Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35046049

RESUMEN

Cancer immunotherapy frequently fails because most carcinomas have few T cells, suggesting that cancers can suppress T cell infiltration. Here, we show that cancer cells of human pancreatic ductal adenocarcinoma (PDA), colorectal cancer, and breast cancer are coated with transglutaminase-2 (TGM2)-dependent covalent CXCL12-keratin-19 (KRT19) heterodimers that are organized as filamentous networks. Since a dimeric form of CXCL12 suppresses the motility of human T cells, we determined whether this polymeric CXCL12-KRT19 coating mediated T cell exclusion. Mouse tumors containing control PDA cells exhibited the CXCL12-KRT19 coating, excluded T cells, and did not respond to treatment with anti-PD-1 antibody. Tumors containing PDA cells not expressing either KRT19 or TGM2 lacked the CXCL12-KRT19 coating, were infiltrated with activated CD8+ T cells, and growth was suppressed with anti-PD-1 antibody treatment. Thus, carcinomas assemble a CXCL12-KRT19 coating to evade cancer immune attack.


Asunto(s)
Carcinoma/etiología , Carcinoma/metabolismo , Quimiocina CXCL12/metabolismo , Citotoxicidad Inmunológica , Queratina-19/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Neoplasias de la Mama , Carcinoma/patología , Línea Celular Tumoral , Quimiocina CXCL12/química , Femenino , Humanos , Queratina-19/química , Masculino , Ratones , Repeticiones de Microsatélite , Neoplasias Pancreáticas , Unión Proteica , Multimerización de Proteína , Neoplasias Pancreáticas
2.
PLoS Biol ; 18(4): e3000656, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32271748

RESUMEN

Chemokines and their receptors are orchestrators of cell migration in humans. Because dysregulation of the receptor-chemokine system leads to inflammation and cancer, both chemokines and receptors are highly sought therapeutic targets. Yet one of the barriers for their therapeutic targeting is the limited understanding of the structural principles behind receptor-chemokine recognition and selectivity. The existing structures do not include CXC subfamily complexes and lack information about the receptor distal N-termini, despite the importance of the latter in signaling, regulation, and bias. Here, we report the discovery of the geometry of the complex between full-length CXCR4, a prototypical CXC receptor and driver of cancer metastasis, and its endogenous ligand CXCL12. By comprehensive disulfide cross-linking, we establish the existence and the structure of a novel interface between the CXCR4 distal N-terminus and CXCL12 ß1-strand, while also recapitulating earlier findings from nuclear magnetic resonance, modeling and crystallography of homologous receptors. A cross-linking-informed high-resolution model of the CXCR4-CXCL12 complex pinpoints the interaction determinants and reveals the occupancy of the receptor major subpocket by the CXCL12 proximal N terminus. This newly found positioning of the chemokine proximal N-terminus provides a structural explanation of CXC receptor-chemokine selectivity against other subfamilies. Our findings challenge the traditional two-site understanding of receptor-chemokine recognition, suggest the possibility of new affinity and signaling determinants, and fill a critical void on the structural map of an important class of therapeutic targets. These results will aid the rational design of selective chemokine-receptor targeting small molecules and biologics with novel pharmacology.


Asunto(s)
Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Receptores CXCR4/química , Receptores CXCR4/metabolismo , Animales , Sitios de Unión , Western Blotting , Quimiocina CXCL12/genética , Cisteína/química , Cisteína/genética , Disulfuros/química , Citometría de Flujo , Células HEK293 , Humanos , Insectos/citología , Modelos Moleculares , Mutación , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores CXCR4/genética , beta-Arrestinas/metabolismo
3.
Nucleic Acids Res ; 49(D1): D687-D693, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33084904

RESUMEN

Despite antibiotic resistance being a matter of growing concern worldwide, the bacterial mechanisms of pathogenesis remain underexplored, restraining our ability to develop new antimicrobials. The rise of high-throughput sequencing technology has made available a massive amount of transcriptomic data that could help elucidate the mechanisms underlying bacterial infection. Here, we introduce the DualSeqDB database, a resource that helps the identification of gene transcriptional changes in both pathogenic bacteria and their natural hosts upon infection. DualSeqDB comprises nearly 300 000 entries from eight different studies, with information on bacterial and host differential gene expression under in vivo and in vitro conditions. Expression data values were calculated entirely from raw data and analyzed through a standardized pipeline to ensure consistency between different studies. It includes information on seven different strains of pathogenic bacteria and a variety of cell types and tissues in Homo sapiens, Mus musculus and Macaca fascicularis at different time points. We envisage that DualSeqDB can help the research community in the systematic characterization of genes involved in host infection and help the development and tailoring of new molecules against infectious diseases. DualSeqDB is freely available at http://www.tartaglialab.com/dualseq.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Interacciones Huésped-Patógeno/genética , Infecciones/genética , Análisis de Secuencia de ARN , Secuencia de Aminoácidos , Quimiocina CXCL12/química , Regulación de la Expresión Génica , Humanos
4.
J Biol Chem ; 295(40): 13927-13939, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-32788219

RESUMEN

The human chemokine family consists of 46 protein ligands that induce chemotactic cell migration by activating a family of 23 G protein-coupled receptors. The two major chemokine subfamilies, CC and CXC, bind distinct receptor subsets. A sequence motif defining these families, the X position in the CXC motif, is not predicted to make significant contacts with the receptor, but instead links structural elements associated with binding and activation. Here, we use comparative analysis of chemokine NMR structures, structural modeling, and molecular dynamic simulations that suggested the X position reorients the chemokine N terminus. Using CXCL12 as a model CXC chemokine, deletion of the X residue (Pro-10) had little to no impact on the folded chemokine structure but diminished CXCR4 agonist activity as measured by ERK phosphorylation, chemotaxis, and Gi/o-mediated cAMP inhibition. Functional impairment was attributed to over 100-fold loss of CXCR4 binding affinity. Binding to the other CXCL12 receptor, ACKR3, was diminished nearly 500-fold. Deletion of Pro-10 had little effect on CXCL12 binding to the CXCR4 N terminus, a major component of the chemokine-GPCR interface. Replacement of the X residue with the most frequent amino acid at this position (P10Q) had an intermediate effect between WT and P10del in each assay, with ACKR3 having a higher tolerance for this mutation. This work shows that the X residue helps to position the CXCL12 N terminus for optimal docking into the orthosteric pocket of CXCR4 and suggests that the CC/CXC motif contributes directly to receptor selectivity by orienting the chemokine N terminus in a subfamily-specific direction.


Asunto(s)
Quimiocina CXCL12/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Receptores CXCR4/química , Receptores CXCR/química , Secuencias de Aminoácidos , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Humanos , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Relación Estructura-Actividad
5.
J Biol Chem ; 295(41): 14111-14124, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32753481

RESUMEN

The multifaceted adaptor protein ß-arr1 (ß-arrestin1) promotes activation of focal adhesion kinase (FAK) by the chemokine receptor CXCR4, facilitating chemotaxis. This function of ß-arr1 requires the assistance of the adaptor protein STAM1 (signal-transducing adaptor molecule 1) because disruption of the interaction between STAM1 and ß-arr1 reduces CXCR4-mediated activation of FAK and chemotaxis. To begin to understand the mechanism by which ß-arr1 together with STAM1 activates FAK, we used site-directed spin-labeling EPR spectroscopy-based studies coupled with bioluminescence resonance energy transfer-based cellular studies to show that STAM1 is recruited to activated ß-arr1 by binding to a novel surface on ß-arr1 at the base of the finger loop, at a site that is distinct from the receptor-binding site. Expression of a STAM1-deficient binding ß-arr1 mutant that is still able to bind to CXCR4 significantly reduced CXCL12-induced activation of FAK but had no impact on ERK-1/2 activation. We provide evidence of a novel surface at the base of the finger loop that dictates non-GPCR interactions specifying ß-arrestin-dependent signaling by a GPCR. This surface might represent a previously unidentified switch region that engages with effector molecules to drive ß-arrestin signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Complejos de Clasificación Endosomal Requeridos para el Transporte , Sistema de Señalización de MAP Quinasas , Fosfoproteínas , Receptores CXCR4 , beta-Arrestina 1 , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Células HEK293 , Humanos , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estructura Secundaria de Proteína , Receptores CXCR4/química , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , beta-Arrestina 1/química , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo
6.
Biol Chem ; 402(11): 1453-1464, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34218538

RESUMEN

The delivery of chemotactic signaling molecules via customized biomaterials can effectively guide the migration of cells to improve the regeneration of damaged or diseased tissues. Here, we present a novel biohybrid hydrogel system containing two different sulfated glycosaminoglycans (sGAG)/sGAG derivatives, namely either a mixture of short heparin polymers (Hep-Mal) or structurally defined nona-sulfated tetrahyaluronans (9s-HA4-SH), to precisely control the release of charged signaling molecules. The polymer networks are described in terms of their negative charge, i.e. the anionic sulfate groups on the saccharides, using two parameters, the integral density of negative charge and the local charge distribution (clustering) within the network. The modulation of both parameters was shown to govern the release characteristics of the chemotactic signaling molecule SDF-1 and allows for seamless transitions between burst and sustained release conditions as well as the precise control over the total amount of delivered protein. The obtained hydrogels with well-adjusted release profiles effectively promote MSC migration in vitro and emerge as promising candidates for new treatment modalities in the context of bone repair and wound healing.


Asunto(s)
Quimiocina CXCL12/metabolismo , Glicosaminoglicanos/metabolismo , Hidrogeles/metabolismo , Quimiocina CXCL12/química , Glicosaminoglicanos/química , Humanos , Hidrogeles/síntesis química , Hidrogeles/química , Células Madre Mesenquimatosas/metabolismo , Estructura Molecular
7.
Biol Chem ; 402(11): 1337-1355, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-33882203

RESUMEN

The interaction of regulatory proteins with extracellular matrix or cell surface-anchored glycosaminoglycans (GAGs) plays important roles in molecular recognition, wound healing, growth, inflammation and many other processes. In spite of their high biological relevance, protein-GAG complexes are significantly underrepresented in structural databases because standard tools for structure determination experience difficulties in studying these complexes. Co-crystallization with subsequent X-ray analysis is hampered by the high flexibility of GAGs. NMR spectroscopy experiences difficulties related to the periodic nature of the GAGs and the sparse proton network between protein and GAG with distances that typically exceed the detection limit of nuclear Overhauser enhancement spectroscopy. In contrast, computer modeling tools have advanced over the last years delivering specific protein-GAG docking approaches successfully complemented with molecular dynamics (MD)-based analysis. Especially the combination of NMR spectroscopy in solution providing sparse structural constraints with molecular docking and MD simulations represents a useful synergy of forces to describe the structure of protein-GAG complexes. Here we review recent methodological progress in this field and bring up examples where the combination of new NMR methods along with cutting-edge modeling has yielded detailed structural information on complexes of highly relevant cytokines with GAGs.


Asunto(s)
Quimiocina CXCL12/metabolismo , Quimiocinas CXC/metabolismo , Glicosaminoglicanos/metabolismo , Interleucina-10/metabolismo , Quimiocina CXCL12/química , Quimiocinas CXC/química , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Glicosaminoglicanos/química , Humanos , Interleucina-10/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares
8.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769095

RESUMEN

Long-term delivery of growth factors and immunomodulatory agents is highly required to support the integrity of tissue in engineering constructs, e.g., formation of vasculature, and to minimize immune response in a recipient. However, for proteins with a net positive charge at the physiological pH, controlled delivery from negatively charged alginate (Alg) platforms is challenging due to electrostatic interactions that can hamper the protein release. In order to regulate such interactions between proteins and the Alg matrix, we propose to complex proteins of interest in this study - CXCL12, FGF-2, VEGF - with polyanionic heparin prior to their encapsulation into Alg microbeads of high content of α-L-guluronic acid units (high-G). This strategy effectively reduced protein interactions with Alg (as shown by model ITC and SPR experiments) and, depending on the protein type, afforded control over the protein release for at least one month. The released proteins retained their in vitro bioactivity: CXCL12 stimulated the migration of Jurkat cells, and FGF-2 and VEGF induced proliferation and maturation of HUVECs. The presence of heparin also intensified protein biological efficiency. The proposed approach for encapsulation of proteins with a positive net charge into high-G Alg hydrogels is promising for controlled long-term protein delivery under in vivo conditions.


Asunto(s)
Alginatos/química , Quimiocina CXCL12/química , Factor 2 de Crecimiento de Fibroblastos/química , Heparina/química , Factor A de Crecimiento Endotelial Vascular/química , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microesferas , Ingeniería de Tejidos
9.
Molecules ; 25(20)2020 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-33092204

RESUMEN

CXCL12 are small pro-inflammatory chemo-attractant cytokines that bind to a specific receptor CXCR4 with a role in angiogenesis, tumor progression, metastasis, and cell survival. Globally, cancer metastasis is a major cause of morbidity and mortality. In this study, we targeted CXCL12 rather than the chemokine receptor (CXCR4) because most of the drugs failed in clinical trials due to unmanageable toxicities. Until now, no FDA approved medication has been available against CXCL12. Therefore, we aimed to find new inhibitors for CXCL12 through virtual screening followed by molecular dynamics simulation. For virtual screening, active compounds against CXCL12 were taken as potent inhibitors and utilized in the generation of a pharmacophore model, followed by validation against different datasets. Ligand based virtual screening was performed on the ChEMBL and in-house databases, which resulted in successive elimination through the steps of pharmacophore-based and score-based screenings, and finally, sixteen compounds of various interactions with significant crucial amino acid residues were selected as virtual hits. Furthermore, the binding mode of these compounds were refined through molecular dynamic simulations. Moreover, the stability of protein complexes, Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF), and radius of gyration were analyzed, which led to the identification of three potent inhibitors of CXCL12 that may be pursued in the drug discovery process against cancer metastasis.


Asunto(s)
Aminoácidos/antagonistas & inhibidores , Quimiocina CXCL12/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos , Ligandos , Aminoácidos/química , Sitios de Unión/efectos de los fármacos , Quimiocina CXCL12/química , Química Computacional , Humanos , Enlace de Hidrógeno/efectos de los fármacos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad Cuantitativa , Receptores CXCR4/química , Interfaz Usuario-Computador
10.
Arterioscler Thromb Vasc Biol ; 38(7): e117-e134, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29853570

RESUMEN

OBJECTIVE: The objective of this study was to develop small-diameter vascular grafts capable of eluting SDF (stromal cell-derived factor)-1α-derived peptide and SP (substance P) for in situ vascular regeneration. APPROACH AND RESULTS: Polycaprolactone (PCL)/collagen grafts containing SP or SDF-1α-derived peptide were fabricated by electrospinning. SP and SDF-1α peptide-loaded grafts recruited significantly higher numbers of mesenchymal stem cells than that of the control group. The in vivo potential of PCL/collagen, SDF-1, and SP grafts was assessed by implanting them in a rat abdominal aorta for up to 4 weeks. All grafts remained patent as observed using color Doppler and stereomicroscope. Host cells infiltrated into the graft wall and the neointima was formed in peptides-eluting grafts. The lumen of the SP grafts was covered by the endothelial cells with cobblestone-like morphology, which were elongated in the direction of the blood flow, as discerned using scanning electron microscopy. Moreover, SDF-1α and SP grafts led to the formation of a confluent endothelium as evaluated using immunofluorescence staining with von Willebrand factor antibody. SP and SDF-1α grafts also promoted smooth muscle cell regeneration, endogenous stem cell recruitment, and blood vessel formation, which was the most prominent in the SP grafts. Evaluation of inflammatory response showed that 3 groups did not significantly differ in terms of the numbers of proinflammatory macrophages, whereas SP grafts showed significantly higher numbers of proremodeling macrophages than that of the control and SDF-1α grafts. CONCLUSIONS: SDF-1α and SP grafts can be potential candidates for in situ vascular regeneration and are worthy for future investigations.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Aorta Abdominal/cirugía , Implantación de Prótesis Vascular/instrumentación , Prótesis Vascular , Quimiocina CXCL12/farmacología , Materiales Biocompatibles Revestidos , Colágeno Tipo I/química , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Poliésteres/química , Sustancia P/farmacología , Inductores de la Angiogénesis/química , Animales , Aorta Abdominal/diagnóstico por imagen , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/química , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Neointima , Fragmentos de Péptidos/química , Diseño de Prótesis , Ratas Sprague-Dawley , Sustancia P/química , Factores de Tiempo , Ultrasonografía Doppler en Color , Grado de Desobstrucción Vascular , Remodelación Vascular
11.
Biochem J ; 475(4): 723-732, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29301984

RESUMEN

Leukocyte arrest on the endothelial cell surface during leukocyte extravasation is induced by rapid integrin activation by chemokines. We recently reported that fractalkine induces integrin activation without its receptor CX3CR1 through binding to the allosteric site (site 2) of integrins. Peptides from site 2 bound to fractalkine and suppressed integrin activation by fractalkine. We hypothesized that this is not limited to membrane-bound fractalkine. We studied whether stromal cell-derived factor-1 (SDF1), another chemokine that plays a critical role in leukocyte arrest, activates integrins through binding to site 2. We describe here that (1) SDF1 activated soluble integrin αvß3 in cell-free conditions, suggesting that SDF1 can activate αvß3 without CXCR4; (2) site 2 peptide bound to SDF1, suggesting that SDF1 binds to site 2; (3) SDF1 activated integrins αvß3, α4ß1, and α5ß1 on CHO cells (CXCR4-negative) and site 2 peptide suppressed the activation; (4) A CXCR4 antagonist AMD3100 did not affect the site 2-mediated integrin activation by SDF1; (5) Cell-surface integrins were fully activated in 1 min (much faster than activation of soluble αvß3) and the activation lasted at least for 1 h. We propose that the binding of SDF1 to cell-surface proteoglycan facilitates the allosteric activation process; (6) Mutations in the predicted site 2-binding site in SDF1 suppressed integrin activation. These results suggest that SDF1 (e.g. presented on proteoglycans) can rapidly activate integrins in an allosteric manner by binding to site 2 in the absence of CXCR4. The allosteric integrin activation by SDF1 is a novel target for drug discovery.


Asunto(s)
Quimiocina CXCL12/química , Integrinas/química , Receptores CXCR4/química , Sitio Alostérico , Animales , Sitios de Unión , Células CHO , Sistema Libre de Células , Quimiocina CX3CL1/química , Quimiocina CX3CL1/genética , Quimiocina CXCL12/genética , Cricetulus , Humanos , Integrinas/genética , Simulación del Acoplamiento Molecular , Mutación , Unión Proteica , Receptores CXCR4/genética , Transducción de Señal/genética
12.
Proc Natl Acad Sci U S A ; 113(35): 9928-33, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27543332

RESUMEN

The atomic-level mechanisms by which G protein-coupled receptors (GPCRs) transmit extracellular ligand binding events through their transmembrane helices to activate intracellular G proteins remain unclear. Using a comprehensive library of mutations covering all 352 residues of the GPCR CXC chemokine receptor 4 (CXCR4), we identified 41 amino acids that are required for signaling induced by the chemokine ligand CXCL12 (stromal cell-derived factor 1). CXCR4 variants with each of these mutations do not signal properly but remain folded, based on receptor surface trafficking, reactivity to conformationally sensitive monoclonal antibodies, and ligand binding. When visualized on the structure of CXCR4, the majority of these residues form a continuous intramolecular signaling chain through the transmembrane helices; this chain connects chemokine binding residues on the extracellular side of CXCR4 to G protein-coupling residues on its intracellular side. Integrated into a cohesive model of signal transmission, these CXCR4 residues cluster into five functional groups that mediate (i) chemokine engagement, (ii) signal initiation, (iii) signal propagation, (iv) microswitch activation, and (v) G protein coupling. Propagation of the signal passes through a "hydrophobic bridge" on helix VI that coordinates with nearly every known GPCR signaling motif. Our results agree with known conserved mechanisms of GPCR activation and significantly expand on understanding the structural principles of CXCR4 signaling.


Asunto(s)
Conformación Proteica , Receptores CXCR4/química , Receptores CXCR4/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Sitios de Unión/genética , Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Mutación , Unión Proteica , Multimerización de Proteína , Receptores CXCR4/genética , Homología de Secuencia de Aminoácido
13.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 594-603, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28040478

RESUMEN

Glioblastoma (GBM) is the most aggressive primary brain tumor with poor patient survival that is at least partly caused by malignant and therapy-resistant glioma stem-like cells (GSLCs) that are protected in GSLC niches. Previously, we have shown that the chemo-attractant stromal-derived factor-1α (SDF-1α), its C-X-C receptor type 4 (CXCR4) and the cysteine protease cathepsin K (CatK) are localized in GSLC niches in glioblastoma. Here, we investigated whether SDF-1α is a niche factor that through its interactions with CXCR4 and/or its second receptor CXCR7 on GSLCs facilitates their homing to niches. Furthermore, we aimed to prove that SDF-1α cleavage by CatK inactivates SDF-1α and inhibits the invasion of GSLCs. We performed mass spectrometric analysis of cleavage products of SDF-1α after proteolysis by CatK. We demonstrated that CatK cleaves SDF-1α at 3 sites in the N-terminus, which is the region of SDF-1α that binds to its receptors. Confocal imaging of human GBM tissue sections confirmed co-localization of SDF-1α and CatK in GSLC niches. In accordance, 2D and 3D invasion experiments using CXCR4/CXCR7-expressing GSLCs and GBM cells showed that SDF-1α had chemotactic activity whereas CatK cleavage products of SDF-1α did not. Besides, CXCR4 inhibitor plerixafor inhibited invasion of CXCR4/CXCR7-expressing GSLCs. In conclusion, CatK can cleave and inactivate SDF-1α. This implies that CatK activity facilitates migration of GSLCs out of niches. We propose that activation of CatK may be a promising strategy to prevent homing of GSLCs in niches and thus render these cells sensitive to chemotherapy and radiation.


Asunto(s)
Catepsina K/metabolismo , Quimiocina CXCL12/metabolismo , Células Madre Neoplásicas/metabolismo , Neuroglía/metabolismo , Receptores CXCR4/metabolismo , Secuencia de Aminoácidos , Bencilaminas , Catepsina K/genética , Línea Celular Tumoral , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiotaxis , Ciclamas , Expresión Génica , Compuestos Heterocíclicos/farmacología , Humanos , Células Madre Neoplásicas/patología , Neuroglía/patología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Proteolisis , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Nicho de Células Madre/genética
14.
Med Res Rev ; 38(4): 1188-1234, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28768055

RESUMEN

CXCR4 antagonists (e.g., PlerixaforTM ) have been successfully validated as stem cell mobilizers for peripheral blood stem cell transplantation. Applications of the CXCR4 antagonists have heralded the era of cell-based therapy and opened a potential therapeutic horizon for many unmet medical needs such as kidney injury, ischemic stroke, cancer, and myocardial infarction. In this review, we first introduce the central role of CXCR4 in diverse cellular signaling pathways and discuss its involvement in several disease progressions. We then highlight the molecular design and optimization strategies for targeting CXCR4 from a large number of case studies, concluding that polyamines are the preferred CXCR4-binding ligands compared to other structural options, presumably by mimicking the highly positively charged natural ligand CXCL12. These results could be further justified with computer-aided docking into the CXCR4 crystal structure wherein both major and minor subpockets of the binding cavity are considered functionally important. Finally, from the clinical point of view, CXCR4 antagonists could mobilize hematopoietic stem/progenitor cells with long-term repopulating capacity to the peripheral blood, promising to replace surgically obtained bone marrow cells as a preferred source for stem cell transplantation.


Asunto(s)
Quimiocina CXCL12/química , Infecciones por VIH/tratamiento farmacológico , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Receptores CXCR4/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Ensayos Clínicos como Asunto , Cristalografía por Rayos X , Humanos , Inmunoterapia , Ligandos , Isquemia Miocárdica/tratamiento farmacológico , Metástasis de la Neoplasia , Neutrófilos/citología , Péptidos/química , Regeneración , Transducción de Señal , Trasplante de Células Madre
15.
Chembiochem ; 19(5): 448-458, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29239081

RESUMEN

An academic chemical screening approach was developed by using 2D protein-detected NMR, and a 352-chemical fragment library was screened against three different protein targets. The approach was optimized against two protein targets with known ligands: CXCL12 and BRD4. Principal component analysis reliably identified compounds that induced nonspecific NMR crosspeak broadening but did not unambiguously identify ligands with specific affinity (hits). For improved hit detection, a novel scoring metric-difference intensity analysis (DIA)-was devised that sums all positive and negative intensities from 2D difference spectra. Applying DIA quickly discriminated potential ligands from compounds inducing nonspecific NMR crosspeak broadening and other nonspecific effects. Subsequent NMR titrations validated chemotypes important for binding to CXCL12 and BRD4. A novel target, mitochondrial fission protein Fis1, was screened, and six hits were identified by using DIA. Screening these diverse protein targets identified quinones and catechols that induced nonspecific NMR crosspeak broadening, hampering NMR analyses, but are currently not computationally identified as pan-assay interference compounds. The results established a streamlined screening workflow that can easily be scaled and adapted as part of a larger screening pipeline to identify fragment hits and assess relative binding affinities in the range of 0.3-1.6 mm. DIA could prove useful in library screening and other applications in which NMR chemical shift perturbations are measured.


Asunto(s)
Quimiocina CXCL12/metabolismo , Descubrimiento de Drogas/métodos , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Proteínas Nucleares/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Quimiocina CXCL12/química , Humanos , Ligandos , Proteínas de la Membrana/química , Proteínas Mitocondriales/química , Modelos Moleculares , Proteínas Nucleares/química , Análisis de Componente Principal , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Factores de Transcripción/química
16.
J Chem Inf Model ; 58(3): 683-691, 2018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29481075

RESUMEN

Fragment-based drug discovery (FBDD) has become a mainstream approach in drug design because it allows the reduction of the chemical space and screening libraries while identifying fragments with high protein-ligand efficiency interactions that can later be grown into drug-like leads. In this work, we leverage high-throughput molecular dynamics (MD) simulations to screen a library of 129 fragments for a total of 5.85 ms against the CXCL12 monomer, a chemokine involved in inflammation and diseases such as cancer. Our in silico binding assay was able to recover binding poses, affinities, and kinetics for the selected library and was able to predict 8 mM-affinity fragments with ligand efficiencies higher than 0.3. All of the fragment hits present a similar chemical structure, with a hydrophobic core and a positively charged group, and bind to either sY7 or H1S68 pockets, where they share pharmacophoric properties with experimentally resolved natural binders. This work presents a large-scale screening assay using an exclusive combination of thousands of short MD adaptive simulations analyzed with a Markov state model (MSM) framework.


Asunto(s)
Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Descubrimiento de Drogas/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Sitios de Unión , Quimiocina CXCL12/química , Diseño de Fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Simulación del Acoplamiento Molecular/métodos , Simulación de Dinámica Molecular , Bibliotecas de Moléculas Pequeñas/química
17.
J Cell Mol Med ; 21(10): 2298-2307, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28374486

RESUMEN

Endothelial progenitor cells (EPCs) play a capital role in angiogenesis via directly participating in neo-vessel formation and secreting pro-angiogenic factors. Stromal cell-derived factor 1 (SDF-1) and its receptor CXCR4 play a critical role in the retention and quiescence of EPCs within its niche in the bone marrow. Disturbing the interaction between SDF-1 and CXCR4 is an effective strategy for EPC mobilization. We developed a novel CXCR4 antagonist P2G, a mutant protein of SDF-1ß with high antagonistic activity against CXCR4 and high potency in enhancing ischaemic angiogenesis and blood perfusion. However, its direct effects on ischaemic tissue remain largely unknown. In this study, P2G was found to possess a robust capability to promote EPC infiltration and incorporation in neo-vessels, enhance the expression and function of pro-angiogenic factors, such as SDF-1, vascular endothelial growth factor and matrix metalloprotein-9, and activate cell signals involved in angiogenesis, such as proliferating cell nuclear antigen, protein kinase B (Akt), extracellular regulated protein kinases and mammalian target of rapamycin, in ischaemic tissue. Moreover, P2G can attenuate fibrotic remodelling to facilitate the recovery of ischaemic tissue. The capability of P2G in direct augmenting ischaemic environment for angiogenesis suggests that it is a potential candidate for the therapy of ischaemia diseases.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Isquemia/prevención & control , Péptidos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Animales , Vasos Sanguíneos/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Isquemia/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Biol Chem ; 291(30): 15881-95, 2016 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-27226569

RESUMEN

An emerging number of non-chemokine mediators are found to bind to classical chemokine receptors and to elicit critical biological responses. Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine that exhibits chemokine-like activities through non-cognate interactions with the chemokine receptors CXCR2 and CXCR4, in addition to activating the type II receptor CD74. Activation of the MIF-CXCR2 and -CXCR4 axes promotes leukocyte recruitment, mediating the exacerbating role of MIF in atherosclerosis and contributing to the wealth of other MIF biological activities. Although the structural basis of the MIF-CXCR2 interaction has been well studied and was found to engage a pseudo-ELR and an N-like loop motif, nothing is known about the regions of CXCR4 and MIF that are involved in binding to each other. Using a genetic strain of Saccharomyces cerevisiae that expresses a functional CXCR4 receptor, site-specific mutagenesis, hybrid CXCR3/CXCR4 receptors, pharmacological reagents, peptide array analysis, chemotaxis, fluorescence spectroscopy, and circular dichroism, we provide novel molecular information about the structural elements that govern the interaction between MIF and CXCR4. The data identify similarities with classical chemokine-receptor interactions but also provide evidence for a partial allosteric agonist compared with CXCL12 that is possible due to the two binding sites of CXCR4.


Asunto(s)
Quimiocina CXCL12 , Oxidorreductasas Intramoleculares , Factores Inhibidores de la Migración de Macrófagos , Receptores CXCR4 , Regulación Alostérica , Animales , Células CHO , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Cricetinae , Cricetulus , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptores CXCR4/química , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Biochem Biophys Res Commun ; 488(2): 303-310, 2017 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-28487111

RESUMEN

CXCL12 overexpression improves neurobehavioral recovery during post-ischemic stroke through multiple mechanisms including promoting endothelial progenitor cells function in animal models. It has been proposed that the monomer and dimer forms possess differential chemotactic and regulatory function. The aim of present study is to explore whether a monomeric or dimeric CXCL12 plays a different role in the endothelial progenitor cells proliferation, migration, and tube-formation in vitro. In this study, we transferred monomeric, dimeric and wild type CXCL12 gene into endothelial progenitor cells via lentiviral vectors. We investigated endothelial progenitor cells function following the interaction of CXCL12/CXCR4 or CXCL12/CXCR7 and downstream signaling pathways. Our results showed that the monomeric CXCL12 transfected endothelial progenitor cells had enhanced ability in cell proliferation, migration, and tube-formation compared to that in dimeric or wild type controls (p < 0.05). Both CXCR4 and CXCR7 were significantly overexpressed in the monomeric CXCL12 transfected endothelial progenitor cells compared to that in the dimeric or wide type controls (p < 0.05). The function of migration, but not proliferation or tube-formation, was significantly reduced in the monomeric CXCL12 transfected endothelial progenitor cells when the cells were pre-treated with either CXCR4 inhibitor AMD3100 or siCXCR7 (p < 0.05), suggesting this cell function was partially regulated by CXCL12/CXCR4 and CXCL12/CXCR7 signal pathways. Our study demonstrated that monomeric CXCL12 was the fundamental form, which played important roles in endothelial progenitor cells' proliferation, migration, and tube-formation.


Asunto(s)
Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Movimiento Celular , Quimiocina CXCL12/genética , Humanos
20.
Int J Clin Oncol ; 22(6): 991-1000, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29022185

RESUMEN

CXC chemokine ligand 12 (CXCL12) is an important member of the CXC subfamily of chemokines, and has been extensively studied in various human body organs and systems, both in physiological and clinical states. Ligation of CXCL12 to CXCR4 and CXCR7 as its receptors on peripheral immune cells gives rise to pleiotropic activities. CXCL12 itself is a highly effective chemoattractant which conservatively attracts lymphocytes and monocytes, whereas there exists no evidence to show attraction for neutrophils. CXCL12 regulates inflammation, neo-vascularization, metastasis, and tumor growth, phenomena which are all pivotally involved in cancer development and further metastasis. Generation and secretion of CXCL12 by stromal cells facilitate attraction of cancer cells, acting through its cognate receptor, CXCR4, which is expressed by both hematopoietic and non-hematopoietic tumor cells. CXCR4 stimulates tumor progression by different mechanisms and is required for metastatic spread to organs where CXCL12 is expressed, thereby allowing tumor cells to access cellular niches, such as the marrow, which favor tumor cell survival and proliferation. It has also been demonstrated that CXCL12 binds to another seven-transmembrane G-protein receptor or G-protein-coupled receptor, namely CXCR7. These studies indicated critical roles for CXCR4 and CXCR7 mediation of tumor metastasis in several types of cancers, suggesting their contributions as biomarkers of tumor behavior as well as potential therapeutic targets. Furthermore, CXCL12 itself has the capability to stimulate survival and growth of neoplastic cells in a paracrine fashion. CXCL12 is a supportive chemokine for tumor neovascularization via attracting endothelial cells to the tumor microenvironment. It has been suggested that elevated protein and mRNA levels of CXCL12/CXCR4/CXCR7 are associated with human bladder cancer (BC). Taken together, mounting evidence suggests a role for CXCR4, CXCR7, and their ligand CXCL12 during the genesis of BC and its further development. However, a better understanding is still required before exploring CXCL12/CXCR4/CXCR7 targeting in the clinic.


Asunto(s)
Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Neoplasias de la Vejiga Urinaria/etiología , Quimiocina CXCL12/química , Humanos , Terapia Molecular Dirigida/métodos , Neovascularización Patológica , Receptores CXCR/química , Receptores CXCR4/química , Transducción de Señal , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria/irrigación sanguínea , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA