Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 205(8): 2243-2254, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32917786

RESUMO

In IgA nephropathy (IgAN), IgA immune complexes are deposited in the mesangium and drive inflammation and extracellular matrix (ECM) remodelling. The functional links between IgA deposition, inflammation, and matrix remodelling are not well characterized. We recently performed urine liquid chromatography-tandem mass spectrometry proteomics and identified multiple ECM glycoproteins whose expression and function in IgAN is unclear. None of the urine glycoproteins was regulated in IgAN transcriptomics, indicating that tissue remodelling rather than increased expression might contribute to their presence in urine. To investigate this, we examined the IgAN expression profile of metalloproteinases, enzymes involved in the remodelling of ECM proteins, and noted that the proteoglycanase ADAMTS5 was upregulated in IgAN kidneys. ADAMTS5 accumulated in areas of inflammation, and ADAMTS5+ cells were seen in the tubulointerstitium and glomeruli. The enzyme was expressed by CD64+ cells and its expression was increased by IL-1 and LPS. Analysis of myeloid cell transcriptomics revealed that ADAMTS5 is enriched in human classical monocytes. ADAMTS5+ cells were present in areas of matrix remodelling and associated with ECM proteins lumican, versican, and collagen-4. Liquid chromatography-tandem mass spectrometry proteomics of kidney explants digested with ADAMTS5, identified multiple kidney proteins affected by ADAMTS5 and revealed specific proteolysis of complement C3 and fibronectin associated with IgA on immune complexes. ADAMTS5 processing of immune complex proteins reduced binding to cultured mesangial cells. ADAMTS5 is associated with interstitial inflammatory cells in IgAN and other kidney lesions and fragments relevant extracellular proteins. The proteolytic enzyme might be a new translational target relevant to inflammation and scarring in kidney disease.


Assuntos
Proteína ADAMTS5/imunologia , Matriz Extracelular/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Glomerulonefrite por IGA/imunologia , Glomérulos Renais/imunologia , Monócitos/imunologia , Adulto , Idoso , Matriz Extracelular/patologia , Feminino , Glomerulonefrite por IGA/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Glomérulos Renais/patologia , Masculino , Pessoa de Meia-Idade , Monócitos/patologia
2.
Kidney Int ; 99(5): 1127-1139, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33417998

RESUMO

Understanding why certain patients with IgA nephropathy progress to kidney failure while others maintain normal kidney function remains a major unanswered question. To help answer this, we performed miRNome profiling by next generation sequencing of kidney biopsies in order to identify microRNAs specifically associated with the risk of IgA nephropathy progression. Following sequencing and validation in independent cohorts, four microRNAs (-150-5p, -155-5p, -146b-5p, -135a-5p) were found to be differentially expressed in IgA nephropathy progressors compared to non-progressors, and patients with thin membrane nephropathy, lupus nephritis and membranous nephropathy, and correlated with estimated glomerular filtration rate, proteinuria, and the Oxford MEST-C scores (five histological features that are independent predictors of clinical outcome). Each individual microRNA increased the discrimination score of the International IgAN Prediction Tool, although due to the small number of samples the results did not reach statistical significance. miR-150-5p exhibited the largest amplitude of expression between cohorts and displayed the best discrimination between IgA nephropathy progressors and non-progressors by receiver operating curve analysis (AUC: 0.8). However, expression was similarly upregulated in kidneys with established fibrosis and low estimated glomerular filtration rates at the time of biopsy. Consistent with a more generic role in kidney fibrosis, in situ hybridization revealed that miR-150-5p was found in lymphoid infiltrates, and areas of proliferation and fibrosis consistent with the known drivers of progression. Thus, miR-150-5p may be a potential functional mediator of kidney fibrosis that may add value in predicting risk of progression in IgA nephropathy and other kidney diseases.


Assuntos
Glomerulonefrite por IGA , MicroRNAs , Biomarcadores , Progressão da Doença , Taxa de Filtração Glomerular , Glomerulonefrite por IGA/genética , Humanos , Rim , MicroRNAs/genética
3.
J Autoimmun ; 105: 102309, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31402200

RESUMO

IgA nephropathy (IgAN) is the most common glomerulonephritis worldwide and a major cause of chronic kidney disease and failure. IgAN is driven by an autoimmune reaction against galactose-deficient IgA1 that results in the generation of autoantibodies and large IgG-IgA immune complexes. Immune complexes accumulate in the glomerular mesangium causing chronic inflammation and renal scarring. A significant proportion of IgAN patients develop end-stage kidney disease and require dialysis or transplantation. Currently, there are no approved specific therapies that can ameliorate the systemic autoimmune reaction in IgAN and no biomarkers that can predict renal inflammation and scarring. In this study, we used shotgun LC-MS/MS proteomics to compare small volumes of urine from healthy subjects and IgAN patients. We identified multiple urine proteins with unknown renal or IgAN function. Our attention was captured by the increase of phosphatidylethanolamine binding protein-4 (PEBP4) in IgAN urine. The function of PEBP4 in IgAN or renal disease is unknown. Increased levels of urine and serum PEBP4 were subsequently validated in different cohorts of IgAN patients and PEBP4 was linked to declining kidney function in IgAN. Strong PEBP4 staining was sporadically seen in IgAN kidney biopsies, colocalising with IgA in glomeruli and in the lumen of kidney tubules. In a small number of IgAN biopsies, PEBP4 colocalised with IgA and CD19 while the increased excretion of PEBP4 in IgAN urine was accompanied by increased excretion of classic B-cell factors BAFF, BCMA and TACI as well as IgA and IgG. PEBP4 is a new IgAN-related protein with unknown function and a likely renal disease marker in urine and serum.


Assuntos
Linfócitos B/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina A/imunologia , Rim/imunologia , Proteína de Ligação a Fosfatidiletanolamina/imunologia , Adulto , Complexo Antígeno-Anticorpo/imunologia , Autoanticorpos/imunologia , Linfócitos B/metabolismo , Biomarcadores/metabolismo , Biópsia , Estudos de Casos e Controles , Feminino , Galactose/imunologia , Galactose/metabolismo , Mesângio Glomerular/imunologia , Mesângio Glomerular/metabolismo , Glomerulonefrite por IGA/metabolismo , Humanos , Rim/metabolismo , Falência Renal Crônica/imunologia , Falência Renal Crônica/metabolismo , Masculino
4.
Arterioscler Thromb Vasc Biol ; 38(7): 1537-1548, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29622560

RESUMO

OBJECTIVE: Thoracic aortic aneurysm (TAA), a degenerative disease of the aortic wall, is accompanied by changes in the structure and composition of the aortic ECM (extracellular matrix). The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteases has recently been implicated in TAA formation. This study aimed to investigate the contribution of ADAMTS-5 to TAA development. APPROACH AND RESULTS: A model of aortic dilatation by AngII (angiotensin II) infusion was adopted in mice lacking the catalytic domain of ADAMTS-5 (Adamts5Δcat). Adamts5Δcat mice showed an attenuated rise in blood pressure while displaying increased dilatation of the ascending aorta (AsAo). Interestingly, a proteomic comparison of the aortic ECM from AngII-treated wild-type and Adamts5Δcat mice revealed versican as the most upregulated ECM protein in Adamts5Δcat mice. This was accompanied by a marked reduction of ADAMTS-specific versican cleavage products (versikine) and a decrease of LRP1 (low-density lipoprotein-related protein 1). Silencing LRP1 expression in human aortic smooth muscle cells reduced the expression of ADAMTS5, attenuated the generation of versikine, but increased soluble ADAMTS-1. A similar increase in ADAMTS-1 was observed in aortas of AngII-treated Adamts5Δcat mice but was not sufficient to maintain versican processing and prevent aortic dilatation. CONCLUSIONS: Our results support the emerging role of ADAMTS proteases in TAA. ADAMTS-5 rather than ADAMTS-1 is the key protease for versican regulation in murine aortas. Further studies are needed to define the ECM substrates of the different ADAMTS proteases and their contribution to TAA formation.


Assuntos
Proteína ADAMTS5/metabolismo , Aorta Torácica/enzimologia , Aneurisma da Aorta Torácica/enzimologia , Matriz Extracelular/enzimologia , Remodelação Vascular , Proteína ADAMTS1/metabolismo , Proteína ADAMTS5/deficiência , Proteína ADAMTS5/genética , Angiotensina II , Animais , Aorta Torácica/patologia , Aneurisma da Aorta Torácica/induzido quimicamente , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/patologia , Células Cultivadas , Dilatação Patológica , Modelos Animais de Doenças , Matriz Extracelular/patologia , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos Knockout , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Versicanas/metabolismo
5.
Brain ; 141(8): 2362-2381, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29912283

RESUMO

Chondroitinase ABC is a promising preclinical therapy that promotes functional neuroplasticity after CNS injury by degrading extracellular matrix inhibitors. Efficient delivery of chondroitinase ABC to the injured mammalian spinal cord can be achieved by viral vector transgene delivery. This approach dramatically modulates injury pathology and restores sensorimotor functions. However, clinical development of this therapy is limited by a lack of ability to exert control over chondroitinase gene expression. Prior experimental gene regulation platforms are likely to be incompatible with the non-resolving adaptive immune response known to occur following spinal cord injury. Therefore, here we apply a novel immune-evasive dual vector system, in which the chondroitinase gene is under a doxycycline inducible regulatory switch, utilizing a chimeric transactivator designed to evade T cell recognition. Using this novel vector system, we demonstrate tight temporal control of chondroitinase ABC gene expression, effectively removing treatment upon removal of doxycycline. This enables a comparison of short and long-term gene therapy paradigms in the treatment of clinically-relevant cervical level contusion injuries in adult rats. We reveal that transient treatment (2.5 weeks) is sufficient to promote improvement in sensory axon conduction and ladder walking performance. However, in tasks requiring skilled reaching and grasping, only long term treatment (8 weeks) leads to significantly improved function, with rats able to accurately grasp and retrieve sugar pellets. The late emergence of skilled hand function indicates enhanced neuroplasticity and connectivity and correlates with increased density of vGlut1+ innervation in spinal cord grey matter, particularly in lamina III-IV above and below the injury. Thus, our novel gene therapy system provides an experimental tool to study temporal effects of extracellular matrix digestion as well as an encouraging step towards generating a safer chondroitinase gene therapy strategy, longer term administration of which increases neuroplasticity and recovery of descending motor control. This preclinical study could have a significant impact for tetraplegic individuals, for whom recovery of hand function is an important determinant of independence, and supports the ongoing development of chondroitinase gene therapy towards clinical application for the treatment of spinal cord injury.


Assuntos
Condroitina ABC Liase/administração & dosagem , Terapia Genética/métodos , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Condroitina ABC Liase/farmacologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Regeneração Nervosa/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Mutantes , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Transgenes/genética
6.
Ann Rheum Dis ; 77(9): 1372-1380, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29925506

RESUMO

OBJECTIVES: One mechanism by which cartilage responds to mechanical load is by releasing heparin-bound growth factors from the pericellular matrix (PCM). By proteomic analysis of the PCM, we identified connective tissue growth factor (CTGF) and here investigate its function and mechanism of action. METHODS: Recombinant CTGF (rCTGF) was used to stimulate human chondrocytes for microarray analysis. Endogenous CTGF was investigated by in vitro binding assays and confocal microscopy. Its release from cut cartilage (injury CM) was analysed by Western blot under reducing and non-reducing conditions. A postnatal, conditional CtgfcKO mouse was generated for cartilage injury experiments and to explore the course of osteoarthritis (OA) by destabilisation of the medial meniscus. siRNA knockdown was performed on isolated human chondrocytes. RESULTS: The biological responses of rCTGF were TGFß dependent. CTGF displaced latent TGFß from cartilage and both were released on cartilage injury. CTGF and latent TGFß migrated as a single high molecular weight band under non-reducing conditions, suggesting that they were in a covalent (disulfide) complex. This was confirmed by immunoprecipitation. Using CtgfcKO mice, CTGF was required for sequestration of latent TGFß in the matrix and activation of the latent complex at the cell surface through TGFßR3. In vivo deletion of CTGF increased the thickness of the articular cartilage and protected mice from OA. CONCLUSIONS: CTGF is a latent TGFß binding protein that controls the matrix sequestration and activation of TGFß in cartilage. Deletion of CTGF in vivo caused a paradoxical increase in Smad2 phosphorylation resulting in thicker cartilage that was protected from OA.


Assuntos
Artrite Experimental/metabolismo , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Osteoartrite/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Artrite Experimental/patologia , Artrite Experimental/prevenção & controle , Cartilagem Articular/lesões , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo/deficiência , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fator de Crescimento do Tecido Conjuntivo/farmacologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Camundongos Knockout , Osteoartrite/patologia , Osteoartrite/prevenção & controle , Proteoglicanas/metabolismo , Proteômica , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Recombinantes/farmacologia , Proteína Smad2/metabolismo , Técnicas de Cultura de Tecidos
7.
Int J Mol Sci ; 19(5)2018 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-29758010

RESUMO

Spinal cord injury (SCI) causes irreversible tissue damage and severe loss of neurological function. Currently, there are no approved treatments and very few therapeutic targets are under investigation. Here, we combined 4 high-throughput transcriptomics and proteomics datasets, 7 days and 8 weeks following clinically-relevant rat SCI to identify proteins with persistent differential expression post-injury. Out of thousands of differentially regulated entities our combined analysis identified 40 significantly upregulated versus 48 significantly downregulated molecules, which were persistently altered at the mRNA and protein level, 7 days and 8 weeks post-SCI. Bioinformatics analysis was then utilized to identify currently available drugs with activity against the filtered molecules and to isolate proteins with known or unknown function in SCI. Our findings revealed multiple overlooked therapeutic candidates with important bioactivity and established druggability but with unknown expression and function in SCI including the upregulated purine nucleoside phosphorylase (PNP), cathepsins A, H, Z (CTSA, CTSH, CTSZ) and proteasome protease PSMB10, as well as the downregulated ATP citrate lyase (ACLY), malic enzyme (ME1) and sodium-potassium ATPase (ATP1A3), amongst others. This work reveals previously unappreciated therapeutic candidates for SCI and available drugs, thus providing a valuable resource for further studies and potential repurposing of existing therapeutics for SCI.


Assuntos
Biologia Computacional , Perfilação da Expressão Gênica , Proteoma , Proteômica , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Transcriptoma , Animais , Biologia Computacional/métodos , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Proteômica/métodos , Ratos , Traumatismos da Medula Espinal/tratamento farmacológico , Fatores de Tempo
8.
J Neurosci ; 35(23): 8959-69, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26063927

RESUMO

G-protein receptor 84 (GPR84) is an orphan receptor that is induced markedly in monocytes/macrophages and microglia during inflammation, but its pathophysiological function is unknown. Here, we investigate the role of GPR84 in a murine model of traumatic nerve injury. Naive GPR84 knock-out (KO) mice exhibited normal behavioral responses to acute noxious stimuli, but subsequent to partial sciatic nerve ligation (PNL), KOs did not develop mechanical or thermal hypersensitivity, in contrast to wild-type (WT) littermates. Nerve injury increased ionized calcium binding adapter molecule 1 (Iba1) and phosphorylated p38 MAPK immunoreactivity in the dorsal horn and Iba1 and cluster of differentiation 45 expression in the sciatic nerve, with no difference between genotypes. PCR array analysis revealed that Gpr84 expression was upregulated in the spinal cord and sciatic nerve of WT mice. In addition, the expression of arginase-1, a marker for anti-inflammatory macrophages, was upregulated in KO sciatic nerve. Based on this evidence, we investigated whether peripheral macrophages behave differently in the absence of GPR84. We found that lipopolysaccharide-stimulated KO macrophages exhibited attenuated expression of several proinflammatory mediators, including IL-1ß, IL-6, and TNF-α. Forskolin-stimulated KO macrophages also showed greater cAMP induction, a second messenger associated with immunosuppression. In summary, our results demonstrate that GPR84 is a proinflammatory receptor that contributes to nociceptive signaling via the modulation of macrophages, whereas in its absence the response of these cells to an inflammatory insult is impaired.


Assuntos
Regulação da Expressão Gênica/genética , Limiar da Dor/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Ciática/metabolismo , Ciática/fisiopatologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Hipersensibilidade/etiologia , Hipersensibilidade/genética , Inflamação/etiologia , Inflamação/genética , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Medição da Dor , Estimulação Física/efeitos adversos , Receptores Acoplados a Proteínas G/genética , Ciática/patologia , Medula Espinal/metabolismo , Temperatura , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
J Neurosci ; 34(49): 16424-32, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25471580

RESUMO

Chondroitinase ABC (ChABC) has striking effects on promoting neuronal plasticity after spinal cord injury (SCI), but little is known about its involvement in other pathological mechanisms. Recent work showed that ChABC might also modulate the immune response by promoting M2 macrophage polarization. Here we investigate in detail the immunoregulatory effects of ChABC after SCI in rats. Initially, we examined the expression profile of 16 M1/M2 macrophage polarization markers at 3 h and 7 d postinjury. ChABC treatment had a clear effect on the immune signature after SCI. More specifically, ChABC increased the expression of the anti-inflammatory cytokine IL-10, accompanied by a reduction in the proinflammatory cytokine IL-12B in injured spinal tissue. These effects were associated with a distinct, IL-10-mediated anti-inflammatory response in ChABC-treated spinal cords. Mechanistically, we show that IL-10 expression is driven by tissue injury and macrophage infiltration, while the p38 MAPK is the central regulator of IL-10 expression in vivo. These findings provide novel insights into the effects of ChABC in the injured spinal cord and explain its immunoregulatory activity.


Assuntos
Condroitina ABC Liase/fisiologia , Regulação da Expressão Gênica , Imunomodulação/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Interleucina-10/biossíntese , Traumatismos da Medula Espinal/imunologia , Animais , Condroitina ABC Liase/administração & dosagem , Condroitina ABC Liase/farmacologia , Imidazóis/farmacologia , Imunomodulação/fisiologia , Injeções Espinhais , Interleucina-12/biossíntese , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/fisiologia , Masculino , Proteoglicanas/metabolismo , Piridinas/farmacologia , Ratos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
10.
J Neurosci ; 34(14): 4822-36, 2014 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-24695702

RESUMO

Chondroitin sulfate proteoglycans (CSPGs) inhibit repair following spinal cord injury. Here we use mammalian-compatible engineered chondroitinase ABC (ChABC) delivered via lentiviral vector (LV-ChABC) to explore the consequences of large-scale CSPG digestion for spinal cord repair. We demonstrate significantly reduced secondary injury pathology in adult rats following spinal contusion injury and LV-ChABC treatment, with reduced cavitation and enhanced preservation of spinal neurons and axons at 12 weeks postinjury, compared with control (LV-GFP)-treated animals. To understand these neuroprotective effects, we investigated early inflammatory changes following LV-ChABC treatment. Increased expression of the phagocytic macrophage marker CD68 at 3 d postinjury was followed by increased CD206 expression at 2 weeks, indicating that large-scale CSPG digestion can alter macrophage phenotype to favor alternatively activated M2 macrophages. Accordingly, ChABC treatment in vitro induced a significant increase in CD206 expression in unpolarized monocytes stimulated with conditioned medium from spinal-injured tissue explants. LV-ChABC also promoted the remodelling of specific CSPGs as well as enhanced vascularity, which was closely associated with CD206-positive macrophages. Neuroprotective effects of LV-ChABC corresponded with improved sensorimotor function, evident as early as 1 week postinjury, a time point when increased neuronal survival correlated with reduced apoptosis. Improved function was maintained into chronic injury stages, where improved axonal conduction and increased serotonergic innervation were also observed. Thus, we demonstrate that ChABC gene therapy can modulate secondary injury processes, with neuroprotective effects that lead to long-term improved functional outcome and reveal novel mechanistic evidence that modulation of macrophage phenotype may underlie these effects.


Assuntos
Condroitina ABC Liase/genética , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Terapia Genética/métodos , Macrófagos/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Células Cultivadas , Proteoglicanas de Sulfatos de Condroitina/administração & dosagem , Modelos Animais de Doenças , Estimulação Elétrica , Feminino , Regulação da Expressão Gênica/fisiologia , Injeções Espinhais , Proteínas do Tecido Nervoso/metabolismo , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/fisiologia , Desempenho Psicomotor/fisiologia , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Fatores de Tempo
11.
Circ Res ; 113(10): 1138-47, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24006456

RESUMO

RATIONALE: MicroRNAs (miRNAs), in particular miR-29b and miR-30c, have been implicated as important regulators of cardiac fibrosis. OBJECTIVE: To perform a proteomics comparison of miRNA effects on extracellular matrix secretion by cardiac fibroblasts. METHODS AND RESULTS: Mouse cardiac fibroblasts were transfected with pre-/anti-miR of miR-29b and miR-30c, and their conditioned medium was analyzed by mass spectrometry. miR-29b targeted a cadre of proteins involved in fibrosis, including multiple collagens, matrix metalloproteinases, and leukemia inhibitory factor, insulin-like growth factor 1, and pentraxin 3, 3 predicted targets of miR-29b. miR-29b also attenuated the cardiac fibroblast response to transforming growth factor-ß. In contrast, miR-30c had little effect on extracellular matrix production but opposite effects regarding leukemia inhibitory factor and insulin-like growth factor 1. Both miRNAs indirectly affected cardiac myocytes. On transfection with pre-miR-29b, the conditioned medium of cardiac fibroblasts lost its ability to support adhesion of rat ventricular myocytes and led to a significant reduction of cardiac myocyte proteins (α-actinin, cardiac myosin-binding protein C, and cardiac troponin I). Similarly, cardiomyocytes derived from mouse embryonic stem cells atrophied under pre-miR-29 conditioned medium, whereas pre-miR-30c conditioned medium had a prohypertrophic effect. Levels of miR-29a, miR-29c, and miR-30c, but not miR-29b, were significantly reduced in a mouse model of pathological but not physiological hypertrophy. Treatment with antagomiRs to miR-29b induced excess fibrosis after aortic constriction without overt deterioration in cardiac function. CONCLUSIONS: Our proteomic analysis revealed novel molecular targets of miRNAs that are linked to a fibrogenic cardiac phenotype. Such comprehensive screening methods are essential to define the concerted actions of miRNAs in cardiovascular disease.


Assuntos
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , MicroRNAs/fisiologia , Miocárdio/metabolismo , Proteômica , Animais , Proteína C-Reativa/metabolismo , Células Cultivadas , Colágeno/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibrose , Fator de Crescimento Insulin-Like I/metabolismo , Fator Inibidor de Leucemia/metabolismo , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Miocárdio/patologia , Componente Amiloide P Sérico/metabolismo , Fator de Crescimento Transformador beta/farmacologia
12.
Arthritis Rheum ; 65(2): 397-407, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23124605

RESUMO

OBJECTIVE: To investigate whether cartilage injury activates protein tyrosine kinases distinct from fibroblast growth factor (FGF)-related signaling, and whether they contribute to injury-induced gene responses. METHODS: Phosphokinases and protein tyrosine phosphorylation were assayed by Western blotting of cartilage lysates. Immunoprecipitation and Western blotting with 4G10 antibody and immunoprecipitation kinase assay were carried out. Tyrosine-phosphorylated proteins on silver-stained gels of injured cartilage lysates were identified by mass spectrometry. Messenger RNA induction in cartilage explants was assessed by quantitative reverse transcriptase-polymerase chain reaction. RESULTS: Protein tyrosine phosphorylation occurred within seconds of injury to the surface of intact articular cartilage, as did activation of MAPKs and IKK. Activation did not reoccur upon reinjury of cultured explants. The prominent tyrosine-phosphorylated proteins focal adhesion kinase, paxillin, and cortactin were identified as substrates of Src family kinases. The Src family kinase inhibitor PP2 blocked injury-induced tyrosine phosphorylation. It did not prevent activation of the MAPKs and IKK but differentially inhibited 8 of 10 inflammatory response genes that were induced by injury. In contrast, FGF signaling blockade with PD173074 reduced all MAPK and IKK activation by ∼50% and inhibited a different subset of genes but had no effect on Src-like signaling. CONCLUSION: Injury to the surface of intact articular cartilage activates Src-like kinases as well as MAPKs and IKK (implying NF-κB activation). FGF-2 contributes to MAPK/IKK activation but not to Src-like signaling, suggesting that the latter is a parallel pathway that also regulates the injury-induced inflammatory gene response.


Assuntos
Cartilagem Articular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Expressão Gênica/fisiologia , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo , Animais , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/lesões , Inibidores Enzimáticos/farmacologia , Fator 2 de Crescimento de Fibroblastos/genética , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Expressão Gênica/efeitos dos fármacos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Pirimidinas/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Suínos , Quinases da Família src/genética
13.
J Mol Cell Cardiol ; 54: 65-72, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23154128

RESUMO

Smooth muscle cells (SMC) contribute to the development and stability of atherosclerotic lesions. The molecular mechanisms that mediate their properties are incompletely defined. We employed proteomics and in vitro functional assays to identify the unique characteristics of intimal SMC isolated from human carotid endarterectomy specimens and medial SMC from thoracic aortas and carotids. We verified our findings in the Tampere Vascular Study. Human atheroma-derived SMC exhibit decreased expression of mitochondrial proteins ATP Synthase subunit-beta and Aldehyde dehydrogenase 2, and decreased mitochondrial activity when compared to control SMC. Moreover, a comparison between plaque-derived SMC isolated from patients with or without recent acute cerebrovascular symptoms uncovered an increase in Annexin A1, an endogenous anti-inflammatory protein, in the asymptomatic group. The deletion of Annexin A1 or the blockade of its signaling in SMC resulted in increased cytokine production at baseline and after stimulation with the pro-inflammatory cytokine Tumor Necrosis Factor α. In summary, our proteomics and biochemical analysis revealed mitochondrial damage in human plaque-derived SMC as well as a role of Annexin A1 in reducing the production of pro-inflammatory mediators in SMC.


Assuntos
Anexina A1/metabolismo , Aterosclerose/patologia , Doenças das Artérias Carótidas/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteoma/metabolismo , Adulto , Aldeído Desidrogenase/metabolismo , Aldeído-Desidrogenase Mitocondrial , Animais , Doenças das Artérias Carótidas/patologia , Células Cultivadas , Citocinas/metabolismo , Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias Musculares/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Músculo Liso Vascular/patologia , Oxirredução , Peroxirredoxinas/metabolismo , Fenótipo , Análise de Componente Principal , Proteômica
14.
J Biol Chem ; 287(23): 19341-5, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22493487

RESUMO

Atherosclerosis is initiated by the retention of lipoproteins on proteoglycans in the arterial intima. However, the mechanisms leading to proteoglycan accumulation and lipoprotein retention are poorly understood. In this study, we set out to investigate the role of ADAMTS-5 (a disintegrin and metalloprotease with thrombospondin motifs-5) in the vasculature. ADAMTS-5 was markedly reduced in atherosclerotic aortas of apolipoprotein E-null (apoE(-/-)) mice. The reduction of ADAMTS-5 was accompanied by accumulation of biglycan and versican, the major lipoprotein-binding proteoglycans, in atherosclerosis. ADAMTS-5 activity induced the release of ADAMTS-specific versican (DPEAAE(441)) and aggrecan ((374)ALGS) fragments as well as biglycan and link protein from the aortic wall. Fibroblast growth factor 2 (FGF-2) inhibited ADAMTS-5 expression in isolated aortic smooth muscle cells and blocked the spontaneous release of ADAMTS-generated versican and aggrecan fragments from aortic explants. In aortas of ADAMTS-5-deficient mice, DPEAAE(441) versican neoepitopes were not detectable. Instead, biglycan levels were increased, highlighting the role of ADAMTS-5 in the catabolism of vascular proteoglycans. Importantly, ADAMTS-5 proteolytic activity reduced the LDL binding ability of biglycan and released LDL from human aortic lesions. This study provides the first evidence implicating ADAMTS-5 in the regulation of proteoglycan turnover and lipoprotein retention in atherosclerosis.


Assuntos
Proteínas ADAM/metabolismo , Agrecanas/metabolismo , Aorta/enzimologia , Apolipoproteínas E/metabolismo , Aterosclerose/enzimologia , Lipoproteínas LDL/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Proteínas ADAM/genética , Proteína ADAMTS5 , Agrecanas/genética , Animais , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Feminino , Humanos , Lipoproteínas LDL/genética , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Musculares/genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia
15.
Circulation ; 125(6): 789-802, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22261194

RESUMO

BACKGROUND: After myocardial ischemia, extracellular matrix (ECM) deposition occurs at the site of the focal injury and at the border region. METHODS AND RESULTS: We have applied a novel proteomic method for the analysis of ECM in cardiovascular tissues to a porcine model of ischemia/reperfusion injury. ECM proteins were sequentially extracted and identified by liquid chromatography tandem mass spectrometry. For the first time, ECM proteins such as cartilage intermediate layer protein 1, matrilin-4, extracellular adipocyte enhancer binding protein 1, collagen α-1(XIV), and several members of the small leucine-rich proteoglycan family, including asporin and prolargin, were shown to contribute to cardiac remodeling. A comparison in 2 distinct cardiac regions (the focal injury in the left ventricle and the border region close to the occluded coronary artery) revealed a discordant regulation of protein and mRNA levels; although gene expression for selected ECM proteins was similar in both regions, the corresponding protein levels were much higher in the focal lesion. Further analysis based on >100 ECM proteins delineated a signature of early- and late-stage cardiac remodeling with transforming growth factor-ß1 signaling at the center of the interaction network. Finally, novel cardiac ECM proteins identified by proteomics were validated in human left ventricular tissue acquired from ischemic cardiomyopathy patients at cardiac transplantation. CONCLUSION: Our findings reveal a biosignature of early- and late-stage ECM remodeling after myocardial ischemia/reperfusion injury, which may have clinical utility as a prognostic marker and modifiable target for drug discovery.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Proteômica , Animais , Biomarcadores , Cromatografia Líquida , Descoberta de Drogas , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/isolamento & purificação , Fibrose , Perfilação da Expressão Gênica , Transplante de Coração , Humanos , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Prognóstico , RNA Mensageiro/biossíntese , Distribuição Aleatória , Sus scrofa , Suínos , Espectrometria de Massas em Tandem , Fator de Crescimento Transformador beta1/fisiologia , Remodelação Ventricular/genética , Remodelação Ventricular/fisiologia
16.
Mol Cell Proteomics ; 10(8): M111.008128, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21593211

RESUMO

Abdominal aortic aneurysms (AAA) are characterized by pathological remodeling of the aortic extracellular matrix (ECM). However, besides the well-characterized elastolysis and collagenolysis little is known about changes in other ECM proteins. Previous proteomics studies on AAA focused on cellular changes without emphasis on the ECM. In the present study, ECM proteins and their degradation products were selectively extracted from aneurysmal and control aortas using a solubility-based subfractionation methodology and analyzed by gel-liquid chromatography-tandem MS and label-free quantitation. The proteomics analysis revealed novel changes in the ECM of AAA, including increased expression as well as degradation of collagen XII, thrombospondin 2, aortic carboxypeptidase-like protein, periostin, fibronectin and tenascin. Proteomics also confirmed the accumulation of macrophage metalloelastase (MMP-12). Incubation of control aortic tissue with recombinant MMP-12 resulted in the extensive fragmentation of these glycoproteins, most of which are novel substrates of MMP-12. In conclusion, our proteomics methodology allowed the first detailed analysis of the ECM in AAA and identified markers of pathological ECM remodeling related to MMP-12 activity.


Assuntos
Aorta Abdominal/fisiopatologia , Aneurisma da Aorta Abdominal/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteoma/metabolismo , Adulto , Idoso , Aorta Abdominal/patologia , Carboxipeptidases/metabolismo , Estudos de Casos e Controles , Fracionamento Químico , Análise por Conglomerados , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/isolamento & purificação , Feminino , Glicoproteínas/química , Glicoproteínas/isolamento & purificação , Glicoproteínas/metabolismo , Guanidina/química , Humanos , Mediadores da Inflamação/química , Mediadores da Inflamação/isolamento & purificação , Mediadores da Inflamação/metabolismo , Masculino , Metaloproteinase 12 da Matriz/metabolismo , Pessoa de Meia-Idade , Proteoma/química , Proteoma/isolamento & purificação , Proteômica , Proteínas Repressoras/metabolismo , Solubilidade , Adulto Jovem
17.
Mol Cell Proteomics ; 9(9): 2048-62, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20551380

RESUMO

The vascular extracellular matrix (ECM) is essential for the structural integrity of the vessel wall and also serves as a substrate for the binding and retention of secreted products of vascular cells as well as molecules coming from the circulation. Although proteomics has been previously applied to vascular tissues, few studies have specifically targeted the vascular ECM and its associated proteins. Thus, its detailed composition remains to be characterized. In this study, we describe a methodology for the extraction of extracellular proteins from human aortas and their identification by proteomics. The approach is based on (a) effective decellularization to enrich for scarce extracellular proteins, (b) successful solubilization and deglycosylation of ECM proteins, and (c) relative estimation of protein abundance using spectral counting. Our three-step extraction approach resulted in the identification of 103 extracellular proteins of which one-third have never been reported in the proteomics literature of vascular tissues. In particular, three glycoproteins (podocan, sclerostin, and agrin) were identified for the first time in human aortas at the protein level. We also identified extracellular adipocyte enhancer-binding protein 1, the cartilage glycoprotein asporin, and a previously hypothetical protein, retinal pigment epithelium (RPE) spondin. Moreover, our methodology allowed us to screen for proteolysis in the aortic samples based on the identification of proteolytic enzymes and their corresponding degradation products. For instance, we were able to detect matrix metalloproteinase-9 by mass spectrometry and relate its presence to degradation of fibronectin in a clinical specimen. We expect this proteomics methodology to further our understanding of the composition of the vascular extracellular environment, shed light on ECM remodeling and degradation, and provide insights into important pathological processes, such as plaque rupture, aneurysm formation, and restenosis.


Assuntos
Aorta/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Espaço Extracelular/metabolismo , Proteômica , Eletroforese em Gel de Poliacrilamida , Glicosilação , Humanos , Espectrometria de Massas em Tandem
18.
Arterioscler Thromb Vasc Biol ; 30(7): 1325-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20431068

RESUMO

OBJECTIVE: Recent studies on cardiovascular progenitors have led to a new appreciation that paracrine factors may support the regeneration of damaged tissues. METHODS AND RESULTS: We used a shotgun proteomics strategy to compare the secretome of peripheral blood-derived smooth muscle progenitors (SPCs) with human aortic smooth muscle cells. The late-outgrowth SPCs produced fewer proteolytic enzymes and inflammatory cytokines and showed reduced invasive capacity. Similar to smooth muscle cells, SPCs secreted extracellular matrix. However, SPCs produced different matrix proteins, as evidenced by the truncation of proangiogenic domains in collagen alpha-1 (I) and increased production of periostin. Moreover, SPCs retained serum proteins, including proteoglycans, regulating collagen assembly; and pigment epithelium-derived factor, a potent inhibitor of angiogenesis. As a functional consequence, their conditioned medium was less angiogenic, as demonstrated by endothelial tube formation assays in vitro and implantation of Matrigel plugs into nude, severe combined immunodeficient mice (NOD/SCID). CONCLUSIONS: The present study represents an important conceptual development, suggesting that SPCs may contribute to extracellular matrix production.


Assuntos
Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica , Proteômica , Células-Tronco/metabolismo , Animais , Aorta/metabolismo , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Cromatografia de Fase Reversa , Meios de Cultivo Condicionados/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Músculo Liso Vascular/citologia , Comunicação Parácrina , Peptídeo Hidrolases/metabolismo , Proteômica/métodos , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
19.
mSphere ; 5(3)2020 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-32581077

RESUMO

COVID-19 is often related to hyperinflammation that drives lung or multiorgan injury. The immunopathological mechanisms that cause excessive inflammation are under investigation and constantly updated. Here, a gene network approach was used on recently published data sets to identify possible COVID-19 inflammatory mechanisms and bioactive genes. First, network analysis of putative SARS-CoV-2 cellular receptors led to the mining of a neutrophil-response signature and relevant inflammatory genes. Second, analysis of RNA-seq data sets of lung cells infected with SARS-CoV-2 revealed that infected cells expressed neutrophil-attracting chemokines. Third, analysis of RNA-seq data sets of bronchoalveolar lavage fluid cells from COVID-19 patients identified upregulation of neutrophil genes and chemokines. Different inflammatory genes mined here, including TNFR, IL-8, CXCR1, CXCR2, ADAM10, GPR84, MME, ANPEP, and LAP3, might be druggable targets in efforts to limit SARS-CoV-2 inflammation in severe clinical cases. The possible role of neutrophils in COVID-19 inflammation needs to be studied further.


Assuntos
Betacoronavirus/imunologia , Quimiocinas/imunologia , Infecções por Coronavirus/imunologia , Inflamação/patologia , Neutrófilos/imunologia , Pneumonia Viral/imunologia , Líquido da Lavagem Broncoalveolar/citologia , COVID-19 , Quimiocinas/genética , Infecções por Coronavirus/patologia , Humanos , Inflamação/imunologia , Pneumopatias/imunologia , Pneumopatias/patologia , Infiltração de Neutrófilos/imunologia , Pandemias , Pneumonia Viral/patologia , Receptores Virais/genética , SARS-CoV-2
20.
Curr Atheroscler Rep ; 11(3): 188-95, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19361350

RESUMO

Acute coronary syndromes (ACS), such as unstable angina, acute myocardial infarction, and sudden cardiac death, are commonly associated with the presence of vulnerable plaques in coronary arteries. Rupture or erosion of vulnerable plaques results in the formation of luminal thrombi due to the physical contact between platelets and thrombogenic elements within the atherosclerotic lesions. Considering the socioeconomic burden of ACS, it is imperative that the scientific community achieves a clear understanding of the multifaceted pathophysiology of vulnerable atheroma to identify accurate prognostic biomarkers and therapeutic targets. The analytical power of modern proteomic technologies could facilitate our understanding of vulnerable plaques and lead to the discovery of novel therapeutic targets and diagnostic biomarkers.


Assuntos
Síndrome Coronariana Aguda/fisiopatologia , Trombose Coronária/fisiopatologia , Proteômica , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA