Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Proc Natl Acad Sci U S A ; 113(27): E3901-10, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27298374

RESUMO

Treatment for fibrosis represents a critical unmet need, because fibrosis is the leading cause of death in industrialized countries, and there is no effective therapy to counteract the fibrotic process. The development of fibrosis relates to the interplay between vessel injury, immune cell activation, and fibroblast stimulation, which can occur in various tissues. Immunotherapies have provided a breakthrough in the treatment of immune diseases. The glycoprotein OX40-OX40 ligand (OX40L) axis offers the advantage of a targeted approach to costimulatory signals with limited impact on the whole immune response. Using systemic sclerosis (SSc) as a prototypic disease, we report compelling evidence that blockade of OX40L is a promising strategy for the treatment of inflammation-driven fibrosis. OX40L is overexpressed in the fibrotic skin and serum of patients with SSc, particularly in patients with diffuse cutaneous forms. Soluble OX40L was identified as a promising serum biomarker to predict the worsening of lung and skin fibrosis, highlighting the role of this pathway in fibrosis. In vivo, OX40L blockade prevents inflammation-driven skin, lung, and vessel fibrosis and induces the regression of established dermal fibrosis in different complementary mouse models. OX40L exerts potent profibrotic effects by promoting the infiltration of inflammatory cells into lesional tissues and therefore the release of proinflammatory mediators, thereafter leading to fibroblast activation.


Assuntos
Ligante OX40/antagonistas & inibidores , Ligante OX40/sangue , Fibrose Pulmonar/prevenção & controle , Escleroderma Sistêmico/sangue , Pele/patologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Biomarcadores/sangue , Bleomicina , Estudos de Casos e Controles , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Fibrose , Antígeno 2 Relacionado a Fos/genética , Humanos , Hipertensão Pulmonar/prevenção & controle , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Fibrose Pulmonar/genética , Escleroderma Sistêmico/tratamento farmacológico , Pele/metabolismo , Fator de Transcrição AP-1/metabolismo
3.
Ann Rheum Dis ; 75(12): 2175-2183, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26961294

RESUMO

BACKGROUND: The pathogenesis of systemic sclerosis (SSc) involves a distinctive triad of autoimmune, vascular and inflammatory alterations resulting in fibrosis. Evidence suggests that peroxisome proliferator-activated receptors (PPARs) play an important role in SSc-related fibrosis and their agonists may become effective therapeutic targets. OBJECTIVE: To determine the expression of PPARs in human fibrotic skin and investigate the effects of IVA337, a pan PPAR agonist, in in vitro and in vivo models of fibrosis. METHODS: The antifibrotic effects of IVA337 were studied using a bleomycin-induced mouse model of dermal fibrosis. The in vivo effect of IVA337 on wound closure and inflammation were studied using an excisional model of wound healing. RESULTS: Low levels of PPARα and PPARγ were detected in the skin of patients with SSc compared with controls. In mice, IVA337 was associated with decreased extracellular matrix (ECM) deposition and reduced expression of phosphorylated SMAD2/3-intracellular effector of transforming growth factor (TGF)-ß1. Although the antifibrotic effect of pan PPAR was similar to that of PPARγ agonist alone, a significant downregulation of several markers of inflammation was associated with IVA337. Despite its anti-inflammatory and antifibrotic properties, IVA337 did not interfere with wound closure. In vitro effects of IVA337 included attenuation of transcription of ECM genes and alteration of canonical and non-canonical TGF-ß signalling pathways. CONCLUSIONS: These findings indicate that simultaneous activation of all three PPAR isoforms exerts a dampening effect on inflammation and fibrosis, making IVA337 a potentially effective therapeutic candidate in the treatment of fibrotic diseases including SSc.


Assuntos
Anti-Inflamatórios/farmacologia , Benzotiazóis/farmacologia , Fármacos Dermatológicos/farmacologia , PPAR alfa/agonistas , PPAR gama/agonistas , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/patologia , Sulfonamidas/farmacologia , Animais , Bleomicina , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Fibrose , Humanos , Camundongos , PPAR alfa/metabolismo , PPAR gama/metabolismo , Escleroderma Sistêmico/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Cicatrização/efeitos dos fármacos
4.
Ann Rheum Dis ; 75(12): 2142-2149, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26912566

RESUMO

OBJECTIVE: Activated T cells are the main component of the inflammatory skin infiltrates that characterise systemic sclerosis (SSc). Our aim was to investigate the efficacy of abatacept, which tempers T-cell activation, in reducing skin fibrosis in complementary mouse models of SSc. METHODS: The antifibrotic properties of abatacept were evaluated in the mouse models of bleomycin-induced dermal fibrosis and sclerodermatous chronic graft-versus-host disease, reflecting early and inflammatory stages of SSc. Thereafter, we studied the efficacy of abatacept in tight skin (Tsk-1) mice, an inflammation-independent mouse model of skin fibrosis. RESULTS: Abatacept efficiently prevented bleomycin-induced skin fibrosis and was also effective in the treatment of established fibrosis. In this model, abatacept decreased total and activated T-cell, B-cell and monocyte infiltration in the lesional skin. Abatacept did not protect CB17-SCID mice from the development of bleomycin-induced dermal fibrosis, which supports that T cells are necessary to drive the antifibrotic effects of abatacept. Upon bleomycin injections, skin interleukin (IL) 6 and IL-10 levels were significantly reduced upon abatacept treatment. Moreover, treatment with abatacept ameliorated fibrosis in the chronic graft-versus-host disease model, but demonstrated no efficacy in Tsk-1 mice. The tolerance of abatacept was excellent in the three mouse models. CONCLUSIONS: Using complementary models, we demonstrate that inhibition of T-cell activation by abatacept can prevent and induce the regression of inflammation-driven dermal fibrosis. Translation to human disease is now required, and targeting early and inflammatory stages of SSc sounds the most appropriate for positioning abatacept in SSc.


Assuntos
Abatacepte/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/patologia , Dermatopatias/patologia , Dermatopatias/prevenção & controle , Linfócitos T/efeitos dos fármacos , Animais , Linfócitos B/efeitos dos fármacos , Bleomicina , Modelos Animais de Doenças , Fibrose , Doença Enxerto-Hospedeiro/tratamento farmacológico , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Camundongos , Escleroderma Sistêmico/induzido quimicamente , Dermatopatias/induzido quimicamente
5.
Mediators Inflamm ; 2015: 415024, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26640324

RESUMO

Primary myelofibrosis (PMF) is a clonal myeloproliferative neoplasm where severity as well as treatment complexity is mainly attributed to a long lasting disease and presence of bone marrow stroma alterations as evidenced by myelofibrosis, neoangiogenesis, and osteosclerosis. While recent understanding of mutations role in hematopoietic cells provides an explanation for pathological myeloproliferation, functional involvement of stromal cells in the disease pathogenesis remains poorly understood. The current dogma is that stromal changes are secondary to the cytokine "storm" produced by the hematopoietic clone cells. However, despite therapies targeting the myeloproliferation-sustaining clones, PMF is still regarded as an incurable disease except for patients, who are successful recipients of allogeneic stem cell transplantation. Although the clinical benefits of these inhibitors have been correlated with a marked reduction in serum proinflammatory cytokines produced by the hematopoietic clones, further demonstrating the importance of inflammation in the pathological process, these treatments do not address the role of the altered bone marrow stroma in the pathological process. In this review, we propose hypotheses suggesting that the stroma is inflammatory-imprinted by clonal hematopoietic cells up to a point where it becomes "independent" of hematopoietic cell stimulation, resulting in an inflammatory vicious circle requiring combined stroma targeted therapies.


Assuntos
Medula Óssea/fisiologia , Inflamação/complicações , Mielofibrose Primária/etiologia , Metilação de DNA , Mineração de Dados , Células-Tronco Hematopoéticas/fisiologia , Humanos , Células Estromais/fisiologia , Fator de Crescimento Transformador beta/fisiologia
6.
Arthritis Res Ther ; 17: 145, 2015 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-26025306

RESUMO

INTRODUCTION: Systemic sclerosis (SSc) is a connective tissue disorder characterised by the development of skin fibrosis. Our current understanding of the disease pathogenesis is incomplete and the study of SSc is hindered, at least partially, by a lack of animal models that fully replicate the complex state of human disease. Murine model of bleomycin-induced dermal fibrosis encapsulates important events that take place early in the disease course. METHODS: To characterise the optimum in vivo parameters required for the successful induction of dermal fibrosis we subjected three commonly used mouse strains to repeated subcutaneous bleomycin injections. We aimed to identify the effects of genetic background and gender on the severity of skin fibrosis. We used male and female Balb/C, C57BL/6, and DBA/2 strains and assessed their susceptibility to bleomycin-induced fibrosis by measuring dermal thickness, hydroxyproline/collagen content and number of resident myofibroblasts, all of which are important indicators of the severity of skin fibrosis. All data are expressed as mean values ± SEM. The Mann-Whitney U test was used for statistical analysis with GraphPad Prism 6.04 software. RESULTS: Dermal fibrosis was most severe in Balb/C mice compared to C57BL/6 and DBA/2 suggesting that Balb/C mice are more susceptible to bleomycin-induced fibrosis. Analysis of the effect of gender on the severity of fibrosis showed that male Balb/C, C57BL/6, DBA/2 mice had a tendency to develop more pronounced fibrosis phenotype than female mice. Of potential importance, male Balb/C mice developed the most severe fibrosis phenotype compared to male C57BL/6 and male DBA/2 as indicated by significantly increased number of dermal myofibroblasts. CONCLUSION: Our study highlights the importance of genetic background and gender in the induction of murine dermal fibrosis. Robust and reproducible animal models of fibrosis are important research tools used in pharmacological studies which may lead to better understanding of the pathogenesis of fibrotic diseases and assist in identification of new drugs.


Assuntos
Bleomicina/efeitos adversos , Patrimônio Genético , Dermatopatias/genética , Dermatopatias/patologia , Animais , Biópsia por Agulha , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fibrose/induzido quimicamente , Fibrose/patologia , Imuno-Histoquímica , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Distribuição Aleatória , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/patologia , Fatores Sexuais , Dermatopatias/induzido quimicamente , Especificidade da Espécie , Estatísticas não Paramétricas
7.
Arthritis Res Ther ; 17: 71, 2015 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-25880423

RESUMO

INTRODUCTION: Systemic sclerosis (SSc) and primary biliary cirrhosis (PBC) are rare polygenic autoimmune diseases (AIDs) characterized by fibroblast dysfunction. Furthermore, both diseases share some genetic bases with other AIDs, as evidenced by autoimmune gene pleiotropism. The present study was undertaken to investigate whether single-nucleotide polymorphisms (SNPs) identified by a large genome-wide association study (GWAS) in PBC might contribute to SSc susceptibility. METHODS: Sixteen PBC susceptibility SNPs were genotyped in a total of 1,616 patients with SSc and 3,621 healthy controls from two European populations (France and Italy). RESULTS: We observed an association between PLCL2 rs1372072 (odds ratio (OR) = 1.22, 95% confidence interval (CI) 1.12 to 1.33, P adj = 7.22 × 10(-5)), nuclear factor-kappa-B (NF-κB) rs7665090 (OR = 1.15, 95% CI 1.06 to 1.25, P adj = 0.01), and IRF8 rs11117432 (OR = 0.75, 95% CI 0.67 to 0.86, P adj = 2.49 × 10(-4)) with SSc susceptibility. Furthermore, phenotype stratification showed an association between rs1372072 and rs11117432 with the limited cutaneous subgroup (lcSSc) (P adj = 4.45 × 10(-4) and P adj = 0.001), whereas rs7665090 was associated with the diffuse cutaneous subtype (dcSSc) (P adj = 0.003). Genotype-mRNA expression correlation analysis revealed that the IRF8 protective allele was associated with increased interferon-gamma (IFN-γ) expression (P = 0.03) in patients with SSc but decreased type I IFN (IFIT1) expression in patients and controls (P = 0.02). In addition, we found an epistatic interaction between NF-κB and IRF8 (OR = 0.56, 95% CI 0.00 to 0.74, P = 4 × 10(-4)) which in turn revealed that the IRF8 protective effect is dependent on the presence of the NF-κB susceptibility allele. CONCLUSIONS: An analysis of pleiotropic genes identified two new susceptibility genes for SSc (NF-κB and PLCL2) and confirmed the IRF8 locus. Furthermore, the IRF8 variant influenced the IFN signature, and we found an interaction between IRF8 and NF-κB gene variants that might play a role in SSc susceptibility.


Assuntos
Predisposição Genética para Doença/genética , Fatores Reguladores de Interferon/genética , Interferon gama/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , NF-kappa B/genética , Escleroderma Sistêmico/genética , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Fatores Reguladores de Interferon/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Masculino , Pessoa de Meia-Idade , NF-kappa B/biossíntese , Polimorfismo de Nucleotídeo Único/genética , Escleroderma Sistêmico/patologia
8.
Arthritis Rheumatol ; 66(11): 3140-50, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25155144

RESUMO

OBJECTIVE: Nuclear receptors regulate cell growth, differentiation, and homeostasis. Selective nuclear receptors promote fibroblast activation, which leads to tissue fibrosis, the hallmark of systemic sclerosis (SSc). This study was undertaken to investigate the effects of constitutive androstane receptor (CAR)/NR1I3, an orphan nuclear receptor, on fibroblast activation and experimental dermal fibrosis. METHODS: CAR expression was quantified by quantitative polymerase chain reaction, Western blotting, immunohistochemistry, and immunofluorescence. CAR expression was modulated by small molecules, small interfering RNA, forced overexpression, and site-directed mutagenesis. The effects of CAR activation were analyzed in cultured fibroblasts, in bleomycin-induced dermal fibrosis, and in mice overexpressing a constitutively active transforming growth factor ß (TGFß) receptor type I (TßRI-CA). RESULTS: Up-regulation of CAR was detected in the skin and in dermal fibroblasts in SSc patients. Stimulation of healthy fibroblasts with TGFß induced the expression of CAR messenger RNA and protein in a Smad-dependent manner. Pharmacologic activation or overexpression of CAR in healthy fibroblasts significantly increased the stimulatory effects of TGFß on collagen synthesis and myofibroblast differentiation, and amplified the stimulatory effects of TGFß on COL1A2 transcription activity. Treatment with CAR agonist increased the activation of canonical TGFß signaling in murine models of SSc and exacerbated bleomycin-induced and TßRI-CA-induced fibrosis with increased dermal thickening, myofibroblast counts, and collagen accumulation. CONCLUSION: Our findings indicate that CAR is up-regulated in SSc and regulates TGFß signaling. Activation of CAR increases the profibrotic effects of TGFß in cultured fibroblasts and in different preclinical models of SSc. Thus, inactivation of CAR might be a novel approach to target aberrant TGFß signaling in SSc and in other fibrotic diseases.


Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Escleroderma Sistêmico/metabolismo , Pele/metabolismo , Pele/patologia , Fator de Crescimento Transformador beta/metabolismo , Adulto , Idoso , Animais , Bleomicina/efeitos adversos , Células Cultivadas , Colágeno Tipo I/metabolismo , Receptor Constitutivo de Androstano , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/induzido quimicamente , Fibrose/metabolismo , Fibrose/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Escleroderma Sistêmico/patologia , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
9.
Diabetologia ; 57(2): 402-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24292564

RESUMO

AIMS/HYPOTHESIS: Skin lesions and ulcerations are severe complications of diabetes that often result in leg amputations. In this study we investigated the function of the cytoskeletal protein flightless I (FLII) in diabetic wound healing. We hypothesised that overexpression of FLII would have a negative effect on diabetic wound closure and modulation of this protein using specific FLII-neutralising antibodies (FnAb) would enhance cellular proliferation, migration and angiogenesis within the diabetic wound. METHODS: Using a streptozotocin-induced model of diabetes we investigated the effect of altered FLII levels through Flii genetic knockdown, overexpression or treatment with FnAb on wound healing. Diabetic wounds were assessed using histology, immunohistochemistry and biochemical analysis. In vitro and in vivo assays of angiogenesis were used to assess the angiogenic response. RESULTS: FLII levels were elevated in the wounds of both diabetic mice and humans. Reduction in the level of FLII improved healing of murine diabetic wounds and promoted a robust pro-angiogenic response with significantly elevated von Willebrand factor (vWF) and vascular endothelial growth factor (VEGF)-positive endothelial cell infiltration. Diabetic mouse wounds treated intradermally with FnAb showed improved healing and a significantly increased rate of re-epithelialisation. FnAb improved the angiogenic response through enhanced formation of capillary tubes and functional neovasculature. Reducing the level of FLII led to increased numbers of mature blood vessels, increased recruitment of smooth muscle actin-α-positive cells and improved tight junction formation. CONCLUSIONS/INTERPRETATION: Reducing the level of FLII in a wound may be a potential therapeutic approach for the treatment of diabetic foot ulcers.


Assuntos
Proteínas do Citoesqueleto/farmacologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Angiopatias Diabéticas/patologia , Pele/patologia , Cicatrização/imunologia , Indutores da Angiogênese , Animais , Anticorpos Neutralizantes/metabolismo , Proteínas de Transporte , Proliferação de Células , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Angiopatias Diabéticas/imunologia , Feminino , Humanos , Imuno-Histoquímica , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos , Pele/lesões , Transativadores , Úlcera/patologia
10.
Biomed Res Int ; 2013: 389792, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555084

RESUMO

Impaired wound healing and ulceration represent a serious complication of both type 1 and type 2 diabetes. Cytoskeletal protein Flightless I (Flii) is an important inhibitor of wound repair, and reduced Flii gene expression in fibroblasts increased migration, proliferation, and adhesion. As such it has the ability to influence all phases of wound healing including inflammation, remodelling and angiogenesis. Flii has the potential to modulate inflammation through its interaction with MyD88 which it an adaptor protein for TLR4. To assess the effect of Flii on the inflammatory response of diabetic wounds, we used a murine model of streptozocin-induced diabetes and Flii genetic mice. Increased levels of Flii were detected in Flii transgenic murine wounds resulting in impaired healing which was exacerbated when diabetes was induced. When Flii levels were reduced in diabetic wounds of Flii-deficient mice, healing was improved and decreased levels of TLR4 were observed. In contrast, increasing the level of Flii in diabetic mouse wounds led to increased TLR4 and NF- κ B production. Treatment of murine diabetic wounds with neutralising antibodies to Flii led to an improvement in healing with decreased expression of TLR4. Decreasing the level of Flii in diabetic wounds may therefore reduce the inflammatory response and improve healing.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Complicações do Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Proteínas de Transporte , Proteínas do Citoesqueleto/genética , Complicações do Diabetes/genética , Complicações do Diabetes/patologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/metabolismo , Camundongos , Proteínas dos Microfilamentos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Receptor 4 Toll-Like/genética , Transativadores , Cicatrização/genética , Ferimentos e Lesões/induzido quimicamente
11.
J Invest Dermatol ; 133(4): 1008-16, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23223144

RESUMO

Epidermolysis bullosa (EB) is a chronic inheritable disease that leads to severe blistering and fibrosis. Previous studies have shown that the actin cytoskeletal protein flightless I (Flii) impairs wound healing associated with EB. Using a mouse model of EB acquisita (EBA), the effect of "mopping up" Flii using Flii-neutralizing antibodies (FnAbs) before, during, and after blister formation was determined. FnAbs, incorporated into a cream vehicle and applied topically to the skin, penetrated into the basal epidermis and upper papillary dermis but were not detected in serum or other organs and did not alter neutrophil or macrophage infiltration into the blistered skin. Histological assessment of blister severity showed that treatment of early-stage blisters with FnAb cream reduced their severity and improved their rate of healing. Treatment of established blisters with FnAb cream also improved healing and restored the skin's tensile strength toward that of normal skin. Repeated application of FnAbs to EBA skin before the onset of blistering reduced the severity of skin blistering. Independent of when the FnAbs were applied, skin barrier function and wound healing were improved and skin fragility was reduced, suggesting that FnAbs could potentially improve healing of patients with EB.


Assuntos
Anticorpos Neutralizantes/farmacologia , Proteínas do Citoesqueleto/imunologia , Epidermólise Bolhosa Adquirida/tratamento farmacológico , Epidermólise Bolhosa Adquirida/imunologia , Cicatrização/imunologia , Administração Tópica , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/imunologia , Vesícula/tratamento farmacológico , Vesícula/imunologia , Vesícula/patologia , Proteínas de Transporte , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Epidermólise Bolhosa Adquirida/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos , Índice de Gravidade de Doença , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Creme para a Pele/administração & dosagem , Creme para a Pele/farmacologia , Sus scrofa , Resistência à Tração/fisiologia , Transativadores , Cicatrização/efeitos dos fármacos
12.
Eur J Dermatol ; 22(6): 740-50, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23178274

RESUMO

Chronic non-healing wounds form a medical need which will expand as the population ages and the obesity epidemic grows. Whilst the complex mechanisms underlying wound repair are not fully understood, remodelling of the actin cytoskeleton plays a critical role. Elevated expression of the actin cytoskeletal protein Flightless I (Flii) is known to impair wound outcomes. To determine if Flii is involved in the impaired healing observed in chronic wounds, its expression in non-healing human wounds from patients with venous leg ulcers was determined and compared to its expression in acute wounds and unwounded skin. Increased expression of Flii was observed in both chronic and acute wounds with wound fluid and plasma also containing secreted Flii protein. Inflammation is a key aspect of wound repair and fluorescence-activated cell sorting (FACS) analysis revealed Flii was located in neutrophils within the blood and that it co-localised with CD16+ neutrophils in chronic wounds. The function of secreted Flii was investigated as both chronic wound fluid and Flii have previously been shown to inhibit fibroblast proliferation. To determine if the inhibitory effect of wound fluid was due in part to the presence of Flii, wound fluids were depleted of Flii using Flii-specific neutralizing antibodies (FnAb). Flii depleted chronic wound fluid no longer inhibited fibroblast proliferation, suggesting that Flii may contribute to the inhibitory effect of chronic wound fluid on fibroblast function. Application of FnAbs to chronic wounds may therefore be a novel approach used to improve the local environment of non-healing wounds and potentially improve healing outcomes.


Assuntos
Úlcera da Perna/metabolismo , Proteínas dos Microfilamentos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Cicatrização/fisiologia , Ferimentos e Lesões/metabolismo , Adulto , Idoso de 80 Anos ou mais , Análise de Variância , Anticorpos Neutralizantes/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Doença Crônica , Feminino , Fibroblastos , Humanos , Úlcera da Perna/patologia , Masculino , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/sangue , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/sangue , Receptores de IgG/metabolismo , Pele/metabolismo , Pele/patologia , Transativadores , Ferimentos e Lesões/patologia
13.
J Cell Sci ; 125(Pt 18): 4288-96, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22718342

RESUMO

Flightless (Flii) is upregulated in response to wounding and has been shown to function in wound closure and scarring. In macrophages intracellular Flii negatively modulates Toll-Like Receptor (TLR) signalling and dampens cytokine production. We now show that Flii is constitutively secreted from macrophages and fibroblasts and is present in human plasma. Secretion from fibroblasts is upregulated in response to scratch wounding and lipopolysaccharide (LPS)-activated macrophages also temporally upregulate their secretion of Flii. Using siRNA, and wild-type and mutant proteins, we show that Flii is secreted by means of a late endosomal/lysosomal pathway that is regulated by Rab7 and Stx11. Flii contains 11 leucine-rich repeat domains in its N-terminus that have nearly 50% similarity to those in the extracellular pathogen binding portion of Toll-like receptor 4 (TLR4). We show secreted Flii can also bind LPS and has the ability to alter macrophage activation. LPS activation of macrophages in Flii-depleted conditioned medium leads to enhanced macrophage activation and increased TNF secretion compared with cells activated in the presence of Flii. These results show secreted Flii binds to LPS and in doing so alters macrophage activation and cytokine secretion, suggesting that like the intracellular pool of Flii, secreted Flii also has the ability to alter inflammation.


Assuntos
Citocinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Endossomos/metabolismo , Lipopolissacarídeos/metabolismo , Lisossomos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Proteínas de Transporte , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Ativação de Macrófagos , Fusão de Membrana , Camundongos , Células NIH 3T3 , Ligação Proteica , Transporte Proteico , Proteínas Qa-SNARE/metabolismo , Pele/metabolismo , Transativadores , Fator de Necrose Tumoral alfa/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
14.
Commun Integr Biol ; 5(6): 546-9, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23336022

RESUMO

Intracellular Flightless I (Flii), a gelsolin family member, has been found to have roles modulating actin regulation, transcriptional regulation and inflammation. In vivo Flii can regulate wound healing responses. We have recently shown that a pool of Flii is secreted by fibroblasts and macrophages, cells typically found in wounds, and its secretion can be upregulated upon wounding. We show that secreted Flii can bind to the bacterial cell wall component lipopolysaccharide and has the potential to regulate inflammation. We now show that secreted Flii is present in both acute and chronic wound fluid.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...