Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 134(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33408247

RESUMO

The migration of circulating neutrophils towards damaged or infected tissue is absolutely critical to the inflammatory response. L-selectin is a cell adhesion molecule abundantly expressed on circulating neutrophils. For over two decades, neutrophil L-selectin has been assigned the exclusive role of supporting tethering and rolling - the initial stages of the multi-step adhesion cascade. Here, we provide direct evidence for L-selectin contributing to neutrophil transendothelial migration (TEM). We show that L-selectin co-clusters with PECAM-1 - a well-characterised cell adhesion molecule involved in regulating neutrophil TEM. This co-clustering behaviour occurs specifically during TEM, which serves to augment ectodomain shedding of L-selectin and expedite the time taken for TEM (TTT) to complete. Blocking PECAM-1 signalling (through mutation of its cytoplasmic tail), PECAM-1-dependent adhesion or L-selectin shedding, leads to a significant delay in the TTT. Finally, we show that co-clustering of L-selectin with PECAM-1 occurs specifically across TNF- but not IL-1ß-activated endothelial monolayers - implying unique adhesion interactomes forming in a cytokine-specific manner. To our knowledge, this is the first report to implicate a non-canonical role for L-selectin in regulating neutrophil TEM.


Assuntos
Movimento Celular , Selectina L , Neutrófilos , Migração Transendotelial e Transepitelial , Adesão Celular , Endotélio Vascular , Humanos , Selectina L/genética
2.
Biochem Biophys Res Commun ; 606: 10-16, 2022 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-35338853

RESUMO

BACKGROUND: There is compelling evidence implicating dysregulated inflammation in the mechanism of ventricular remodeling and heart failure (HF) after MI. The transcription factor nuclear factor erythroid-derived 2-like 2 (Nrf2, encoded by Nfe2l2) is a promising target in this context since it impedes transcriptional upregulation of pro-inflammatory cytokines and is anti-inflammatory in various murine models. OBJECTIVES: We aimed to investigate the contribution of Nrf2 to the inflammatory response after experimental myocardial infarction (MI). METHODS: We subjected Nrf2-/- mice and wild type (WT) controls to permanent left coronary artery (LCA) ligation. The inflammatory response was investigated with fluorescence-activated cell sorting (FACS) analysis of peripheral blood and heart cell suspensions, together with qRT-PCR of infarcted tissue for chemokines and their receptors. To investigate whether Nrf2-mediated transcription is a dedicated function of leukocytes, we interrogated publicly available RNA-sequencing (RNA-seq) data from mouse hearts after permanent LCA ligation for Nrf2-regulated gene (NRG) expression. RESULTS: FACS analysis demonstrated a profoundly inflamed phenotype in the hearts of global Nrf2-/- mice as compared to WT mice after MI. Moreover, infarcted tissue from Nrf2-/- mice displayed higher expression of mRNA coding for inflammatory cytokines, chemokines, and their receptors, including IL-6, Ccl2, and Cxcr4. RNA-seq analysis showed upregulated NRG expression in WT mice after MI compared to naive mice, which was significantly higher in bioinformatically isolated CCR2+ cells. CONCLUSIONS: Taken together, the results suggest that Nrf2 signalling in leukocytes, and possibly CCR2+ monocytes and monocyte-derived cardiac resident macrophages, may be potential targets to prevent post-MI ventricular remodeling.


Assuntos
Infarto do Miocárdio , Fator 2 Relacionado a NF-E2/metabolismo , Remodelação Ventricular , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Fator 2 Relacionado a NF-E2/genética , Remodelação Ventricular/fisiologia
3.
J Cell Sci ; 131(13)2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29777033

RESUMO

Leukocyte transendothelial migration (TEM) is absolutely fundamental to the inflammatory response, and involves initial pseudopod protrusion and subsequent polarised migration across inflamed endothelium. Ezrin/radixin/moesin (ERM) proteins are expressed in leukocytes and mediate cell shape changes and polarity. The spatio-temporal organisation of ERM proteins with their targets, and their individual contribution to protrusion during TEM, has never been explored. Here, we show that blocking binding of moesin to phosphatidylinositol 4,5-bisphosphate (PIP2) reduces its C-terminal phosphorylation during monocyte TEM, and that on-off cycling of ERM activity is essential for pseudopod protrusion into the subendothelial space. Reactivation of ERM proteins within transmigrated pseudopods re-establishes their binding to targets, such as L-selectin. Knockdown of ezrin, but not moesin, severely impaired the recruitment of monocytes to activated endothelial monolayers under flow, suggesting that this protein plays a unique role in the early recruitment process. Ezrin binds preferentially to L-selectin in resting cells and during early TEM. The moesin-L-selectin interaction increases within transmigrated pseudopods as TEM proceeds, facilitating localised L-selectin ectodomain shedding. In contrast, a non-cleavable L-selectin mutant binds selectively to ezrin, driving multi-pseudopodial extensions. Taken together, these results show that ezrin and moesin play mutually exclusive roles in modulating L-selectin signalling and shedding to control protrusion dynamics and polarity during monocyte TEM.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Endotélio/citologia , Selectina L/metabolismo , Proteínas dos Microfilamentos/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Linhagem Celular , Movimento Celular , Proteínas do Citoesqueleto/genética , Endotélio/metabolismo , Humanos , Selectina L/genética , Proteínas dos Microfilamentos/genética , Ligação Proteica
5.
J Biol Chem ; 292(16): 6703-6714, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28235798

RESUMO

L-selectin regulates leukocyte adhesion and rolling along the endothelium. Proteins binding to the cytoplasmic tail of L-selectin regulate L-selectin functions. We used L-selectin cytoplasmic tail peptide pulldown assays combined with high sensitivity liquid chromatography/mass spectrometry to identify novel L-selectin tail-binding proteins. Incubation of the L-selectin tail with cell extracts from phorbol 12-myristate 13-acetate-stimulated Raw 264.7 macrophages resulted in the binding of µ1A of the clathrin-coated vesicle AP-1 complex. Furthermore, full-length GST-µ1A and the GST-µ1A C-terminal domain, but not the GST-µ1A N-terminal domain, bind to L-selectin tail peptide, and the intracellular pool of L-selectin colocalizes with AP-1 at the trans-Golgi network. We identified a novel basic protein motif consisting of a cluster of three dibasic residues (356RR357, 359KK360, and 362KK363) in the membrane-proximal domain of the L-selectin tail as well as a doublet of aspartic acid residues (369DD370) in the membrane-distal end of the L-selectin tail involved in µ1A binding. Stimulation of Raw 264.7 macrophages with PMA augmented the amount of µ1A associated with anti-L-selectin immunoprecipitates. However, full-length GST-µ1A did not bind to the phospho-L-selectin tail or phospho-mimetic S364D L-selectin tail. Accordingly, we propose that phosphorylation of µ1A is required for interaction with the L-selectin tail and that L-selectin tail phosphorylation may regulate this interaction in vivo Molecular docking of the L-selectin tail to µ1A was used to identify the µ1A surface domain binding the L-selectin tail and to explain how phosphorylation of the L-selectin tail abrogates µ1A interaction. Our findings indicate that L-selectin is transported constitutively by the AP-1 complex, leading to the formation of a trans-Golgi network reserve pool and that phosphorylation of the L-selectin tail blocks AP-1-dependent retrograde transport of L-selectin.


Assuntos
Complexo 1 de Proteínas Adaptadoras/química , Subunidades mu do Complexo de Proteínas Adaptadoras/química , Selectina L/química , Motivos de Aminoácidos , Animais , Ácido Aspártico/química , Cristalografia por Raios X , Citoplasma/metabolismo , Endotélio Vascular/metabolismo , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Imunoprecipitação , Macrófagos/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Monócitos/metabolismo , Fosforilação , Ligação Proteica , Domínios Proteicos , Proteômica , Células RAW 264.7 , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Serina/química , Rede trans-Golgi/metabolismo
6.
Cell Tissue Res ; 371(3): 437-453, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29353325

RESUMO

L-selectin is a type I transmembrane cell adhesion molecule expressed on most circulating leukocytes, including neutrophils. Engagement of L-selectin with endothelial-derived ligands initiates neutrophil tethering and rolling behaviour along luminal walls of post-capillary venules, constituting the first step of the multi-step adhesion cascade. There is a large body of evidence to suggest that signalling downstream of L-selectin can influence neutrophil behaviour: adhesion, migration and priming. This review will cover aspects of L-selectin form and function and introduce the "triad of L-selectin regulation", highlighting the inextricable links between adhesion, signalling and ectodomain shedding and also highlighting the cytosolic proteins that interconnect them. Recent advances in how L-selectin impacts priming, transendothelial migration (TEM) and cell polarity will also be discussed.


Assuntos
Selectina L/metabolismo , Neutrófilos/citologia , Sequência de Aminoácidos , Animais , Polaridade Celular , Humanos , Selectina L/química , Neutrófilos/metabolismo , Domínios Proteicos , Transdução de Sinais
7.
Proc Natl Acad Sci U S A ; 112(12): E1461-70, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25775539

RESUMO

L-selectin is a cell adhesion molecule that tethers free-flowing leukocytes from the blood to luminal vessel walls, facilitating the initial stages of their emigration from the circulation toward an extravascular inflammatory insult. Following shear-resistant adhesion to the vessel wall, L-selectin has frequently been reported to be rapidly cleaved from the plasma membrane (known as ectodomain shedding), with little knowledge of the timing or functional consequence of this event. Using advanced imaging techniques, we observe L-selectin shedding occurring exclusively as primary human monocytes actively engage in transendothelial migration (TEM). Moreover, the shedding was localized to transmigrating pseudopods within the subendothelial space. By capturing monocytes in midtransmigration, we could monitor the subcellular distribution of L-selectin and better understand how ectodomain shedding might contribute to TEM. Mechanistically, L-selectin loses association with calmodulin (CaM; a negative regulator of shedding) specifically within transmigrating pseudopods. In contrast, L-selectin/CaM interaction remained intact in nontransmigrated regions of monocytes. We show phosphorylation of L-selectin at Ser 364 is critical for CaM dissociation, which is also restricted to the transmigrating pseudopod. Pharmacological or genetic inhibition of L-selectin shedding significantly increased pseudopodial extensions in transmigrating monocytes, which potentiated invasive behavior during TEM and prevented the establishment of front/back polarity for directional migration persistence once TEM was complete. We conclude that L-selectin shedding directly regulates polarity in transmigrated monocytes, which affirms an active role for this molecule in driving later stages of the multistep adhesion cascade.


Assuntos
Polaridade Celular , Selectina L/metabolismo , Monócitos/citologia , Sequência de Aminoácidos , Adesão Celular , Movimento Celular , Citoplasma/metabolismo , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Leucócitos/metabolismo , Microscopia Eletrônica de Transmissão , Microscopia de Vídeo , Dados de Sequência Molecular , Monócitos/metabolismo , Fosforilação , Serina/química
8.
Circulation ; 127(16): 1712-22, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23529610

RESUMO

BACKGROUND: Vascular endothelial cell growth factor plays a pivotal role in angiogenesis via regulating endothelial cell proliferation. The X-box binding protein 1 (XBP1) is believed to be a signal transducer in the endoplasmic reticulum stress response. It is unknown whether there is crosstalk between vascular endothelial cell growth factor signaling and XBP1 pathway. METHODS AND RESULTS: We found that vascular endothelial cell growth factor induced the kinase insert domain receptor internalization and interaction through C-terminal domain with the unspliced XBP1 and the inositol requiring enzyme 1 α in the endoplasmic reticulum, leading to inositol requiring enzyme 1 α phosphorylation and XBP1 mRNA splicing, which was abolished by siRNA-mediated knockdown of kinase insert domain receptor. Spliced XBP1 regulated endothelial cell proliferation in a PI3K/Akt/GSK3ß/ß-catenin/E2F2-dependent manner and modulated the cell size increase in a PI3K/Akt/GSK3ß/ß-catenin/E2F2-independent manner. Knockdown of XBP1 or inositol requiring enzyme 1 α decreased endothelial cell proliferation via suppression of Akt/GSK3ß phosphorylation, ß-catenin nuclear translocation, and E2F2 expression. Endothelial cell-specific knockout of XBP1 (XBP1ecko) in mice retarded the retinal vasculogenesis in the first 2 postnatal weeks and impaired the angiogenesis triggered by ischemia. Reconstitution of XBP1 by Ad-XBP1s gene transfer significantly improved angiogenesis in ischemic tissue in XBP1ecko mice. Transplantation of bone marrow from wild-type o XBP1ecko mice could also slightly improve the foot blood reperfusion in ischemic XBP1ecko mice. CONCLUSIONS: These results suggest that XBP1 can function via growth factor signaling pathways to regulate endothelial proliferation and angiogenesis.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Animais , Aorta/citologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/irrigação sanguínea , Estresse do Retículo Endoplasmático/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Terapia Genética , Transplante de Células-Tronco Hematopoéticas , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Isquemia/fisiopatologia , Isquemia/terapia , Perna (Membro)/irrigação sanguínea , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Quimera por Radiação , Fatores de Transcrição de Fator Regulador X , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/crescimento & desenvolvimento , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Proteína 1 de Ligação a X-Box
9.
Cell Adh Migr ; 18(1): 1-3, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38860721

RESUMO

In September 2023 members of the cell adhesion and cell migration research community came together to share their latest research and consider how our work might be translated for clinical practice. Alongside invited speakers, selected speakers and poster presentations, the meeting also included a round table discussion of how we might overcome the challenges associated with research translation. This meeting report seeks to highlight the key outcomes of that discussion and spark interest in the cell adhesions and cell migration research community to cross the perceived valley of death and translate our work into therapeutic benefit.


Assuntos
Adesão Celular , Movimento Celular , Humanos , Pesquisa Translacional Biomédica , Animais , Neoplasias/patologia , Neoplasias/terapia
10.
Front Cardiovasc Med ; 10: 975012, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36923955

RESUMO

In this study, we addressed the functional significance of co-operative DNA binding of the cytokine-driven transcription factor STAT1 (signal transducer and activator of transcription 1) in an experimental murine model of acute myocardial infarction (MI). STAT1 knock-in mice expressing a phenylalanine-to-alanine substitution at position 77 in the STAT1 amino-terminal domain were examined for the early clinical effects produced by ligation of the left anterior descending coronary artery (LAD), an established model for MI. The F77A mutation has been previously reported to disrupt amino-terminal interactions between adjacent STAT1 dimers resulting in impaired tetramerization and defective co-operative binding on DNA, while leaving other protein functions unaffected. Our results demonstrate that a loss of STAT1 tetramer stabilization improves survival of adult male mice and ameliorates left ventricular dysfunction in female mice, as determined echocardiographically by an increased ejection fraction and a reduced left intra-ventricular diameter. We found that the ratio of STAT3 to STAT1 protein level was higher in the infarcted tissue in knock-in mice as compared to wild-type (WT) mice, which was accompanied by an enhanced infiltration of immune cells in the infarcted area, as determined by histology. Additionally, RNA sequencing of the infarcted tissue 24 h after LAD ligation revealed an upregulation of inflammatory genes in the knock-in mice, as compared to their WT littermates. Concomitantly, genes involved in oxidative phosphorylation and other metabolic pathways showed a significantly more pronounced downregulation in the infarcted tissue from STAT1F77A/F77A mice than in WT animals. Based on these results, we propose that dysfunctional STAT1 signalling owing to a lack of oligomerisation results in a compensatory increase in STAT3 expression and promotes early infiltration of immune cells in the infarcted area, which has beneficial effects on left ventricular remodelling in early MI following LAD ligation.

11.
JID Innov ; 3(1): 100154, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36561914

RESUMO

Inflammatory edema formation and polymorphonuclear leukocyte (neutrophil) accumulation are common components of cutaneous vascular inflammation, and their assessment is a powerful investigative and drug development tool but typically requires independent cohorts of animals to assess each. We have established the use of a mathematical formula to estimate the ellipsoidal-shaped volume of the edematous wheal or bleb after intradermal injections of substances in mice pretreated intravenously with Evans blue dye (which binds to plasma albumin) to act as an edema marker. Whereas previous extraction of Evans blue dye with formamide is suitable for all strains of mice, we report this quicker and more reliable assessment of edema volume in situ. This therefore allows neutrophil accumulation to be assessed from the same mouse using the myeloperoxidase assay. Importantly, we examined the influence of Evans blue dye on the spectrometry readout at the wavelength at which myeloperoxidase activity is measured. The results indicate that it is feasible to quantify edema formation and neutrophil accumulation in the same mouse skin site. Thus, we show techniques that can assess edema formation and neutrophil accumulation at the same site in the same mouse, allowing paired measurements and reducing the total use of mice by 50%.

12.
Blood ; 115(26): 5355-65, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20354175

RESUMO

Leukocytes rely on dynamic actin-dependent changes in cell shape to pass through blood vessels, which is fundamental to immune surveillance. Wiskott-Aldrich Syndrome protein (WASp) is a hematopoietic cell-restricted cytoskeletal regulator important for modulating cell shape through Arp2/3-mediated actin polymerization. A recently identified WASp(I294T) mutation was shown to render WASp constitutively active in vivo, causing increased filamentous (F)-actin polymerization, high podosome turnover in macrophages, and myelodysplasia. The aim of this study was to determine the effect of WASp(I294T) expression in lymphocytes. Here, we report that lymphocytes isolated from a patient with WASp(I294T), and in a cellular model of WASp(I294T), displayed abnormal microvillar architecture, associated with an increase in total cellular F-actin. Microvillus function was additionally altered as lymphocytes bearing the WASp(I294T) mutation failed to roll normally on L-selectin ligand under flow. This was not because of defects in L-selectin expression, shedding, cytoskeletal anchorage, or membranal positioning; however, under static conditions of adhesion, WASp(I294T)-expressing lymphocytes exhibited altered dynamic interaction with L-selectin ligand, with a significantly reduced rate of adhesion turnover. Together, our results demonstrate that WASp(I294T) significantly affects lymphocyte membrane topography and L-selectin-dependent adhesion, which may be linked to defective hematopoiesis and leukocyte function in affected patients.


Assuntos
Adesão Celular , Doenças Genéticas Ligadas ao Cromossomo X/genética , Leucopenia/genética , Linfócitos/citologia , Microvilosidades/ultraestrutura , Mutação , Proteína da Síndrome de Wiskott-Aldrich/genética , Actinas/metabolismo , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Células Cultivadas , Expressão Gênica , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Humanos , Selectina L/genética , Selectina L/metabolismo , Leucócitos Mononucleares/citologia , Leucopenia/metabolismo , Linfócitos/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
13.
Blood ; 113(16): 3696-705, 2009 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-19060239

RESUMO

Localization of primed T cells to antigenic tissue is essential for the development of effective immunity. Together with tissue-selective homing molecules, T-cell receptor (TCR)- and CD28-mediated signals have been shown to promote transendothelial migration of specific T cells into nonlymphoid antigen-rich tissue. However, the cellular and molecular requirements for T-cell accumulation to target tissue following their recruitment are largely undefined. The guanine nucleotide exchange factor (GEF) Vav1 has an integral role in coupling TCR and CD28 to signaling pathways that regulate T-cell activation and migration. Here, we have investigated the contribution of TCR- and CD28-induced Vav1 activity to the trafficking and localization of primed HY-specific CD4(+) T cells to antigenic sites. Severe migratory defects displayed by Vav1(-/-) T cells in vitro were fully compensated by a combination of shear flow and chemokines, leading to normal recruitment of Vav1(-/-) T cells in vivo. In contrast, Vav1(-/-) T-cell retention into antigen-rich tissue was severely impaired, reflecting T cells' inability to engage in sustained TCR- and CD28-mediated interactions with tissue-resident antigen-presenting cells (APCs). This novel function of APC-induced, and TCR- and CD28-mediated Vav1 activity in the regulation of effector T-cell immunity highlights its potential as a therapeutic target in T cell-mediated tissue damage.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígenos CD28/imunologia , Comunicação Celular/fisiologia , Proteínas Proto-Oncogênicas c-vav/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD28/genética , Movimento Celular/imunologia , Quimiocinas/genética , Quimiocinas/imunologia , Feminino , Imunidade Celular/fisiologia , Ativação Linfocitária/fisiologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-vav/genética , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais/fisiologia
14.
Arterioscler Thromb Vasc Biol ; 30(12): 2467-74, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20884873

RESUMO

OBJECTIVE: To use protein kinase C (PKC) δ-knockout mice to investigate the role of PKCδ in lesion development and to understand the underlying mechanism of the vascular disease. METHODS AND RESULTS: PKCδ functions as a signal transducer mediating several essential functions of cell proliferation and apoptosis. However, the effect of PKCδ on neointimal formation in wire-injured vessels is unknown. Three weeks after wire injury of femoral arteries, neointimal lesions were significantly increased in PKCδ(-/-) mice compared with PKCδ(+/+) animals. Immunohistochemical staining revealed that total numbers of smooth muscle cells and macrophages in the lesions of PKCδ(-/-) mice were markedly elevated without changing the ratio of these 2 cell types. To further elucidate the mechanisms of PKCδ-mediated increase in the lesion, an in vivo endothelial migration model was established to evaluate endothelial wound healing after wire injury. Data showed that reendothelialization of the injured vessel was markedly delayed in PKCδ(-/-) mice; this coincided with more severe intimal hyperplasia. Migration of endothelial cells cultivated from cardiac tissue was markedly reduced in the absence of PKCδ, whereas no difference in proliferation or apoptosis was detected. Inhibition of PKCδ activity or protein expression by small hairpin RNA (shRNA) in cultured endothelial cells confirmed the defective migratory phenotype. Interestingly, vasohibin-1, an antiangiogenesis protein, was elevated in endothelial cells derived from PKCδ(-/-) mice, which was mainly because of delayed protein degradation mediated by PKCδ. Downregulation of vasohibin-1 restored the migration rate of PKCδ(-/-) endothelial cells to a similar level as PKCδ(+/+) cells. CONCLUSIONS: PKCδ deficiency enhances neointimal formation, which is associated with delayed reendothelialization and involves increased cellular vasohibin-1 accumulation.


Assuntos
Arteriopatias Oclusivas/enzimologia , Proteínas de Ciclo Celular/metabolismo , Células Endoteliais/enzimologia , Artéria Femoral/enzimologia , Miócitos de Músculo Liso/enzimologia , Proteína Quinase C-delta/deficiência , Túnica Íntima/enzimologia , Cicatrização , Animais , Apoptose , Arteriopatias Oclusivas/etiologia , Arteriopatias Oclusivas/patologia , Proteínas de Ciclo Celular/genética , Movimento Celular , Proliferação de Células , Células Cultivadas , Constrição Patológica , Modelos Animais de Doenças , Células Endoteliais/patologia , Artéria Femoral/lesões , Artéria Femoral/patologia , Genótipo , Hiperplasia , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Fenótipo , Proteína Quinase C-delta/genética , Interferência de RNA , Fatores de Tempo , Túnica Íntima/lesões , Túnica Íntima/patologia
15.
Biochem J ; 428(2): 293-304, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20331435

RESUMO

L-selectin mediates the initial tethering and subsequent rolling of leucocytes along luminal walls of inflamed venules. TACE [TNFalpha (tumour necrosis factor alpha)-converting enzyme] is responsible for cleaving the membrane-proximal extracellular domain of L-selectin (also known as shedding), which reduces the efficiency of leucocyte recruitment to sites of inflammation. Many reports have highlighted roles for PKC (protein kinase C) and p38 MAPK (mitogen-activated protein kinase) in promoting L-selectin shedding with little insight into the mechanism involved. By using PMA and the phosphatase inhibitors cantharidin and calyculin A, we could selectively activate PKC or p38 MAPK respectively to promote TACE-dependent shedding of L-selectin. Interestingly, the intracellular mechanisms leading to the shedding event differed dramatically. For example, regulatory elements within the L-selectin cytoplasmic tail, such as ERM (ezrin/radixin/moesin)-binding and serine residues, were important for PKC- but not p38 MAPK-dependent shedding. Also, increased and sustained cell surface levels of TACE, and phosphorylation of its cytoplasmic tail (a hallmark of TACE activation), occurred in lymphocytes and monocytes following p38 MAPK activation. Finally, we showed that TNFalpha-induced shedding of L-selectin in monocytes was strikingly similar to cantharidin-induced shedding and suggest that this newly characterized mechanism could be physiologically relevant in inflammatory cells.


Assuntos
Proteínas ADAM/metabolismo , Selectina L/metabolismo , Proteína Quinase C/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas ADAM/química , Proteína ADAM17 , Animais , Cantaridina/farmacologia , Linhagem Celular , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Selectina L/química , Toxinas Marinhas , Proteínas de Membrana/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Oxazóis/farmacologia , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Células U937
16.
ESC Heart Fail ; 8(2): 1427-1437, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33511759

RESUMO

AIMS: Endothelial activation and inflammatory cell infiltration have important roles in the development of cardiac fibrosis induced by renin-angiotensin system activation. NADPH oxidases (Nox proteins) are expressed in endothelial cells (ECs) and alter their function. Previous studies indicated that Nox2 in ECs contributes to angiotensin II (AngII)-induced cardiac fibrosis. However, the effects of EC Nox4 on cardiac fibrosis are unknown. METHODS AND RESULTS: Transgenic (TG) mice overexpressing endothelial-restricted Nox4 were studied alongside wild-type (WT) littermates as controls. At baseline, Nox4 TG mice had significantly enlarged hearts compared with WT, with elongated cardiomyocytes (increased by 18.5%, P < 0.01) and eccentric hypertrophy but well-preserved cardiac function by echocardiography and in vivo pressure-volume analysis. Animals were subjected to a chronic AngII infusion (AngII, 1.1 mg/kg/day) for 14 days. Whereas WT/AngII developed a 2.1-fold increase in interstitial cardiac fibrosis as compared with WT/saline controls (P < 0.01), TG/AngII mice developed significant less fibrosis (1.4-fold increase, P > 0.05), but there were no differences in cardiac hypertrophy or contractile function between the two groups. TG hearts displayed significantly decreased inflammatory cell infiltration with reduced levels of vascular cell adhesion molecule 1 in both the vasculature and myocardium compared with WT after AngII treatment. TG microvascular ECs stimulated with AngII in vitro supported significantly less leukocyte adhesion than WT ECs. CONCLUSIONS: A chronic increase in endothelial Nox4 stimulates physiological cardiac hypertrophy and protects against AngII-induced cardiac fibrosis by inhibiting EC activation and the recruitment of inflammatory cells.


Assuntos
Células Endoteliais , Miocárdio/patologia , NADPH Oxidase 4 , Angiotensina II/efeitos adversos , Animais , Fibrose , Inflamação , Camundongos , Camundongos Transgênicos
17.
JCI Insight ; 6(18)2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34375309

RESUMO

Nox2 is a ROS-generating enzyme, deficiency of which increases suppression by Tregs in vitro and in an in vivo model of cardiac remodeling. As Tregs have emerged as a candidate therapy in autoimmunity and transplantation, we hypothesized that Nox2 deficiency in Tregs in recipient mice may improve outcomes in a heart transplant model. We generated a potentially novel B6129 mouse model with Treg-targeted Nox2 deletion (Nox2fl/flFoxP3Cre+ mice) and transplanted with hearts from CB6F1 donors. As compared with those of littermate controls, Nox2fl/flFoxP3Cre+ mice had lower plasma levels of alloantibodies and troponin-I, reduced levels of IFN-γ in heart allograft homogenates, and diminished cardiomyocyte necrosis and allograft fibrosis. Single-cell analyses of allografts revealed higher absolute numbers of Tregs and lower CD8+ T cell infiltration in Nox2-deficient recipients compared with Nox2-replete mice. Mechanistically, in addition to a greater suppression of CD8+CD25- T effector cell proliferation and IFN-γ production, Nox2-deficient Tregs expressed higher levels of CCR4 and CCR8, driving cell migration to allografts; this was associated with increased expression of miR-214-3p. These data indicate that Nox2 deletion in Tregs enhances their suppressive ability and migration to heart allografts. Therefore, Nox2 inhibition in Tregs may be a useful approach to improve their therapeutic efficacy.


Assuntos
Aloenxertos/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração , NADPH Oxidase 2/genética , Linfócitos T Reguladores/imunologia , Aloenxertos/metabolismo , Aloenxertos/patologia , Animais , Linfócitos T CD8-Positivos/fisiologia , Movimento Celular , Proliferação de Células , Feminino , Fibrose , Rejeição de Enxerto/sangue , Interferon gama/metabolismo , Isoanticorpos/sangue , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Miócitos Cardíacos/patologia , Necrose , Receptores CCR4/metabolismo , Receptores CCR8/metabolismo , Linfócitos T Reguladores/metabolismo , Transplante Homólogo , Troponina I/sangue
18.
Shock ; 56(2): 268-277, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34276040

RESUMO

ABSTRACT: Leukocyte Nox2 is recognized to have a fundamental microbicidal function in sepsis but the specific role of Nox2 in endothelial cells (EC) remains poorly elucidated. Here, we tested the hypothesis that endothelial Nox2 participates in the pathogenesis of systemic inflammation and hypotension induced by LPS. LPS was injected intravenously in mice with Tie2-targeted deficiency or transgenic overexpression of Nox2. Mice with Tie2-targeted Nox2 deficiency had increased circulating levels of TNF-α, enhanced numbers of neutrophils trapped in lungs, and aggravated hypotension after LPS injection, as compared to control LPS-injected animals. In contrast, Tie2-driven Nox2 overexpression attenuated inflammation and prevented the hypotension induced by LPS. Because Tie2-Cre targets both EC and myeloid cells we generated bone marrow chimeric mice with Nox2 deletion restricted to leukocytes or ECs. Mice deficient in Nox2 either in leukocytes or ECs had reduced LPS-induced neutrophil trapping in the lungs and lower plasma TNF-α levels as compared to control LPS-injected mice. However, the pronounced hypotensive response to LPS was present only in mice with EC-specific Nox2 deletion. Experiments in vitro with human vein or aortic endothelial cells (HUVEC and HAEC, respectively) treated with LPS revealed that EC Nox2 controls NF-κB activation and the transcription of toll-like receptor 4 (TLR4), which is the recognition receptor for LPS. In conclusion, these results suggest that endothelial Nox2 limits NF-κB activation and TLR4 expression, which in turn attenuates the severity of hypotension and systemic inflammation induced by LPS.


Assuntos
Células Endoteliais/fisiologia , Endotoxemia/etiologia , Hipotensão/etiologia , Inflamação/etiologia , NADPH Oxidase 2/fisiologia , Receptor 4 Toll-Like/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Cardiovasc Res ; 116(6): 1101-1112, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31841135

RESUMO

Improvements in early interventions after acute myocardial infarction (AMI), notably, the increased use of timely reperfusion therapy, have increased survival dramatically in recent decades. Despite this, maladaptive ventricular remodelling and subsequent heart failure (HF) following AMI remain a significant clinical challenge, particularly because several pre-clinical strategies to attenuate remodelling have failed to translate into clinical practice. Monocytes and macrophages, pleiotropic cells of the innate immune system, are integral in both the initial inflammatory response to injury and subsequent wound healing in many tissues, including the heart. However, maladaptive immune cell behaviour contributes to ventricular remodelling in mouse models, prompting experimental efforts to modulate the immune response to prevent the development of HF. Seminal work in macrophage biology defined macrophages as monocyte-derived cells that are comprised of two populations, pro-inflammatory M1 macrophages and reparative M2 macrophages, and initial investigations into cardiac macrophage populations following AMI suggested they aligned well to this model. However, more recent data, in the heart and other tissues, demonstrate remarkable heterogeneity and plasticity in macrophage development, phenotype, and function. These recent insights into macrophage biology may explain the failure of non-specific immunosuppressive strategies and offer novel opportunities for therapeutic targeting to prevent HF following AMI. Here, we summarize the traditional monocyte-macrophage paradigm, experimental evidence for the significance of these cells in HF after AMI, and the potential relevance of emerging evidence that refutes canonical models of monocyte and macrophage biology.


Assuntos
Macrófagos/patologia , Monócitos/patologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , Remodelação Ventricular , Animais , Humanos , Macrófagos/metabolismo , Monócitos/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA