Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
FASEB J ; 33(10): 10966-10972, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31287960

RESUMO

Vorapaxar-dependent protease-activated receptor (PAR)-1 inhibition diminishes diabetic nephropathy in experimental type 1 diabetes. As most patients with diabetic nephropathy suffer from type 2 diabetes, the aim of this study was to investigate whether PAR-1 inhibition also limits diabetic nephropathy in experimental type 2 diabetes. Consequently, leptin-deficient black and tan brachyuric (BTBRob/ob) mice were randomly assigned to vorapaxar (1.75 mg/kg; twice weekly via oral gavage) or vehicle treatment, whereas matched wild-type (WT) BTBR (BTBRWT) mice served as nondiabetic controls. Weight and (nonfasting) blood glucose levels were monitored for up to 18 wk, after which kidney function and histologic damage was evaluated postmortem. We show that blood glucose levels and body weight increased in diabetic BTBRob/ob mice compared with nondiabetic BTBRWT controls. Vorapaxar-dependent PAR-1 inhibition reduced but did not normalize blood glucose levels in BTBRob/ob mice, whereas it potentiated the increase in body weight. Vorapaxar did not, however, preserve kidney function, whereas it only minimally reduced histopathological signs of kidney injury. Overall, we thus show that PAR-1 inhibition reduces blood glucose levels during the progression of diabetic nephropathy in experimental type 2 diabetes but does not improve renal function. This is in contrast to the therapeutic potential of vorapaxar in type 1 diabetes-induced nephropathy, highlighting the importance of disease-dependent treatment modalities.-Waasdorp, M., Florquin, S., Duitman, J., Spek, C. A. Pharmacological PAR-1 inhibition reduces blood glucose levels but does not improve kidney function in experimental type 2 diabetic nephropathy.


Assuntos
Glicemia/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Rim/efeitos dos fármacos , Lactonas/uso terapêutico , Piridinas/uso terapêutico , Receptor PAR-1/antagonistas & inibidores , Animais , Diabetes Mellitus Tipo 2/genética , Nefropatias Diabéticas/metabolismo , Feminino , Rim/fisiopatologia , Lactonas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/farmacologia , Receptores para Leptina/deficiência , Receptores para Leptina/genética
2.
J Cell Mol Med ; 23(2): 1268-1279, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30485646

RESUMO

End-stage renal disease, the final stage of all chronic kidney disorders, is associated with renal fibrosis and inevitably leads to renal failure and death. Transition of tubular epithelial cells (TECs) into mesenchymal fibroblasts constitutes a proposed mechanism underlying the progression of renal fibrosis and here we assessed whether protease-activated receptor (PAR)-1, which recently emerged as an inducer of epithelial-to-mesenchymal transition (EMT), aggravates renal fibrosis. We show that PAR-1 activation on TECs reduces the expression of epithelial markers and simultaneously induces mesenchymal marker expression reminiscent of EMT. We next show that kidney damage was reduced in PAR-1-deficient mice during unilateral ureter obstruction (UUO) and that PAR-1-deficient mice develop a diminished fibrotic response. Importantly, however, we did hardly observe any signs of mesenchymal transition in both wild-type and PAR-1-deficient mice suggesting that diminished fibrosis in PAR-1-deficient mice is not due to reduced EMT. Instead, the accumulation of macrophages and fibroblasts was significantly reduced in PAR-1-deficient animals which were accompanied by diminished production of MCP-1 and TGF-ß. Overall, we thus show that PAR-1 drives EMT of TECs in vitro and aggravates UUO-induced renal fibrosis although this is likely due to PAR-1-dependent pro-fibrotic cytokine production rather than EMT.


Assuntos
Injúria Renal Aguda/etiologia , Fibrose/etiologia , Nefropatias/fisiopatologia , Nefrite Intersticial/etiologia , Receptor PAR-1/fisiologia , Obstrução Ureteral/complicações , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Doença Crônica , Transição Epitelial-Mesenquimal , Fibrose/metabolismo , Fibrose/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite Intersticial/metabolismo , Nefrite Intersticial/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
3.
Nucleic Acids Res ; 40(18): 9272-85, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22821563

RESUMO

Cells release RNA-carrying vesicles and membrane-free RNA/protein complexes into the extracellular milieu. Horizontal vesicle-mediated transfer of such shuttle RNA between cells allows dissemination of genetically encoded messages, which may modify the function of target cells. Other studies used array analysis to establish the presence of microRNAs and mRNA in cell-derived vesicles from many sources. Here, we used an unbiased approach by deep sequencing of small RNA released by immune cells. We found a large variety of small non-coding RNA species representing pervasive transcripts or RNA cleavage products overlapping with protein coding regions, repeat sequences or structural RNAs. Many of these RNAs were enriched relative to cellular RNA, indicating that cells destine specific RNAs for extracellular release. Among the most abundant small RNAs in shuttle RNA were sequences derived from vault RNA, Y-RNA and specific tRNAs. Many of the highly abundant small non-coding transcripts in shuttle RNA are evolutionary well-conserved and have previously been associated to gene regulatory functions. These findings allude to a wider range of biological effects that could be mediated by shuttle RNA than previously expected. Moreover, the data present leads for unraveling how cells modify the function of other cells via transfer of specific non-coding RNA species.


Assuntos
Pequeno RNA não Traduzido/análise , Vesículas Transportadoras/química , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/química , Células Dendríticas/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , MicroRNAs/análise , MicroRNAs/química , Pequeno RNA não Traduzido/química , Pequeno RNA não Traduzido/fisiologia , RNA de Transferência/análise , RNA de Transferência/química , Sequências Repetitivas de Ácido Nucleico , Análise de Sequência de RNA , Linfócitos T/química , Linfócitos T/imunologia
4.
Pflugers Arch ; 465(2): 247-59, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23192368

RESUMO

Drug-induced ion channel trafficking disturbance can cause cardiac arrhythmias. We showed that the antiprotozoic pentamidine decreased K(IR)2.x carried I(K1) current and that inhibiting protein degradation in the lysosome increased intracellular K(IR)2.1 levels. In this study, we aim to identify and then inhibit preceding steps in clathrin-mediated endocytosis of K(IR)2.1 to further restore normal levels of functional K(IR)2.1 channels. K(IR)2.1 trafficking in HEK293 cells was studied by live cell imaging, immunofluorescence microscopy, and Western blot following pharmacological intervention with dynasore (Dyn), chlorpromazine (CPZ), bafilomycin A1 (Baf), or chloroquine (CQ). K(IR)2.1 function was determined by patch-clamp electrophysiology. CQ induced lysosomal build-up of full length (3.8 ± 0.8-fold) and N-terminal cleaved K(IR)2.1 protein. Baf induced late endosomal build-up of full length protein only (6.1 ± 1.6-fold). CPZ and Dyn increased plasma membrane-localized channel and protein levels (2.6 ± 0.4- and 4.2 ± 1.1-fold, respectively). Dyn increased I(K1) (at -60 mV) from 31 ± 6 to 55 ± 7 pA/pF (N = 9 and 13 respectively, p < 0.05), while the CPZ effect on current density was not testable due to acute I(K1) block. Baf and CQ did not significantly enhance I(K1) densities. Pentamidine (10 µM, 48 h) reduced K(IR)2.1 levels to 0.6 ± 0.1-fold, which could be rescued by Baf (3.2 ± 0.9), CPZ (1.2 ± 0.3), or Dyn (1.2 ± 0.3). Taken together, the clathrin-mediated endocytosis pathway functions in K(IR)2.1 degradation. Pentamidine-induced downregulation of K(IR)2.1 can be rescued at the level of the plasma membrane, implying that acquired trafficking defects can be rescued.


Assuntos
Antiprotozoários/farmacologia , Vesículas Revestidas por Clatrina/metabolismo , Regulação para Baixo , Endocitose/efeitos dos fármacos , Pentamidina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Potenciais de Ação , Membrana Celular/metabolismo , Cloroquina/farmacologia , Clorpromazina/farmacologia , Células HEK293 , Humanos , Hidrazonas/farmacologia , Macrolídeos/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Transporte Proteico/efeitos dos fármacos , Proteólise
5.
Cells ; 12(3)2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36766687

RESUMO

Burn injury induces a complex inflammatory response, both locally and systemically, and is not yet completely unravelled and understood. In order to enable the development of accurate treatment options, it is of paramount importance to fully understand post-burn immunology. Research in the last decades describes insights into the prolonged and excessive inflammatory response that could exist after both severe and milder burn trauma and that this response differs from that of none-burn acute trauma. Persistent activity of complement, acute phase proteins and pro- and anti-inflammatory mediators, changes in lymphocyte activity, activation of the stress response and infiltration of immune cells have all been related to post-burn local and systemic pathology. This "narrative" review explores the current state of knowledge, focusing on both the local and systemic immunology post-burn, and further questions how it is linked to the clinical outcome. Moreover, it illustrates the complexity of post-burn immunology and the existing gaps in knowledge on underlying mechanisms of burn pathology.


Assuntos
Proteínas do Sistema Complemento , Linfócitos , Proteínas de Fase Aguda , Mediadores da Inflamação
6.
Cell Rep ; 38(1): 110189, 2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986347

RESUMO

Fibrosis is a major cause of mortality worldwide, characterized by myofibroblast activation and excessive extracellular matrix deposition. Systemic sclerosis is a prototypic fibrotic disease in which CXCL4 is increased and strongly correlates with skin and lung fibrosis. Here we aim to elucidate the role of CXCL4 in fibrosis development. CXCL4 levels are increased in multiple inflammatory and fibrotic mouse models, and, using CXCL4-deficient mice, we demonstrate the essential role of CXCL4 in promoting fibrotic events in the skin, lungs, and heart. Overexpressing human CXCL4 in mice aggravates, whereas blocking CXCL4 reduces, bleomycin-induced fibrosis. Single-cell ligand-receptor analysis predicts CXCL4 to affect endothelial cells and fibroblasts. In vitro, we confirm that CXCL4 directly induces myofibroblast differentiation and collagen synthesis in different precursor cells, including endothelial cells, by stimulating endothelial-to-mesenchymal transition. Our findings identify a pivotal role of CXCL4 in fibrosis, further substantiating the potential role of neutralizing CXCL4 as a therapeutic strategy.


Assuntos
Matriz Extracelular/patologia , Miofibroblastos/metabolismo , Fator Plaquetário 4/metabolismo , Fibrose Pulmonar/patologia , Escleroderma Sistêmico/patologia , Animais , Bleomicina/toxicidade , Linhagem Celular , Colágeno/biossíntese , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/citologia , Pericitos/metabolismo , Fator Plaquetário 4/genética , Células Estromais/citologia , Células Estromais/metabolismo
7.
Biomolecules ; 11(8)2021 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-34439831

RESUMO

Wound healing is an essential process to restore tissue integrity after trauma. Large skin wounds such as burns often heal with hypertrophic scarring and contractures, resulting in disfigurements and reduced joint mobility. Such adverse healing outcomes are less common in the oral mucosa, which generally heals faster compared to skin. Several studies have identified differences between oral and skin wound healing. Most of these studies however focus only on a single stage of wound healing or a single cell type. The aim of this review is to provide an extensive overview of wound healing in skin versus oral mucosa during all stages of wound healing and including all cell types and molecules involved in the process and also taking into account environmental specific factors such as exposure to saliva and the microbiome. Next to intrinsic properties of resident cells and differential expression of cytokines and growth factors, multiple external factors have been identified that contribute to oral wound healing. It can be concluded that faster wound closure, the presence of saliva, a more rapid immune response, and increased extracellular matrix remodeling all contribute to the superior wound healing and reduced scar formation in oral mucosa, compared to skin.


Assuntos
Matriz Extracelular/imunologia , Microbiota/imunologia , Mucosa Bucal/lesões , Pele/lesões , Cicatrização/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Matriz Extracelular/química , Fibroblastos/imunologia , Fibroblastos/microbiologia , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Queratinócitos/imunologia , Queratinócitos/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Mucosa Bucal/imunologia , Mucosa Bucal/microbiologia , Mucosa Bucal/patologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Especificidade de Órgãos , Saliva/imunologia , Saliva/microbiologia , Transdução de Sinais , Pele/imunologia , Pele/microbiologia , Pele/patologia
8.
Oncotarget ; 9(31): 21655-21662, 2018 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-29774092

RESUMO

BACKGROUND: Twenty years after the onset of diabetes, up to 40% of patients develop diabetic nephropathy. Protease-activated receptor-1 (PAR-1) has recently been shown to aggravate the development of experimental diabetic nephropathy. PAR-1 deficient mice develop less albuminuria and glomerular lesions and PAR-1 stimulation induces proliferation and fibronectin production in mesangial cells in vitro. Vorapaxar is a clinically available PAR-1 inhibitor which is currently used for secondary prevention of ischemic events. OBJECTIVES: The aim of this study was to investigate in a preclinical setting whether vorapaxar treatment may be a novel strategy to reduce diabetes-induced kidney damage. RESULTS: While control treated diabetic mice developed significant albuminuria, mesangial expansion and glomerular fibronectin deposition, diabetic mice on vorapaxar treatment did not show any signs of kidney damage despite having similar levels of hyperglycemia. CONCLUSIONS: These data show that PAR-1 inhibition by vorapaxar prevents the development of diabetic nephropathy in this preclinical animal model for type I diabetes and pinpoint PAR-1 as a novel therapeutic target to pursue in the setting of diabetic nephropathy. MATERIALS AND METHODS: 22 C57Bl/6 mice were made diabetic using multiple low-dose streptozotocin injections (50 mg/kg) and 22 littermates served as non-diabetic controls. Four weeks after the induction of diabetes, 11 mice of each group were assigned to control or vorapaxar treatment. Mice were sacrificed after 20 weeks of treatment and kidney damage was evaluated.

9.
Am J Transl Res ; 10(7): 2220, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30093959

RESUMO

[This corrects the article on p. 4512 in vol. 9, PMID: 29118913.].

10.
Biochem Biophys Rep ; 10: 152-156, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29114573

RESUMO

BACKGROUND: Protease-activated receptor-1 (PAR-1) potentiates diabetic nephropathy (DN) as evident from reduced kidney injury in diabetic PAR-1 deficient mice. Although thrombin is the prototypical PAR-1 agonist, anticoagulant treatment does not limit DN in experimental animal models suggesting that thrombin is not the endogenous PAR-1 agonist driving DN. OBJECTIVES: To identify the endogenous PAR-1 agonist potentiating diabetes-induced nephropathy. METHODS: Unbiased protease expression profiling in glomeruli from human kidneys with DN was performed using publically available microarray data. The identified prime candidate PAR-1 agonist was subsequently analysed for PAR-1-dependent induction of fibrosis in vitro. RESULTS: Of the 553 proteases expressed in the human genome, 247 qualified as potential PAR-1 agonists of which 71 were significantly expressed above background in diabetic glomeruli. The recently identified PAR-1 agonist plasmin(ogen), together with its physiological activator tissue plasminogen activator, were among the highest expressed proteases. Plasmin did however not induce mesangial proliferation and/or fibronectin deposition in vitro. In a PAR-1 independent manner, plasmin even reduced fibronectin deposition. CONCLUSION: Expression profiling identified plasmin as potential endogenous PAR-1 agonist driving DN. Instead of inducing fibronectin expression, plasmin however reduced mesangial fibronectin deposition in vitro. Therefore we conclude that plasmin may not be the endogenous PAR-1 agonist potentiating DN.

11.
Am J Transl Res ; 9(10): 4512-4520, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29118913

RESUMO

Diabetic nephropathy is a major microvascular complication of diabetes mellitus, and the leading cause of end stage renal disease worldwide. The pathogenesis of diabetic nephropathy is complex, making the development of novel treatments that stop or reverse the progression of microalbuminuria into end stage renal disease a challenge. Protease activated receptor (PAR)-2 has recently been shown to aggravate disease progression in diabetic nephropathy based upon which it was suggested that PAR-2 would be a potential target for the treatment of diabetic nephropathy. To fully appreciate the translational potential of PAR-2 in diabetic nephropathy, we evaluated the effect of PAR-2 deficiency on the development of diabetic nephropathy in a streptozotocin-induced diabetes model characteristic of type 1 diabetes. Although diabetic PAR-2 deficient mice showed reduced albuminuria compared to diabetic wild type mice, an increase in mesangial expansion was evident in the PAR-2 deficient mice. No differences were observed in blood glucose levels, podocyte numbers or in glomerular vascular density. These results show that PAR-2 plays a dual role in the development of streptozotocin-induced diabetic nephropathy and may thus not be the eagerly awaited novel target to combat diabetic nephropathy. Targeting PAR-2 should consequently only be pursued with great care in a clinical setting.

12.
Sci Rep ; 6: 33030, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27618774

RESUMO

Endogenously administered activated protein C ameliorates diabetic nephropathy (DN) in a protease-activated receptor-1 (PAR-1)-dependent manner, suggesting that PAR-1 activation limits the progression of DN. Activation of PAR-1 in fibroblast-like cells, however, induces proliferation and extracellular matrix production, thereby driving fibrotic disease. Considering the key role of mesangial proliferation and extracellular matrix production during DN, PAR-1 may in fact potentiate diabetes-induced kidney injury. To determine the net effect of PAR-1 in DN, streptozotocin-induced DN was studied in wild type and PAR-1 deficient mice. Subsequent mechanistic insight was obtained by assessing profibrotic responses of mesangial and tubular epithelial cells in vitro, following PAR-1 stimulation and inhibition. Despite having similar glucose levels, PAR-1 deficient mice developed less kidney damage after induction of diabetes, as evidenced by diminished proteinuria, plasma cystatin C levels, expansion of the mesangial area, and tubular atrophy. In vitro, PAR-1 signaling in mesangial cells led to increased proliferation and expression of matrix proteins fibronectin and collagen IV. Conversely, a reduction in both proliferation and fibronectin deposition was observed in diabetic PAR-1 deficient mice. Overall, we show that PAR-1 plays an important role in the development of DN and PAR-1 might therefore be an attractive therapeutic target to pursue in DN.


Assuntos
Proliferação de Células , Diabetes Mellitus Experimental/prevenção & controle , Nefropatias Diabéticas/prevenção & controle , Túbulos Renais/metabolismo , Células Mesangiais/metabolismo , Receptor PAR-1/deficiência , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Túbulos Renais/lesões , Túbulos Renais/patologia , Células Mesangiais/patologia , Camundongos , Camundongos Knockout
13.
Oncotarget ; 6(34): 35304-14, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26474459

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a destructive disease in part resulting from premature or mature cellular aging. Protease-activated receptor-1 (PAR-1) recently emerged as a critical component in the context of fibrotic lung diseases. Therefore, we aimed to study the role of macrophages in PAR-1-mediated idiopathic pulmonary fibrosis. The number of macrophages were significantly reduced in lungs of PAR-1 antagonist (P1pal-12) treated animals upon bleomycin instillation. In line with these data, PAR-1 stimulation increased monocyte / macrophage recruitment in response to epithelium injury in in vitro trans-well assays. Moreover, macrophages induced fibroblasts migration, differentiation and secretion of collagen, which were inhibited in the presence of TGF-ß receptor inhibitors. Interestingly, these profibrotic effects were partially inhibited by treatment with the PAR-1 inhibitor P1pal-12. Using shRNA mediated PAR-1 knock down in fibroblasts, we demonstrate that fibroblast PAR-1 contributes to TGF-ß activation and production. Finally, we show that the macrophage-dependent induction of PAR-1 driven TGF-ß activation was mediated by FXa. Our data identify novel mechanisms by which PAR-1 stimulation on different cell types can contribute to IPF and identify macrophages as key players in PAR-1 dependent development of this devastating disease. IPF may result from cellular senescence mediated by macrophages in the lung.


Assuntos
Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Receptor PAR-1/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Fibrose Pulmonar Idiopática/genética , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Células RAW 264.7 , Receptor PAR-1/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA