (dermal layer) has become more widely accepted. CONCLUSIONS: In animal studies, it is important to simultaneously evaluate lung tissues in addition to skin fibrosis induced in mice by subcutaneous BLM application, following the 3R (replacement, reduction, and refinement) principle to avoid cruelty to animals.


Pulmonary Fibrosis , Scleroderma, Systemic , Skin Diseases , Humans , Animals , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Bleomycin/toxicity , Skin/pathology , Fibrosis , Scleroderma, Systemic/pathology , Skin Diseases/pathology , Disease Models, Animal
16.
Matrix Biol ; 128: 31-38, 2024 Apr.
Article En | MEDLINE | ID: mdl-38423396

The largest mammalian organ, skin, consisting of a dermal connective tissue layer that underlies and supports the epidermis, acts as a protective barrier that excludes external pathogens and disseminates sensory signals emanating from the local microenvironment. Dermal connective tissue is comprised of a collagen-rich extracellular matrix (ECM) that is produced by connective tissue fibroblasts resident within the dermis. When wounded, a tissue repair program is induced whereby fibroblasts, in response to alterations in the microenvironment, produce new ECM components, resulting in the formation of a scar. Failure to terminate the normal tissue repair program causes fibrotic conditions including: hypertrophic scars, keloids, and the systemic autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). Histological and single-cell RNA sequencing (scRNAseq) studies have revealed that fibroblasts are heterogeneous and highly plastic. Understanding how this diversity contributes to dermal homeostasis, wounding, fibrosis, and cancer may ultimately result in novel anti-fibrotic therapies and personalized medicine. This review summarizes studies supporting this concept.


Cicatrix, Hypertrophic , Scleroderma, Systemic , Animals , Epidermis/pathology , Fibroblasts/pathology , Fibrosis , Mammals , Scleroderma, Systemic/genetics , Scleroderma, Systemic/pathology , Skin/pathology
17.
Zhonghua Nei Ke Za Zhi ; 63(2): 176-182, 2024 Feb 01.
Article Zh | MEDLINE | ID: mdl-38326044

Objectives: Primary cardiac involvement (SSc-PHI) in systemic sclerosis is an important prognostic factor. We aimed to characterize and identify subclinical SSc-PHI using cardiovascular MRI to determine whether disease severity and serum biomarkers are associated with subclinical SSc-PHI. Methods: A total of 26 patients with SSc who had no history of cardiovascular disease or pulmonary hypertension underwent 3 T-enhanced cardiovascular MRI. Measurements included native T1, extracellular volume, advanced gadolinium enhancement, T2 mapping, and left ventricular volume function. Troponin T and N telencephalic natriuretic peptide precursors were also determined. Results: LGE was observed in 13 of 26 patients (50.0%), suggesting focal fibrosis, and T2 mapping was significantly higher in the dcSSc group than in the lcSSc group (P=0.009). Left ventricular volume and function were within the normal range in all patients, but final systolic left ventricular volume was significantly higher in dcSSc than in lcSSc (P=0.021). The modified Rodnan skin score (mRSS) was significantly higher in patients with LGE focal fibrosis (P=0.019). Logistic regression analysis confirmed the association between mRSS and LGE (OR=1.224, P=0.037). In multivariate analysis, T2 mapping was negatively correlated with disease course, and was correlated with dcSSc and fingertip ulcer (R2=0.711, P=0.018, P=0.013, P=0.030). Troponin T was correlated with T2 mapping (r=0.555, P=0.049). Conclusions: Subclinical SSc-PHI is characterized by diffuse and focal myocardial fibrosis, but preserves myocardial systolic function. Subclinical SSC-Phi is associated with TNT, SSc disease severity, and complex peripheral vascular disease. These data provide information for identifying individuals at risk of SSc-PHI.


Contrast Media , Scleroderma, Systemic , Humans , Troponin T , Gadolinium , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology , Fibrosis , Magnetic Resonance Imaging , Myocardium/pathology
18.
Clin Rheumatol ; 43(2): 733-741, 2024 Feb.
Article En | MEDLINE | ID: mdl-38190091

INTRODUCTION: Juvenile Sjögren's disease (jSjD) is a rare autoimmune disease characterized by exocrine gland involvement and systemic manifestations, including small vessel vasculitis and Raynaud's phenomenon (RP). We aimed to investigate the microvascular status in jSjD patients by nailfold videocapillaroscopy (NVC) and the potential correlations with clinical and serological features. METHODS: Clinical data from thirteen consecutive jSjD patients (11 females and 2 males), with a mean age of 16 ± 4 years, diagnosed before 16 years of age (mean age at diagnosis 12 ± 3) according to the 2016 American College of Rheumatology/EULAR criteria for adult SjD, were collected including age- and sex-matched healthy controls (HCs). Clinical, laboratory, and instrumental data were collected, together with NVC examination. Non-specific and specific NVC parameters were investigated, such as capillary density, capillary dilations, giant capillaries, microhaemorrhages and abnormal shapes. Associations between NVC findings and clinical/serological features were explored and analysed using parametrical and non-parametrical tests. RESULTS: Capillary density reduction correlated significantly with articular involvement (arthralgias) (p = 0.024). Microhaemorrhages correlated with lower C3 levels (p = 0.034). No specific NVC pattern for jSjD was identified, whereas abnormal capillary shapes were significantly higher in jSjD patients than HCs (p = 0.005). NVC abnormalities were not associated with SjD-specific instrumental tests (biopsy, imaging, Schirmer's test). RP was present in 8% of jSjD patients. CONCLUSIONS: The reduction of capillary density, as well as microhaemorrhages at NVC analysis, are significantly associated with some clinical aspects like articular involvement and serum biomarkers (C3 reduction). The NVC is suggested as safe and further analysis in jSjD patients.


Autoimmune Diseases , Raynaud Disease , Scleroderma, Systemic , Sjogren's Syndrome , Male , Adult , Female , Humans , Child , Adolescent , Young Adult , Microscopic Angioscopy/methods , Nails/blood supply , Capillaries/diagnostic imaging , Capillaries/pathology , Autoimmune Diseases/pathology , Sjogren's Syndrome/diagnostic imaging , Sjogren's Syndrome/pathology , Raynaud Disease/pathology , Scleroderma, Systemic/pathology
19.
Nat Commun ; 15(1): 210, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38172207

Systemic sclerosis (SSc) is a devastating autoimmune disease characterized by excessive production and accumulation of extracellular matrix, leading to fibrosis of skin and other internal organs. However, the main cellular participants in SSc skin fibrosis remain incompletely understood. Here using differentiation trajectories at a single cell level, we demonstrate a dual source of extracellular matrix deposition in SSc skin from both myofibroblasts and endothelial-to-mesenchymal-transitioning cells (EndoMT). We further define a central role of Hippo pathway effectors in differentiation and homeostasis of myofibroblast and EndoMT, respectively, and show that myofibroblasts and EndoMTs function as central communication hubs that drive key pro-fibrotic signaling pathways in SSc. Together, our data help characterize myofibroblast differentiation and EndoMT phenotypes in SSc skin, and hint that modulation of the Hippo pathway may contribute in reversing the pro-fibrotic phenotypes in myofibroblasts and EndoMTs.


Hippo Signaling Pathway , Scleroderma, Systemic , Humans , Fibrosis , Scleroderma, Systemic/pathology , Myofibroblasts/metabolism , Endothelial Cells/metabolism , Skin/pathology , Fibroblasts/metabolism
20.
Camb Q Healthc Ethics ; 33(1): 135-136, 2024 Jan.
Article En | MEDLINE | ID: mdl-37605983

"Scleroderma," the rheumatologist said after examining my stiff swollen arms and legs. "Unfortunately, given your biomarkers, it's likely to get worse before it gets better, but you never know." She gave a quick rundown of what I might expect-rapidly progressive skin and joint tightening, GI symptoms, high likelihood of multi-organ involvement…. "Let's hope for the best." She paused, then asked if I had any questions.


Scleroderma, Systemic , Humans , Scleroderma, Systemic/diagnosis , Scleroderma, Systemic/pathology
...