Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
1.
Proteins ; 92(1): 76-95, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37646459

RESUMO

Cell invasion is an important process in cancer progression and recurrence. Invasion and implantation of cancer cells from their original place to other tissues, by disabling vital organs, challenges the treatment of cancer patients. Given the importance of the matter, many molecular treatments have been developed to inhibit cancer cell invasion. Because of their low production cost and ease of production, peptides are valuable therapeutic molecules for inhibiting cancer cell invasion. In recent years, advances in the field of computational biology have facilitated the design of anti-cancer peptides. In our investigation, using computational biology approaches such as evolutionary analysis, residue scanning, protein-peptide interaction analysis, molecular dynamics, and free energy analysis, our team designed a peptide library with about 100 000 candidates based on A6 (acetyl-KPSSPPEE-amino) sequence which is an anti-invasion peptide. During computational studies, two of the designed peptides that give the highest scores and showed the greatest sequence similarity to A6 were entered into the experimental analysis workflow for further analysis. In experimental analysis steps, the anti-metastatic potency and other therapeutic effects of designed peptides were evaluated using MTT assay, RT-qPCR, zymography analysis, and invasion assay. Our study disclosed that the IK1 (acetyl-RPSFPPEE-amino) peptide, like A6, has great potency to inhibit the invasion of cancer cells.


Assuntos
Receptores de Ativador de Plasminogênio Tipo Uroquinase , Ativador de Plasminogênio Tipo Uroquinase , Humanos , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/uso terapêutico , Peptídeos/farmacologia , Invasividade Neoplásica
2.
Bioorg Med Chem ; 95: 117499, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37879145

RESUMO

The inhibition of human urokinase-type plasminogen activator (huPA), a serine protease that plays an important role in pericellular proteolysis, is a promising strategy to decrease the invasive and metastatic activity of tumour cells. However, the generation of selective small molecule huPA inhibitors has proven to be challenging due to the high structural similarity of huPA to other paralogue serine proteases. Efforts to generate more specific therapies have led to the development of cyclic peptide-based inhibitors with much higher selectivity against huPA. While this latter property is desired, the sparing of the orthologue murine poses difficulties for the testing of the inhibitor in preclinical mouse model. In this work, we have applied a Darwinian evolution-based approach to identify phage-encoded bicyclic peptide inhibitors of huPA with better cross-reactivity towards murine uPA (muPA). The best selected bicyclic peptide (UK132) inhibited huPA and muPA with Ki values of 0.33 and 12.58 µM, respectively. The inhibition appears to be specific for uPA, as UK132 only weakly inhibits a panel of structurally similar serine proteases. Removal or substitution of the second loop with one not evolved in vitro led to monocyclic and bicyclic peptide analogues with lower potency than UK132. Moreover, swapping of 1,3,5-tris-(bromomethyl)-benzene with different small molecules not used in the phage selection, resulted in an 80-fold reduction of potency, revealing the important structural role of the branched cyclization linker. Further substitution of an arginine in UK132 to a lysine resulted in a bicyclic peptide UK140 with enhanced inhibitory potency against both huPA (Ki = 0.20 µM) and murine orthologue (Ki = 2.79 µM). By combining good specificity, nanomolar affinity and a low molecular mass, the bicyclic peptide inhibitor developed in this work may provide a novel human and murine cross-reactive lead for the development of a potent and selective anti-metastatic therapy.


Assuntos
Peptídeos , Ativador de Plasminogênio Tipo Uroquinase , Camundongos , Humanos , Animais , Ativador de Plasminogênio Tipo Uroquinase/química , Peptídeos/farmacologia , Peptídeos/química , Serina Proteases , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/química
3.
Cancer Lett ; 559: 216117, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36889376

RESUMO

The fibrinolytic system is involved in many physiological functions, among which the important members can interact with each other, either synergistically or antagonistically to participate in the pathogenesis of many diseases. Plasminogen activator inhibitor 1 (PAI-1) acts as a crucial element of the fibrinolytic system and functions in an anti-fibrinolytic manner in the normal coagulation process. It inhibits plasminogen activator, and affects the relationship between cells and extracellular matrix. PAI-1 not only involved in blood diseases, inflammation, obesity and metabolic syndrome but also in tumor pathology. Especially PAI-1 plays a different role in different digestive tumors as an oncogene or cancer suppressor, even a dual role for the same cancer. We term this phenomenon "PAI-1 paradox". PAI-1 is acknowledged to have both uPA-dependent and -independent effects, and its different actions can result in both beneficial and adverse consequences. Therefore, this review will elaborate on PAI-1 structure, the dual value of PAI-1 in different digestive system tumors, gene polymorphisms, the uPA-dependent and -independent mechanisms of regulatory networks, and the drugs targeted by PAI-1 to deepen the comprehensive understanding of PAI-1 in digestive system tumors.


Assuntos
Neoplasias do Sistema Digestório , Neoplasias Gastrointestinais , Humanos , Neoplasias do Sistema Digestório/genética , Inflamação , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
4.
Biophys J ; 121(20): 3940-3949, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36039386

RESUMO

Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases produced during the coagulation cascade and inflammation. Previous studies showed that NM was a highly safe drug for the treatment of different cancers, but the precise functions and mechanisms of NM are not clear. In this study, we determined a series of crystal structures of NM and its hydrolysates in complex with a serine protease (urokinase-type plasminogen activator [uPA]). These structures reveal that NM was cleaved by uPA and that a hydrolyzed product (4-guanidinobenzoic acid [GBA]) remained covalently linked to Ser195 of uPA, and the other hydrolyzed product (6-amidino-2-naphthol [6A2N]) released from uPA. Strikingly, in the inactive uPA (uPA-S195A):NM structure, the 6A2N side of intact NM binds to the specific pocket of uPA. Molecular dynamics simulations and end-point binding free-energy calculations show that the conf1 of NM (6A2N as P1 group) in the uPA-S195A:NM complex may be more stable than conf2 of NM (GBA as P1 group). Moreover, in the structure of uPA:NM complex, the imidazole group of His57 flips further away from Ser195 and disrupts the stable canonical catalytic triad conformation. These results not only reveal the inhibitory mechanism of NM as an efficient serine protease inhibitor but also might provide the structural basis for the further development of serine protease inhibitors.


Assuntos
Inibidores de Serina Proteinase , Ativador de Plasminogênio Tipo Uroquinase , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Inibidores de Serina Proteinase/farmacologia , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/metabolismo , Serina Proteases , Imidazóis
5.
Angew Chem Int Ed Engl ; 59(24): 9398-9402, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32176815

RESUMO

Secondary structures tend to be recognizable because they have repeating structural motifs, but mimicry of these does not have to follow such well-defined patterns. Bioinformatics studies to match side-chain orientations of a novel hydantoin triazole chemotype (1) to protein-protein interfaces revealed it tends to align well across parallel and antiparallel sheets, like rungs on a ladder. One set of these overlays was observed for the protein-protein interaction uPA⋅uPAR. Consequently, chemotype 1 was made with appropriate side-chains to mimic uPA at this interface. Biophysical assays indicate these compounds did in fact bind uPAR, and elicit cellular responses that affected invasion, migration, and wound healing.


Assuntos
Materiais Biomiméticos/química , Proteínas/química , Materiais Biomiméticos/farmacologia , Estrutura Secundária de Proteína , Triazóis/química , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
6.
Phys Chem Chem Phys ; 22(6): 3570-3583, 2020 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-31995079

RESUMO

Despite being recognized as a therapeutic target in the processes of cancer cell proliferation and metastasis for over 50 years, the interaction of the urokinase plasminogen activator uPA with its receptor uPAR still needs an improved understanding. High resolution crystallographic data (PDB ) of the uPA-uPAR binding geometry was used to perform quantum biochemistry computations within the density functional theory (DFT) framework. A divide to conquer methodology considering a mixed homogeneous/inhomogeneous dielectric model and explicitly taking water molecules into account was employed to obtain a large set of uPA-uPAR residue-residue interaction energies. In order of importance, not only were Phe25 > Tyr24 > Trp30 > Ile28 shown to be the most relevant uPA residues binding it to uPAR, but the residues Lys98 > His87 > Gln40 > Asn22 > Lys23 > Val20 also had significant interaction energies, which helps to explain published experimental mutational data. Furthermore, the results obtained with the uPA-uPAR in/homogeneous dielectric function show that a high dielectric constant value ε = 40 is adequate to take into account the electrostatic environment at the interface between the proteins, while using a smaller value of ε (<10) leads to an overestimation of the uPA-uPAR binding energy. Hot spots of the uPA-uPAR binding domain were identified and a quantum biochemistry description of the uPAR blockers uPA21-30 and cyclo21,29uPA21-29[(S21C;H29C)] was performed, demonstrating that cyclization improves the stability of mimetic peptides without compromising their binding energies to uPAR.


Assuntos
Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/química , Sequência de Aminoácidos , Aminoácidos/química , Teoria da Densidade Funcional , Peptídeos Cíclicos/química , Ligação Proteica , Conformação Proteica , Eletricidade Estática , Relação Estrutura-Atividade , Termodinâmica
7.
Ann Clin Lab Sci ; 49(6): 710-721, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31882421

RESUMO

The SLURP1 (secreted LY6/urokinase type plasminogen activator receptor related protein-1) belongs to the gene family of urokinase, a type of plasminogen activator receptor (uPAR). Mutations in the SLURP1 have been reported to cause serious genetic problems of skin, Mal De Meleda, and malignancies. With the advancement of computational tools, it became possible to predict the potential impact of gene variants on the structure and function of protein. Therefore, in present study, we aimed to perform in-silico analyses of the disease causing SLURP1 mutations using online tools. In-total, 21 variants occurring in coding and non-coding regions of SLURP1 were found from public databases. In curated data, we have found 57.14% (12/21) missense, 23.81% (5/21) splice site, 9.52% (2/21) nonsense, 4.76% (1/21) deletion, and 4.76% (1/21) frameshift mutations. Moreover, heterogeneity in genotypes and phenotypes, along with 7 hotspot points in SLURP1 has been noted. In-silico analyses of the subjected variants have depicted a range of pathogenicity by combinatorial predictions of different tools from being lowly to highly pathogenic. Thus, the present study paves a platform to link computational analyses of mutations for important regulatory genes that can be undertaken for their phenotypes and their correlation with the disease status in case control studies.


Assuntos
Antígenos Ly/genética , Ceratodermia Palmar e Plantar/genética , Mutação , Ativador de Plasminogênio Tipo Uroquinase/genética , Substituição de Aminoácidos , Animais , Antígenos Ly/química , Antígenos Ly/metabolismo , Humanos , Melanoma/genética , Taxa de Mutação , Mutação de Sentido Incorreto , Sítios de Splice de RNA , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
8.
Cell Death Dis ; 10(10): 775, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31601791

RESUMO

Uterine angiogenesis and vascular remodeling play critical roles in determing the normal menstrual cycle and successful pregnancy. Poor uterine angiogenesis usually results in pregnancy failure. Protein O-fucosyltransferase 1 (poFUT1) is the key enzyme responsible for O-fucosylated glycan biosynthesis on glycoproteins. However, the dynamic expression and regulation of poFUT1 on the uterine angiogenesis and vascular remodeling remain unknown. Here, we showed that the enlargement of the vascular lumen in the secretory phase was greater than that in the proliferative phase of the uterine endometrium during menstrual cycle; whereas there was a narrower vessel lumen and fewer blood vessels in the decidua from miscarriage patients than in that from healthy pregnancy women. Additionally, the expression of poFUT1 was increased in the uterine endometrium during the secretory phase compared with that in the proliferation phase, and its expression was decreased in the uterus of miscarriage patients compared with that of the healthy pregnancy women. Using hESCs and a mouse model, we demonstrated that poFUT1 increased the O-fucosylation on uPA, and activated of the RhoA signaling pathway, thus facilitating uterine angiogenesis and vascular remodeling. We also provide evidence that poFUT1 promotes hESCs angiogenesis by the decreased stemness of hESCs. These findings reveal a new insight into the uterine angiogenesis and vascular remodeling. The study suggests that poFUT1 could be seen as a novel potential diagnostic and therapeutic target for miscarriage.


Assuntos
Aborto Espontâneo/enzimologia , Endométrio/irrigação sanguínea , Endométrio/enzimologia , Glucosiltransferases/metabolismo , Neovascularização Patológica/enzimologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Aborto Espontâneo/metabolismo , Animais , Endométrio/citologia , Endométrio/metabolismo , Feminino , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Glucosiltransferases/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/genética , Gravidez , Transdução de Sinais/fisiologia , Células Estromais/enzimologia , Células Estromais/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Remodelação Vascular/genética , Remodelação Vascular/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo
9.
ACS Appl Mater Interfaces ; 11(42): 38417-38428, 2019 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-31556584

RESUMO

Triple negative breast cancer (TNBC) is insensitive to either chemotherapy or endocrine therapy because of the powerful DNA reparation and the negative expression of surface antigens, which urgently claims for an effective approach to improve the prognosis. Herein, DNA repair blocker BRCA1 small interfering RNA (siRNA) was introduced with cisplatin (Pt) into the elaborately designed pH-sensitive shell-core platform to enhance the chemotherapeutic treatment effect by silencing the DNA repair related gene. In this platform, BRCA1 siRNA and Pt prodrug (Pro-Pt) were separately encapsulated in the porous outer shell and hydrophobic inner core with extremely high encapsulation efficiency and stability effectively preventing them from degradation during circulation. Suitable size and urokinase plasminogen activator analogues (uPA) with high affinity for the uPA receptor (uPAR) realized an excellent dual passive and active tumor targeting ability. Moreover, the exposed PEG hydrophilic chain prevented the nanoparticles (NPs) from precipitating by serum protein or inactivating by nuclease in the blood cycle. Most importantly, the degradable CaP (calcium ions and phosphate ions) shell with smart pH sensitivity would dissipate from NPs in the lysosomes to burst the lysosome membranes so as to guarantee the lysosomal escape and the sequential release of the siRNA and Pro-Pt where the BRCA1 siRNA blocked the DNA repairing pathway followed by reducing Pro-Pt to Pt for irreversible DNA damage. Hence, the uPA-SP@CaP NPs provided a promising strategy for high-efficiency treatment of TNBC along with bringing new hope for more patients.


Assuntos
Proteína BRCA1/metabolismo , Dano ao DNA/efeitos dos fármacos , Nanopartículas/química , Pró-Fármacos/farmacologia , RNA Interferente Pequeno/metabolismo , Animais , Proteína BRCA1/antagonistas & inibidores , Proteína BRCA1/genética , Cálcio/química , Linhagem Celular Tumoral , Cisplatino/química , Cisplatino/farmacologia , Feminino , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Camundongos Endogâmicos BALB C , Fosfatos/química , Porosidade , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , RNA Interferente Pequeno/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
10.
Adv Mater ; 31(46): e1904742, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31560141

RESUMO

Chemotherapy is widely used in the clinic though its benefits are controversial owing to low cancer specificity. Nanovehicles capable of selectively transporting drugs to cancer cells have been energetically pursued to remodel cancer treatment. However, no active targeting nanomedicines have succeeded in clinical translation to date, partly due to either modest targetability or complex fabrication. CD44-specific A6 short peptide (KPSSPPEE) functionalized polymersomal epirubicin (A6-PS-EPI), which boosts targetability and anticancer efficacy toward human multiple myeloma (MM) in vivo, is described. A6-PS-EPI encapsulating 11 wt% EPI is small (≈55 nm), robust, reduction-responsive, and easy to fabricate. Of note, A6 decoration markedly augments the uptake and anticancer activity of PS-EPI in CD44-overexpressing LP-1 MM cells. A6-PS-EPI displays remarkable targeting ability to orthotopic LP-1 MM, causing depleted bone damage and striking survival benefits compared to nontargeted PS-EPI. Overall, A6-PS-EPI, as a simple and intelligent nanotherapeutic, demonstrates high potential for clinical translation.


Assuntos
Epirubicina/química , Epirubicina/farmacologia , Receptores de Hialuronatos/metabolismo , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Sinergismo Farmacológico , Humanos , Camundongos , Mieloma Múltiplo/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/uso terapêutico
11.
J Labelled Comp Radiopharm ; 62(8): 483-494, 2019 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-30970388

RESUMO

The treatment of cancer remains a major challenge, especially after tumour cell dissemination and metastases formation. Expression of the urokinase-type plasminogen activation system including urokinase (uPA) and its receptor (uPAR) has been associated with the complex process of cell migration, a tumour's invasive potential as well as a reduced overall and disease-free survival of patients with solid cancers and haematological disorders. A cyclic peptide cyclo[21,29][d-Cys21 ,Cys29 ]-uPA21-30 was designed from the growth factor-like domain (GFD) of urokinase whose binding to uPAR was found to inhibit tumour growth and spread of human ovarian cancer cells in mice. With the aim of visualising uPAR expression using PET imaging to attempt an estimate on the tumour's aggressiveness, the cyclic peptide was modified with an either C- or N-terminally attached variable spacer and chelator. The free ligands were evaluated for their binding affinities to the isolated human uPAR and labelled with 68 Ga and 177 Lu to assess their lipophilicities and stabilities in human serum. Although retaining the full binding potential displayed by cyclo[21,29][d-Cys21 ,Cys29 ]-uPA21-30 to its target was found to be a challenging task upon both C- and N-terminal modification, chelator-bearing ligands were identified that can serve as promising starting points in the development of uPAR-addressing PET tracers.


Assuntos
Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ligação Competitiva , Humanos , Marcação por Isótopo , Traçadores Radioativos
12.
J Biol Chem ; 294(18): 7403-7418, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30894413

RESUMO

The urokinase receptor (uPAR) is a founding member of a small protein family with multiple Ly6/uPAR (LU) domains. The motif defining these LU domains contains five plesiotypic disulfide bonds stabilizing its prototypical three-fingered fold having three protruding loops. Notwithstanding the detailed knowledge on structure-function relationships in uPAR, one puzzling enigma remains unexplored. Why does the first LU domain in uPAR (DI) lack one of its consensus disulfide bonds, when the absence of this particular disulfide bond impairs the correct folding of other single LU domain-containing proteins? Here, using a variety of contemporary biophysical methods, we found that reintroducing the two missing half-cystines in uPAR DI caused the spontaneous formation of the corresponding consensus 7-8 LU domain disulfide bond. Importantly, constraints due to this cross-link impaired (i) the binding of uPAR to its primary ligand urokinase and (ii) the flexible interdomain assembly of the three LU domains in uPAR. We conclude that the evolutionary deletion of this particular disulfide bond in uPAR DI may have enabled the assembly of a high-affinity urokinase-binding cavity involving all three LU domains in uPAR. Of note, an analogous neofunctionalization occurred in snake venom α-neurotoxins upon loss of another pair of the plesiotypic LU domain half-cystines. In summary, elimination of the 7-8 consensus disulfide bond in the first LU domain of uPAR did have significant functional and structural consequences.


Assuntos
Evolução Biológica , Deleção de Sequência , Sulfetos/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Fenômenos Biofísicos , Quimotripsina/metabolismo , Glicosilação , Cinética , Ligantes , Dobramento de Proteína , Proteólise , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Ativador de Plasminogênio Tipo Uroquinase/química
14.
J Pharm Biomed Anal ; 164: 241-248, 2019 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-30396051

RESUMO

The urokinase plasminogen activator (uPA) is regarded as the crucial trigger for plasmin generation, which is involved in several diseases especially for neoplasm metastasis. In this study, an efficient approach integrating ultrafiltration, LC/MS, bioassay and in silico docking, was proposed for rapidly detecting uPA ligands from Traditional Chinese Medicines (TCMs). Forty-two TCMs were initially assessed, and as illustrative case studies, Galla Chinensis and Sanguisorbae Radix, which appeared significant inhibitory activities on uPA, were chosen to develpe and verify the strategy. A total of seven uPA ligands were successfully detected and identified. Two of them, pentagalloylglucose and 28-O-ß-d-glucopyranosyl pomolic acid, were demonstrated to be potential inhibitors, with IC50 at 1.639 µM and 37.82 µM repectively. Furthermore, a combinatorial compound library screening combined with in silico docking assay, was revealed that ursolic acid (IC50 = 2.623 µM) was also speculated to be a potent parent structure for inhibition of uPA. This approach offers a multidimensional perspective to discover uPA-binding leading compounds from TCMs or other complex mixtures, which would provide an efficient route for drug discovery.


Assuntos
Descoberta de Drogas/métodos , Medicamentos de Ervas Chinesas/análise , Ensaios Enzimáticos/métodos , Inibidores Enzimáticos/análise , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Cromatografia Líquida de Alta Pressão/instrumentação , Cromatografia Líquida de Alta Pressão/métodos , Descoberta de Drogas/instrumentação , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Ensaios Enzimáticos/instrumentação , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Rhus/química , Sanguisorba/química , Espectrometria de Massas em Tandem/instrumentação , Espectrometria de Massas em Tandem/métodos , Triterpenos/análise , Triterpenos/química , Triterpenos/farmacologia , Ultrafiltração/instrumentação , Ultrafiltração/métodos , Ativador de Plasminogênio Tipo Uroquinase/química , Ácido Ursólico
15.
Chembiochem ; 20(1): 46-50, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30225958

RESUMO

Urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) are two serine proteases that contribute to initiating fibrinolysis by activating plasminogen. uPA is also an important tumour-associated protease due to its role in extracellular matrix remodelling. Overexpression of uPA has been identified in several different cancers and uPA inhibition has been reported as a promising therapeutic strategy. Although several peptide-based uPA inhibitors have been developed, the extent to which uPA tolerates different tetrapeptide sequences that span the P1-P4 positions remains to be thoroughly explored. In this study, we screened a sequence-defined peptide aldehyde library against uPA and tPA. Preferred sequences from the library screen yielded potent inhibitors for uPA, led by Ac-GTAR-H (Ki =18 nm), but not for tPA. Additionally, synthetic peptide substrates corresponding to preferred inhibitor sequences were cleaved with high catalytic efficiency by uPA but not by tPA. These findings provide new insights into the binding specificity of uPA and tPA and the relative activity of tetrapeptide inhibitors and substrates against these enzymes.


Assuntos
Aldeídos/química , Inibidores Enzimáticos/química , Peptídeos/química , Ativador de Plasminogênio Tecidual/química , Ativador de Plasminogênio Tipo Uroquinase/química , Aldeídos/síntese química , Domínio Catalítico , Inibidores Enzimáticos/síntese química , Humanos , Biblioteca de Peptídeos , Peptídeos/síntese química , Especificidade por Substrato , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores
16.
Cell Commun Signal ; 16(1): 62, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30241478

RESUMO

BACKGROUND: TEM8 is a cell membrane protein predominantly expressed in tumor endothelium, which serves as a receptor for the protective antigen (PA) of anthrax toxin. However, the physiological ligands for TEM8 remain unknown. RESULTS: Here we identified uPA as an interacting partner of TEM8. Binding of uPA stimulated the phosphorylation of TEM8 and augmented phosphorylation of EGFR and ERK1/2. Finally, TEM8-Fc, a recombinant fusion protein comprising the extracellular domain of human TEM8 linked to the Fc portion of human IgG1, efficiently abrogated the interaction between uPA and TEM8, blocked uPA-induced migration of HepG2 cells in vitro and inhibited the growth and metastasis of human MCF-7 xenografts in vivo. uPA, TEM8 and EGFR overexpression and ERK1/2 phosphorylation were found co-located on frozen cancer tissue sections. CONCLUSIONS: Taken together, our data provide evidence that TEM8 is a novel receptor for uPA, which may play a significant role in the regulation of tumor growth and metastasis.


Assuntos
Receptores ErbB/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Proliferação de Células , Humanos , Cinética , Proteínas dos Microfilamentos , Metástase Neoplásica , Fosforilação , Domínios Proteicos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química
17.
Phys Chem Chem Phys ; 20(35): 22818-22830, 2018 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-30151512

RESUMO

Urokinase plasminogen activator (uPA) is a biomarker and therapeutic target for several cancer types whose inhibition has been shown to slow tumor growth and metastasis. In this work, crystallographic data of uPA complexed with distinct ligands (PDB id: 1SQA, 1SQO, and 1FV9) were used to perform quantum biochemistry calculations based on the framework of density functional theory (DFT) and within the molecular fractionation with conjugated caps (MFCC) scheme. Our calculations revealed a total energy interaction of -107.30, -99.5, and -35.30 kcal mol-1 for two naphthamidine-based compounds (Ul1 and UI2) and 2-amino-5-hydroxybenzimidazole (172), respectively, which are in good agreement with known inhibitory experiments. Residues Asp189, Ser190, Cys191-Cys220, Gln192, Trp 215, Gly216, and Gly219 were identified as the main interacting amino acid residues with interaction energy contributions lower than -4.0 kcal mol-1 for uPA/UI1 and UPA/UI2 complexes. In the case of compound 172, our calculations have shown that the most important interactions occur with residues Asp189, Cys191-Cys220, and Ser190. Our results highlight the relevance of the protonation state of ligands and residues and that the naphthamidine scaffold of UI1 and UI2 is the main determinant of their potency, followed by their aminopyrimidine substitution. Altogether, the results of this work contribute to the understanding of the uPA binding mechanisms of the inhibitory compounds Ul1 and 172, stimulating the use of quantum biochemistry theoretical approaches for the development of new uPA inhibitors as new medicines for cancer treatment.


Assuntos
Benzimidazóis/química , Naftalenos/química , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/química , Humanos , Modelos Moleculares , Ligação Proteica , Teoria Quântica
18.
Acta Biomater ; 70: 227-236, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29412186

RESUMO

Thrombosis, a critical event in blood vessels, not only is associated with myocardial infarction and stroke, but also accounts for considerable morbidity and mortality. Thrombolytic drugs are usually applied to the treatment of acute myocardial infarction, acute cerebral infarction and pulmonary embolism. However, thrombolytic drugs show limited efficacy in clinical practice because of the short half-life in plasma and systemic side effects. In this study, the cyclic RGD (cRGD) functionalized liposomes were prepared to encapsulate urokinase, a cheap and widely used thrombolytic drug in clinic and better thrombolysis efficacy was achieved. The flow cytometry analysis showed that the cRGD liposomes could bind to the activated platelets while not to the resting platelets. In vitro release study revealed that the release percentage reached plateau in about 5 h with 60% urokinase being released from liposomes. Results from the in vitro thrombolysis experiments demonstrated a good thrombolysis potential of the cRGD urokinase liposomes. The in vivo thrombolysis study demonstrated that the cRGD liposomes could significantly reduce the dose of urokinase by 75% while achieving the equivalent thrombolysis effect as the free urokinase in mouse mesenteric thrombosis model. In conclusion, the cRGD liposomes encapsulating urokinase hold great promise in clinic for better thrombolytic efficacy. STATEMENT OF SIGNIFICANCE: In this paper, the cRGD liposomes were prepared to encapsulate urokinase for targeted thrombolysis therapy. The cRGD liposomes could specifically bind to the activated platelets and could stably and continuously release its loaded urokinase. The mouse mesenteric thrombosis model was established to evaluate the thrombolysis effect of the cRGD urokinase liposomes. The results demonstrated that the cRGD liposomes could improve the thrombolytic efficacy by almost 4-fold over free urokinase. In conclusion, the cRGD liposomes encapsulating urokinase had great potential for the clinical treatment of thrombosis.


Assuntos
Enzimas Imobilizadas , Peptídeos Cíclicos , Terapia Trombolítica , Trombose/tratamento farmacológico , Ativador de Plasminogênio Tipo Uroquinase , Animais , Modelos Animais de Doenças , Enzimas Imobilizadas/química , Enzimas Imobilizadas/farmacologia , Humanos , Lipossomos , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Trombose/metabolismo , Trombose/patologia , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/farmacologia
19.
J Comput Biol ; 24(11): 1134-1137, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28570103

RESUMO

We analyzed 78 binding pockets of the human urokinase plasminogen activator (uPA) catalytic domain extracted from a data set of crystallized uPA-ligand complexes. These binding pockets were computed with an original geometric method that does NOT involve any arbitrary parameter, such as cutoff distances, angles, and so on. We measured the deviation from convexity of each pocket shape with the pocket convexity index (PCI). We defined a new pocket descriptor called distributional sphericity coefficient (DISC), which indicates to which extent the protein atoms of a given pocket lie on the surface of a sphere. The DISC values were computed with the freeware PCI. The pocket descriptors and their high correspondences with ligand descriptors are crucial for polypharmacology prediction. We found that the protein heavy atoms lining the urokinases binding pockets are either located on the surface of their convex hull or lie close to this surface. We also found that the radii of the urokinases binding pockets and the radii of their ligands are highly correlated (r = 0.9).


Assuntos
Modelos Moleculares , Software , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sítios de Ligação , Humanos , Ligantes , Ligação Proteica , Conformação Proteica
20.
Sci Rep ; 7(1): 3385, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28611361

RESUMO

Although trypsin-like serine proteases have flexible surface-exposed loops and are known to adopt higher and lower activity conformations, structural determinants for the different conformations have remained largely obscure. The trypsin-like serine protease, urokinase-type plasminogen activator (uPA), is central in tissue remodeling processes and also strongly implicated in tumor metastasis. We solved five X-ray crystal structures of murine uPA (muPA) in the absence and presence of allosteric molecules and/or substrate-like molecules. The structure of unbound muPA revealed an unsuspected non-chymotrypsin-like protease conformation in which two ß-strands in the core of the protease domain undergoes a major antiparallel-to-parallel conformational transition. We next isolated two anti-muPA nanobodies; an active-site binding nanobody and an allosteric nanobody. Crystal structures of the muPA:nanobody complexes and hydrogen-deuterium exchange mass spectrometry revealed molecular insights about molecular factors controlling the antiparallel-to-parallel equilibrium in muPA. Together with muPA activity assays, the data provide valuable insights into regulatory mechanisms and conformational flexibility of uPA and trypsin-like serine proteases in general.


Assuntos
Conformação Proteica , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Especificidade de Anticorpos , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Camundongos , Modelos Moleculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA