Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nat Commun ; 15(1): 2476, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509088

RESUMEN

Cellular entry of the hepatitis B and D viruses (HBV/HDV) requires binding of the viral surface polypeptide preS1 to the hepatobiliary transporter Na+-taurocholate co-transporting polypeptide (NTCP). This interaction can be blocked by bulevirtide (BLV, formerly Myrcludex B), a preS1 derivative and approved drug for treating HDV infection. Here, to elucidate the basis of this inhibitory function, we determined a cryo-EM structure of BLV-bound human NTCP. BLV forms two domains, a plug lodged in the bile salt transport tunnel of NTCP and a string that covers the receptor's extracellular surface. The N-terminally attached myristoyl group of BLV interacts with the lipid-exposed surface of NTCP. Our structure reveals how BLV inhibits bile salt transport, rationalizes NTCP mutations that decrease the risk of HBV/HDV infection, and provides a basis for understanding the host specificity of HBV/HDV. Our results provide opportunities for structure-guided development of inhibitors that target HBV/HDV docking to NTCP.


Asunto(s)
Hepatitis B , Lipopéptidos , Simportadores , Humanos , Virus de la Hepatitis B/fisiología , Antivirales/uso terapéutico , Receptores Virales/metabolismo , Ácidos y Sales Biliares/metabolismo , Virus de la Hepatitis Delta/fisiología , Simportadores/metabolismo , Internalización del Virus , Hepatocitos/metabolismo
2.
Biol Chem ; 404(7): 673-690, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37103224

RESUMEN

Na+/taurocholate cotransporting polypeptide (NTCP) is a member of the solute carrier (SLC) family 10 transporters (gene symbol SLC10A1) and is responsible for the sodium-dependent uptake of bile salts across the basolateral membrane of hepatocytes. In addition to its primary transporter function, NTCP is the high-affinity hepatic receptor for hepatitis B (HBV) and hepatitis D (HDV) viruses and, therefore, is a prerequisite for HBV/HDV virus entry into hepatocytes. The inhibition of HBV/HDV binding to NTCP and internalization of the virus/NTCP receptor complex has become a major concept in the development of new antiviral drugs called HBV/HDV entry inhibitors. Hence, NTCP has emerged as a promising target for therapeutic interventions against HBV/HDV infections in the last decade. In this review, recent findings on protein-protein interactions (PPIs) between NTCP and cofactors relevant for entry of the virus/NTCP receptor complex are summarized. In addition, strategies aiming to block PPIs with NTCP to dampen virus tropism and HBV/HDV infection rates are discussed. Finally, this article suggests novel directions for future investigations evaluating the functional contribution of NTCP-mediated PPIs in the development and progression of HBV/HDV infection and subsequent chronic liver disorders.


Asunto(s)
Hepatitis B , Simportadores , Humanos , Antivirales/farmacología , Células Hep G2 , Hepatitis B/tratamiento farmacológico , Hepatitis B/metabolismo , Virus de la Hepatitis B , Virus de la Hepatitis Delta/metabolismo , Hepatocitos/metabolismo , Péptidos , Simportadores/metabolismo , Ácido Taurocólico/metabolismo , Ácido Taurocólico/uso terapéutico , Internalización del Virus
3.
Viruses ; 14(6)2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35746730

RESUMEN

Na+/taurocholate cotransporting polypeptide (NTCP, gene symbol SLC10A1) is a hepatic bile acid uptake carrier participating in the enterohepatic circulation of bile acids. Apart from its transporter function, NTCP acts as the high-affinity liver-specific receptor for the hepatitis B virus (HBV), which attaches via its preS1-peptide domain of the large surface protein to NTCP, subsequently leading to endocytosis of the virus/NTCP-receptor complex. Although the process of NTCP-dependent HBV infection of hepatocytes has received much attention over the last decade, the precise molecular sites of the virus/NTCP interaction have not been fully identified. Inspection of the primary protein sequence of human NTCP revealed 139YIYSRGIY146 as a highly conserved tyrosine-rich motif. To study the role of Y139, Y141 and Y146 amino acids in NTCP biology, the aforementioned residues were substituted with alanine, phenylalanine or glutamate (mimicking phosphorylation) using site-directed mutagenesis. Similar to wt NTCP, the Y139A, Y141A, Y146A, Y141F, Y146F, and Y146E mutants were expressed at the plasma membrane of HEK293 cells and exhibited intact bile acid transport function. Y146A, Y146E, and Y146F demonstrated transport kinetics comparable to wild-type NTCP with Km values of 57.3-112.4 µM and Vmax values of 6683-7579 pmol/mg protein/min. Only Y141E was transport deficient, most likely due to an intracellular accumulation of the mutant protein. Most importantly, Y146A and Y146E mutation completely abrogated binding of the viral preS1-peptide to NTCP, while the Y146F mutant of NTCP showed some residual binding competence for preS1. Consequently, the NTCP mutants Y146A and Y146E, when expressed in HepG2 hepatoma cells, showed complete loss of susceptibility for in vitro HBV infection. In conclusion, tyrosine 146, and to some extent tyrosine 141, both belonging to the tyrosine-rich motif 139YIYSRGIY146 of human NTCP, are newly identified amino acid residues that play an essential role in the interaction of HBV with its receptor NTCP and, thus, in the process of virus entry into hepatocytes.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Ácidos y Sales Biliares/metabolismo , Células HEK293 , Células Hep G2 , Virus de la Hepatitis B/fisiología , Hepatocitos , Humanos , Receptores Virales/metabolismo , Ácido Taurocólico , Tirosina/metabolismo , Internalización del Virus
4.
Biomedicines ; 10(1)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35052874

RESUMEN

Hepatitis B virus (HBV) infections are among the major public health concerns worldwide with more than 250 million of chronically ill individuals. Many of them are additionally infected with the Hepatitis D virus, a satellite virus to HBV. Chronic infection frequently leads to serious liver diseases including cirrhosis and hepatocellular carcinoma, the most common type of liver cancer. Although current antiviral therapies can control HBV replication and slow down disease progress, there is an unmet medical need to identify therapies to cure this chronic infectious disease. Lately, a noteworthy progress in fighting against HBV has been made by identification of the high-affinity hepatic host receptor for HBV and HDV, namely Na+/taurocholate cotransporting polypeptide (NTCP, gene symbol SLC10A1). Next to its primary function as hepatic uptake transporter for bile acids, NTCP is essential for the cellular entry of HBV and HDV into hepatocytes. Due to this high-ranking discovery, NTCP has become a valuable target for drug development strategies for HBV/HDV-infected patients. In this review, we will focus on a newly predicted three-dimensional NTCP model that was generated using computational approaches and discuss its value in understanding the NTCP's membrane topology, substrate and virus binding taking place in plasma membranes. We will review existing data on structural, functional, and biological consequences of amino acid residue changes and mutations that lead to loss of NTCP's transport and virus receptor functions. Finally, we will discuss new directions for future investigations aiming at development of new NTCP-based HBV entry blockers that inhibit HBV tropism in human hepatocytes.

5.
Front Immunol ; 10: 664, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31019507

RESUMEN

Interleukin-1ß (IL-1ß) is a potent, pro-inflammatory cytokine of the innate immune system that plays an essential role in host defense against infection. However, elevated circulating levels of IL-1ß can cause life-threatening systemic inflammation. Hence, mechanisms controlling IL-1ß maturation and release are of outstanding clinical interest. Secretory leukocyte protease inhibitor (SLPI), in addition to its well-described anti-protease function, controls the expression of several pro-inflammatory cytokines on the transcriptional level. In the present study, we tested the potential involvement of SLPI in the control of ATP-induced, inflammasome-dependent IL-1ß maturation and release. We demonstrated that SLPI dose-dependently inhibits the ATP-mediated inflammasome activation and IL-1ß release in human monocytic cells, without affecting the induction of pro-IL-1ß mRNA by LPS. In contrast, the ATP-independent IL-1ß release induced by the pore forming bacterial toxin nigericin is not impaired, and SLPI does not directly modulate the ion channel function of the human P2X7 receptor heterologously expressed in Xenopus laevis oocytes. In human monocytic U937 cells, however, SLPI efficiently inhibits ATP-induced ion-currents. Using specific inhibitors and siRNA, we demonstrate that SLPI activates the calcium-independent phospholipase A2ß (iPLA2ß) and leads to the release of a low molecular mass factor that mediates the inhibition of IL-1ß release. Signaling involves nicotinic acetylcholine receptor subunits α7, α9, α10, and Src kinase activation and results in an inhibition of ATP-induced caspase-1 activation. In conclusion, we propose a novel anti-inflammatory mechanism induced by SLPI, which inhibits the ATP-dependent maturation and secretion of IL-1ß. This novel signaling pathway might lead to development of therapies that are urgently needed for the prevention and treatment of systemic inflammation.


Asunto(s)
Adenosina Trifosfato/metabolismo , Interleucina-1beta/metabolismo , Monocitos/citología , Monocitos/metabolismo , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Animales , Línea Celular , Células Cultivadas , Citocinas/biosíntesis , Expresión Génica , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1816-1827, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29501774

RESUMEN

OBJECTIVES: Enolase-1-dependent cell surface proteolysis plays an important role in cell invasion. Although enolase-1 (Eno-1), a glycolytic enzyme, has been found on the surface of various cells, the mechanism responsible for its exteriorization remains elusive. Here, we investigated the involvement of post-translational modifications (PTMs) of Eno-1 in its lipopolysaccharide (LPS)-triggered trafficking to the cell surface. RESULTS: We found that stimulation of human lung adenocarcinoma cells with LPS triggered the monomethylation of arginine 50 (R50me) within Eno-1. The Eno-1R50me was confirmed by its interaction with the tudor domain (TD) from TD-containing 3 (TDRD3) protein recognizing methylarginines. Substitution of R50 with lysine (R50K) reduced Eno-1 association with epithelial caveolar domains, thereby diminishing its exteriorization. Similar effects were observed when pharmacological inhibitors of arginine methyltransferases were applied. Protein arginine methyltransferase 5 (PRMT5) was identified to be responsible for Eno-1 methylation. Overexpression of PRMT5 and caveolin-1 enhanced levels of membrane-bound extracellular Eno-1 and, conversely, pharmacological inhibition of PRMT5 attenuated Eno-1 cell-surface localization. Importantly, Eno-1R50me was essential for cancer cell motility since the replacement of Eno-1 R50 by lysine or the suppression of PRMT 5 activity diminished Eno-1-triggered cell invasion. CONCLUSIONS: LPS-triggered Eno-1R50me enhances Eno-1 cell surface levels and thus potentiates the invasive properties of cancer cells. Strategies to target Eno-1R50me may offer novel therapeutic approaches to attenuate tumor metastasis in cancer patients.


Asunto(s)
Adenocarcinoma/enzimología , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias Pulmonares/enzimología , Proteínas de Neoplasias/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Células A549 , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Biomarcadores de Tumor/genética , Caveolina 1/genética , Caveolina 1/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Lipopolisacáridos/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas de Neoplasias/genética , Fosfopiruvato Hidratasa/genética , Transporte de Proteínas/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Supresoras de Tumor/genética
7.
Mediators Inflamm ; 2017: 1434872, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28757683

RESUMEN

Chemokines and ATP are among the mediators of inflammatory sites that can enter the circulation via damaged blood vessels. The main function of chemokines is leukocyte mobilization, and ATP typically triggers inflammasome assembly. IL-1ß, a potent inflammasome-dependent cytokine of innate immunity, is essential for pathogen defense. However, excessive IL-1ß may cause life-threatening systemic inflammation. Here, we hypothesize that chemokines control ATP-dependent secretion of monocytic IL-1ß. Lipopolysaccharide-primed human monocytic U937 cells were stimulated with the P2X7 agonist BzATP for 30 min to induce IL-1ß release. CCL3, CCL4, and CCL5 dose dependently inhibited BzATP-stimulated release of IL-1ß, whereas CXCL16 was ineffective. The effect of CCL3 was confirmed for primary mononuclear leukocytes. It was blunted after silencing CCR1 or calcium-independent phospholipase A2 (iPLA2) by siRNA and was sensitive to antagonists of nicotinic acetylcholine receptors containing subunits α7 and α9. U937 cells secreted small factors in response to CCL3 that mediated the inhibition of IL-1ß release. We suggest that CCL chemokines inhibit ATP-induced release of IL-1ß from U937 cells by a triple-membrane-passing mechanism involving CCR, iPLA2, release of small mediators, and nicotinic acetylcholine receptor subunits α7 and α9. We speculate that whenever chemokines and ATP enter the circulation concomitantly, systemic release of IL-1ß is minimized.


Asunto(s)
Adenosina Trifosfato/farmacología , Quimiocina CCL3/farmacología , Quimiocina CCL4/farmacología , Quimiocina CCL5/farmacología , Quimiocinas/farmacología , Interleucina-1beta/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Humanos , Células U937
8.
FASEB J ; 31(5): 1916-1928, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28148565

RESUMEN

Pirfenidone is an antifibrotic drug, recently approved for the treatment of patients with idiopathic pulmonary fibrosis (IPF). Although pirfenidone exhibits anti-inflammatory, antioxidant, and antifibrotic properties, the molecular mechanism underlying its protective effects remains unknown. Here, we link pirfenidone action with the regulation of the profibrotic hedgehog (Hh) signaling pathway. We demonstrate that pirfenidone selectively destabilizes the glioma-associated oncogene homolog (GLI)2 protein, the primary activator of Hh-mediated gene transcription. Consequently, pirfenidone decreases overall Hh pathway activity in patients with IPF and in patient-derived primary lung fibroblasts and leads to diminished levels of Hh target genes, such as GLI1, Hh receptor Patched-1, α-smooth muscle actin, and fibronectin, and to reduced cell migration and proliferation. Interestingly, Hh-triggered TGF-ß1 expression potentiated Hh responsiveness of primary lung fibroblasts by elevating the available pool of glioma-associated oncogene homolog (GLI)1/GLI2, thus creating a vicious cycle of amplifying fibrotic processes. Because GLI transcription factors are not only crucial for Hh-mediated changes but are also required as mediators of TGF-ß signaling, our findings suggest that pirfenidone exerts its clinically beneficial effects through dual Hh/TGF-ß inhibition by targeting the GLI2 protein.-Didiasova, M., Singh, R., Wilhelm, J., Kwapiszewska, G., Wujak, L., Zakrzewicz, D., Schaefer, L., Markart, P., Seeger, W., Lauth, M., Wygrecka, M. Pirfenidone exerts antifibrotic effects through inhibition of GLI transcription factors.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Piridonas/farmacología , Adulto , Anciano , Femenino , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Masculino , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteína Gli2 con Dedos de Zinc
9.
Am J Respir Crit Care Med ; 196(2): 186-199, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28005404

RESUMEN

RATIONALE: Acute respiratory distress syndrome is characterized by alveolar epithelial cell injury, edema formation, and intraalveolar contact phase activation. OBJECTIVES: To explore whether C1 esterase inhibitor (C1INH), an endogenous inhibitor of the contact phase, may protect from lung injury in vivo and to decipher the possible underlying mechanisms mediating protection. METHODS: The ability of C1INH to control the inflammatory processes was studied in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: Here, we demonstrate that application of C1INH alleviates bleomycin-induced lung injury via direct interaction with extracellular histones. In vitro, C1INH was found to bind all histone types. Interaction with histones was independent of its protease inhibitory activity, as demonstrated by the use of reactive-center-cleaved C1INH, but dependent on its glycosylation status. C1INH sialylated-N- and -O-glycans were not only essential for its interaction with histones but also to protect against histone-induced cell death. In vivo, histone-C1INH complexes were detected in bronchoalveolar lavage fluid from patients with acute respiratory distress syndrome and multiple models of lung injury. Furthermore, reactive-center-cleaved C1INH attenuated pulmonary damage evoked by intravenous histone instillation. CONCLUSIONS: Collectively, C1INH administration provides a new therapeutic option for disorders associated with histone release.


Asunto(s)
Proteína Inhibidora del Complemento C1/farmacología , Histonas/metabolismo , Lesión Pulmonar/prevención & control , Síndrome de Dificultad Respiratoria/fisiopatología , Animales , Líquido del Lavado Bronquioalveolar , Proteína Inhibidora del Complemento C1/metabolismo , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , Pulmón/fisiopatología , Lesión Pulmonar/fisiopatología , Ratones , Ratones Endogámicos C57BL
10.
Sci Rep ; 6: 37758, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27892961

RESUMEN

Streptococcus pneumoniae is the most frequent cause of community-acquired pneumonia. The infection process involves bacterial cell surface receptors, which interact with host extracellular matrix components to facilitate colonization and dissemination of bacteria. Here, we investigated the role of host-derived extracellular RNA (eRNA) in the process of pneumococcal alveolar epithelial cell infection. Our study demonstrates that eRNA dose-dependently increased S. pneumoniae invasion of alveolar epithelial cells. Extracellular enolase (Eno), a plasminogen (Plg) receptor, was identified as a novel eRNA-binding protein on S. pneumoniae surface, and six Eno eRNA-binding sites including a C-terminal 15 amino acid motif containing lysine residue 434 were characterized. Although the substitution of lysine 434 for glycine (K434G) markedly diminished the binding of eRNA to Eno, the adherence to and internalization into alveolar epithelial cells of S. pneumoniae strain carrying the C-terminal lysine deletion and the mutation of internal Plg-binding motif were only marginally impaired. Accordingly, using a mass spectrometric approach, we identified seven novel eRNA-binding proteins in pneumococcal cell wall. Given the high number of eRNA-interacting proteins on pneumococci, treatment with RNase1 completely inhibited eRNA-mediated pneumococcal alveolar epithelial cell infection. Our data support further efforts to employ RNAse1 as an antimicrobial agent to combat pneumococcal infectious diseases.


Asunto(s)
Adhesión Bacteriana/efectos de los fármacos , Células Endoteliales/microbiología , Células Epiteliales/microbiología , Espacio Extracelular/metabolismo , ARN/farmacología , Streptococcus pneumoniae/citología , Células A549 , Secuencias de Aminoácidos , Animales , Sitios de Unión , Bovinos , ADN/metabolismo , Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Glicina/metabolismo , Humanos , Pulmón/patología , Lisina/metabolismo , Mutación/genética , Nucleótidos/metabolismo , Fosfopiruvato Hidratasa/química , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Ribonucleasa Pancreática/metabolismo , Streptococcus pneumoniae/efectos de los fármacos
11.
Biochim Biophys Acta ; 1852(12): 2678-88, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26391253

RESUMEN

OBJECTIVE: Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by epithelial cell injury, fibroblast activation and excessive extracellular matrix deposition. Although protein arginine methyltransferase 1 (PRMT1) was found to regulate cell proliferation, differentiation and migration, its role in the development/progression of IPF has not yet been described. RESULTS: Expression of PRMT1 was elevated in lung homogenates from IPF patients. Significant upregulation of PRMT1 expression was also observed in the lungs of bleomycin-treated mice. Immunohistochemical analysis revealed PRMT1-positive staining in fibroblasts/myofibroblasts and alveolar type II cells of IPF lungs and in fibrotic lesions of bleomycin-injured lungs. Fibroblasts isolated from IPF lungs demonstrated increased PRMT1 expression. Interleukin-4 (IL-4), a profibrotic cytokine, enhanced the expression of PRMT1 and the migration of donor and IPF fibroblasts. Interference with the expression or the activity of PRMT1 diminished the migration of the cells in response to IL-4. Strikingly, even though the incubation of donor and IPF fibroblasts with IL-4 did not affect their proliferation, depletion, but not blockage of PRMT1 activity suppressed cell growth. CONCLUSIONS: PRMT1 can contribute to the development of pulmonary fibrosis by regulating fibroblast activities. Thus, interference with its expression and/or activity may provide a novel therapeutic option for patients with IPF.

12.
J Biol Chem ; 290(19): 11983-99, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25805497

RESUMEN

Tumor cells use broad spectrum proteolytic activity of plasmin to invade tissue and form metastatic foci. Cell surface-associated enolase-1 (ENO-1) enhances plasmin formation and thus participates in the regulation of pericellular proteolysis. Although increased levels of cell surface bound ENO-1 have been described in different types of cancer, the molecular mechanism responsible for ENO-1 exteriorization remains elusive. In the present study, increased ENO-1 protein levels were found in ductal breast carcinoma and on the cell surface of highly metastatic breast cancer cell line MDA-MB-231. Elevated cell surface-associated ENO-1 expression correlated with augmented MDA-MB-231 cell migratory and invasive properties. Exposure of MDA-MB-231 cells to LPS potentiated translocation of ENO-1 to the cell surface and its release into the extracellular space in the form of exosomes. These effects were independent of de novo protein synthesis and did not require the classical endoplasmic reticulum/Golgi pathway. LPS-triggered ENO-1 exteriorization was suppressed by pretreatment of MDA-MB-231 cells with the Ca(2+) chelator BAPTA or an inhibitor of endoplasmic reticulum Ca(2+)-ATPase pump, cyclopiazonic acid. In line with these observations, the stromal interaction molecule (STIM) 1 and the calcium release-activated calcium modulator (ORAI) 1-mediated store-operated Ca(2+) entry were found to regulate LPS-induced ENO-1 exteriorization. Pharmacological blockage or knockdown of STIM1 or ORAI1 reduced ENO-1-dependent migration of MDA-MB-231 cells. Collectively, our results demonstrate the pivotal role of store-operated Ca(2+) channel-mediated Ca(2+) influx in the regulation of ENO-1 exteriorization and thus in the modulation of cancer cell migratory and invasive properties.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Canales de Calcio/metabolismo , Calcio/metabolismo , Membrana Celular/enzimología , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Fosfopiruvato Hidratasa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Biotinilación , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quelantes/química , Ácido Egtácico/análogos & derivados , Ácido Egtácico/química , Exosomas/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Indoles/química , Inflamación , Células MCF-7 , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteína ORAI1 , Molécula de Interacción Estromal 1 , Ácido Tricloroacético/química
13.
Transplantation ; 99(5): 925-34, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25719257

RESUMEN

BACKGROUND: In addition to its well-described role in lipid metabolism, apolipoprotein E (ApoE) exerts immunomodulatory functions. A protective role of ApoE and ApoE-mimetic peptide (ApoE(133-149)) application was documented in several inflammatory disorders. In this study, we test the hypothesis that ApoE(133-149) promotes renal allograft survival. METHODS: Dark Agouti, Brown Norway, and Fischer 344 kidneys were transplanted to Lewis rats to investigate fatal and reversible acute rejection. Apolipoprotein E expression was assessed in intravascular leukocytes of renal grafts, in graft tissue and in recipient blood plasma. Recipients of Brown Norway kidneys were treated with ApoE(133-149), and graft survival was monitored until day 100. Graft infiltration, cytokine, and chemokine production were analyzed. RESULTS: Intravascular graft leukocytes and renal tissue obtained from animals undergoing reversible acute rejection expressed increased levels of ApoE mRNA, whereas during fatal rejection, ApoE expression was reduced or remained unchanged. Animals treated with ApoE(133-149) showed prolonged allograft survival, which was associated with a reduced infiltration of CD8 and α/ß T-cell receptor-expressing cells, diminished Granzyme B mRNA expression, and decreased caspase-3 activation. CONCLUSIONS: Endogenous ApoE overexpression and exogenous application of ApoE(133-149) seem to protect renal allografts from fatal acute rejection. This effect was associated with a reduced influx of cytotoxic T cells.


Asunto(s)
Apolipoproteínas E/uso terapéutico , Rechazo de Injerto/prevención & control , Fragmentos de Péptidos/uso terapéutico , Enfermedad Aguda , Secuencia de Aminoácidos , Animales , Apolipoproteínas E/sangre , Movimiento Celular , Quimiocinas/genética , Citocinas/genética , Leucocitos Mononucleares/fisiología , Masculino , Datos de Secuencia Molecular , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344 , Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Linfocitos T/inmunología , Transcriptoma
14.
J Biol Chem ; 290(11): 7027-39, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25589788

RESUMEN

Hageman factor (FXIIa) initiates the intrinsic coagulation pathway and triggers the kallikrein-kinin and the complement systems. In addition, it functions as a growth factor by expressing promitogenic activities toward several cell types. FXIIa binds to the cell surface via a number of structurally unrelated surface receptors; however, the underlying mechanisms are not yet fully understood. Here, we demonstrate that FXIIa utilizes cell membrane-bound glycosaminoglycans to interact with the cell surface of human lung fibroblasts (HLF). The combination of enzymatic, inhibitory, and overexpression approaches identified a heparan sulfate (HS) component of proteoglycans as an important determinant of the FXIIa binding capacity of HLF. Moreover, cell-free assays and competition experiments revealed preferential binding of FXIIa to HS and heparin over dextran sulfate, dermatan sulfate, and chondroitin sulfate A and C. Finally, we demonstrate that fibroblasts isolated from the lungs of the patients suffering from idiopathic pulmonary fibrosis (IPF) exhibit enhanced FXIIa binding capacity. Increased sulfation of HS resulting from elevated HS 6-O-sulfotransferase-1 expression in IPF HLF accounted, in part, for this phenomenon. Application of RNA interference technology and inhibitors of intracellular sulfation revealed the cooperative action of cell surface-associated HS and urokinase-type plasminogen activator receptor in the accumulation of FXIIa on the cell surface of IPF HLF. Moreover, FXIIa stimulated IPF HLF migration, which was abrogated by pretreatment of cells with heparinase I. Collectively, our study uncovers a novel role of HS-type glycosaminoglycans in a local accumulation of FXIIa on the cell membrane. The enhanced association of FXIIa with IPF HLF suggests its contribution to fibrogenesis.


Asunto(s)
Factor XIIa/metabolismo , Fibroblastos/patología , Proteoglicanos de Heparán Sulfato/metabolismo , Pulmón/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Células Cultivadas , Factor XIIa/análisis , Fibroblastos/metabolismo , Proteoglicanos de Heparán Sulfato/análisis , Humanos , Pulmón/metabolismo , Unión Proteica
15.
Int J Mol Sci ; 15(11): 21229-52, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25407528

RESUMEN

Cell surface-associated proteolysis mediated by plasmin (PLA) is an essential feature of wound healing, angiogenesis and cell invasion, processes that are dysregulated in cancer development, progression and systemic spread. The generation of PLA, initiated by the binding of its precursor plasminogen (PLG) to the cell surface, is regulated by an array of activators, inhibitors and receptors. In this review, we will highlight the importance of the best-characterized components of the PLG/PLA cascade in the pathogenesis of cancer focusing on the role of the cell surface-PLG receptors (PLG-R). PLG-R overexpression has been associated with poor prognosis of cancer patients and resistance to chemotherapy. We will also discuss recent findings on the molecular mechanisms regulating cell surface expression and distribution of PLG-R.


Asunto(s)
Fibrinolisina/metabolismo , Neoplasias/metabolismo , Plasminógeno/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Anexina A2/genética , Anexina A2/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Queratina-8/genética , Queratina-8/metabolismo , Neoplasias/genética , Neoplasias/patología , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Transporte de Proteínas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
16.
Biochem J ; 460(2): 295-307, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24628430

RESUMEN

Cell-surface-associated proteolysis plays a crucial role in embryonic development, monocyte/macrophage recruitment and tumour cell invasion. The glycolytic enzyme ENO-1 (enolase-1) is translocated from the cytoplasm to the cell surface, where it binds PLG (plasminogen) to enhance pericellular plasmin production and cell motility. In the present study, ENO-1 was found to localize to a specialized subset of lipid rafts called caveolae as demonstrated by fluorescence confocal microscopy and sucrose gradient ultracentrifugation. Co-immunoprecipitation studies revealed that ENO-1 interacts with Cav-1 (caveolin-1), but not with Cav-2, via the CSD (Cav-scaffolding domain). Moreover, an evolutionarily conserved CBM (Cav-binding motif) F296DQDDWGAW304 was identified within ENO-1. The point mutation W301A within the ENO-1 CBM was, however, not sufficient to disrupt ENO-1-Cav-1 interaction, whereas the mutations F296A and W304A markedly affected ENO-1 protein expression. Furthermore, ENO-1 was found associated with Annx2 (annexin 2), representing another caveolar protein, and this interaction was dependent on Cav-1 expression. Knockdown of Cav-1 and Annx2 markedly decreased cell surface expression of ENO-1. ENO-1 overexpression increased cell migration and invasion in a Cav-1-dependent manner. Thus the differential association of ENO-1 with caveolar proteins regulates ENO-1 subcellular localization and, consequently, ENO-1-dependent cell migration and invasion.


Asunto(s)
Anexina A2/metabolismo , Biomarcadores de Tumor/metabolismo , Caveolas/metabolismo , Caveolina 1/metabolismo , Proteínas de Unión al ADN/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Movimiento Celular , Células HEK293 , Humanos , Ratones , Plasminógeno/metabolismo , Transporte de Proteínas , Células Tumorales Cultivadas
17.
Thromb Haemost ; 110(2): 295-307, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23739922

RESUMEN

Extravascular activation of the coagulation cascade in the lung is commonly observed in pulmonary fibrosis. Coagulation proteases may exert profibrotic cellular effects via protease-activated receptors (PARs)-1 and -2. Here, we investigated the potential role of two other members of the PAR family, namely PAR-3 and PAR-4, in the pathobiology of lung fibrosis. Elevated expression of PAR-3, but not PAR-4, was detected in the lungs of idiopathic pulmonary fibrosis (IPF) patients and in bleomycin-induced lung fibrosis in mice. Increased PAR-3 expression in fibrotic lungs was mainly attributable to alveolar type II (ATII) cells. Stimulation of primary mouse ATII, MLE15 and A549 cells with thrombin (FIIa) - that may activate PAR-1, PAR-3 and PAR-4 - induced epithelial-mesenchymal transition (EMT), a process that has been suggested to be a possible mechanism underlying the expanded (myo)fibroblast pool in lung fibrosis. EMT was evidenced by morphological alterations, expression changes of epithelial and mesenchymal phenotype markers, and functional changes. Single knockdown of FIIa receptors, PAR-1, PAR-3, or PAR-4, had no major impact on FIIa-induced EMT. Simultaneous depletion of PAR-1 and PAR-3, however, almost completely inhibited this process, whereas only a partial effect on FIIa-mediated EMT was observed when PAR-1 and PAR-4, or PAR-3 and PAR-4 were knocked down. PAR-1 and PAR-3 co-localise within ATII cells with both being predominantely plasma membrane associated. In conclusion, our study indicates that PARs synergise to mediate FIIa-induced EMT and provides first evidence that PAR-3 via its ability to potentiate FIIa-triggered EMT could potentially contribute to the pathogenesis of pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Fibrosis Pulmonar/etiología , Receptor PAR-1/metabolismo , Receptores Proteinasa-Activados/metabolismo , Receptores de Trombina/metabolismo , Animales , Bleomicina/toxicidad , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Protrombina/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/genética , Receptores Proteinasa-Activados/antagonistas & inhibidores , Receptores Proteinasa-Activados/genética , Receptores de Trombina/antagonistas & inhibidores , Receptores de Trombina/genética , Trombina/metabolismo
18.
Am J Pathol ; 182(6): 2094-108, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23562441

RESUMEN

Mast cell (MC) accumulation has been demonstrated in the lungs of idiopathic pulmonary fibrosis (IPF) patients. Mediators released from MCs may regulate tissue remodeling processes, thereby contributing to IPF pathogenesis. We investigated the role of MC-fibroblast interaction in the progression of lung fibrosis. Increased numbers of activated MCs, in close proximity to fibroblast foci and alveolar type II cells, were observed in IPF lungs. Correspondingly elevated tryptase levels were detected in IPF lung tissue samples. Coculture of human lung MCs with human lung fibroblasts (HLFs) induced MC activation, as evinced by tryptase release, and stimulated HLF proliferation; IPF HLFs exhibited a significantly higher growth rate, compared with control. Tryptase stimulated HLF growth in a PAR-2/PKC-α/Raf-1/p44/42-dependent manner and potentiated extracellular matrix production, but independent of PKC-α, Raf-1, and p44/42 activities. Proproliferative properties of tryptase were attenuated by knockdown or pharmacological inhibition of PAR-2, PKC-α, Raf-1, or p44/42. Expression of transmembrane SCF, but not soluble SCF, was elevated in IPF lung tissue and in fibroblasts isolated from IPF lungs. Coculture of IPF HLFs with MCs enhanced MC survival and proliferation. These effects were cell-contact dependent and could be inhibited by application of anti-SCF antibody or CD117 inhibitor. Thus, fibroblasts and MCs appear to work in concert to perpetuate fibrotic processes and so contribute to lung fibrosis progression.


Asunto(s)
Fibroblastos/fisiología , Mastocitos/fisiología , Fibrosis Pulmonar/patología , Comunicación Celular/fisiología , Recuento de Células , Degranulación de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/patología , Mastocitos/metabolismo , Proteínas Quinasas/fisiología , Fibrosis Pulmonar/metabolismo , Receptor PAR-2/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Células Madre/fisiología , Triptasas/farmacología , Triptasas/fisiología
19.
Int J Mol Sci ; 13(10): 12383-400, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23202904

RESUMEN

Protein arginine methylation is a novel posttranslational modification that plays a pivotal role in a variety of intracellular events, such as signal transduction, protein-protein interaction and transcriptional regulation, either by the direct regulation of protein function or by metabolic products originating from protein arginine methylation that influence nitric oxide (NO)-dependent processes. A growing body of evidence suggests that both mechanisms are implicated in cardiovascular and pulmonary diseases. This review will present and discuss recent research on PRMTs and the methylation of non-histone proteins and its consequences for the pathogenesis of various lung disorders, including lung cancer, pulmonary fibrosis, pulmonary hypertension, chronic obstructive pulmonary disease and asthma. This article will also highlight novel directions for possible future investigations to evaluate the functional contribution of arginine methylation in lung homeostasis and disease.


Asunto(s)
Enfermedades Pulmonares/enzimología , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Asma/enzimología , Asma/patología , Humanos , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/patología , Enfermedades Pulmonares/patología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfermedad Pulmonar Obstructiva Crónica/patología , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología
20.
Am J Respir Cell Mol Biol ; 47(5): 614-27, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22771387

RESUMEN

The disturbance of hemostatic balance, associated with increased tissue factor (TF) expression and activity, occurs in the lungs of patients with idiopathic pulmonary fibrosis (IPF). However, the molecular mechanisms responsible for the regulation of TF expression under profibrotic conditions have not been assessed. We found that transforming growth factor-ß1 (TGF-ß1) markedly enhanced TF expression in primary human lung fibroblasts (HLFs), whereas platelet-derived growth factor (PDGF)-BB and IGF (insulin-like growth factor)-1 showed only a moderate effect, and PDGB-CC exerted no effect. TGF-ß1-induced TF expression correlated with its elevated cell-surface activity, it required de novo gene transcription and protein synthesis, and it was dependent on JNK and Akt activity, because pharmacological inhibition or the knockdown of the previously mentioned kinases prevented TF synthesis. Exposure of HLFs to TGF-ß1 activated JNK in a PI3K-dependent manner and induced Akt phosphorylation at threonine 308 and serine 473, but did not change the phosphorylation status of threonine 450. Akt phosphorylation at serine 473 correlated with JNK activity, and co-immunoprecipitation studies revealed a direct interaction between JNK and Akt. Furthermore, TGF-ß1-induced TF expression required the recruitment of c-Fos and JunD into a heterodimeric activator protein (AP)-1 complex. Moreover, strong immunoreactivity for phosphorylated Akt and JNK as well as c-Fos and JunD was observed in fibroblasts and myofibroblasts in IPF lungs. In conclusion, PI3K/JNK/Akt and AP-1 synergize to induce TF expression in HLFs after TGF-ß1 challenge. Our findings provide new insights into the molecular mechanisms responsible for the regulation of TF expression, and open new perspectives on the treatment of pulmonary fibrosis and other diseases characterized by the inappropriate expression of this cell-surface receptor.


Asunto(s)
Fibroblastos/metabolismo , Pulmón/patología , Transducción de Señal , Tromboplastina/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Células Cultivadas , Regulación de la Expresión Génica , Semivida , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Cultivo Primario de Células , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Estabilidad del ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Tromboplastina/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA