Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Connect Tissue Res ; 65(2): 161-169, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38436275

RESUMO

OBJECTIVE: The COL1A1 proximal promoter contains two GC-rich regions and two inverted CCAAT boxes. The transcription factors Sp1 and CBF bind to the GC sequence at -122 to -115 bp and the inverted CCAAT box at -101 to -96 bp, respectively, and stimulate COL1A1 transcriptional activity. METHODS: To further define the regulatory mechanisms controlling COL1A1 expression by Sp1 and CBF, we introduced 2, 4, 6, or 8 thymidine nucleotides (T-tracts) at position -111 bp of the COL1A1 gene promoter to increase the physical distance between these two binding sites and examined in vitro the transcriptional activities of the resulting constructs and their response to TGF-ß1.`. RESULTS: Insertion of 2 or 4 nucleotides decreased COL1A1 promoter activity by up to 70%. Furthermore, the expected increase in COL1A1 transcription in response to TGF-ß1 was abolished. Computer modeling of the modified DNA structure indicated that increasing the physical distance between the Sp1 and CBF binding sites introduces a rotational change in the DNA topology that disrupts the alignment of Sp1 and CBF binding sites and likely alters protein-protein interactions among these transcription factors or their associated co-activators. CONCLUSION: The topology of the COL1A1 proximal promoter is crucial in determining the transcriptional activity of the gene and its response to the stimulatory effects of TGF-ß1.


Assuntos
Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/metabolismo , DNA , Nucleotídeos
2.
Curr Rheumatol Rev ; 2023 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-37921216

RESUMO

Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology with a highly complex pathogenesis that despite extensive investigation is not completely understood. The clinical and pathologic manifestations of the disease result from three distinct processes: 1) Severe and frequently progressive tissue fibrosis causing exaggerated and deleterious accumulation of interstitial collagens and other extracellular matrix molecules in the skin and various internal organs; 2) extensive fibroproliferative vascular lesions affecting small arteries and arterioles causing tissue ischemic alterations; and 3) cellular and humoral immunity abnormalities with the production of numerous autoantibodies, some with very high specificity for SSc. The fibrotic process in SSc is one of the main causes of disability and high mortality of the disease. Owing to its essentially universal presence and the severity of its clinical effects, the mechanisms involved in the development and progression of tissue fibrosis have been extensively investigated, however, despite intensive investigation, the precise molecular mechanisms have not been fully elucidated. Several recent studies have suggested that cellular transdifferentiation resulting in the phenotypic conversion of various cell types into activated myofibroblasts may be one important mechanism. Here, we review the potential role that cellular transdifferentiation may play in the development of severe and often progressive tissue fibrosis in SSc.

3.
Rheumatology (Oxford) ; 62(3): 999-1008, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35944210

RESUMO

SSc is a systemic autoimmune disease of unknown etiology characterized by frequently progressive cutaneous and internal organ fibrosis causing severe disability, organ failure and high mortality. A remarkable feature of SSc is the extension of the fibrotic alterations to nonaffected tissues. The mechanisms involved in the extension of fibrosis have remained elusive. We propose that this process is mediated by exosome microvesicles released from SSc-affected cells that induce an activated profibrotic phenotype in normal or nonaffected cells. Exosomes are secreted microvesicles involved in an intercellular communication system. Exosomes can transfer their macromolecular content to distant target cells and induce paracrine effects in the recipient cells, changing their molecular pathways and gene expression. Confirmation of this hypothesis may identify the molecular mechanisms responsible for extension of the SSc fibrotic process from affected cells to nonaffected cells and may allow the development of novel therapeutic approaches for the disease.


Assuntos
Exossomos , Escleroderma Sistêmico , Humanos , Fibrose , Fibroblastos/metabolismo , Fenótipo
4.
PLoS One ; 17(12): e0279461, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36548367

RESUMO

BACKGROUND: A major unmet need for Systemic Sclerosis (SSc) clinical management is the lack of biomarkers for the early diagnosis of patients with Raynaud's Phenomenon at high risk of evolving into SSc. OBJECTIVE: To identify proteins contained within serum exosomes employing an aptamer proteomic analysis that may serve to reveal patients with Raynaud's Phenomenon at risk of developing SSc. METHODS: Exosomes were isolated from serum samples from patients with Primary Raynaud's Phenomenon and from patients with Raynaud's Phenomenon harbouring serum antinuclear antibodies (ANA) who may be at high risk of evolving into SSc. The expression of 1,305 proteins was quantified using SOMAscan aptamer proteomics, and associations of the differentially elevated or reduced proteins with the clinical subsets of Raynaud's Phenomenon were assessed. RESULTS: Twenty one differentially elevated and one differentially reduced (absolute fold change >|1.3|) proteins were identified. Principal component analysis using these 22 most differentially expressed proteins resulted in excellent separation of the two Raynaud's Phenomenon clinical subsets. Remarkably, the most differentially elevated proteins are involved in enhanced inflammatory responses, immune cell activation and cell migration, and abnormal vascular functions. CONCLUSION: Aptamer proteomic analysis of circulating exosomes identified differentially elevated or reduced proteins between Raynaud's Phenomenon at high risk of evolving into SSc and Primary Raynaud's Phenomenon patients. Some of these proteins are involved in relevant biological pathways that may play a role in SSc pathogenesis including enhanced inflammatory responses, immune cell activation, and endothelial cell and vascular abnormalities.


Assuntos
Exossomos , Doença de Raynaud , Escleroderma Sistêmico , Humanos , Proteômica , Escleroderma Sistêmico/complicações , Biomarcadores
5.
J Clin Med ; 10(20)2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34682914

RESUMO

Numerous clinical and research investigations conducted during the last two decades have implicated excessive oxidative stress caused by high levels of reactive oxygen species (ROS) in the development of the severe and frequently progressive fibrotic process in Systemic Sclerosis (SSc). The role of excessive oxidative stress in SSc pathogenesis has been supported by the demonstration of increased levels of numerous biomarkers, indicative of cellular and molecular oxidative damage in serum, plasma, and other biological fluids from SSc patients, and by the demonstration of elevated production of ROS by various cell types involved in the SSc fibrotic process. However, the precise mechanisms mediating oxidative stress development in SSc and its pathogenetic effects have not been fully elucidated. The participation of the NADPH oxidase NOX4, has been suggested and experimentally supported by the demonstration that SSc dermal fibroblasts display constitutively increased NOX4 expression and that reduction or abrogation of NOX4 effects decreased ROS production and the expression of genes encoding fibrotic proteins. Furthermore, NOX4-stimulated ROS production may be involved in the development of certain endothelial and vascular abnormalities and may even participate in the generation of SSc-specific autoantibodies. Collectively, these observations suggest NOX4 as a novel therapeutic target for SSc.

6.
Sci Rep ; 11(1): 20435, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34650102

RESUMO

Myofibroblasts are the key effector cells responsible for the exaggerated tissue fibrosis in Systemic Sclerosis (SSc). Despite their importance to SSc pathogenesis, the specific transcriptome of SSc myofibroblasts has not been described. The purpose of this study was to identify transcriptome differences between SSc myofibroblasts and non-myofibroblastic cells. Alpha smooth muscle actin (α-SMA) expressing myofibroblasts and α-SMA negative cells were isolated employing laser capture microdissection from dermal cell cultures from four patients with diffuse SSc of recent onset. Total mRNA was extracted from both cell populations, amplified and analyzed employing microarrays. Results for specific genes were validated by Western blots and by immunohistochemistry. Transcriptome analysis revealed 97 differentially expressed transcripts in SSc myofibroblasts compared with non-myofibroblasts. Annotation clustering of the SSc myofibroblast-specific transcripts failed to show a TGF-ß signature. The most represented transcripts corresponded to several different genes from the Neuroblastoma Breakpoint Family (NBPF) of genes. NBPF genes are highly expanded in humans but are not present in murine or rat genomes. In vitro studies employing cultured SSc dermal fibroblasts and immunohistochemistry of affected SSc skin confirmed increased NBPF expression in SSc. These results indicate that SSc myofibroblasts represent a unique cell lineage expressing a specific transcriptome that includes very high levels of transcripts corresponding to numerous NBPF genes. Elevated expression of NBPF genes in SSc myofibroblasts suggests that NBPF gene products may play a role in SSc pathogenesis and may represent a novel therapeutic target.


Assuntos
Miofibroblastos/metabolismo , Proteínas de Neoplasias/genética , Escleroderma Sistêmico/metabolismo , Adulto , Western Blotting , Estudos de Casos e Controles , Feminino , Perfilação da Expressão Gênica , Genes Neoplásicos , Humanos , Microdissecção e Captura a Laser , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Escleroderma Sistêmico/genética , Pele/metabolismo , Fator de Crescimento Transformador beta/metabolismo
7.
Transl Res ; 231: 139-158, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33422651

RESUMO

Systemic sclerosis (SSc) is an idiopathic autoimmune disease with a heterogeneous clinical phenotype ranging from limited cutaneous involvement to rapidly progressive diffuse SSc. The most severe SSc clinical and pathologic manifestations result from an uncontrolled fibrotic process involving the skin and various internal organs. The molecular mechanisms responsible for the initiation and progression of the SSc fibrotic process have not been fully elucidated. Recently it has been suggested that tyrosine protein kinases play a role. The implicated kinases include receptor-activated tyrosine kinases and nonreceptor tyrosine kinases. The receptor kinases are activated following specific binding of growth factors (platelet-derived growth factor, fibroblast growth factor, or vascular endothelial growth factor). Other receptor kinases are the discoidin domain receptors activated by binding of various collagens, the ephrin receptors that are activated by ephrins and the angiopoetin-Tie-2s receptors. The nonreceptor tyrosine kinases c-Abl, Src, Janus, and STATs have also been shown to participate in SSc-associated tissue fibrosis. Currently, there are no effective disease-modifying therapies for SSc-associated tissue fibrosis. Therefore, extensive investigation has been conducted to examine whether tyrosine kinase inhibitors (TKIs) may exert antifibrotic effects. Here, we review the role of receptor and nonreceptor tyrosine kinases in the pathogenesis of the frequently progressive cutaneous and systemic fibrotic alterations in SSc, and the potential of TKIs as SSc disease-modifying antifibrotic therapeutic agents.


Assuntos
Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Escleroderma Sistêmico/enzimologia , Escleroderma Sistêmico/patologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Tirosina Quinases/genética , Escleroderma Sistêmico/tratamento farmacológico
8.
Clin Exp Rheumatol ; 39(6): 1298-1306, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33253099

RESUMO

OBJECTIVES: Systemic sclerosis (SSc) is characterised by severe fibroproliferative vasculopathy, fibrosis in skin and multiple internal organs, and humoral, cellular and innate immunity abnormalities. Vascular alterations are the earliest and most severe SSc manifestations, however, the mechanisms responsible have remained elusive. To investigate the molecular abnormalities involved in SSc-vasculopathy we examined global gene expression differences between highly purified lung microvascular endothelial cells (MVECs) from patients with SSc-interstitial lung disease (SSc-ILD) and normal lung MVECs. METHODS: MVECs were isolated from fresh transplanted lungs from patients with SSc-ILD. Sequential CD31 and CD102 immunopurification was performed to obtain highly purified CD31+/CD102+ lung MVECs. Global gene expression analysis was successfully performed in CD31+/CD102+ MVEC from two SSc-ILD patients and from two normal lungs. RT-PCR, Western blots, and indirect immunofluorescence validated the gene expression results. RESULTS: Numerous interferon-regulated genes (IRGs) including IFI44, IFI44L, IFI6, IFIH1, IFIT1, ISG-15, BST-2/Tetherin, and RSAD2/Viperin, genes encoding innate immunity antiviral responses (OAS1, OAS2, OAS3, OASL) and antiviral MX1 and MX2 proteins, and mesenchymal cell-specific genes were significantly overexpressed in CD31+/CD102+ SSc-ILD lung MVECs. CONCLUSIONS: Highly purified CD31+/CD102+ MVECs from lungs from SSc patients with end stage SSc-ILD displayed remarkable overexpression of numerous IRGs and of genes encoding antiviral innate immune response and antiviral proteins. These observations suggest that interferon-induced and antiviral response proteins may participate in the pathogenesis of SSc vasculopathy and SSc-ILD. The CD31+/CD102+ lung MVECs from SSc-ILD also showed elevated expression of mesenchymal cell-specific genes confirming the presence of endothelial to mesenchymal transition in SSc-ILD.


Assuntos
Fatores de Restrição Antivirais/genética , Interferons , Doenças Pulmonares Intersticiais , Escleroderma Sistêmico , Células Endoteliais , Humanos , Pulmão , Doenças Pulmonares Intersticiais/genética , Escleroderma Sistêmico/genética
9.
Semin Arthritis Rheum ; 50(6): 1226-1237, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33059296

RESUMO

Numerous drugs and chemical substances are capable of inducing exaggerated tissue fibrotic responses. The vast majority of these agents cause localized fibrotic tissue reactions or fibrosis confined to specific organs. Although much less frequent, chemically-induced systemic fibrotic disorders have been described, sometimes occurring as temporally confined outbreaks. These include the Toxic Oil Syndrome (TOS), the Eosinophilia-Myalgia Syndrome (EMS), and Nephrogenic Systemic Fibrosis (NSF). Although each of these disorders displays some unique characteristics, they all share crucial features with Systemic Sclerosis (SSc), the prototypic idiopathic systemic fibrotic disease, including vasculopathy, chronic inflammatory cell infiltration of affected tissues, and cutaneous and visceral tissue fibrosis. The study of the mechanisms and molecular alterations involved in the development of the chemically-induced systemic fibrotic disorders has provided valuable clues that may allow elucidation of SSc etiology and pathogenesis. Here, we review relevant aspects of the TOS, EMS, and NSF epidemic outbreaks of chemically-induced systemic fibrosing disorders that provide strong support to the hypothesis that SSc is caused by a toxic or biological agent that following its internalization by endothelial cells induces in genetically predisposed individuals a series of molecular alterations that result in the development of SSc clinical and pathological alterations.


Assuntos
Eosinofilia , Escleroderma Sistêmico , Células Endoteliais , Fibrose , Humanos , Escleroderma Sistêmico/induzido quimicamente , Pele
10.
Rheumatology (Oxford) ; 59(10): 3092-3098, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32442272

RESUMO

OBJECTIVE: SSc is a systemic fibrotic disease affecting skin, numerous internal organs and the microvasculature. The molecular pathogenesis of SSc tissue fibrosis has not been fully elucidated, although TGF-ß1 plays a crucial role. The Hic-5 protein encoded by the TGF-ß1-inducible HIC-5 gene participates in numerous TGF-ß-mediated pathways, however, the role of Hic-5 in SSc fibrosis has not been investigated. The aim of this study was to examine HIC-5 involvement in SSc tissue fibrosis. METHODS: Affected skin from three patients with diffuse SSc and dermal fibroblasts cultured from affected and non-affected SSc skin were examined for HIC-5 and COL1A1 gene expression. Real-time PCR, IF microscopy, western blotting and small interfering RNA-mediated HIC-5 were performed. RESULTS: HIC-5 and COL1A1 transcripts and Hic-5, type 1 collagen (COL1) and α-smooth muscle actin (α-SMA) protein levels were increased in clinically affected SSc skin compared with normal skin and in cultured dermal fibroblasts from affected SSc skin compared with non-affected skin fibroblasts from the same patients. HIC-5 knockdown caused a marked reduction of COL1 production in SSc dermal fibroblasts. CONCLUSION: HIC-5 expression is increased in affected SSc skin compared with skin from normal individuals. Affected SSc skin fibroblasts display increased HIC-5 and COL1A1 expression compared with non-affected skin fibroblasts from the same patients. Hic-5 protein was significantly increased in cultured SSc dermal fibroblasts. HIC-5 mRNA knockdown in SSc fibroblasts caused >50% reduction of COL1 production. Although these are preliminary results owing to the small number of skin samples studied, they indicate that Hic-5 plays a role in the profibrotic activation of SSc dermal fibroblasts and may represent a novel molecular target for antifibrotic therapy in SSc.


Assuntos
Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Escleroderma Sistêmico/metabolismo , Actinas/metabolismo , Colágeno/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/efeitos dos fármacos , Proteínas com Domínio LIM/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Escleroderma Sistêmico/genética , Pele/metabolismo , Fator de Crescimento Transformador beta/farmacologia
11.
Physiol Rev ; 99(2): 1281-1324, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30864875

RESUMO

Numerous studies have demonstrated that endothelial cells are capable of undergoing endothelial to mesenchymal transition (EndMT), a newly recognized type of cellular transdifferentiation. EndMT is a complex biological process in which endothelial cells adopt a mesenchymal phenotype displaying typical mesenchymal cell morphology and functions, including the acquisition of cellular motility and contractile properties. Endothelial cells undergoing EndMT lose the expression of endothelial cell-specific proteins such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and initiate the expression of mesenchymal cell-specific genes and the production of their encoded proteins including α-smooth muscle actin, extra domain A fibronectin, N-cadherin, vimentin, fibroblast specific protein-1, also known as S100A4 protein, and fibrillar type I and type III collagens. Transforming growth factor-ß1 is considered the main EndMT inducer. However, EndMT involves numerous molecular and signaling pathways that are triggered and modulated by multiple and often redundant mechanisms depending on the specific cellular context and on the physiological or pathological status of the cells. EndMT participates in highly important embryonic development processes, as well as in the pathogenesis of numerous genetically determined and acquired human diseases including malignant, vascular, inflammatory, and fibrotic disorders. Despite intensive investigation, many aspects of EndMT remain to be elucidated. The identification of molecules and regulatory pathways involved in EndMT and the discovery of specific EndMT inhibitors should provide novel therapeutic approaches for various human disorders mediated by EndMT.


Assuntos
Doença , Transição Epitelial-Mesenquimal/fisiologia , Animais , Desenvolvimento Embrionário , Transição Epitelial-Mesenquimal/genética , Humanos
12.
Clin Exp Rheumatol ; 36 Suppl 113(4): 36-44, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30277861

RESUMO

OBJECTIVES: To examine the effects of simultaneous inhibition of c-Abl and Src kinases on the gene expression and in vitro production of profibrotic molecules by dermal fibroblasts from patients with diffuse systemic sclerosis (SSc) of recent onset. METHODS: Dermal fibroblasts from normal individuals or from patients with diffuse cutaneous SSc fulfilling the American College of Rheumatology/EULAR SSc classification criteria were cultured in media containing increasing concentrations of the dual c-Abl and Src kinase inhibitor Bosutinib for 24 h. Total soluble collagen in cell culture supernatants was quantified. Western blots were performed for quantitative assessment of type I collagen, fibronectin, and α-smooth muscle actin (α-SMA) production. Quantitative PCR was performed to examine the effects of Bosutinib on the expression of profibrotic and TGF-ß-responsive genes in cultured SSc dermal fibroblasts. RESULTS: Simultaneous inhibition of c-Abl and Src kinases with Bosutinib reduced the expression of numerous fibrosis-associated genes including COL1A1, COL1A3, FN, and TGFß and the production of the corresponding proteins by SSc dermal fibroblasts. Bosutinib also decreased the transition of normal dermal fibroblasts into activated myofibroblasts induced by TGF-ß as evidenced by reduction of α-SMA in cell extracts from normal and SSc dermal fibroblasts. CONCLUSIONS: Simultaneous inhibition of c-Abl and Src kinases with Bosutinib abrogates the exaggerated expression of genes encoding fibrillar collagens, fibronectin, and TGF-ß-responsive genes and reduces type I collagen, fibronectin and α-SMA production by SSc dermal fibroblasts in vitro. Bosutinib also abrogates TGF- ß-induced transition of normal fibroblasts to activated myofibroblasts. These results indicate that inhibition of c-Abl and Src kinases activity may be an effective disease modifying antifibrotic therapeutic intervention for SSc.


Assuntos
Compostos de Anilina/farmacologia , Fibroblastos/efeitos dos fármacos , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Quinolinas/farmacologia , Esclerodermia Difusa/tratamento farmacológico , Pele/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores , Estudos de Casos e Controles , Células Cultivadas , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/enzimologia , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Esclerodermia Difusa/enzimologia , Esclerodermia Difusa/genética , Esclerodermia Difusa/patologia , Transdução de Sinais/efeitos dos fármacos , Pele/enzimologia , Pele/patologia , Quinases da Família src/metabolismo
13.
Nat Rev Rheumatol ; 14(7): 421-432, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29789665

RESUMO

The discovery and validation of biomarkers resulting from technological advances in the analysis of genomic, transcriptomic, lipidomic and metabolomic pathways involved in the pathogenesis of complex human diseases have led to the development of personalized and rationally designed approaches for the clinical management of such disorders. Although some of these approaches have been applied to systemic sclerosis (SSc), an unmet need remains for validated, non-invasive biomarkers to aid in the diagnosis of SSc, as well as in the assessment of disease progression and response to therapeutic interventions. Advances in global transcriptomic technology over the past 15 years have enabled the assessment of microRNAs that circulate in the blood of patients and the analysis of the macromolecular content of a diverse group of lipid bilayer membrane-enclosed extracellular vesicles, such as exosomes and other microvesicles, which are released by all cells into the extracellular space and circulation. Such advances have provided new opportunities for the discovery of biomarkers in SSc that could potentially be used to improve the design and evaluation of clinical trials and that will undoubtedly enable the development of personalized and individualized medicine for patients with SSc.


Assuntos
Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo , Escleroderma Sistêmico/diagnóstico , Biomarcadores/metabolismo , Ensaios Clínicos como Assunto , Progressão da Doença , Diagnóstico Precoce , Humanos , Medicina de Precisão , Escleroderma Sistêmico/sangue , Escleroderma Sistêmico/tratamento farmacológico , Resultado do Tratamento
14.
Rheumatology (Oxford) ; 57(10): 1698-1706, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29140474

RESUMO

There is an important unmet need for clinically validated non-invasive biomarkers for SSc diagnosis, assessment of disease activity, extent of internal organ involvement, therapeutic response and prognosis. There is also an unmet need for biomarkers to accurately differentiate primary RP from recent onset RP evolving into SSc. The lack of sensitive and specific biomarkers for SSc and SSc-associated RP is a limitation for the optimal clinical management of these patients. The development of highly sensitive and specific proteomic analysis employing aptamers and the expansion in the number of proteins that can be specifically identified by aptamer proteomics have opened new horizons for biomarker discovery. Here, we review the background and rationale for aptamer proteomic analysis for the identification of novel non-invasive biomarkers for SSc and recent onset RP evolving into SSc. Large scale application of aptamer proteomic platforms for this purpose will be of substantial value for the precision and personalized medical care of SSc patients. These studies will be placed in context by comparison with proteomic biomarker studies performed for other rheumatological inflammatory and autoimmune diseases.


Assuntos
Proteômica/métodos , Escleroderma Sistêmico/diagnóstico , Biomarcadores/análise , Humanos
15.
Nat Commun ; 8(1): 1060, 2017 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-29057934

RESUMO

Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of skin and internal organs. Protein tyrosine phosphatases have received little attention in the study of SSc or fibrosis. Here, we show that the tyrosine phosphatase PTP4A1 is highly expressed in fibroblasts from patients with SSc. PTP4A1 and its close homolog PTP4A2 are critical promoters of TGFß signaling in primary dermal fibroblasts and of bleomycin-induced fibrosis in vivo. PTP4A1 promotes TGFß signaling in human fibroblasts through enhancement of ERK activity, which stimulates SMAD3 expression and nuclear translocation. Upstream from ERK, we show that PTP4A1 directly interacts with SRC and inhibits SRC basal activation independently of its phosphatase activity. Unexpectedly, PTP4A2 minimally interacts with SRC and does not promote the SRC-ERK-SMAD3 pathway. Thus, in addition to defining PTP4A1 as a molecule of interest for TGFß-dependent fibrosis, our study provides information regarding the functional specificity of different members of the PTP4A subclass of phosphatases.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Tirosina Fosfatases/metabolismo , Escleroderma Sistêmico/enzimologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Linhagem Celular , Derme/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Escleroderma Sistêmico/metabolismo , Escleroderma Sistêmico/patologia , Proteína Smad3/metabolismo
16.
Methods Mol Biol ; 1627: 1-23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836191

RESUMO

Human fibrotic diseases constitute a major health problem worldwide owing to the large number of affected individuals, the incomplete knowledge of the fibrotic process pathogenesis, the marked heterogeneity in their etiology and clinical manifestations, the absence of appropriate and fully validated biomarkers, and, most importantly, the current void of effective disease-modifying therapeutic agents. The fibrotic disorders encompass a wide spectrum of clinical entities including systemic fibrotic diseases such as systemic sclerosis (SSc), sclerodermatous graft vs. host disease, and nephrogenic systemic fibrosis, as well as numerous organ-specific disorders including radiation-induced fibrosis and cardiac, pulmonary, liver, and kidney fibrosis. Although their causative mechanisms are quite diverse and in several instances have remained elusive, these diseases share the common feature of an uncontrolled and progressive accumulation of fibrotic tissue in affected organs causing their dysfunction and ultimate failure. Despite the remarkable heterogeneity in the etiologic mechanisms responsible for the development of fibrotic diseases and in their clinical manifestations, numerous studies have identified activated myofibroblasts as the common cellular element ultimately responsible for the replacement of normal tissues with nonfunctional fibrotic tissue. Critical signaling cascades, initiated primarily by transforming growth factor-ß (TGF-ß), but also involving numerous cytokines and signaling molecules which stimulate profibrotic reactions in myofibroblasts, offer potential therapeutic targets. Here, we briefly review the current knowledge of the molecular mechanisms involved in the development of tissue fibrosis and point out some of the most important challenges to research in the fibrotic diseases and to the development of effective therapeutic approaches for this often fatal group of disorders. Efforts to further clarify the complex pathogenetic mechanisms of the fibrotic process should be encouraged to attain the elusive goal of developing effective therapies for these serious, untreatable, and often fatal disorders.


Assuntos
Fibrose/etiologia , Fibrose/metabolismo , Animais , Biomarcadores , Proteínas Morfogenéticas Ósseas/metabolismo , Suscetibilidade a Doenças , Matriz Extracelular/metabolismo , Fibrose/diagnóstico , Fibrose/terapia , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Terapia de Alvo Molecular , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Estresse Oxidativo , Pesquisa , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
17.
Lab Invest ; 97(7): 806-818, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28346399

RESUMO

In this study, we tested the hypothesis that constitutive endothelial cell-specific activation of TGF-ß signaling induces tissue fibrosis and vasculopathy resembling the characteristic fibrotic and vascular alterations of systemic sclerosis. Transgenic mice with inducible expression of a constitutively active TGF-ß receptor I specifically in endothelial cells were generated by intercrossing mice harboring a constitutively active TGF-ß receptor I with a mouse strain containing the endothelial cell-specific Cdh5 gene promoter directing the tamoxifen-inducible expression of the Cre-ERT2 cassette. Administration of tamoxifen to these mice would result in constitutive TGF-ß activation and signaling confined to endothelial lineage cells. The effects of constitutive TGF-ß endothelial cell activation were assessed by histopathological examination of skin and various internal organs, tissue hydroxyproline analysis, and assessment of expression of myofibroblast differentiation and TGF-ß signaling genes employing real-time PCR and immunohistochemical staining of lung vessels for endothelial- and myofibroblast-specific proteins. Constitutive TGFß-1 signaling in endothelial cells resulted in cutaneous and visceral fibrosis with prominent fibrotic involvement of the lungs and severe perivascular and subendothelial fibrosis of small arterioles. A marked increase in the expression of fibrosis-associated genes and of genes indicative of myofibroblast activation was also found. Confocal microscopy of lung vessels showed evidence consistent with the induction of endothelial-to-mesenchymal transition (EndoMT). Taken together, our data indicate that transgenic mice with constitutive endothelial cell-specific activation of TGF-ß signaling display severe cutaneous, pulmonary, and microvascular fibrosis resembling the fibrotic and microvascular alterations characteristic of systemic sclerosis.


Assuntos
Células Endoteliais/metabolismo , Fibrose/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Hidroxiprolina , Imuno-Histoquímica , Pulmão/química , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Especificidade de Órgãos , Tamoxifeno , Fator de Crescimento Transformador beta/genética
18.
Clin Rheumatol ; 36(5): 1173-1178, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28220270

RESUMO

The levels of 63 cytokines, chemokines, and growth factors were measured in the serum of four patients with idiopathic morphea and of one patient with vitamin K1-induced morphea employing a multiplex assay to identify the role of inflammatory/immunologic events in their pathogenesis. Full-thickness skin biopsies of affected skin were analyzed by histopathology. Luminex assays for 63 cytokines, chemokines, and growth factors were performed in the sera from four patients with idiopathic morphea and in two different samples of serum obtained in two separate occasions from one patient with vitamin K1-induced morphea. The serum values of numerous inflammatory cytokines and growth factors including IL-2, IL-4, IL-6, and IFNß were markedly increased in the serum of patients with idiopathic morphea, whereas, these values were normal in the serum of the patient with vitamin K1-induced morphea. In contrast, serum eotaxin levels were greater than threefold higher in the patient with vitamin K1-induced morphea compared to patients with idiopathic morphea. The results demonstrated remarkable increases in the levels of numerous cytokines and chemokines in the serum samples of all patients with idiopathic morphea indicative of a prominent role of inflammatory/immunologic events in its pathogenesis. The results also showed statistically significant differences between idiopathic morphea and vitamin K1-induced morphea suggesting that their development involves different pathogenetic mechanisms.


Assuntos
Quimiocinas/sangue , Citocinas/sangue , Esclerodermia Localizada/sangue , Pele/patologia , Vitamina K 1/efeitos adversos , Idoso , Biomarcadores/sangue , Biópsia , Feminino , Humanos , Injeções Intramusculares , Esclerodermia Localizada/etiologia , Esclerodermia Localizada/patologia , Pele/efeitos dos fármacos , Vitamina K 1/administração & dosagem , Vitaminas/administração & dosagem , Vitaminas/efeitos adversos
19.
Clin Exp Rheumatol ; 35 Suppl 106(4): 21-30, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28094758

RESUMO

OBJECTIVES: Exosomes are lipid bilayer-bound microvesicles containing various macromolecules including numerous microRNA (miRNA). Exosomes mediate intercellular communication by fusing and releasing their macromolecular content into target cells. Here, we analysed the content of profibrotic and antifibrotic miRNAs in exosomes isolated from the serum of systemic sclerosis (SSc) patients and tested their ability to induce a profibrotic phenotype in normal human dermal fibroblasts in vitro. METHODS: Exosomes were isolated from serum from patients with limited cutaneous or diffuse cutaneous SSc and were characterised by Nanosight Particle Tracking Analysis, exosome antibody arrays, and transmission electron microscopy. The content of nine profibrotic and eighteen antifibrotic miRNA was assessed in the isolated exosomes by semiquantitative real time PCR. The effects of the isolated exosomes on cultured normal human dermal fibroblasts were assessed by real time PCR and Western blotting. RESULTS: The isolated serum exosomes displayed the expected exosome size and morphology and contained characteristic exosome proteins. Six profibrotic miRNAs were increased and ten antifibrotic miRNAs were decreased in SSc serum exosomes compared to normal serum exosomes. The levels of eight miRNA were significantly different between exosomes from limited and diffuse SSc. Exosomes isolated from both limited or diffuse SSc patients caused dose-dependent stimulation of profibrotic gene expression and type I collagen and fibronectin production and secretion in normal human dermal fibroblasts in vitro. CONCLUSIONS: Serum exosomes from SSc patients contain miRNA displaying a markedly profibrotic profile and induce a profibrotic phenotype in target normal fibroblasts in vitro suggesting a plausible mechanism for the extension of the fibrotic SSc process to non-affected tissues.


Assuntos
Exossomos/metabolismo , MicroRNAs/análise , Escleroderma Sistêmico/patologia , Adulto , Células Cultivadas , Colágeno Tipo I/genética , Exossomos/ultraestrutura , Feminino , Fibroblastos/metabolismo , Fibronectinas/biossíntese , Fibrose , Humanos , Pessoa de Meia-Idade , Fenótipo , Escleroderma Sistêmico/sangue , Pele/citologia , Pele/metabolismo
20.
J Clin Med ; 5(4)2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-27077889

RESUMO

Fibrotic diseases encompass a wide spectrum of clinical entities including systemic fibrotic diseases such as systemic sclerosis, sclerodermatous graft versus host disease, nephrogenic systemic fibrosis, and IgG4-associated sclerosing disease, as well as numerous organ-specific disorders including radiation-induced fibrosis, and cardiac, pulmonary, liver, and kidney fibrosis. Although their causative mechanisms are quite diverse, these diseases share the common feature of an uncontrolled and progressive accumulation of fibrous tissue macromolecules in affected organs leading to their dysfunction and ultimate failure. The pathogenesis of fibrotic diseases is complex and despite extensive investigation has remained elusive. Numerous studies have identified myofibroblasts as the cells responsible for the establishment and progression of the fibrotic process. Tissue myofibroblasts in fibrotic diseases originate from several sources including quiescent tissue fibroblasts, circulating CD34+ fibrocytes, and the phenotypic conversion of various cell types including epithelial and endothelial cells into activated myofibroblasts. However, the role of the phenotypic transition of endothelial cells into mesenchymal cells (Endothelial to Mesenchymal Transition or EndoMT) in the pathogenesis of fibrotic disorders has not been fully elucidated. Here, we review the evidence supporting EndoMT's contribution to human fibrotic disease pathogenesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA