Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Biochem Biophys Res Commun ; 701: 149552, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38335918

RESUMO

The Interleukin-17 (IL17) family is a group of cytokines implicated in the etiology of several inflammatory diseases. Interleukin-17 receptor D (IL17RD), also known as Sef (similar expression to fibroblast growth factor) belonging to the family of IL17 receptors, has been shown to modulate IL17A-associated inflammatory phenotypes. The objective of this study was to test the hypothesis that IL17RD promotes endothelial cell activation and consequent leukocyte adhesion. We utilized primary human aortic endothelial cells and demonstrated that RNAi targeting of IL17RD suppressed transcript levels by 83 % compared to non-targeted controls. Further, RNAi knockdown of IL17RD decreased the adhesion of THP-1 monocytic cells onto a monolayer of aortic endothelial cells in response to IL17A. Additionally, we determined that IL17A did not significantly enhance the activation of canonical MAPK and NFκB pathways in endothelial cells, and further did not significantly affect the expression of VCAM-1 and ICAM-1 in aortic endothelial cells, which is contrary to previous findings. We also determined the functional relevance of our findings in vivo by comparing the expression of endothelial VCAM-1 and ICAM-1 and leukocyte infiltration in the aorta in Western diet-fed Il17rd null versus wild-type mice. Our results showed that although Il17rd null mice do not have significant alteration in aortic expression of VCAM-1 and ICAM-1 in endothelial cells, they exhibit decreased accumulation of proinflammatory monocytes and neutrophils, suggesting that endothelial IL17RD induced in vivo myeloid cell accumulation is not dependent on upregulation of VCAM-1 and ICAM-1 expression. We further performed proteomics analysis to identify potential molecular mediators of the IL17A/IL17RD signaling axis. Collectively, our results underscore a critical role for Il17rd in the regulation of aortic myeloid cell infiltration in the context of Western diet feeding.


Assuntos
Células Endoteliais , Molécula 1 de Adesão Intercelular , Humanos , Animais , Camundongos , Molécula 1 de Adesão Intercelular/metabolismo , Células Endoteliais/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Dieta Ocidental , Aorta/metabolismo , Células Mieloides/metabolismo , Monócitos/metabolismo , Adesão Celular , Receptores de Interleucina/metabolismo
2.
Transfusion ; 63 Suppl 3: S168-S176, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37070378

RESUMO

INTRODUCTION: Tranexamic acid (TXA) is widely used as an antifibrinolytic agent in hemorrhagic trauma patients. The beneficial effects of TXA exceed the suppression of blood loss and include the ability to decrease inflammation and edema. We found that TXA suppresses the release of mitochondrial DNA and enhances mitochondrial respiration. These results allude that TXA could operate through plasmin-independent mechanisms. To address this hypothesis, we compared the effects of TXA on lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines in plasminogen (Plg) null and Plg heterozygous mice. METHODS: Plg null and Plg heterozygous mice were injected with LPS and TXA or LPS only. Four hours later, mice were sacrificed and total RNA was prepared from livers and hearts. Real time quantitative polymerase chain reaction with specific primers was used to assess the effects of LPS and TXA on the expression of pro-inflammatory cytokines. RESULTS: LPS enhanced the expression of Tnfα in the livers and hearts of recipient mice. The co-injection of TXA significantly decreased the effect of LPS both in Plg null and heterozygous mice. A similar trend was observed with LPS-induced Il1α expression in hearts and livers. CONCLUSIONS: The effects of TXA on the endotoxin-stimulated expression of Tnfα and Il1α in mice do not depend on the inhibition of plasmin generation. These results indicate that TXA has other biologically important target(s) besides plasminogen/plasmin. Fully understanding the molecular mechanisms behind the extensive beneficial effects of TXA and future identification of its targets may lead to improvement in the use of TXA in trauma, cardiac, and orthopedic surgical patients.


Assuntos
Antifibrinolíticos , Ácido Tranexâmico , Camundongos , Animais , Ácido Tranexâmico/farmacologia , Fibrinolisina , Fibrinolíticos , Endotoxinas , Fator de Necrose Tumoral alfa/genética , Lipopolissacarídeos/farmacologia , Antifibrinolíticos/farmacologia , Plasminogênio/genética , Plasminogênio/metabolismo , Hemorragia
3.
Transfusion ; 62 Suppl 1: S301-S312, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35834488

RESUMO

Tranexamic acid (TXA) is a popular antifibrinolytic drug widely used in hemorrhagic trauma patients and cardiovascular, orthopedic, and gynecological surgical patients. TXA binds plasminogen and prevents its maturation to the fibrinolytic enzyme plasmin. A number of studies have demonstrated the broad life-saving effects of TXA in trauma, superior to those of other antifibrinolytic agents. Besides preventing fibrinolysis and blood loss, TXA has been reported to suppress posttraumatic inflammation and edema. Although the efficiency of TXA transcends simple inhibition of fibrinolysis, little is known about its mechanisms of action besides the suppression of plasmin maturation. Understanding the broader effects of TXA at the cell, organ, and organism levels are required to elucidate its potential mechanisms of action transcending antifibrinolytic activity. In this article, we provide a brief review of the current clinical use of TXA and then focus on the effects of TXA beyond antifibrinolytics such as its anti-inflammatory activity, protection of the endothelial and epithelial monolayers, stimulation of mitochondrial respiration, and suppression of melanogenesis.


Assuntos
Antifibrinolíticos , Transtornos da Coagulação Sanguínea , Ácido Tranexâmico , Antifibrinolíticos/farmacologia , Antifibrinolíticos/uso terapêutico , Fibrinolisina/farmacologia , Fibrinolisina/uso terapêutico , Fibrinólise , Hemorragia , Humanos , Ácido Tranexâmico/farmacologia , Ácido Tranexâmico/uso terapêutico
4.
J Mol Cell Cardiol ; 152: 1-16, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33259856

RESUMO

BACKGROUND: Myeloid cells play an important role in a wide variety of cardiovascular disorders, including both ischemic and non-ischemic cardiomyopathies. Neuregulin-1 (NRG-1)/ErbB signaling has recently emerged as an important factor contributing to the control of inflammatory activation of myeloid cells after an ischemic injury. However, the role of ErbB signaling in myeloid cells in non-ischemic cardiomyopathy is not fully understood. This study investigated the role of ErbB3 receptors in the regulation of early adaptive response using a mouse model of transverse aortic constriction (TAC) for non-ischemic cardiomyopathy. METHODS AND RESULTS: TAC surgery was performed in groups of age- and sex-matched myeloid cell-specific ErbB3-deficient mice (ErbB3MyeKO) and control animals (ErbB3MyeWT). The number of cardiac CD45 immune cells, CD11b myeloid cells, Ly6G neutrophils, and Ly6C monocytes was determined using flow cytometric analysis. Five days after TAC, survival was dramatically reduced in male but not female ErbB3MyeKO mice or control animals. The examination of lung weight to body weight ratio suggested that acute pulmonary edema was present in ErbB3MyeKO male mice after TAC. To determine the cellular and molecular mechanisms involved in the increased mortality in ErbB3MyeKO male mice, cardiac cell populations were examined at day 3 post-TAC using flow cytometry. Myeloid cells accumulated in control but not in ErbB3MyeKO male mouse hearts. This was accompanied by increased proliferation of Sca-1 positive non-immune cells (endothelial cells and fibroblasts) in control but not ErbB3MyeKO male mice. No significant differences in intramyocardial accumulation of myeloid cells or proliferation of Sca-1 cells were found between the groups of ErbB3MyeKO and ErbB3MyeWT female mice. An antibody-based protein array analysis revealed that IGF-1 expression was significantly downregulated only in ErbB3MyeKO mice hearts compared to control animals after TAC. CONCLUSION: Our data demonstrate the crucial role of myeloid cell-specific ErbB3 signaling in the cardiac accumulation of myeloid cells, which contributes to the activation of cardiac endothelial cells and fibroblasts and development of an early adaptive response to cardiac pressure overload in male mice.


Assuntos
Adaptação Fisiológica , Cardiomegalia/prevenção & controle , Modelos Animais de Doenças , Hipertrofia Ventricular Esquerda/prevenção & controle , Células Mieloides/imunologia , Receptor ErbB-3/fisiologia , Animais , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Feminino , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Masculino , Camundongos , Camundongos Knockout , Células Mieloides/metabolismo
5.
Circulation ; 142(19): 1831-1847, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-32972203

RESUMO

BACKGROUND: Cardiac fibroblasts (CFs) have a central role in the ventricular remodeling process associated with different types of fibrosis. Recent studies have shown that fibroblasts do not respond homogeneously to heart injury. Because of the limited set of bona fide fibroblast markers, a proper characterization of fibroblast population heterogeneity in response to cardiac damage is lacking. The purpose of this study was to define CF heterogeneity during ventricular remodeling and the underlying mechanisms that regulate CF function. METHODS: Collagen1α1-GFP (green fluorescent protein)-positive CFs were characterized after myocardial infarction (MI) by single-cell and bulk RNA sequencing, assay for transposase-accessible chromatin sequencing, and functional assays. Swine and patient samples were studied using bulk RNA sequencing. RESULTS: We identified and characterized a unique CF subpopulation that emerges after MI in mice. These activated fibroblasts exhibit a clear profibrotic signature, express high levels of Cthrc1 (collagen triple helix repeat containing 1), and localize into the scar. Noncanonical transforming growth factor-ß signaling and different transcription factors including SOX9 are important regulators mediating their response to cardiac injury. Absence of CTHRC1 results in pronounced lethality attributable to ventricular rupture. A population of CFs with a similar transcriptome was identified in a swine model of MI and in heart tissue from patients with MI and dilated cardiomyopathy. CONCLUSIONS: We report CF heterogeneity and their dynamics during the course of MI and redefine the CFs that respond to cardiac injury and participate in myocardial remodeling. Our study identifies CTHRC1 as a novel regulator of the healing scar process and a target for future translational studies.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , RNA-Seq , Análise de Célula Única , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/genética , Fibroblastos/patologia , Humanos , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/patologia
6.
J Surg Res ; 251: 287-295, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32199337

RESUMO

BACKGROUND: The endothelial glycocalyx (EG) is involved in critical regulatory mechanisms that maintain endothelial vascular integrity. We hypothesized that prolonged cardiopulmonary bypass (CPB) may be associated with EG degradation. We performed an analysis of soluble syndecan-1 levels in relation to duration of CPB, as well as factors associated with cell stress and damage, such as mitochondrial DNA (mtDNA) and inflammation. METHODS: Blood samples from subjects undergoing cardiac surgery with CPB (n = 54) were obtained before and during surgery, 4-8 h and 24 h after completion of CPB, and on postoperative day 4. Flow cytometry was used to determine subpopulations of white blood cells. Plasma levels of mtDNA were determined using quantitative polymerase chain reaction and plasma content of shed syndecan-1 was measured. To determine whether syndecan-1 was signaling white blood cells, the effect of recombinant syndecan-1 on mobilization of neutrophils from bone marrow was tested in mice. RESULTS: CPB is associated with increased mtDNA during surgery, increased syndecan-1 blood levels at 4-8 h, and increased white blood cell count at 4-8 h and 24 h. Correlation analysis revealed significant positive associations between time on CPB and syndecan-1 (rs = 0.488, P < 0.001) and level of syndecan-1 and neutrophil count (rs = 0.351, P = 0.038) at 4-8 h. Intravenous administration of recombinant syndecan-1 in mice resulted in a 2.5-fold increase in the number of circulating neutrophils, concurrent with decreased bone marrow neutrophil number. CONCLUSIONS: Longer duration of CPB is associated with increased plasma levels of soluble syndecan-1, a signal for EG degradation, which can induce neutrophil egress from the bone marrow. Development of therapy targeting EG shedding may be beneficial in patients with prolonged CPB.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Endotélio/ultraestrutura , Glicocálix/fisiologia , Duração da Cirurgia , Idoso , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Ponte Cardiopulmonar/métodos , DNA Mitocondrial/sangue , Feminino , Humanos , Interleucina-6/sangue , Contagem de Leucócitos , Masculino , Camundongos , Pessoa de Meia-Idade , Neutrófilos/patologia , Proteínas Recombinantes/farmacologia , Sindecana-1/sangue , Sindecana-1/farmacologia
7.
J Cell Physiol ; 234(11): 19121-19129, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30941770

RESUMO

Damage-associated molecular patterns, including mitochondrial DNA (mtDNA) are released during hemorrhage resulting in the development of endotheliopathy. Tranexamic acid (TXA), an antifibrinolytic drug used in hemorrhaging patients, enhances their survival despite the lack of a comprehensive understanding of its cellular mechanisms of action. The present study is aimed to elucidate these mechanisms, with a focus on mitochondria. We found that TXA inhibits the release of endogenous mtDNA from granulocytes and endothelial cells. Furthermore, TXA attenuates the loss of the endothelial monolayer integrity induced by exogenous mtDNA. Using the Seahorse XF technology, it was demonstrated that TXA strongly stimulates mitochondrial respiration. Studies using Mitotracker dye, cells derived from mito-QC mice, and the ActivSignal IPAD assay, indicate that TXA stimulates biogenesis of mitochondria and inhibits mitophagy. These findings open the potential for improvement of the strategies of TXA applications in trauma patients and the development of more efficient TXA derivatives.


Assuntos
DNA Mitocondrial/efeitos dos fármacos , Hemorragia/tratamento farmacológico , Ácido Tranexâmico/farmacologia , Ferimentos e Lesões/tratamento farmacológico , Animais , Dano ao DNA/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Granulócitos/efeitos dos fármacos , Hemorragia/genética , Hemorragia/patologia , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Ferimentos e Lesões/genética , Ferimentos e Lesões/patologia
8.
J Cell Biochem ; 119(3): 2636-2645, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29058774

RESUMO

Highly variable expression of mesoderm-specific transcript (Mest) in adipose tissue among genetically homogeneous mice fed an obesogenic diet, and its positive association with fat mass expansion, suggests that Mest is an epigenetic determinant for the development of obesity. Although the mechanisms by which MEST augments fat accumulation in adipocytes have not been elucidated, it has sequence homology and catalytic peptide motifs which suggests that it functions as an epoxide hydrolase or as a glycerol- or acylglycerol-3-phosphate acyltransferase. To better understand MEST function, detailed studies were performed to precisely define the intracellular organelle localization of MEST using immunofluorescence confocal microscopy. Lentiviral-mediated expression of a C-terminus Myc-DDK-tagged MEST fusion protein expressed in 3T3-L1 preadipocytes/adipocytes, and ear-derived mesenchymal stem cells (EMSC) from mice was observed in the endoplasmic reticulum (ER) membranes and is consistent with previous studies showing endogenous MEST in the membrane fraction of adipose tissue. MEST was not associated with the Golgi apparatus or mitochondria; however, frequent contacts were observed between MEST-positive ER and mitochondria. MEST-positive domains were also shown on the plasma membrane (PM) of non-permeabilized cells but they did not co-localize with ER-PM bridges. Post-adipogenic differentiated 3T3-L1 adipocytes and EMSC showed significant co-localization of MEST with the lipid droplet surface marker perilipin at contact points between the ER and lipid droplet. Identification of MEST as an ER-specific protein that co-localizes with lipid droplets in cells undergoing adipogenic differentiation supports a function for MEST in the facilitation of lipid accumulation and storage in adipocytes.


Assuntos
Adipócitos/metabolismo , Retículo Endoplasmático/metabolismo , Gotículas Lipídicas/metabolismo , Proteínas/metabolismo , Células 3T3-L1 , Adipócitos/patologia , Animais , Diferenciação Celular/fisiologia , Hipertrofia/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Knockout , Obesidade/metabolismo
9.
Biochemistry ; 55(7): 1159-67, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26836284

RESUMO

Fibroblast growth factor 1 (FGF1), a ubiquitously expressed pro-angiogenic protein that is involved in tissue repair, carcinogenesis, and maintenance of vasculature stability, is released from the cells via a stress-dependent nonclassical secretory pathway. FGF1 secretion is a result of transmembrane translocation of this protein. It correlates with the ability of FGF1 to permeabilize membranes composed of acidic phospholipids. Like several other nonclassically exported proteins, FGF1 exhibits ß-barrel folding. To assess the role of folding of FGF1 in its secretion, we applied targeted mutagenesis in combination with a complex of biophysical methods and molecular dynamics studies, followed by artificial membrane permeabilization and stress-induced release experiments. It has been demonstrated that a mutation of proline 135 located in the C-terminus of FGF1 results in (i) partial unfolding of FGF1, (ii) a decrease in FGF1's ability to permeabilize bilayers composed of phosphatidylserine, and (iii) drastic inhibition of stress-induced FGF1 export. Thus, folding of FGF1 is critical for its nonclassical secretion.


Assuntos
Permeabilidade da Membrana Celular , Fator 1 de Crescimento de Fibroblastos/química , Modelos Moleculares , Dobramento de Proteína , Substituição de Aminoácidos , Animais , Varredura Diferencial de Calorimetria , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Cinética , Bicamadas Lipídicas/química , Membranas Artificiais , Camundongos , Simulação de Dinâmica Molecular , Mutação , Células NIH 3T3 , Permeabilidade , Fosfatidilserinas/química , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
10.
J Cell Physiol ; 231(3): 650-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26218437

RESUMO

Transient FGF stimulation of various cell types results in FGF memory--a sustained blockage of efficient proliferative response to FGF and other growth factors. FGF memory establishment requires HDAC activity, indicating its epigenetic character. FGF treatment stimulates proinflammatory NFκB signaling, which is also critical for FGF memory formation. The search for FGF-induced mediators of FGF memory revealed that FGF stimulates HDAC-dependent expression of the inflammatory cytokine IL1α. Similarly to FGF, transient cell treatment with recombinant IL1α inhibits the proliferative response to further FGF and EGF stimulation, but does not prevent FGF receptor-mediated signaling. Interestingly, like cells pretreated with FGF1, cells pretreated with IL1α exhibit enhanced restructuring of actin cytoskeleton and increased migration in response to FGF stimulation. IRAP, a specific inhibitor of IL 1 receptor, and a neutralizing anti-IL1α antibody prevent the formation of FGF memory and rescue an efficient proliferative response to FGF restimulation. A similar effect results following treatment with the anti-inflammatory agents aspirin and dexamethasone. Thus, FGF memory is mediated by proinflammatory IL1 signaling. It may play a role in the limitation of proliferative response to tissue damage and prevention of wound-induced hyperplasia.


Assuntos
Anti-Inflamatórios/farmacologia , Proliferação de Células/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Interleucina-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas , Fatores de Crescimento de Fibroblastos/metabolismo , Camundongos , NF-kappa B/metabolismo
11.
J Cell Biochem ; 117(9): 2182-93, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26910604

RESUMO

Our objective was to characterize lipid profiles in cell models of adipocyte differentiation in comparison to mouse adipose tissues in vivo. A novel lipid extraction strategy was combined with global lipid profiling using direct infusion and sequential precursor ion fragmentation, termed MS/MS(ALL) . Perirenal and inguinal white adipose tissue and interscapular brown adipose tissues from adult C57BL/6J mice were analyzed. 3T3-L1 preadipocytes, ear mesenchymal progenitor cells, and brown adipose-derived BAT-C1 cells were also characterized. Over 3000 unique lipid species were quantified. Principal component analysis showed that perirenal versus inguinal white adipose tissues varied in lipid composition of triacyl- and diacylglycerols, sphingomyelins, glycerophospholipids and, notably, cardiolipin CL 72:3. In contrast, hexosylceramides and sphingomyelins distinguished brown from white adipose. Adipocyte differentiation models showed broad differences in lipid composition among themselves, upon adipogenic differentiation, and with adipose tissues. Palmitoyl triacylglycerides predominate in 3T3-L1 differentiation models, whereas cardiolipin CL 72:1 and SM 45:4 were abundant in brown adipose-derived cell differentiation models, respectively. MS/MS(ALL) data suggest new lipid biomarkers for tissue-specific lipid contributions to adipogenesis, thus providing a foundation for using in vitro models of adipogenesis to reflect potential changes in adipose tissues in vivo. J. Cell. Biochem. 117: 2182-2193, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Adipócitos Brancos/metabolismo , Adipogenia/fisiologia , Tecido Adiposo Branco/metabolismo , Metabolismo dos Lipídeos/fisiologia , Modelos Biológicos , Células 3T3-L1 , Adipócitos Brancos/citologia , Tecido Adiposo Branco/citologia , Animais , Camundongos
12.
Biochim Biophys Acta ; 1844(12): 2155-63, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25224745

RESUMO

Fibroblast growth factor 1 (FGF1) is a heparin-binding proangiogenic protein. FGF1 lacks the conventional N-terminal signal peptide required for secretion through the endoplasmic reticulum (ER)-Golgi secretory pathway. FGF1 is released through a Cu(2+)-mediated nonclassical secretion pathway. The secretion of FGF1 involves the formation of a Cu(2+)-mediated multiprotein release complex (MRC) including FGF1, S100A13 (a calcium-binding protein) and p40 synaptotagmin (Syt1). It is believed that the binding of Cu(2+) to the C2B domain is important for the release of FGF1 into the extracellular medium. In this study, using a variety of biophysical studies, Cu(2+) and lipid interactions of the C2B domain of Syt1 were characterized. Isothermal titration calorimetry (ITC) experiments reveal that the C2B domain binds to Cu(2+) in a biphasic manner involving an initial endothermic and a subsequent exothermic phase. Fluorescence energy transfer experiments using Tb(3+) show that there are two Cu(2+)-binding pockets on the C2B domain, and one of these is also a Ca(2+)-binding site. Lipid-binding studies using ITC demonstrate that the C2B domain preferentially binds to small unilamellar vesicles of phosphatidyl serine (PS). Results of the differential scanning calorimetry and limited trypsin digestion experiments suggest that the C2B domain is marginally destabilized upon binding to PS vesicles. These results, for the first time, suggest that the main role of the C2B domain of Syt1 is to serve as an anchor for the FGF1 MRC on the membrane bilayer. In addition, the binding of the C2B domain to the lipid bilayer is shown to significantly decrease the binding affinity of the protein to Cu(2+). The study provides valuable insights on the sequence of structural events that occur in the nonclassical secretion of FGF1.

13.
J Cell Biochem ; 116(8): 1522-31, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25560297

RESUMO

FGF1 is a nonclassically released growth factor that regulates carcinogenesis, angiogenesis, and inflammation. In vitro and in vivo, FGF1 export is stimulated by cell stress. Upon stress, FGF1 is transported to the plasma membrane where it localizes prior to transmembrane translocation. To determine which proteins participate in the submembrane localization of FGF1 and its export, we used immunoprecipitation mass spectrometry to identify novel proteins that associate with FGF1 during heat shock. The heat shock-dependent association of FGF1 with the large protein AHNAK2 was observed. Heat shock induced the translocation of FGF1 and AHNAK2 to the cytoskeletal fraction. In heat-shocked cells, FGF1 and the C-terminal fragment of AHNAK2 colocalized with F-actin in the vicinity of the cell membrane. Depletion of AHNAK2 resulted in a drastic decrease of stress-induced FGF1 export but did not affect spontaneous FGF2 export and FGF1 release induced by the inhibition of Notch signaling. Thus, AHNAK2 is an important element of the FGF1 nonclassical export pathway.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Via Secretória , Estresse Fisiológico , Actinas/metabolismo , Animais , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/química , Humanos , Espectrometria de Massas , Camundongos , Células NIH 3T3 , Temperatura
14.
Curr Atheroscler Rep ; 17(6): 509, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25813213

RESUMO

Despite their discovery as angiogenic factors and mitogens for endothelial cells more than 30 years ago, much remains to be determined about the role of fibroblast growth factors (FGFs) and their receptors in vascular development, homeostasis, and disease. In vitro studies show that members of the FGF family stimulate growth, migration, and sprouting of endothelial cells, and growth, migration, and phenotypic plasticity of vascular smooth muscle cells. Recent studies have revealed important roles for FGFs and their receptors in the regulation of endothelial cell sprouting and vascular homeostasis in vivo. Furthermore, recent work has revealed roles for FGFs in atherosclerosis, vascular calcification, and vascular dysfunction. The large number of FGFs and their receptors expressed in endothelial and vascular smooth muscle cells complicates these studies. In this review, we summarize recent studies in which new and unanticipated roles for FGFs and their receptors in the vasculature have been revealed.


Assuntos
Células Endoteliais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/fisiologia , Animais , Vasos Sanguíneos/metabolismo , Humanos
15.
J Biol Chem ; 288(4): 2721-33, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23209292

RESUMO

IL-1ß is a potent pro-inflammatory cytokine produced in response to infection or injury. It is synthesized as an inactive precursor that is activated by the protease caspase-1 within a cytosolic molecular complex called the inflammasome. Assembly of this complex is triggered by a range of structurally diverse damage or pathogen associated stimuli, and the signaling pathways through which these act are poorly understood. Ubiquitination is a post-translational modification essential for maintaining cellular homeostasis. It can be reversed by deubiquitinase enzymes (DUBs) that remove ubiquitin moieties from the protein thus modifying its fate. DUBs present specificity toward different ubiquitin chain topologies and are crucial for recycling ubiquitin molecules before protein degradation as well as regulating key cellular processes such as protein trafficking, gene transcription, and signaling. We report here that small molecule inhibitors of DUB activity inhibit inflammasome activation. Inhibition of DUBs blocked the processing and release of IL-1ß in both mouse and human macrophages. DUB activity was necessary for inflammasome association as DUB inhibition also impaired ASC oligomerization and caspase-1 activation without directly blocking caspase-1 activity. These data reveal the requirement for DUB activity in a key reaction of the innate immune response and highlight the therapeutic potential of DUB inhibitors for chronic auto-inflammatory diseases.


Assuntos
Caspase 1/metabolismo , Endopeptidases/fisiologia , Interleucina-1beta/metabolismo , Animais , Carboxipeptidases/metabolismo , Endopeptidases/química , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Hidrazonas/farmacologia , Hidroxiureia/análogos & derivados , Hidroxiureia/farmacologia , Imunidade Inata , Inflamação , Interleucina-1alfa/metabolismo , Interleucinas/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Ubiquitina Tiolesterase
16.
J Cell Biochem ; 115(5): 874-88, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24375433

RESUMO

FGF applied as a single growth factor to quiescent mouse fibroblasts induces a round of DNA replication, however continuous stimulation results in arrest in the G1 phase of the next cell cycle. We hypothesized that FGF stimulation induces the establishment of cell memory, which prevents the proliferative response to repeated or continuous FGF application. When a 2-5 days quiescence period was introduced between primary and repeated FGF treatments, fibroblasts failed to efficiently replicate in response to secondary FGF application. The establishment of "FGF memory" during the first FGF stimulation did not require DNA synthesis, but was dependent on the activity of FGF receptors, MEK, p38 MAPK and NFκB signaling, and protein synthesis. While secondary stimulation resulted in strongly decreased replication rate, we did not observe any attenuation of morphological changes, Erk1/2 phosphorylation and cyclin D1 induction. However, secondary FGF stimulation failed to induce the expression of cyclin A, which is critical for the progression from G1 to S phase. Treatment of cells with a broad range histone deacetylase inhibitor during the primary FGF stimulation rescued the proliferative response to the secondary FGF treatment suggesting that the establishment of "FGF memory" may be based on epigenetic changes. We suggest that "FGF memory" can prevent the hyperplastic response to cell damage and inflammation, which are associated with an enhanced FGF production and secretion. "FGF memory" may present a natural obstacle to the efficient application of recombinant FGFs for the treatment of ulcers, ischemias, and wounds.


Assuntos
Ciclo Celular/genética , Movimento Celular/genética , Proliferação de Células , Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Ciclina D1/genética , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Fatores de Crescimento de Fibroblastos/administração & dosagem , Fase G1/genética , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Burns ; 50(4): 947-956, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38336496

RESUMO

Burn wound conversion is the observed process where superficial partial thickness burns convert into deep partial or full thickness burn injuries. This conversion process often involves surgical excision to achieve timely wound healing. Unfortunately, the pathophysiology of this phenomenon is multifactorial and poorly understood. Thus, a therapeutic intervention that may prevent secondary progression and cell death in burn-injured tissue is desirable. Recent work by our group and others has established that tranexamic acid (TXA) has significant anti-inflammatory properties in addition to its well-known anti-fibrinolytic effects. This study investigates TXA as a novel therapeutic treatment to mitigate burn wound conversion and reduce systemic inflammation. Sprague-Dawley rats were subjected to a hot comb burn contact injury. A subset of animals underwent a similar comb burn with an adjacent 30%TBSA contact injury. The interspaces represent the ischemic zones simulating the zone of stasis. The treatment group received injections of TXA (100 mg/kg) immediately after injury and once daily until euthanasia. Animals were harvested for analyses at 6 h and 7 days after injury. Full-thickness biopsies from the ischemic zones and lung tissue were assessed with established histological techniques. Plasma was collected for measurement of damage associated molecular patterns (DAMPs), and liver samples were used to study inflammatory cytokines expression. Treatment with TXA was associated with reduced burn wound conversion and decreased burn-induced systemic inflammatory response syndrome (SIRS). Lung inflammation and capillary leak were also significantly reduced in TXA treated animals. Future research will elucidate the underlying anti-inflammatory properties of TXA responsible for these findings.


Assuntos
Antifibrinolíticos , Queimaduras , Modelos Animais de Doenças , Edema , Inflamação , Ratos Sprague-Dawley , Ácido Tranexâmico , Animais , Ácido Tranexâmico/farmacologia , Ácido Tranexâmico/uso terapêutico , Queimaduras/tratamento farmacológico , Queimaduras/complicações , Queimaduras/patologia , Ratos , Antifibrinolíticos/farmacologia , Antifibrinolíticos/uso terapêutico , Inflamação/tratamento farmacológico , Edema/tratamento farmacológico , Masculino , Cicatrização/efeitos dos fármacos , Pele/efeitos dos fármacos , Pele/patologia , Pele/lesões , Fígado/efeitos dos fármacos , Fígado/patologia , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo
18.
Int J Cancer ; 133(11): 2577-86, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729168

RESUMO

Prostate cancer (PC) is still the second cause of cancer-related death among men. Although patients with metastatic presentation have an ominous outcome, the vast majority of PCs are diagnosed at an early stage. Nonetheless, even among patients with clinically localized disease the outcome may vary considerably. Other than androgen sensitivity, little is known about which other signaling pathways are deranged in aggressive, localized cancers. The elucidation of such pathways may help to develop innovative therapies aimed at specific molecular targets. We report that in a hormone-sensitive PC cell line, LNCaP, Notch3 was activated by hypoxia and sustained cell proliferation and colony formation in soft agar. Hypoxia also modulated cellular cholesterol content and the number and size of lipid rafts, causing a coalescence of small rafts into bigger clusters; under this experimental condition, Notch3 migrated from the non-raft into the raft compartment where it colocalized with the γ-secretase complex. We also looked at human PC biopsies and found that expression of Notch3 positively correlated with Gleason score and with expression of carbonic anhydrase IX, a marker of hypoxia. In conclusion, hypoxia triggers the activation of Notch3, which, in turn, sustains proliferation of PC cells. Notch3 pathway represents a promising target for adjuvant therapy in patients with PC.


Assuntos
Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Notch/genética , Antígenos de Neoplasias/metabolismo , Biópsia , Anidrase Carbônica IX , Anidrases Carbônicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Colesterol/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Masculino , Microdomínios da Membrana/metabolismo , Terapia de Alvo Molecular , Estadiamento de Neoplasias , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/patologia , Neoplasias da Próstata/genética , Receptor Notch3 , Receptores Notch/biossíntese
19.
Angiogenesis ; 16(4): 861-75, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23955631

RESUMO

Angiogenesis is mediated by signaling through receptor tyrosine kinases (RTKs), Src family kinases and adhesion receptors such as integrins, yet the mechanism how these signaling pathways regulate one another remains incompletely understood. The RTK modulator, Sprouty4 (Spry4) inhibits endothelial cell functions and angiogenesis, but the mechanisms remain to be fully elucidated. In this study, we demonstrate that Spry4 regulates angiogenesis in part by regulating endothelial cell migration. Overexpression of Spry4 in human endothelial cells inhibited migration and adhesion on vitronectin (VTN), whereas knockdown of Spry4 enhanced these behaviors. These activities were shown to be c-Src-dependent and Ras-independent. Spry4 disrupted the crosstalk between vascular endothelial growth factor-2 and integrin αVß3, the receptor for VTN. Spry4 overexpression resulted in decreased integrin ß3 protein levels in a post-transcriptional manner in part by modulating its tyrosine phosphorylation by c-Src. Conversely, knockdown of Spry4 resulted in increased integrin ß3 protein levels and tyrosine phosphorylation. Moreover, in vivo analysis revealed that Spry4 regulated integrin ß3 levels in murine embryos and yolk sacs. Our findings identify an unanticipated role for Spry4 in regulating c-Src activity and integrin ß3 protein levels, which contributes to the regulation of migration and adhesion of endothelial cells. Thus, targeting Spry4 may be exploited as a target in anti-angiogenesis therapies.


Assuntos
Células Endoteliais/citologia , Integrina beta3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neovascularização Fisiológica/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Quinases da Família src/metabolismo , Animais , Aorta/citologia , Proteína Tirosina Quinase CSK , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Embrião de Mamíferos/citologia , Células Endoteliais/metabolismo , Ativação Enzimática , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina alfaVbeta3/fisiologia , Integrina beta3/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fosforilação , Fosfotirosina/metabolismo , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vasos Retinianos/crescimento & desenvolvimento , Regulação para Cima , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Vitronectina/metabolismo , Saco Vitelino/citologia
20.
Langmuir ; 29(25): 7966-74, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23705855

RESUMO

Model cellular membranes enable the study of biological processes in a controlled environment and reduce the traditional challenges associated with live or fixed cell studies. However, model membrane systems based on the air/water or oil/solution interface do not allow for incorporation of transmembrane proteins or for the study of protein transport mechanisms. Conversely, a phospholipid bilayer deposited via the Langmuir-Blodgett/Langmuir-Schaefer method on a hydrogel layer is potentially an effective mimic of the cross section of a biological membrane and facilitates both protein incorporation and transport studies. Prior to application, however, such membranes must be fully characterized, particularly with respect to the phospholipid bilayer phase transition temperature. Here we present a detailed characterization of the phase transition temperature of the inner and outer leaflets of a chitosan supported model membrane system. Specifically, the lateral diffusion coefficient of each individual leaflet has been determined as a function of temperature. Measurements were performed utilizing z-scan fluorescence correlation spectroscopy (FCS), a technique that yields calibration-free diffusion information. Analysis via the method of Wawrezinieck and co-workers revealed that phospholipid diffusion changes from raftlike to free diffusion as the temperature is increased-an insight into the dynamic behavior of hydrogel supported membranes not previously reported.


Assuntos
Membranas Artificiais , Fosfolipídeos/química , Espectrometria de Fluorescência/métodos , Difusão
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa