Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 11(1): 21906, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34753989

RESUMO

Ground based research modalities of microgravity have been proposed as innovative methods to investigate the aetiology of chronic age-related conditions such as cardiovascular disease. Dry Immersion (DI), has been effectively used to interrogate the sequelae of physical inactivity (PI) and microgravity on multiple physiological systems. Herein we look at the causa et effectus of 3-day DI on platelet phenotype, and correlate with both miRomic and circulating biomarker expression. The miRomic profile of platelets is reflective of phenotype, which itself is sensitive and malleable to the exposome, undergoing responsive transitions in order to fulfil platelets role in thrombosis and haemostasis. Heterogeneous platelet subpopulations circulate at any given time, with varying degrees of sensitivity to activation. Employing a DI model, we investigate the effect of acute PI on platelet function in 12 healthy males. 3-day DI resulted in a significant increase in platelet count, plateletcrit, platelet adhesion, aggregation, and a modest elevation of platelet reactivity index (PRI). We identified 15 protein biomarkers and 22 miRNA whose expression levels were altered after DI. A 3-day DI model of microgravity/physical inactivity induced a prothrombotic platelet phenotype with an unique platelet miRNA signature, increased platelet count and plateletcrit. This correlated with a unique circulating protein biomarker signature. Taken together, these findings highlight platelets as sensitive adaptive sentinels and functional biomarkers of epigenetic drift within the cardiovascular compartment.


Assuntos
Plaquetas/citologia , Proteínas Sanguíneas/metabolismo , MicroRNAs/genética , Modelos Biológicos , Ausência de Peso , Adulto , Biomarcadores/sangue , Hemostasia , Humanos , Masculino , Trombose/metabolismo
3.
Mar Pollut Bull ; 173(Pt A): 112981, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34600169

RESUMO

The toxic benthic dinoflagellate Ostreopsis ovata causes harmful algal blooms. During five years, citizens have monitored blooms of O. cf. ovata along the coast of Molfetta city facing the Adriatic Sea. Coscinodiscus spp., Licmophora spp., Gyrosigma spp. and Achnantes spp. diatoms were also quantified. O. cf. ovata was detected from spring up to winter, however, blooms always occurred in summer. Correlation with nine weather parameters was relatively strong with seawater temperature, dew point and air temperature. Dew point has never been reported before as key parameter. Blooms of O. cf. ovata were preceded by lag period during which conditions permitted proliferation but no proliferation observed. Furthermore, dew point, seawater and air temperature only moderately correlated with proliferation of Coscinodiscus spp. However, correlation between blooms of O. cf. ovata and Coscinodiscus spp. was relatively strong. Correlation between proliferation of O. cf. ovata and Gyrosigma spp. was very weak, while moderate and negative with Licmophora spp. or Achnantes spp.


Assuntos
Ciência do Cidadão , Diatomáceas , Dinoflagellida , Proliferação Nociva de Algas , Água do Mar
5.
Burns ; 46(7): 1585-1602, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32475797

RESUMO

BACKGROUND: Burn injuries are a major cause of morbidity and mortality worldwide. Despite advances in therapeutic strategies for the management of patients with severe burns, the sequelae are pathophysiologically profound, up to the systemic and metabolic levels. Management of patients with a severe burn injury is a long-term, complex process, with treatment dependent on the degree and location of the burn and total body surface area (TBSA) affected. In adverse conditions with limited resources, efficient triage, stabilisation, and rapid transfer to a specialised intensive care burn centre is necessary to provide optimal outcomes. This initial lag time and the form of primary treatment initiated, from injury to specialist care, is crucial for the burn patient. This study aims to investigate the efficacy of a novel visco-elastic burn dressing with a proprietary bio-stimulatory marine mineral complex (MXC) as a primary care treatment to initiate a healthy healing process prior to specialist care. METHODS: A new versatile emergency burn dressing saturated in a >90% translucent water-based, sterile, oil-free gel and carrying a unique bio-stimulatory marine mineral complex (MXC) was developed. This dressing was tested using LabSkin as a burn model platform. LabSkin a novel cellular 3D-dermal organotypic full thickness human skin equivalent, incorporating fully-differentiated dermal and epidermal components that functionally models skin. Cell and molecular analysis was carried out by in vitro Real-Time Cellular Analysis (RTCA), thermal analysis, and focused transcriptomic array profiling for quantitative gene expression analysis, interrogating both wound healing and fibrosis/scarring molecular pathways. In vivo analysis was also performed to assess the bio-mechanical and physiological effects of this novel dressing on human skin. RESULTS: This hybrid emergency burn dressing (EBD) with MXC was hypoallergenic, and improved the barrier function of skin resulting in increased hydration up to 24 h. It was demonstrated to effectively initiate cooling upon application, limiting the continuous burn effect and preventing local tissue from damage and necrosis. xCELLigence RTCA® on primary human dermal cells (keratinocyte, fibroblast and micro-vascular endothelial) demonstrated improved cellular function with respect to tensegrity, migration, proliferation and cell-cell contact (barrier formation) [1]. Quantitative gene profiling supported the physiological and cellular function finding. A beneficial quid pro quo regulation of genes involved in wound healing and fibrosis formation was observed at 24 and 48 h time points. CONCLUSION: Utilisation of this EBD + MXC as a primary treatment is an effective and easily applicable treatment in cases of burn injury, proving both a cooling and hydrating environment for the wound. It regulates inflammation and promotes healing in preparation for specialised secondary burn wound management. Moreover, it promotes a healthy remodelling phenotype that may potentially mitigate scarring. Based on our findings, this EBD + MXC is ideal for use in all pre-hospital, pre-surgical and resource limited settings.


Assuntos
Curativos Hidrocoloides , Queimaduras , Cicatriz , Produtos Biológicos/uso terapêutico , Queimaduras/patologia , Queimaduras/terapia , Cicatriz/patologia , Humanos , Técnicas In Vitro , Pele/patologia , Cicatrização
6.
Front Physiol ; 11: 383, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32431622

RESUMO

Venoconstrictive thigh cuffs are used by cosmonauts to ameliorate symptoms associated with cephalad fluid shift. A ground simulation of microgravity, using the dry immersion (DI) model, was performed to assess the effects of thigh cuffs on body fluid changes and dynamics, as well as on cardiovascular deconditioning. Eighteen healthy men (25-43 years), randomly divided into two groups, (1) control group or (2) group with thigh cuffs worn 10 h/day, underwent 5-day DI. Cardiovascular responses to orthostatic challenge were evaluated using the lower body negative pressure (LBNP) test; body fluid changes were assessed by bio-impedance and hormonal assay; plasma volume evolution was estimated using hemoglobin-hematocrit; subjective tolerance was assessed by questionnaires. DI induced a decrease in plasma volume of 15-20%. Reduction in total body water of 3-6% stabilized toward the third day of DI. This reduction was derived mostly from the extracellular compartment. During the acute phase of DI, thigh cuffs limited the decrease in renin and the increase in N-terminal prohormone of brain natriuretic peptide (NT-proBNP), the loss in total body water, and tended to limit the loss in calf volume, extracellular volume and plasma volume. At the later stable phase of DI, a moderate protective effect of thigh cuffs remained evident on the body fluids. Orthostatic tolerance time dropped after DI without significant difference between groups. Thigh cuff countermeasure slowed down and limited the loss of body water and tended to limit plasma loss induced by DI. These observed physiological responses persisted during periods when thigh cuffs were removed. However, thigh cuffs did not counteract decreased tolerance to orthostatic challenge.

7.
Data Brief ; 22: 903-908, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30723759

RESUMO

The data presented in this article are connected to our research article entitled "D2A-Ala peptide derived from the urokinase receptor exerts anti-tumoural effects in vitro and in vivo" (Furlan et al., 2018). These data further extend our understanding of the inhibitory effects of D2A-Ala peptide. Dose-response curve using a wide range of concentrations of D2A-Ala shows that this peptide has no effects per se on proliferation of rat smooth muscle cells (RSMC). However, D2A-Ala dose-dependently inhibits epidermal growth factor (EGF)-induced RSMC proliferation. Kinetics lasting up to seven days revealed that D2A-Ala peptide completely blocked EGF-promoted RSMC proliferation. Moreover, D2A-Ala peptide inhibited invasion of HT 1080 cells towards RSMC.

8.
Peptides ; 101: 17-24, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29273518

RESUMO

D2A-Ala is a synthetic peptide that has been created by introducing mutations in the original D2A sequence, 130IQEGEEGRPKDDR142 of human urokinase receptor (uPAR). In vitro, D2A-Ala peptide displays strong anti-tumoural properties inhibiting EGF-induced chemotaxis, invasion and proliferation of a human fibrosarcoma cell line, HT 1080, and a human colorectal adenocarcinoma cell line, HT 29. D2A-Ala exerts its effects by preventing EGF receptor (EGFR) phosphorylation. To test D2A-Ala in vivo, this peptide was PEGylated generating polyethyleneglycol (PEG)-D2A-Ala peptide. PEGylation did not alter the inhibitory properties of D2A-Ala. Human tumour xenografts in the immunodeficient nude mice using HT 1080 and HT 29 cell lines showed that PEG-D2A-Ala significantly prevents tumour growth decreasing size, weight and density of tumours. The most efficient doses of the peptide were 5 and 10 mg/kg, thereby relevant for possible development of the peptide into a drug against cancer in particular tumours expressing EGFR.


Assuntos
Adenossarcoma/tratamento farmacológico , Antineoplásicos/farmacologia , Quimiotaxia/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Fibrossarcoma/tratamento farmacológico , Oligopeptídeos/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Adenossarcoma/metabolismo , Adenossarcoma/patologia , Animais , Antineoplásicos/química , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Feminino , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Células HT29 , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Oligopeptídeos/química , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cell Mol Life Sci ; 75(10): 1889-1907, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29184982

RESUMO

The urokinase receptor (uPAR) stimulates cell proliferation by forming a macromolecular complex with αvß3 integrin and the epidermal growth factor receptor (EGFR, ErbB1 or HER1) that we name the uPAR proliferasome. uPAR transactivates EGFR, which in turn mediates uPAR-initiated mitogenic signal to the cell. EGFR activation and EGFR-dependent cell growth are blocked in the absence of uPAR expression or when uPAR activity is inhibited by antibodies against either uPAR or EGFR. The mitogenic sequence of uPAR corresponds to the D2A motif present in domain 2. NMR analysis revealed that D2A synthetic peptide has a particular three-dimensional structure, which is atypical for short peptides. D2A peptide is as effective as EGF in promoting EGFR phosphorylation and cell proliferation that were inhibited by AG1478, a specific inhibitor of the tyrosine kinase activity of EGFR. Both D2A and EGF failed to induce proliferation of NR6-EGFR-K721A cells expressing a kinase-defective mutant of EGFR. Moreover, D2A peptide and EGF phosphorylate ERK demonstrating the involvement of the MAP kinase signalling pathway. Altogether, this study reveals the importance of sequence D2A of uPAR, and the interdependence of uPAR and EGFR.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Integrina alfaVbeta3/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Modelos Moleculares , Fosforilação , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química
10.
Data Brief ; 15: 600-605, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29085875

RESUMO

The data presented herein are connected to our research article (doi: 10.1016/j.biocel.2017.04.012) [1], in which we investigated the functional connections between the urokinase receptor (uPAR), and the ezrin/radixin/moesin (ERM) proteins, moesin and merlin [1]. Firstly, a model of action is proposed that enlightens how uPAR regulates distal integrins. In addition, data show the effects of expressing wild-type moesin or permanently active T558D mutant of moesin on angiogenesis and morphology of human aortic endothelial cells (HAEC). Additional data compare the effects of urokinase (uPA, the main ligand of uPAR) on the same cells. Lastly, we provide technical data demonstrating the effects of specific siRNA for moesin and merlin on moesin and merlin expression, respectively.

11.
Int J Biochem Cell Biol ; 88: 14-22, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28473293

RESUMO

The glycosyl-phosphatidyl-inositol (GPI)-anchored urokinase receptor (uPAR) has no intracellular domain, but nevertheless initiates signalling through proximal interactions with other membrane receptors including integrins. The relationships between uPAR and ezrin/radixin/moesin (ERM) proteins, moesin and merlin have never been explored. Moesin and merlin are versatile membrane-actin links and regulators of receptors signalling, respectively. We show that uPAR controls moesin and merlin, which propagate uPAR-initiated signals and modulate integrin functions, thereby regulating uPAR activity. uPAR rapidly de-phosphorylates moesin and phosphorylates merlin inactivating both proteins, and enhancing cell migration and angiogenesis. Moesin behaves as a molecular switch turning either on or off uPAR signalling through cycles of de-activation/activation, or sustained activation, respectively. Furthermore, moesin is at the crossroads of uPAR-initiated outside-in and inside-out signalling promoting integrin-dependent cell adhesion suggesting that uPAR also activates integrins distally through moesin. Knocking down merlin expression enhanced cell migration and adhesion through different regulation of fibronectin- and vitronectin-binding integrins.


Assuntos
Adesão Celular , Quimiotaxia , Células Endoteliais/citologia , Proteínas dos Microfilamentos/metabolismo , Neovascularização Fisiológica , Neurofibromina 2/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Células Endoteliais/metabolismo , Humanos
12.
Curr Pharm Des ; 17(19): 1872-3, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21711231

RESUMO

The story that led to the discovery of urokinase receptor (uPAR) system started in 1947 with the report of MacFarlane and Pilling who identified but did not named urokinase (uPA). Today, the uPAR system is recognized as one very important actor in tumourigenesis and is even considered as a valuable tumour marker. Its critical functions justify the important effort of translational research that has produced many inhibitors which unfortunately failed to be transferred in the clinic. However, the role of the uPAR system in cancer development should not shade the vital functions of this system in hematopoietic stem cells mobilization, cognitive functions including language development, inflammation, innate immunity, coagulation and fibrinolysis. All these topics are covered in this special theme issue of the journal Current Pharmaceutical Design that comprises 9 reviews written by leading scientists. Other aspects are also embraced by additional articles including the first attempt to depict the complete atlas of the uPAR interactome, and the regulation of the uPAR system by the LDL receptor-related protein-1 (LRP-1).


Assuntos
Antineoplásicos/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Biomarcadores Tumorais , Sistema Nervoso Central/metabolismo , Desenho de Fármacos , Humanos , Invasividade Neoplásica , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Células-Tronco/fisiologia
13.
Curr Pharm Des ; 17(19): 1874-89, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21711237

RESUMO

The urokinase receptor (uPAR) was originally identified as the membrane receptor of the serine protease urokinase (uPA), thereby implicated in the plasminogen activation cascade and regulation of pericellular proteolysis. Later on, vitronectin was showed to be another major ligand providing uPAR with a role in cell adhesion. Other unrelated ligands have been subsequently reported including for example factor XII and SRPX2 expanding the functions of uPAR to unexpected biological areas such as the initiation of the coagulation cascade or the regulation of language development. Due to its glycosylphosphatidylinositol (GPI) anchor, uPAR has no intracellular domain and thus exerts its signaling capacity through lateral interactions with other components of the plasma membrane that actually mediate uPAR-induced signals. As yet, a total 42 proteins interacting directly with uPAR can be numbered comprising 9 soluble ligands and 33 lateral partners. The fact that uPAR interacts with members of three major families of membrane receptors i.e. G protein-coupled receptors, receptor tyrosine kinases, and integrins implies that the actual number of components constituting the uPAR interacome is extremely high. For example, 156 factors belong to the integrin adhesome. Moreover, in the light of the wide diversity of the components of the uPAR interactome, uPAR appears to be an essential player of major biological systems including the blood coagulation, complement and plasma kallikrein-kinin cascades. This review describes the soluble ligands and lateral partners of the uPAR interactome, the mechanisms regulating uPAR interactions and their proved and/or potential biological functions.


Assuntos
Regulação da Expressão Gênica/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Glicosilfosfatidilinositóis/metabolismo , Humanos , Ligantes , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transdução de Sinais
14.
CNS Neurol Disord Drug Targets ; 10(2): 271-94, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20874700

RESUMO

The urokinase receptor (uPAR) is a multifunctional glycosylphosphatidylinositol-anchored protein that regulates important processes such as gene expression, cell proliferation, adhesion, migration, invasion, and metastasis. uPAR is an essential component of the plasminogen activation cascade, a protease receptor that binds the urokinase-type plasminogen activator. uPAR is also an adhesion-modulating receptor, and a signalling receptor transmitting signals to the cell through lateral interactions with a wide array of membrane receptors. Altogether, the external ligands and membrane-bound partners of uPAR constitute a rich uPAR interactome. Recently, a new ligand of uPAR has been identified as the SRPX2 protein which is essential in language and cognitive development. SRPX2 is the second identified gene involved in language disorders. However, previous studies revealed cognitive disorders and defects in the development of the GABAergic interneurons in uPAR null mice. In addition, the expression of uPAR correlates with important human diseases such as epilepsy, autism, multiple sclerosis, Alzheimer's, AIDS dementia, cerebral malaria, and brain tumours. Therefore, uPAR has unexpectedly become a significant receptor in the central nervous system and made a few steps into philosophy. Language is indeed intimately linked to human culture. This in-depth review presents the structure and the sequences of uPAR that are essential for drug design and the generation of new inhibitors. In addition, we summarize all the inhibitors of uPAR that have been created so far. Finally, we discuss the functions of uPAR in the development, functioning, and pathology of the central nervous system.


Assuntos
Sistema Nervoso Central/fisiopatologia , Glicosilfosfatidilinositóis/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiologia , Glicosilfosfatidilinositóis/genética , Humanos , Proteínas de Membrana/biossíntese , Camundongos , Terapia de Alvo Molecular , Proteínas de Neoplasias/biossíntese , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
15.
Int J Biochem Cell Biol ; 41(3): 578-85, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18703159

RESUMO

We have previously reported that the serpin plasminogen activator inhibitor-1 activates the Janus kinase (Jak)/signal transducer and activator of transcription (Stat) signalling pathway and stimulates cell migration by binding to the low-density lipoprotein receptor-related protein. All the free forms (cleaved, latent or active) of this inhibitor were shown to be motogenic. However, the plasminogen activator inhibitor-1 can also interact with vitronectin which acts as a cofactor by increasing the half-life of the active form of the serpin. Since vitronectin influences most of the biological functions of the plasminogen activator inhibitor-1, we explored the effects of vitronectin on signalling and cell migration induced by this serpin. We found that the interaction between vitronectin and the plasminogen activator inhibitor-1 suppressed signalling and cell migration. In fact, a purified vitronectin(1-97)/plasminogen activator inhibitor-1 complex was not chemotactic. Vitronectin interaction with the plasminogen activator inhibitor-1 blocks the binding of this serpin to its motogenic receptor, the low-density lipoprotein receptor-related protein. Consequently, vitronectin inhibits the activation of the Janus kinase/signal transducer and activator of transcription signalling pathway by the plasminogen activator inhibitor-1 and subsequent cell migration. In conclusion, we have unveiled a new inhibitory role of vitronectin, which turns off the intracellular signalling and migration-promoting activity of the plasminogen activator inhibitor-1. Thus, the motogenic (cleaved, latent or active) and non-motogenic (in complex with vitronectin) forms of the plasminogen activator inhibitor-1 have different properties that may explain the rather contrasting physiological and pathological roles of this serpin.


Assuntos
Proteínas Relacionadas a Receptor de LDL/metabolismo , Miócitos de Músculo Liso/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Vitronectina/metabolismo , Animais , Ensaios de Migração Celular , Movimento Celular , Células Cultivadas , Retroalimentação Fisiológica , Humanos , Janus Quinases/metabolismo , Miócitos de Músculo Liso/citologia , Ligação Proteica , Ratos , Transdução de Sinais , Ressonância de Plasmônio de Superfície
16.
BMC Bioinformatics ; 9 Suppl 2: S8, 2008 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-18387210

RESUMO

BACKGROUND: Urokinase, its receptor and the integrins are functionally associated and involved in regulation of cell signaling, migration, adhesion and proliferation. No structural information is available on this potential multimolecular complex. However, the tri-dimensional structure of urokinase, urokinase receptor and integrins is known. RESULTS: We have modeled the interaction of urokinase on two integrins, alphaIIbbeta3 in the open configuration and alphavbeta3 in the closed configuration. We have found that multiple lowest energy solutions point to an interaction of the kringle domain of uPA at the boundary between alpha and beta chains on the surface of the integrins. This region is not far away from peptides that have been previously shown to have a biological role in urokinase receptor/integrins dependent signaling. CONCLUSIONS: We demonstrated that in silico docking experiments can be successfully carried out to identify the binding mode of the kringle domain of urokinase on the scaffold of integrins in the open and closed conformation. Importantly we found that the binding mode was the same on different integrins and in both configurations. To get a molecular view of the system is a prerequisite to unravel the complex protein-protein interactions underlying urokinase/urokinase receptor/integrin mediated cell motility, adhesion and proliferation and to design rational in vitro experiments.


Assuntos
Integrinas/química , Integrinas/ultraestrutura , Modelos Químicos , Modelos Moleculares , Mapeamento de Interação de Proteínas/métodos , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/ultraestrutura , Sítios de Ligação , Simulação por Computador , Ligação Proteica , Conformação Proteica
17.
Cancer Cell ; 13(3): 221-34, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18328426

RESUMO

In vivo tumor cell migration through integrin-dependent pathways is key to the metastatic behavior of malignant cells. Using quantitative in vivo assays and intravital imaging, we assessed the impact of cell migration, regulated by the integrin-associated tetraspanin CD151, on spontaneous human tumor cell metastasis. We demonstrate that promoting immobility through a CD151-specific metastasis blocking mAb prevents tumor cell dissemination by inhibiting intravasation without affecting primary tumor growth, tumor cell arrest, extravasation, or growth at the secondary site. In vivo, this loss of migration is the result of enhanced tumor cell-matrix interactions, promoted by CD151, which prevent dissociation by individual cells and leads to a subsequent inhibition of invasion and intravasation at the site of the primary tumor.


Assuntos
Antígenos CD/metabolismo , Movimento Celular , Membrana Corioalantoide/imunologia , Neoplasias Experimentais/imunologia , Animais , Anticorpos Monoclonais , Antígenos CD/genética , Antígenos CD/imunologia , Sítios de Ligação de Anticorpos , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/metabolismo , Membrana Corioalantoide/patologia , Matriz Extracelular/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos SCID , Microscopia de Fluorescência , Microscopia de Vídeo , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Reação em Cadeia da Polimerase/métodos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Tetraspanina 24 , Fatores de Tempo , Transfecção
18.
J Biol Chem ; 280(26): 24792-803, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15863511

RESUMO

We investigated the interaction between the urokinase receptor (uPAR) and the integrin alphavbeta3. Vitronectin (VN) induces cell migration by binding to alphavbeta3, but expression of the uPAR boosts its efficacy. Thus, uPAR may regulate VN-induced cell migration by interacting laterally with alphavbeta3. In contrast, cells expressing a uPAR mutant lacking domain 2 do not migrate in response to VN. This effect is overcome by D2A, a synthetic peptide derived from the sequence of domain 2. In addition, D2A has chemotactic activity that requires alphavbeta3 and activates alphavbeta3-dependent signaling pathways such as the Janus kinase/Stat pathway. Moreover, D2A disrupts uPAR-alphavbeta3 and uPAR-alpha5beta1 co-immunoprecipitation, indicating that it can bind both of these integrins. We also identify the chemotactically active epitope harbored by peptide D2A. Mutating two glutamic acids into two alanines generates peptide D2A-Ala, which lacks chemotactic activity but inhibits VN-, FN-, and collagen-dependent cell migration. In fact, the GEEG peptide has potent chemotactic activity, and the GAAG sequence has inhibitory capacities. In summary, we have identified an integrin-interacting sequence located in domain 2 of uPAR, which is also a new chemotactic epitope that can activate alphavbeta3-dependent signaling pathways and stimulate cell migration. This sequence thus plays a pivotal role in the regulation of uPAR-integrin interactions. Moreover, we describe a novel, very potent inhibitor of integrin-dependent cell migration.


Assuntos
Integrinas/química , Receptores de Superfície Celular/química , Adenoviridae/genética , Alanina/química , Animais , Aorta/metabolismo , Adesão Celular , Linhagem Celular , Movimento Celular , Quimiotaxia , Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Epitopos/química , Ácido Glutâmico/química , Humanos , Imunoprecipitação , Integrina alfaVbeta3/química , Camundongos , Microscopia de Fluorescência , Mutação , Miócitos de Músculo Liso/citologia , Células NIH 3T3 , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais , Temperatura , Fatores de Tempo , Transfecção , Tirfostinas/farmacologia
19.
J Biol Chem ; 279(21): 22595-604, 2004 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-15001579

RESUMO

Although plasminogen activator inhibitor-1 (PAI-1) is known to stimulate cell migration, little is known about underlying mechanisms. We show that both active and inactive (e.g. cleaved) PAI-1 can activate the Jak/Stat signaling system and stimulate cell migration in chemotaxis, haptotaxis, chemokinesis, and wound healing assays. Moreover, antibodies to the LDL receptor-related protein (LRP) and an LRP antagonist (RAP) blocked these motogenic effects of PAI-1, while a PAI-1 mutant that did not bind to LRP failed to activate the Jak/Stat signaling pathway or to stimulate cell migration. PAI-1 had no chemotactic effect on LRP-deficient cells. These results indicate that LRP is a signaling molecule, that it mediates the migration-promoting activity of PAI-1, and that this activity does not require intact, biologically active PAI-1. Activation of this LRP-dependent signaling pathway by PAI-1 may begin to explain how the inhibitor stimulates cell migration in a variety of normal and pathological processes.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Actinas/metabolismo , Transporte Ativo do Núcleo Celular , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Células Cultivadas , Quimiotaxia , Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Humanos , Cinética , Microscopia de Fluorescência , Mutação , Fosfotirosina/química , Testes de Precipitina , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Tirosina/química , Cicatrização
20.
FEBS Lett ; 553(1-2): 11-7, 2003 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-14550538

RESUMO

The chromosomal protein HMGB1 is now regarded as a proinflammatory cytokine. Importantly, HMGB1 has chemotactic activity suggesting its involvement in the early and late events of the inflammatory reaction. Therefore, HMGB1 has all the hallmarks of a chemokine (chemotactic cytokine). We propose to classify HMGB1 into a new group of proteins unrelated structurally to chemokines but having chemokine-like functions, and to name this class CLF (chemokine-like functions). The CLF class should include other unrelated molecules such as urokinase and its receptor, cytokines macrophage migration inhibitory factor (MIF) and interleukin (IL)-6, anaphylatoxin C5a, ribosomal protein S19, and thioredoxin that have similar chemokine-like activities. This innovative concept may lead to the identification of new therapeutic targets.


Assuntos
Quimiocinas/metabolismo , Proteína HMGB1/metabolismo , DNA/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA